Published online Sep 28, 2025. doi: 10.3748/wjg.v31.i36.110742
Revised: July 4, 2025
Accepted: August 22, 2025
Published online: September 28, 2025
Processing time: 97 Days and 17.8 Hours
Gastrointestinal and hepatic disorders exhibit significant heterogeneity, characterized by complex and diverse clinical phenotypes. Most lesions present without typical symptoms in their early stages, which poses substantial challenges for early clinical identification and intervention. As an interdisciplinary field at the forefront of technology, artificial intelligence (AI) integrates theoretical inno
Core Tip: Artificial intelligence (AI) demonstrates transformative potential across gastrointestinal and hepatic disorders. It enhances early detection of subtle lesions (e.g., Barrett's esophagus) by analyzing diverse clinical data, optimizes treatment decisions (e.g., therapy response in liver cancer) via integrated clinical data assessment (including multimodal integration where applicable), and refines prognostic prediction (e.g., recurrence risk in liver cancer). This translational AI enables intelligent clinical decision-making for diagnosis, personalized treatment, and prognosis assessment throughout the patient journey.
- Citation: Ren SQ, Chen JM, Cai C. Translational artificial intelligence in gastrointestinal and hepatic disorders: Advancing intelligent clinical decision-making for diagnosis, treatment, and prognosis. World J Gastroenterol 2025; 31(36): 110742
- URL: https://www.wjgnet.com/1007-9327/full/v31/i36/110742.htm
- DOI: https://dx.doi.org/10.3748/wjg.v31.i36.110742
Gastrointestinal and hepatic diseases present a considerable challenge to global public health due to their alarmingly high incidence rates, complex clinical manifestations, and heavy economic burden[1]. According to a report by BCC Research, United States, the ed $86.0 billion in 2023 and is projected to surge to $122.5 billion by 2028[2]. Despite this continuous expansion of the global gastrointestinal diagnostics and therapeutics market, the reality is plagued by multiple challenges: (1) Critical diagnostic resource shortages and inequitable distribution: Severe scarcity and uneven allocation of essential resources such as endoscopy lead to diagnostic delays. Moreover, traditional reliance on manual interpretation introduces subjective variability and high miss rates (particularly for early-stage lesions)[3]; (2) Difficulties in personalized treatment: High disease heterogeneity complicates individualized therapeutic decision-making. Risks associated with invasive procedures and steep learning curves limit the widespread adoption of advanced techniques. In addition, inaccurate prediction of treatment response results in ineffective therapies[4]; and (3) Limitations in prognostic models and follow-ups: Existing prognostic models demonstrate insufficient accuracy in predicting recurrence, complications, and survival. Long-term follow-up for high-risk populations suffers from poor adherence and entails substantial costs[5]. The uneven distribution of global healthcare resources further exacerbates these challenges, creating a significant gap in medical treatment, especially in resource-limited settings[6].
In light of these challenges, artificial intelligence (AI) technologies, particularly-machine learning (ML) and deep learning (DL)-are attracting increasing research interest as they offer unprecedented opportunities to revolutionize the approach for addressing core persistent challenges in gastroenterology and hepatology by leveraging their formidable capabilities in data processing, pattern recognition, and prediction. DL models, particularly convolutional neural networks (CNNs), have shown significant potential in medical image analysis, often rivaling or even surpassing the performance of human experts[7-9]. This potential of AI technologies is rapidly translating into diverse practical applications, ranging from enhancing diagnostic accuracy and efficiency (e.g., AI-aided endoscopy, histopathological interpretation, and imaging analysis) and optimizing treatment decision-making (e.g., predicting therapeutic response, assisting surgical planning, and guiding robotic procedures) to enabling prognosis assessment and risk prediction (e.g., developing individualized models for recurrence, complication, and survival prediction). This review aims to systematically delineate the recent advances, proven applications, and future directions of AI in the diagnosis, treatment, and prognosis of gastrointestinal and hepatic diseases.
We conducted a systematic literature search using the PubMed database for studies published between January 2019 and May 2025. The search strategy employed Boolean operators with the following key terms:
AI-related terms: “Artificial intelligence” OR “AI” OR “deep learning” OR “machine learning” OR “computer aided diagnosis” OR “computational intelligence” OR “convolutional neural network” OR “neural network”.
Disease-related terms: “Barrett’s esophagus” OR “esophageal squamous cell carcinoma” OR “Helicobacter pylori” OR “gastric polyp” OR “gastric cancer” OR “Crohn's disease” OR “small intestinal neuroendocrine neoplasm” OR “ulcerative colitis” OR “colorectal polyp” OR “colorectal cancer” OR “hepato-cirrhosis” OR “hepatic encephalopathy” OR “hepatocellular carcinoma” OR “liver cancer” OR “gastrointestinal disease”.
The final query combined both sets: (AI-related terms) AND (disease-related terms), restricted to titles, abstracts, and keywords of English-language publications.
Studies that met all of the following criteria were included: (1) Population: Focused on AI algorithms for diagnosing, guiding treatment, or predicting outcomes in digestive diseases (esophagus, stomach, intestines, and liver); (2) Study design: Clinical studies (prospective/retrospective cohorts, case-control, and randomized trials) or technical validation studies; (3) Data: Reported explicit performance metrics [e.g., area under the curve (AUC), sensitivity, specificity, and accuracy]; and (4) Publication date: January 2019 or later.
Studies were excluded if they had the following characteristics: (1) Type: Conference abstracts, editorials, case reports, letters, reviews, or non-English publications; (2) Data quality: Lacked key performance metrics or had sample sizes < 50 (except for technical validation studies); (3) Duplication: Were redundant publications (only the most comprehensive dataset was retained); and (4) Relevance: Were purely algorithmic development studies without clinical validation, or animal/cell-based research.
Literature screening and data extraction had the following steps: (1) Initial screening: Two investigators (Ren SQ and Chen JM) independently screened titles/abstracts to exclude irrelevant records; (2) Full-text review: Potentially eligible articles underwent full-text assessment against inclusion criteria; and (3) Data extraction: A standardized form was employed to capture: Study design, population characteristics, sample size; AI algorithm type and application (diagnosis/treatment/prognosis); performance metrics (AUC, sensitivity, specificity); and validation method (internal/external). Owing to the widespread practice of incompletely reporting technical details (e.g., specific data augmentation techniques, hyperparameter optimization processes, and detailed validation set splitting strategies) in the original literature - a key challenge to reproducibility in AI research-this review focused on summarizing key parameters wherever feasible (e.g., dataset scale, model architecture, and primary performance metrics). Any discrepancies were resolved through consensus or third-party adjudication.
AI and Barrett’s esophagus: Barrett’s esophagus (BE) is the most significant risk factor for the development of esophageal adenocarcinoma (EAC). Consequently, high-definition white-light endoscopy surveillance has become the preferred method for identifying early neoplastic lesions associated with BE[10,11]. Patients with BE require regular endoscopic examinations to detect EAC at a treatable stage[10,11]. However, early Barrett’s neoplasia is frequently missed because lesions are typically flat and exhibit only subtle morphological changes. Furthermore, the low progression rate of BE means most endoscopists encounter these early lesions infrequently in clinical practice. This lack of familiarity with their endoscopic manifestations further increases the risk of missed diagnosis[12,13].
Recent studies have suggested that AI-assisted detection technology can show promising results in response to the aforementioned clinical challenges. De Groof et al[14] employed a prospective approach to conduct a real-time preliminary assessment of a DL-assisted computer-aided detection (CAD) system for identifying BE-associated neoplasia during live endoscopy. This study captured white-light images at 2 cm intervals during endoscopies in 20 patients (10 with non-dysplastic BE and the other 10 with neoplasia) for real-time analysis by using the CAD system. The results demonstrated a system accuracy of 90% (sensitivity 91%, specificity 89%) in a per-level analysis. In other words, the system successfully identified nine out of ten patients with neoplasia with few false positives (only one non-dysplastic patient was misclassified), providing the initial evidence for its high-precision real-time detection capability and supporting its progression to multicenter clinical trials. Similarly, Struyvenberg et al[15] developed and validated a CAD algorithm based on volumetric laser endomicroscopy (VLE) for real-time identification of BE neoplasia. Their prospective multicenter trial enrolled 47 patients and evaluated 318 biopsy-validated VLE targets. The results showed that the algorithm achieved an accuracy of 85% (sensitivity 91%, specificity 82%) on an independent test set, which is statistically superior to the 77% accuracy achieved by ten VLE experts. These studies indicate that AI-assisted technologies can achieve rapid and highly sensitive detection of neoplastic lesions in BE. This holds promise for effectively guiding targeted biopsies, thereby significantly reducing the risk of missed diagnoses associated with traditional random biopsy strategies.
AI and esophageal squamous cell carcinoma: esophageal squamous cell carcinoma (ESCC), the most common type of esophageal cancer, constitutes approximately 80% of global esophageal cancer cases[16]. Its poor prognosis in advanced stages highlights the critical role of early detection and treatment for improving patient outcomes. Although advanced endoscopic techniques such as narrow-band imaging (NBI) and blue light imaging have enhanced detection capabilities, variability in endoscopist experience remains a significant risk factor for missed early lesions. To address this issue, AI has been introduced to augment the diagnostic performance. Waki et al[17] developed a DL system that focused on the screening performance in scenarios with a high missed-diagnosis risk. Trained on 17336 images, this DL system was validated using 100 rigorously simulated videos reflecting rapid non-magnifying endoscopy (50 videos containing ESCC). The DL system achieved a sensitivity of 85.7% (detecting 54 out of 63 Lesions) when the single-frame threshold was ≥ 0.6 and three consecutive positive frames were required for confirmation. However, it had a specificity of only 40%. This difference in specificity primarily stemmed from rapid-pass artifacts specific to fast endoscopic examination, such as those occurring at the esophagogastric junction. Notably, when used as an adjunctive tool, the AI significantly improved the sensitivity of 21 endoscopists from 75.0% to 77.7% (P = 0.00696) while maintaining a high specificity at 91.6%. This result confirms its efficacy in reducing missed diagnoses during screening without increasing the risk of overdiagnosis.
In a methodological contrast, Everson et al[18] aimed for precise stratification of early-stage ESCC. Their clinically interpretable CNN, trained on 67742 magnifying endoscopy with NBI (ME-NBI) images, employed five-fold cross-validation. It predicted the histological status by identifying intrapapillary capillary loop (IPCL) morphology. On an independent test set of 158 images, the model achieved an overall F1 score of 94% (sensitivity 93.7%, specificity 92.4%, AUC 95.8%), which is comparable to the diagnostic performance of expert endoscopists from both Asia and Europe (average F1 scores 96.5%-98%). A key innovation was the integration of class activation map technology, which visualized the decision rationale of the model with the CNN. This integration confirmed the focus of the model on IPCL microvascular structures, which aligned with clinical understanding and enhanced its credibility for clinical application. These differences in performance metrics between the two studies are mainly due to their divergent target scenarios and the use of different validation methodologies. Waki et al[17] prioritized screening sensitivity (85.7%) to address missed diagnoses, utilizing unedited continuous videos simulating real-world screening conditions. However, the lower specificity reported by Waki et al[17] reflects the inherent challenges of this demanding setting. Conversely, the Everson et al’s study, based on static, high-quality ME-NBI images and optimized via cross-validation, aimed to replicate expert-level stratification capabilities for early lesions, achieving strong overall metrics (F1 score 94%, AUC 95.8%)[18]. This complementary nature demonstrates that AI technology can be directionally optimized according to distinct clinical needs-large-scale screening vs precise diagnosis-providing complementary technical support for early ESCC detection. Future prospective clinical trials should aim to definitively establish the real-world clinical utility of these different AI models. The results of key studies evaluating AI applications the diagnosis of esophageal diseases over the past 6 years are summarized in Table 1[19-27].
Disease | Application | Ref. | Study design | Region/country | Modality | Test set | AI model | Main findings |
BE | Diagnosis | Rosenfeld et al[19] | R | United Kingdom | Questionnaires | 1299 patients | ML | ML model with 8 factors (e.g., age) predicts BE (AUC 0.86/0.81), facilitating high-risk screening |
Diagnosis | Abdelrahim et al[20] | P | Europe | WLI | 75 patients | CNN | An AI system detected Barrett's neoplasia in real-time endoscopy with 93.8% sensitivity, significantly higher than endoscopists (63.5%) | |
Diagnosis | Struyvenberg et al[21] | R | Europe | NBI | 157 videos | CNN | Developed a DL-based CAD system for Barrett's neoplasia in NBI videos: 83% accuracy, 85% sensitivity, 83% specificity, processing at 38 fps | |
Diagnosis | Hashimoto et al[22] | R | United States | WLI, NBI | 1832 images | CNN | AI detects Barrett's early neoplasia at 95.4% accuracy, 96.4% sensitivity via CNN, with real-time lesion localization | |
Esophageal carcinoma, ESCC | Diagnosis | Tokai et al[23] | R | Japan | WLI, NBI | 2042 images | CNN | AI outperformed 13 endoscopists in assessing ESCC invasion depth (accuracy: 80.9%; AUC 0.7873), demonstrating superior diagnostic capability |
Diagnosis | Fukuda et al[24] | R | Japan | NBI, BLI | 28333 images | CNN | AI outperformed endoscopists in ESCC detection sensitivity (91% vs 79%) and characterization accuracy (88% vs 75%) | |
Diagnosis | Li et al[25] | R | China | WLI, NBI | 759 patients | DL | CAD-NBI surpasses CAD-WLI in accuracy/specificity for early ESCC (P < 0.05). Endoscopist combination yields optimal diagnosis (94.9% accuracy, 92.4% sensitivity, 96.7% specificity) | |
Diagnosis | Guo et al[26] | R | China | NBI | 13144 images | DL | This DL model demonstrates high sensitivity (image 98.04%, video 96.1%) and specificity (image 95.03%, video 99.9%) in real-time diagnosis of esophageal precancerous and early SCC | |
Diagnosis | Ohmori et al[27] | R | Japan | WLI, NBI/BLI, ME | 21597 images | CNN | AI detected ESCC via non-magnifying endoscopy (NBI/BLI) with 100% sensitivity. With magnification, accuracy reached 83%, comparable to expert endoscopists |
AI and Helicobacter pylori infection: Helicobacter pylori (H. pylori) infection is closely associated with the development of gastric cancer (GC). Therefore, eradication of H. pylori is an effective measure against GC[28]. Although endoscopic findings of H. pylori infection may include features such as enlarged gastric folds and the disappearance of regular collecting venules, the overall accuracy of human visual assessment is limited. Routine diagnosis of GC still relies on biopsy and/or breath tests; however, the latter carries a certain risk of false-negative results[29,30]. To enhance the diagnostic performance of tests for GC, AI-assisted tools are being actively developed. Klein et al[31] proposed a DL-based decision support algorithm combining image processing techniques (such as threshold segmentation and morphological operations) with a CNN (VGG architecture) to detect H. pylori in gastric biopsy tissue sections [Giemsa and hematoxylin & eosin (H&E) stained]. Evaluated on 87 samples validated by PCR/immunohistochemistry, this DL-based algorithm demonstrated 100% sensitivity (significantly higher than the 68.4% sensitivity achieved via microscopic diagnosis) and 66.2% specificity, highlighting its potential as a high-sensitivity screening tool for pathological diagnosis.
Yasuda et al[32] developed an explainable AI system based on a support vector machine (SVM) for diagnosing H. pylori infection using linked color imaging (LCI) endoscopic images. Their method first grouped LCI images based on hue and extracted feature values, and then trained an SVM classifier for diagnosis. The results showed an overall accuracy of 87.6%, which was comparable to that of experienced endoscopists (accuracy rates of 89.5%-90.5%) and that of superior to less-experienced physicians.
AI and gastric polyps: Gastric polyps, defined as protuberant lesions originating from the gastric epithelium or submucosa and projecting into the lumen, exhibit diverse malignant potential. Their presence may indicate an increased risk of intestinal or extra-intestinal malignancies. Adenomatous polyps are considered true neoplastic lesions, with a malignant transformation rate ranging from 6% to 47%[33]. Consequently, the timely diagnosis, biopsy, and resection of gastric polyps are crucial. This facilitates the early elimination of adenomatous lesions, thereby reducing the subsequent risk of carcinogenesis[34]. As most gastric polyps are clinically asymptomatic and often remain undetected prior to routine examinations, it becomes crucial to develop highly effective diagnostic tools. In recent years, AI has provided some novel approaches to enhance the detection and differential diagnosis of gastric polyps. He et al[35] developed a DL-based framework, MultiAttentiveScopeNet, for gastroscopic image analysis. By integrating and fusing features from different layers of ResNet50 after channel normalization, this DL framework significantly improved the analysis of complex gastroscopic images. It achieved a high accuracy of 0.9308 in the differential diagnosis of gastric polyps and protruding lesions, outperforming junior endoscopists and providing a preliminary solution to diagnostic challenges in this field.
Chen et al[36] further improved detection performance with an automatic detection algorithm based on Single Shot MultiBox Detector (SSD), modifying the VGG-16 network architecture. Their algorithm achieved a mean Average Precision of 95.74% in detecting polyps within gastrointestinal endoscopic images. This represents a 12.4% improvement over manual detection and a 5.7% increase over Mask R-CNN while operating 8.41 times faster. These results demonstrate that the AI technology can effectively address the high rates of missed diagnoses and misdiagnoses of polyps in gastrointestinal endoscopic images to a significant extent.
AI and GC: GC, often presenting with insidious and non-specific symptoms, is frequently diagnosed at an advanced stage with a poor prognosis, and has become the fourth leading cause of cancer-related deaths globally[37]. Consequently, early detection and appropriate treatment are crucial for reducing GC-related mortality. With the widespread application of endoscopy in GC diagnosis, there is a growing demand in the medical community for the development of non-invasive early diagnostic methods. However, endoscopic diagnosis of early GC demands substantial expertise. AI holds promise for assisting in achieving more accurate diagnoses and improving image interpretation efficiency[38,39].
Feng et al[40] developed a real-time diagnostic system based on a deep CNN (DCNN) aimed at enhancing endoscopists’ diagnostic accuracy for early GC. They collected test sets comprised images (1289 images) and videos (130 patients) to compare the performance of 12 endoscopists with and without the system’s assistance. The results demonstrated that the DCNN achieved AUCs of 0.917 and 0.930 in the image and video tests, respectively. With AI assistance, endoscopists’ diagnostic accuracy significantly improved (that of novices increased from 81.33% to 95.22%, reaching expert levels), and the diagnosis time decreased. This observation robustly validates the effectiveness of the DCNN system in enhancing GC diagnostic performance.
Beyond diagnosis, AI also exhibits substantial potential in treatment decision-making. You et al[41] applied radiomics analysis of dual-energy computed tomography (DECT) iodine maps to differentiate between GC T1/2 and T3/4a stages, thereby informing neoadjuvant chemotherapy decisions. They data collected from 263 patients across multiple centers, extracted radiomics features from DECT iodine maps, and constructed a nomogram model by integrating ML algorithms (such as logistic regression) with clinical parameters. This model demonstrated excellent performance across the training set, internal validation set, and external test cohort, achieving AUC values of 0.821-0.894. Hence, the ML-based auxiliary method can effectively identify T3/4a stage tumors, thereby enhancing the precision of individualized treatment.
Furthermore, in the field of prognostic assessment, Li et al[42] developed and validated an ML model for predicting lymph node metastasis in GC patients. This study trained the model using retrospective data from 500 patients at a Chinese hospital and performed external validation using data from 824 Asian American patients in the United States National Cancer Institute’s Surveillance, Epidemiology, and End Results (SEER) database. Eight algorithms, including logistic regression and artificial neural networks (ANN), were evaluated. The results demonstrated that the ANN afforded the best performance (accuracy 0.722), with the depth of tumor invasion, size, and Lauren classification identified as key predictors. The researchers also developed a corresponding web application. This accurate and reliable model holds clinical utility for guiding decisions regarding surgical extent (e.g., degree of lymphadenectomy) and personalized postoperative management in GC. These advances demonstrate the guiding role of AI in the diagnostic prediction, treatment guidance, and prognostic assessment of GC. Table 2 summarizes the results of key studies from the past 6 years evaluating AI in diagnosing common gastric diseases[39,43-56].
Disease | Application | Ref. | Study design | Country/region | Modality | Test set | AI model | Main findings |
H. pylori infection | Diagnosis | Martin et al[43] | R | United States | Gastric biopsy | 406 patients | CNN | DCNNs accurately recognize gastric pathology damage patterns, especially H. pylori gastritis, and serve as effective screening tools |
Diagnosis | Mohan et al[44] | R | China, Japan | WLI, BLI, LCI | - | CNN | For H. pylori infection diagnosis, CNN achieved 87% accuracy, sensitivity, and specificity, comparable to endoscopists (82.9% accuracy) | |
Diagnosis | Nakashima et al[45] | R | Japan | LCI, WLI | 515 patients | CNN | Developed LCI/DL-based CAD classifying H. pylori infection into uninfected, active, and post-eradication statuses with 84.2%, 82.5%, 79.2% accuracy. Outperforms WLI and matches expert endoscopists | |
Gastric polyp | Diagnosis | Yuan et al[46] | R | China | WLI | 9443 patients | DCNNs | AI achieved 96.2% accuracy and 88.0% sensitivity for gastric polyps. With AI, junior endoscopists' accuracy significantly improved (96.9%→97.6%), matching seniors |
Diagnosis | Cao et al[47] | R | China | Gastroscopic imaging | 2270 images | DL | Improved YOLOv3 with feature fusion boosts small polyp detection in gastroscopic images to 91.6% accuracy, resolving complex background interference | |
GC | Diagnosis | Horiuchi et al[48] | R | Japan | ME-NBI | 2828 images | CNN | CNN system distinguishes EGC from gastritis (sensitivity 95.4%, NPV 91.7%, accuracy 85.3%), aiding clinical diagnosis |
Diagnosis | Li et al[39] | P & R | China | ME-NBI | 20341 images | CNN | ME-NBI-based CNN achieves 90.91% accuracy for early GC; 91.18% sensitivity (superior to experts), 90.64% specificity (comparable); overall outperforms non-experts | |
Diagnosis | Bu et al[49] | P | China | Liquid biopsy | 150 samples | ML | Developing NanoFisher for efficient plasma EV isolation, combining metabolomics and machine learning, achieves 92% accuracy in EGC diagnosis | |
Treatment | Wang et al[50] | R | China | CECT | 244 patients | ML | CT radiomics distinguishes T2 from T3/T4 GC, guiding neoadjuvant chemotherapy selection | |
Treatment | Shang et al[51] | R | China | CECT | 311 images | DL | A nomogram built from radiomic and DL features via automated spleen segmentation effectively predicts GC serosal invasion, providing a noninvasive tool for surgical planning | |
Treatment | Kang et al[52] | R | South Korea | CT, WLI, biopsy | 2927 patients | DL | Developed a Transformer-based multimodal AI system integrating endoscopic images and clinical data. Accurately predicts EGC lymph node metastasis risk (AUC 0.908), guiding treatment decisions | |
Treatment | Chen et al[53] | R | China | Laparoscopic surgery video | 2460 images | DL | Develop AI models to accurately identify perigastric vessels, enhancing safety and reducing bleeding risks in laparoscopic gastrectomy | |
Prognosis | Zhang et al[54] | R | China | CT | 669 patients | DCNN | Developed CT-based radiomics nomogram integrating radiomics and clinical factors (e.g., CEA) effectively predicts early recurrence in advanced GC preoperatively (AUC 0.806-0.831) | |
Prognosis | Dong et al[55] | R | China, Italy | CECT | 730 patients | DL | DLRN accurately predicts GC lymph node metastasis number (C-index 0.797-0.822), outperforming clinical staging and correlating significantly with survival | |
Prognosis | Huang et al[56] | R | China | CECT | 205 patients | ML | Developed a ML nomogram combining clinical factors (T/N stage) and CT radiomics to predict gastric cancer PNI (validation AUC 0.885), aiding prognosis |
AI and Crohn's disease: Crohn's disease (CD), a heterogeneous chronic inflammatory bowel disease, presents numerous challenges in clinical management, including intermittent flares, the need for medication adjustments, potential surgical risks, and significant psychological burden[57,58]. As with many chronic diseases, accurately predicting the disease trajectory, treatment response, and prognosis of CD is crucial for individualized precision medicine[59]. To this end, researchers are actively developing AI-assisted tools.
In the field of disease progression prediction, Kim et al[60] integrated clinical variables with transcriptome-wide association study PrediXcan models based on population-specific terminal ileum gene expression data from a Korean cohort. They successfully developed a model predicting the progression of CD in Korean patients to the stenosing (B2) or penetrating (B3) phenotype within 24 months. Analyzing 430 patients, the model achieved AUCs of 0.788 and 0.785 for predicting B2 and B3 progression, respectively, and identified key genes such as CCDC154 and PUS7. This result provides a molecular basis for early intervention in high-risk patients.
Regarding treatment decision optimization, Udristoiu et al[61] focused on the assessment of mucosal healing. They developed a DL algorithm (a CNN-LSTM model) using confocal laser endomicroscopy (CLE) images. Analyzing 6205 CLE images (80% training set, 20% test set), their DL algorithm demonstrated excellent performance on the test set (accuracy 95.3%, sensitivity 92.78%, specificity 94.6%, AUC 0.98), effectively distinguishing inflamed mucosa from the normal one. This result provides an objective, quantitative basis for treatment adjustments, such as medication selection.
For the assessment of disease prognosis, Ferreira et al[62] pioneered the application of a CNN-based DL model for automated detection of intestinal ulcers and erosions in images from PillCam™ Crohn’s Capsule (PCC), a novel panoramic capsule endoscopy system. The model demonstrated high sensitivity (90.0%) and specificity (96.0%), achieving the first automated detection of lesions in PCC images and significantly optimizing the monitoring efficiency and diagnostic performance for CD. This study provides an innovative tool for AI-assisted diagnosis of CD. By enhancing the diagnostic efficacy of the PCC system, this model holds promise for further advancing the precision of mucosal healing assessment and disease staging.
AI and small intestinal neuroendocrine neoplasms: Neuroendocrine neoplasms (NENs) originate from neuroendocrine cells and are commonly found in the gastrointestinal tract and pancreas. Small intestinal NENs (SINENs) are relatively rare[63]. According to the SEER database, the incidence of SINENs has increased by approximately sixfold between 1973 and 2012, with an increase of 1.05 per 100000 in the average incidence rate from 2000 to 2012[64]. Blazevic et al[65] combined two approaches for the diagnosis of SINENs: A systematic visual assessment performed by five clinicians, and the construction of an ML model using 1128 radiomic features extracted from the mesenteric mass and the surrounding tissue. The clinicians’ achieved an AUC of 0.62-0.85 (with a low interobserver agreement), while the best-performing radiomics model, built using features from the surrounding mesentery (SM), achieved a mean AUC of 0.77. This performance was comparable to the assessment by a team of multidisciplinary experts (sensitivity 0.64/specificity 0.68). Hence, it can be suggested that models built using radiomic features from the SM can objectively identify high-risk patients, which provides a basis for prophylactic surgical decision-making. Table 3 summarizes the results of key studies from the past 6 years evaluating the application of AI in diagnosing common diseases of the small intestinal[66-74].
Disease | Application | Ref. | Study design | Country/region | Modality | Test set | AI model | Main findings |
Crohn's disease | Diagnosis | Li et al[66] | R | China | CTE, histopathology | 167 patients | ML | A validated CTE-based radiomics model accurately distinguishes moderate-severe from non-mild fibrosis in Crohn's bowel walls, significantly outperforming radiologists' visual assessments |
Diagnosis | Majtner et al[67] | P | Denmark | pan-CE | 7744 images | DL | Auto-detects Crohn's ulcers with 98.4% accuracy. Comparable small/Large bowel accuracy (98.5% vs 98.1%). Distinguishes severity (κ = 0.72) | |
Diagnosis | Klang et al[68] | R | Israel | CE | 27892 images | CNN | DL model detects Crohn's enteric strictures at 93.5% accuracy (AUC 0.989), precisely distinguishing strictures from ulcers (including severe), enabling automated CE diagnosis | |
Treatment | Konikoff et al[69] | R | Israel | APCT | 101 patients | ML | Developed an ML model using indicators, such as NLR, to predict Crohn's complication risk in emergency CT (AUC 0.774), enabling risk stratification to reduce unnecessary scans | |
Treatment | Con et al[70] | R | Australia | Serum biomarkers | 146 patients | DL | RNN with serial biomarkers (AUC 0.754) outperforms logistic regression (AUC 0.659) in predicting biochemical remission (CRP < 5 mg/L) at 12 months post-anti-TNF therapy in Crohn's disease | |
Prognosis | Ungaro et al[71] | P | United States, Canada | PEA | 265 patients | ML | ML model identified 5 plasma protein markers for penetrating (B3) and 4 for stricturing (B2) complications, outperforming traditional models (B3 AUC 0.79) | |
Prognosis | Stidham et al[72] | R | United States | Laboratory examination | 2809 patients | ML | Machine learning leveraging routine longitudinal lab data predicts Crohn's surgical risk (AUC 0.78) | |
SINENs | Diagnosis | Kjellman et al[73] | P | Northern Europe | PET/CT, MRI etc. | 278 patients | ML | Multi-plasma protein markers with Random Forest boost SI-NETs diagnosis (Sensitivity 89%, Specificity 91%, AUC 0.99) |
Diagnosis | Clift et al[74] | R | United Kingdom | EHR | 382 patients | ML | XGBoost using primary care EHRs effectively identifies undiagnosed high-risk SI-NET patients (AUC 0.869) |
AI and ulcerative colitis: Ulcerative colitis (UC) is a chronic immune-mediated disease characterized by alternating periods of flares and remission[75]. It currently has no cure, requires lifelong medical therapy[76]. It may even progress to multiple organ failure[77]. Endoscopy plays a central role in UC management, and is crucial for establishing the initial diagnosis, assessing disease extent and activity, and identifying complications, serving as an endpoint in clinical trials for novel therapies[78,79]. However, existing UC endoscopic scoring systems exhibit significant heterogeneity and subjectivity, leading to insufficient interobserver agreement[80]. Recent advances in AI technology have substantially enhanced the objectivity and efficiency of endoscopic assessment in UC.
At the diagnostic level, Wang et al[81] developed a DL-based classification model for colonoscopy images for differentiating CD from UC. Their model achieved an accuracy of 93.35% for UC image classification, outperforming that reported by the best-performing physician (92.39%). This result confirms the value of CNNs in improving the diagnostic efficacy of colonoscopy for UC and offering novel approaches for clinical decision support.
Regarding therapeutic decision support, Higuchi et al[82] utilized a ResNet50 CNN to build an automated system for assessing disease severity via colon capsule endoscopy in patients with UC. This system achieved a training accuracy of 99.2% and a validation accuracy of 98.3%. The spatial distribution maps of colonic disease activity generated by the system not only assist physicians in objectively evaluating the treatment efficacy and formulating individualized treatment strategies but also significantly reduce the physician image review time by approximately 80%. In the domain of prognostic assessment, Bhambhvani and Zamora[83] pioneered the exploration of DL for the automated assessment of Mayo Endoscopic Subscore (MES 1, 2, 3) in patients with UC. Using a ResNeXt-101 CNN to process 777 endoscopic images, their model achieved AUC values of 0.96, 0.86, and 0.89 for MES 3, MES 2, and MES 1, respectively, on the test set, with an overall accuracy of 77.2%. This result indicates the effectiveness of the model in performing individual-level MES classification, improving the quality of endoscopic assessment, and holding promise for optimizing prognostic evaluation in UC patients through precise MES grading.
AI and colorectal polyps: Colorectal cancer (CRC) predominantly originates from colorectal polyps, especially the adenomatous ones. Although initially benign, these polyps may undergo malignant transformation over time if left unmanaged[84]. Colonoscopy is widely used as the core method for screening and preventing polyp progression to cancer[85]. However, this technique is highly operator-dependent. Recent clinical studies have revealed that as high as 22%-28% of polyps are missed during colonoscopy examinations[86]. These missed polyps can cause delayed cancer diagnosis, leading to significantly reduced survival rates (which can be as low as 10%)[87].
AI shows promise in reducing polyp-miss rates and enhancing detection capabilities. Ozawa et al[88] developed a CNN-based AI system for the automated detection and classification of colorectal polyps during colonoscopy. Trained on 16418 polyp images using the SSD architecture, their system achieved a detection sensitivity of 92% and an overall classification accuracy of 83% (achieving 97% adenoma recognition rate under white light imaging), along with demonstrated high-speed processing capability (20 millisecond per frame). These characteristics robustly validate the value of their system in assisting early CRC diagnosis and optimizing treatment decisions. Similarly, Song et al[89] constructed a DL-based CAD system that focused on the histological classification of colorectal polyps to support clinical treatment decisions. This CAD system, trained on a dataset containing 12480 NBI close-focus image patches using ResNet-50 and DenseNet-201 models, achieved test accuracies ranging from 81.3% to 82.4% (kappa values 0.614-0.642). Its performance was significantly superior to that of novice endoscopists (accuracy 63.8%-71.8%) and was on par with expert-level performance (accuracy 82.4%-87.3%). These results demonstrate that AI-driven CAD systems can effectively augment the diagnostic capabilities of endoscopists and serve as powerful tools for real-time optimization of treatment decisions.
AI and CRC: CRC, a clinically common malignancy, is of significant concern because of its increasing incidence and high mortality rates. CRC is identified as the second leading cause of cancer-related deaths, with a five-year survival rate of only 30%-40%, primarily owing to postoperative recurrence and metastasis[37]. In China, CRC ranks third in incidence and fifth in mortality among all malignancies. Although highly curable in its early stages and detectable in asymptomatic individuals through screening methods such as fecal tests, sigmoidoscopy, or colonoscopy[90,91], current screening approaches are often limited by factors like high cost, the need for cumbersome bowel preparation [required for endoscopy and computed tomography (CT) colonography], and requiring patients to handle stool samples themselves (e.g., gFOBT, FIT, and multi-target stool DNA tests)[92,93]. Therefore, the ability to more accurately identify high-risk individuals and tailor treatment plans and prognosis predictions based on their needs is crucial for efficient clinical decision-making, as it can effectively reduce both incidence and mortality related to CRC[94,95].
For early identification, Schneider et al[96] developed an ML algorithm based on routine blood test indicators and demographic data to identify CRC risk in a community. Their algorithm achieved a sensitivity of 35.4% (95%CI: 33.8-36.7) and an AUC of 0.78 (95%CI: 0.77-0.78) for predicting CRC within 6 months. In addition, it identified 3% highest-risk individuals [odds ratio (OR) = 17.7], with a particularly high predictive power for proximal cancer (OR = 34.7) and advanced-stage cancer (OR = 24.8). Their algorithm offers a low-cost, accessible complementary strategy for early CRC screening, which is particularly valuable for high-risk groups. For treatment decision-making, Ito et al[97] utilized DL (CNN) to build an endoscopic diagnostic support system distinguishing cT1b-stage CRC (requiring surgery) from other early-stage lesions (cTis/cT1a). Trained on 190 white-light colonoscopy images (from 41 patients) using AlexNet architecture and three-fold cross-validation, the system successfully diagnosed cT1b with a sensitivity of 67.5%, a specificity of 89.0%, an accuracy of 81.2%, and an AUC of 0.871. This result indicates the potential of their system to effectively assist endoscopists in evaluating tumor invasion depth and optimizing treatment decisions. For prognostic assessment, Jiang et al[98] combined CNN and an ML classifier (Gradient Boosting-Colon) to analyze H&E-stained tissue sections. Their model effectively stratified patients into high- vs low-recurrence risk groups [hazard ratio (HR) = 10.273] and good- vs poor-prognosis groups (HR = 10.687), achieving a prediction accuracy of 75%-77%. Their model could economically and non-invasively assist in formulating personalized chemotherapy regimens (e.g., extending treatment cycles for high-risk patients) by directly extracting prognostic information from routine pathological slides, providing a novel tool for precision medicine. Table 4 summarizes the results of key studies from the past 6 years evaluating AI in common colorectal diseases[89,99-125].
Disease | Application | Ref. | Study design | Country/region | Modality | Test set | AI model | Main findings |
Ulcerative colitis | Diagnosis | Sutton et al[99] | R | Norway | Endoscopy | 8000 images | CNN | AI (especially DenseNet121 model) accurately distinguishes UC from non-UC pathology (AUC 0.999) and grades endoscopic activity (mild/severe, AUC 0.90) |
Diagnosis | Lo et al[100] | R | Denmark | WLI | 1484 images | CNN | The CNN model achieved 84% accuracy in distinguishing UC endoscopic severity (Mayo score 0-3), significantly outperforming existing models and standardizing clinical assessment | |
Diagnosis | Ruan et al[101] | R | China | Colonoscope | 1772 patients | CNN | AI model detects UC/CD at 99.1% accuracy vs physicians' 78%-92.2%, enhancing clinical efficiency | |
Diagnosis | Gutierrez Becker et al[102] | R | Europe et al | Endoscopy | 1672 videos | CNN | This model directly analyzes raw colonoscopy videos, automatically assessing UC severity (MCES) with high accuracy (AUC 0.84-0.85), reducing manual annotation needs | |
Treatment | Bossuyt et al[103] | P | Belgium, Japan | Endoscopy | 100 patients | ML | The RD algorithm objectively evaluates UC endoscopic and histologic activity, closely correlated with RHI (r = 0.74), and is sensitive for monitoring therapeutic response | |
Treatment | Iacucci et al[104] | P | Europe, North America | HD-WLE, VCE | 283 patients | CNN | AI system accurately differentiates UC endoscopic activity/remission (AUC 0.94) and predicts histological remission (83% accuracy), comparable to physicians | |
Prognosis | Huang et al[105] | R | China | Colonoscope | 856 images | DL, ML | DL/ML-CAD diagnoses mucosal healing (MES 0-1) with 94.5% accuracy and complete healing (MES 0) at 89.0% | |
Prognosis | Popa et al[106] | R | Romania | Colonoscope | 55 patients | ML | ML models accurately predict endoscopic disease activity one year after anti-TNFα therapy in UC patients (90% accuracy in test set, 100% in validation set) | |
Prognosis | Takenaka et al[107] | P | Japan | Endoscopy | 2012 patients | DNN | DNN achieves 90.1% accuracy for endoscopic remission and 92.9% for histological remission (UC), reducing biopsy needs | |
Prognosis | Maeda et al[108] | P | Japan | Endocytoscope, NBI | 145 patients | ML | Real-time AI endoscopy predicts relapse risk in UC remission by analyzing mucosal microvessels (AI-Active 28.4% vs AI-Healing 4.9%, P < 0.001) | |
Colorectal polyps | Diagnosis | Wang et al[109] | R | China | Colonoscope | 1600 patients | CNN | Enhanced GAP model achieves > 98% accuracy (TPR > 96%, TNR > 98%) for colon polyp detection with reduced parameters, enabling lightweight yet accurate diagnosis |
Diagnosis | Song et al[89] | P & R | South Korea | NBI | 1169 samples | DL | CAD with NBI predicts polyp histology at 81.3%-82.4% accuracy, outperforming junior physicians (63.8-71.8%) and matching experts (82.4-87.3%), enhancing junior diagnostic performance | |
Diagnosis | Jin et al[110] | P & R | South Korea | NBI | 2450 images | CNN | AI assistance significantly boosts endoscopists' (especially novices') accuracy for small polyps (< 5 mm) (73.8%→85.6%) and reduces time (3.92→3.37 seconds/polyp) | |
Diagnosis | Sakamoto et al[111] | R | Japan | WLI, LCI, BLI | 1788 images | DL | CADe sensitive > 94% (WLI/LCI), CADx accuracy > 93% (WLI/BLI), rivals expert endoscopists | |
Diagnosis | Zachariah et al[112] | R | USA | WLI, NBI | 6223 images | CNN | CNN real-time prediction of colorectal polyp pathology meets PIVI standards: 97% adenoma NPV, > 93% surveillance interval concordance | |
Treatment | Wickstrøm et al[113] | R | Europe | Colonoscope | 912 images | FCNs | CNNs rely on polyp shape/edges for segmentation; error risk rises significantly in uncertain areas. FCNs combine uncertainty with interpretability visualization, helping doctors pinpoint high-risk regions fast | |
Treatment | Su et al[114] | P | China | Colonoscope | 659 patients | DCNN | AQCS significantly boosts adenoma detection (28.9% vs 16.5%, P < 0.001), polyp detection, and optimizes withdrawal time and bowel prep during colonoscopy | |
CRC | Diagnosis | Luo et al[115] | P & R | China | Liquid biopsy | 3315 patients | ML | Plasma ctDNA methylation markers (e.g., cg10673833) enable early CRC diagnosis (AUC 0.96) and high-risk group screening (Sensitivity 89.7%) |
Diagnosis | Arabameri et al[116] | R | France, USA, Austria | Fecal microbiota analysis | 350 patients | ML | Combining GRNN and DBFS (new feature selection) identified 6 key microbial markers (e.g., Clostridium), enabling high-precision CRC detection (AUC 0.911) but insufficient adenoma sensitivity (AUC 0.724) | |
Diagnosis | Zeng et al[117] | P | USA | OCT | 26000 images | CNN | PR-OCT system using OCT and RetinaNet distinguishes CRC from normal tissue in real-time with high accuracy (sensitivity 100%, specificity 99.7%, AUC 0.998) | |
Treatment | Wang et al[118] | R | China | MRI | 240 patients | Faster R-CNN | Faster R-CNN detects positive CRM on rectal cancer pre-op high-resolution MRI with 93.2% accuracy (AUC 0.953); 0.2 seconds/image, highly feasible and efficient | |
Treatment | Yang et al[119] | R | China | MRI | 89 patients | ML | Pre-treatment ADC radiomics predicts locally advanced rectal cancer resistance to neoadjuvant chemoradiotherapy (AUC 0.83/91.3% accuracy) | |
Treatment | Fu et al[120] | R | USA | MRI | 43 patients | DL | DL-based radiomics significantly outperform handcrafted features in predicting neoadjuvant chemoradiotherapy response for locally advanced rectal cancer (AUC 0.73 vs 0.64) | |
Prognosis | Xu et al[121] | R | China | CT, MRI et al | 999 patients | ML | GradientBoosting and LightGBM effectively predict stage IV CRC recurrence risk (AUC up to 0.881); key factors: Chemotherapy, age, LogCEA, CEA, anesthesia duration | |
Prognosis | Zhao et al[122] | R | China | Clinical data | 7205 patients | ML | ML-based NCDB nomogram predicts metastatic rectal cancer 3-year OS (C-index > 0.77, internal/external validation), outperforming prior models | |
Prognosis | Reichling et al[123] | R | France | IHC, WSI | 1018 patients | ML | DGMuneS integrating tumor-stroma/CD3+/tumor features better predicts stage III colon cancer recurrence than traditional immune scores (C-index 0.601 vs 0.578) | |
Prognosis | Skrede et al[124] | R | Norway, United Kingdom | H&E staining | 2467 patients | CNN | Developed a DL-based prognostic biomarker (DoMore v1-CRC) using only routine H&E-stained slides, effectively stratifying Stage II/III CRC risk and outperforming existing markers | |
Prognosis | Väyrynen et al[125] | P | USA | H&E staining | 1504 samples | ML | ML on H&E slides links dense stromal lymphocytes/eosinophils and their peri-tumoral localization to significantly improved CRC-specific survival |
AI and hepato-cirrhosis: Hepatic disease, recognized as one of the most life-threatening conditions is a significant global burden, as confirmed by World Health Organization reports[126]. Among them, hepato-cirrhosis-end-stage progressive liver fibrosis-has a high incidence and mortality rates in Asian countries[127]. However, as most patients typically do not exhibit significant clinical symptoms of hepatic diseases before they progress to the decompensated stage, cirrhosis remains often underdiagnosed and is not identified in time[128]. Therefore, it is essential to develop efficient methods for early detection of liver fibrosis. At present, liver biopsy remains the gold standard for diagnosing fibrosis. However, as an invasive procedure, liver biopsy carries risks of complications such as hemorrhage, biliary peritonitis, and pneumothorax[129,130]. Consequently, in clinical practice, experts favor non-invasive and easily accessible ultrasonography over CT or MRI for the initial assessment[131]. However, the diagnostic accuracy of ultrasound is susceptible to the experience and skill level of the operator[132]. AI technology, leveraging its powerful brain-like algorithms, has the potential to enhance the accuracy of cirrhosis diagnosis. Sarvestany et al[133] conducted a retrospective cohort study and developed an integrated ML algorithm for detecting advanced liver fibrosis, including cirrhosis. Six algorithms were trained and validated by utilizing liver biopsy data. Key results showed that the algorithm achieved an AUC of 0.870, outperforming traditional serum biomarkers (such as APRI and FIB-4) and delivering 100% interpretable results, with the performance approaching expert-level proficiency. This advance provides an automated tool for the efficient identification of advanced liver fibrosis, enabling early screening of high-risk populations for cirrhosis and facilitating timely clinical intervention.
AI and hepatic encephalopathy: Hepatic encephalopathy (HE) is defined as a syndrome of the central nervous system dysfunction caused by metabolic disturbances arising from severe liver disease[134,135]. Although minimal HE (MHE), the mildest form of HE, lacks identifiable clinical symptoms, it is characterized by various subtle neurocognitive and psychomotor deficits, such as psychomotor slowing, shortened attention span, executive dysfunction, and memory impairment[134]. Consequently, early diagnosis and effective treatment of MHE are crucial for preventing its progression to overt HE and for improving the quality of life. Zhang et al[136] performed an fMRI dynamic functional connectivity analysis along with a multi-layer modularity algorithm. Their study revealed significant abnormalities in node disjointness within higher-order cognitive networks (e.g., the default mode network and ventral attention network) of patients with MHE[136]. Importantly, these abnormalities are closely correlated with a decline in the attention and visual memory functions of patients. Critically, an SVM model based on this node disjointness metric achieved an accuracy of 88.71% in individually distinguishing patients with MHE from those with cirrhosis without HE. This finding suggests that dynamic brain network node disjointness may serve as an effective biomarker for MHE, providing a novel approach for the early diagnosis of HE.
AI and liver cancer: According to the 2020 global cancer statistics, liver cancer ranks as the sixth most common cancer worldwide and is the third leading cause of cancer-related deaths[36]. Clinically encountered liver cancers are predominantly primary, mainly comprising hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma[137]. The liver is also a frequent site for metastatic tumors[138]. Given the significant differences in treatment strategies for different subtypes of liver tumors[139], multiphasic contrast-enhanced computed tomography has emerged as a critical tool for preoperative diagnosis[140].
However, the differential diagnosis of liver cancer remains challenging, and preoperative misdiagnosis can directly lead to errors in therapeutic decision-making. AI is progressively enhancing various aspects of liver cancer diagnosis and prognosis. In early diagnosis, the HnAIM DL model developed by Cheng et al[141], which processes whole-slide images using an ensemble approach, achieved a high AUC of 0.935 in external validation. Its diagnostic performance even surpassed pathologists' assessments of biopsy samples, significantly improving early HCC detection rates and patient risk stratification capabilities. For prognostic assessment of HCC, Qu et al[142] developed a DL model based on dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). This model integrates radiomic features with clinical indicators to accurately identify proliferative HCC subtypes and stratify the risk of early recurrence following curative resection. Their retrospective study analyzed data from 355 patients and employed a hierarchical fusion strategy to build a joint predictive model: (1) Deep feature extraction: It utilizes a hybrid 3D DenseNet-Transformer architecture to automatically learn features reflecting tumor heterogeneity (e.g., vascular invasion patterns) from multi-phase DCE-MRI sequences; (2) Multimodal fusion: It combines the selected imaging features with key clinical indicators [e.g., alpha-fetoprotein (AFP) level and HBV status] via feature-level concatenation. Logistic regression assigns modality-specific weights (e.g., imaging features OR = 22.48 vs AFP OR = 2.22), quantifying their relative contribution to predicting the proliferative phenotype; and (3) Non-linear interaction optimization: The transformer’s multi-head self-attention mechanism dynamically captures complex interactions between imaging features and clinical indicators (e.g., the synergistic effect between elevated AFP levels and delayed tumor enhancement). The model achieved AUCs of 0.98, 0.89, and 0.83 on the training set, internal validation set, and external validation set, respectively. This performance significantly surpassed that of models based solely on clinical data (AUC = 0.77) or imaging data (AUC = 0.55). Apart from enhancing the discriminative power, this integration enabled personalized recurrence risk stratification by generating a composite risk score (e.g., DL prediction probability + AFP threshold). This stratification revealed significantly different 2-year recurrence rates between proliferative (56.0%) and non-proliferative (36.1%) subtypes (P < 0.001), providing a quantitative basis for tailoring follow-up strategies and adjuvant treatment decisions.
AI in precision therapy for liver cancer: AI is revolutionizing precision therapy for liver cancer, and has demonstrated significant potential in predicting treatment efficacy for both targeted therapies and immunotherapies. Recent studies have leveraged multimodal data integration and DL models for accurate treatment response prediction and patient stratification, thereby establishing a new paradigm for individualized therapeutic decision-making.
Radiomics-based predictive models have emerged as a research focus in targeted therapy. Xu et al[143] developed a DL radiomic nomogram (DLRN) that automatically segments CT imaging features to predict responses to FOLFOX-based hepatic arterial infusion chemotherapy in advanced HCC (AUC = 0.988). This model integrates radiomic features from arterial/portal venous phases, DL signatures, and clinical variables (e.g., peritumoral arterial enhancement), significantly outperforming conventional clinical models (P < 0.001). Crucially, the DLRN enables effective survival risk stratification, with a median overall survival (OS) of 26.0 months in predicted responders vs 12.3 months in non-responders (P < 0.001). Its fully automated workflow (processing time < 30 seconds per case) enhances the clinical translation efficiency.
Multimodal AI models have achieved breakthroughs in predicting outcomes of combination immunotherapies. Hua et al[144] constructed a CT radiomics model extracting 1223 quantitative features to predict objective response rates to lenvatinib plus programmed cell death 1 inhibitors with interventional therapy in unresectable HCC (training cohort AUC = 0.900). Notably, 100% of the 14 key predictive features comprised Laplacian of Gaussian-filtered and wavelet-transformed parameters, revealing tumor heterogeneity undetectable by visual assessment. The model stratified patients into high- and low-risk groups with significant differences in progression-free survival (HR = 2.347) and OS (HR = 2.592), providing a quantitative basis for therapeutic selection.
ML-based subclassification systems are advancing precision management. Han et al’s CLAM-C model restratified patients with Barcelona Clinic Liver Cancer stage C HCC using XGBoost, achieving superior AUCs (0.800/0.831/0.715) for 6/12/24-month survival prediction vs those achieved using conventional models (P < 0.001)[145]. Patients were classified into four risk tiers, with median OS ranging from 26.5 months (low-risk) to 3.0 months (very-high-risk). Critically, the model guided treatment optimization: Immune checkpoint inhibitors or transarterial therapies significantly outperformed tyrosine kinase inhibitors (TKIs) in low-risk groups (P < 0.05), while atezolizumab-bevacizumab provided superior survival benefit vs TKIs in high-risk subgroups (median OS: 5.9 months vs 4.2 months; P = 0.004).
The results of key studies evaluating AI applications in common liver diseases over the past 6 years are summarized in Table 5[143,144,146-164].
Disease | Application | Ref. | Study design | Country/region | Modality | Test set | AI model | Main findings |
Hepato-cirrhosis | Diagnosis | Rhyou and Yoo[146] | R | South Korea | US | 4950 images | DL | A patch-based DL network for ultrasound cirrhosis diagnosis using synthetic image augmentation, achieving 99.95% accuracy, 100% sensitivity, and 99.9% specificity |
Diagnosis | Luetkens et al[147] | R | Germany | MRI | 465 patients | CNN | ResNet50 distinguishes alcoholic from nonalcoholic cirrhosis on MRI: AUC 0.82, 75% accuracy | |
Diagnosis | Chang et al[148] | R | United States | Liver biopsy, FibroScan etc. | 1370 patients | ML | ML models (especially Random Forest) outperform traditional non-invasive tests (e.g., FibroScan, FIB-4) in identifying significant fibrosis and cirrhosis in NAFLD patients | |
Diagnosis | Mazumder et al[149] | R | United States | CT, liver biopsy etc. | 351 patients | DL, ML | Combining AI-extracted CT radiomics with routine lab data boosts liver cirrhosis prediction accuracy (AUC 0.84-0.85) | |
Diagnosis | Guo et al[150] | P | United Kingdom | NMR spectroscopy | 64005 patients | ML | A plasma metabolomics and ML-based nomogram accurately predicts 10-year hepatic cirrhosis complication risk (AUC 0.861), outperforming conventional metrics | |
Hepatic encephalopathy, HE | Diagnosis | Calvo Córdoba et al[151] | P | Spain | VOG | 47 patients | SVM | Automated VOG with SVM detects MHE in 7-10 minutes (93% sensitivity/specificity), outperforming PHES (25-40 minutes) |
Diagnosis | Sparacia et al[152] | R | Italy | MRI | 124 patients | ML | MRI radiomics with KNN predicts HE presence (76.5% accuracy); MLP predicts HE severity (≥ stage 2, 94.1%), demonstrating potential for HE diagnosis and staging | |
Diagnosis | Chen et al[153] | R | China | MRI | 53 patients | SVM | SVM model using gray matter volume discriminates cirrhosis patients with/without MHE at 83.02% accuracy | |
Treatment | Liu et al[154] | R | China | TIPS | 218 patients | ML | Developed logistic regression model (AUC 0.825) accurately predicts OHE post-TIPS | |
Treatment | Zhong et al[155] | R | China | TIPS | 207 patients | ANN | ANN model accurately predicts post-TIPS OHE (C-index = 0.863), with 15.9% incidence within 3 months, providing a clinical stratification tool | |
Liver cancer | Diagnosis | Gao et al[156] | R | China | CECT | 723 patients | DL | This model effectively distinguishes malignant liver tumors (HCC, ICC, metastases), achieving 72.6% test-set accuracy and improving physician ICC sensitivity by 26.9% |
Diagnosis | Xu et al[157] | R | China | CT | 1049 patients | DL | Swin-Transformer model simplifies LI-RADS classification, effectively distinguishes HCC from non-HCC, and enhances diagnostic performance with clinical data | |
Diagnosis | Li et al[158] | R | China | DECT | 262 patients | DL | Dual-energy CT deep-learning radiomics nomogram noninvasively predicts HCC MTM subtype, outperforming clinical-radiologic models (AUC 0.87-0.91) | |
Diagnosis | Ma et al[159] | R | China | CT, MRI | 211 patients | ML | CT/MRI radiomics + clinical features (SVM) achieves highest HCC diagnostic accuracy (82.4%), significantly outperforms single-modality models, and distinguishes HCC vs non-HCC | |
Treatment | Hua et al[144] | R | China | CECT | 151 patients | ML | Developed and validated a pretreatment CT-based radiomics model predicting response and survival outcomes after triple therapy in unresectable HCC to guide clinical decisions | |
Treatment | Xu et al[143] | R | China | CECT | 458 patients | DL | The model significantly outperforms single models (externally validated AUC 0.896), effectively distinguishing survival differences (P < 0.001), providing a tool for personalized treatment | |
Treatment | An et al[160] | R | China | IATs | 2959 patients | ML | Developed MLDSM to risk-stratify unresectable HCC patients undergoing transarterial therapies (alone/combined) for 12-month mortality, guiding clinical treatment decision (e.g. TACE, HAIC) | |
Prognosis | Cao et al[161] | R | China | CT, MRI et al | 466 patients | DL, ML | Developed pre-/post-op dual-phase DeepSurv model predicting HCC recurrence post-liver transplant; outperformed Milan Criteria (C-index 0.765-0.839), guiding individualized surveillance | |
Prognosis | Altaf et al[162] | R | Pakistan | CT, MRI etc. | 192 patients | CNN | AI model with tumor size, AFP, and grade predicts post-transplant HCC recurrence risk (validation AUC 0.77) | |
Prognosis | Dong et al[163] | R | United States | Clinical data | 2038 patients | ML | XGBoost model effectively predicts 1-, 3-, and 5-year survival (AUC > 0.7) in AFP-positive HCC patients, outperforming other algorithms, offering a clinical tool for early intervention | |
Prognosis | Yan et al[164] | R | China | MRI | 285 patients | CNN | Deep learning-based nomogram integrating imaging, MVI, and tumor number significantly outperforms traditional models in predicting early HCC recurrence (AUC 0.949 vs 0.751) |
Although a large body of research confirms the strong clinical potential of AI in processing clinical images, identifying therapeutic targets, and analyzing large datasets, its real-world implementation faces numerous limitations and challenges. A primary concern is the high dependency of AI models on image quality, with performance requiring significant improvement in complex scenarios (e.g., active hemorrhage) and complex lesion recognition[53]. In specific applications such as intestinal endoscopy, AI systems not only need to incorporate bowel preparation assessment but also urgently require enhanced robustness against common interfering factors like bubbles and residue[67].
More critically, insufficient model generalizability constitutes a core barrier, prominently manifested in three dimensions: The first one is the cross-center/device compatibility issues, as illustrated by Krois et al[165]. A DL model for apical lesion detection trained on German dental panoramic radiographs achieved an F1-score of 54.1% in local testing. However, it sharply dropped to 32.7% when validated at an Indian center. Although cross-center mixed training partially improved the generalizability (raising the Indian F1-score to 46.1%), this reveals significant disparities in equipment parameters, image quality, and disease manifestations across regions. Enhancing generalizability necessitates cross-institutional collaborative training, which inevitably encounters data privacy and standardization challenges[124].
The second dimension comprises limitations in the detection of rare diseases. For example, the rarity of pediatric (particularly congenital) cataracts makes it very difficult to obtain high-quality, large-scale datasets for training and validation. A multicenter randomized clinical trial conducted by Lin et al[166] at Zhongshan Ophthalmic Center, China, revealed a critical performance gap. While the model achieved 98.87% diagnostic accuracy under strictly controlled laboratory conditions, the accuracy significantly declined to 87.4% in real-world multicenter clinical applications. This pronounced “lab-to-clinic translation gap” starkly exposes the fragility of AI model generalizability when confronting data-scarce rare diseases.
Third, the lack of standardized and comprehensive reporting of technical details, such as data preprocessing, hyperparameter settings, and validation strategies, significantly contributes to the poor reproducibility of AI models. This deficiency directly impedes the objective assessment of their true generalization capability.
Together, these three fundamental challenges-poor reporting standards, data/condition dependency, and limited generalizability-constitute major barriers hindering the translation of AI models from research into widespread clinical adoption. Addressing these issues requires establishing standardized reporting guidelines, actively promoting multi-center external validation, and developing more robust algorithms.
To address barriers such as hardware costs and lack of specialized personnel in primary care settings, China has pioneered practical solutions including: Cloud-based AI systems (e.g., Wenzhou’s “Health Cloud Imaging Platform” integrating AI-assisted image analysis, covering > 90% of medical institutions and completing nearly 7 million AI-assisted diagnoses) and tele-diagnosis collaboration models within medical consortiums (e.g., Shanxi Province’s AI diagnostic system deployed across 85 township hospitals). These models aim to overcome technical capacity and resource constraints at the primary care level. However, significant implementation constraints persist, as such solutions heavily rely on high-performance hardware and complex network architectures, which severely limit their widespread adoption and sustainable operation in resource-constrained primary healthcare facilities.
Beyond technical challenges, the potential impact of AI on the clinician-patient relationship remains unclear. Regardless of AI-assisted diagnostic outcomes, the ultimate responsibility lies unequivocally with the clinician. We believe that the development of multimodal AI will extend beyond disease detection and diagnosis to encompass clinical decision support, including pathological diagnosis and metastasis prediction. The ultimate goal of multimodal AI is to create a system capable of comprehensively leveraging large-scale clinical data-such as radiological imaging, endoscopic images, histopathology, and genomic/metagenomic information-to enable precise prediction and assessment of disease risk, treatment response, likelihood of recurrence/metastasis, and both short-term and long-term prognoses.
This article reviews the current applications of AI in common gastrointestinal and hepatic diseases. Research on the use of AI in this field is progressively transitioning from an exploratory stage towards building a solid evidence base for clinical practice. Therefore, we believe that AI will become routinely integrated into the clinical management of gastrointestinal and hepatic disorders in the future, particularly in key aspects such as early diagnosis, treatment decision support, and prognostic assessment, where AI will undergo continuous refinement and improvement through application.
1. | Wang Y, Huang Y, Chase RC, Li T, Ramai D, Li S, Huang X, Antwi SO, Keaveny AP, Pang M. Global Burden of Digestive Diseases: A Systematic Analysis of the Global Burden of Diseases Study, 1990 to 2019. Gastroenterology. 2023;165:773-783.e15. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 83] [Cited by in RCA: 75] [Article Influence: 37.5] [Reference Citation Analysis (0)] |
2. | BCC Research. Gastrointestinal Therapeutics and Diagnostics: Propelling Global Markets with Cutting-Edge Technologies. Apr 15, 2024. [cited 23 June 2025]. Available from: https://www.bccresearch.com/pressroom/phm/gastrointestinal-therapeutics-and-diagnostics. |
3. | Nemoto D, Guo Z, Katsuki S, Takezawa T, Maemoto R, Kawasaki K, Inoue K, Akutagawa T, Tanaka H, Sato K, Omori T, Takanashi K, Hayashi Y, Nakajima Y, Miyakura Y, Matsumoto T, Yoshida N, Esaki M, Uraoka T, Kato H, Inoue Y, Peng B, Zhang R, Hisabe T, Matsuda T, Yamamoto H, Tanaka N, Lefor AK, Zhu X, Togashi K. Computer-aided diagnosis of early-stage colorectal cancer using nonmagnified endoscopic white-light images (with videos). Gastrointest Endosc. 2023;98:90-99.e4. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 15] [Cited by in RCA: 12] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
4. | Kleppe A, Skrede OJ, De Raedt S, Hveem TS, Askautrud HA, Jacobsen JE, Church DN, Nesbakken A, Shepherd NA, Novelli M, Kerr R, Liestøl K, Kerr DJ, Danielsen HE. A clinical decision support system optimising adjuvant chemotherapy for colorectal cancers by integrating deep learning and pathological staging markers: a development and validation study. Lancet Oncol. 2022;23:1221-1232. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 25] [Cited by in RCA: 46] [Article Influence: 15.3] [Reference Citation Analysis (0)] |
5. | Foersch S, Glasner C, Woerl AC, Eckstein M, Wagner DC, Schulz S, Kellers F, Fernandez A, Tserea K, Kloth M, Hartmann A, Heintz A, Weichert W, Roth W, Geppert C, Kather JN, Jesinghaus M. Multistain deep learning for prediction of prognosis and therapy response in colorectal cancer. Nat Med. 2023;29:430-439. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 67] [Cited by in RCA: 108] [Article Influence: 54.0] [Reference Citation Analysis (0)] |
6. | GBD 2019 Human Resources for Health Collaborators. Measuring the availability of human resources for health and its relationship to universal health coverage for 204 countries and territories from 1990 to 2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet. 2022;399:2129-2154. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 186] [Cited by in RCA: 166] [Article Influence: 55.3] [Reference Citation Analysis (0)] |
7. | Chahal D, Byrne MF. A primer on artificial intelligence and its application to endoscopy. Gastrointest Endosc. 2020;92:813-820.e4. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 24] [Cited by in RCA: 47] [Article Influence: 9.4] [Reference Citation Analysis (0)] |
8. | Deo RC. Machine Learning in Medicine. Circulation. 2015;132:1920-1930. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1155] [Cited by in RCA: 1965] [Article Influence: 218.3] [Reference Citation Analysis (6)] |
9. | 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: 20107] [Article Influence: 2010.7] [Reference Citation Analysis (0)] |
10. | American Gastroenterological Association; Spechler SJ, Sharma P, Souza RF, Inadomi JM, Shaheen NJ. American Gastroenterological Association medical position statement on the management of Barrett's esophagus. Gastroenterology. 2011;140:1084-1091. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 292] [Cited by in RCA: 383] [Article Influence: 27.4] [Reference Citation Analysis (0)] |
11. | Weusten B, Bisschops R, Coron E, Dinis-Ribeiro M, Dumonceau JM, Esteban JM, Hassan C, Pech O, Repici A, Bergman J, di Pietro M. Endoscopic management of Barrett's esophagus: European Society of Gastrointestinal Endoscopy (ESGE) Position Statement. Endoscopy. 2017;49:191-198. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 321] [Cited by in RCA: 373] [Article Influence: 46.6] [Reference Citation Analysis (0)] |
12. | Hvid-Jensen F, Pedersen L, Drewes AM, Sørensen HT, Funch-Jensen P. Incidence of adenocarcinoma among patients with Barrett's esophagus. N Engl J Med. 2011;365:1375-1383. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 985] [Cited by in RCA: 982] [Article Influence: 70.1] [Reference Citation Analysis (1)] |
13. | Sikkema M, de Jonge PJ, Steyerberg EW, Kuipers EJ. Risk of esophageal adenocarcinoma and mortality in patients with Barrett's esophagus: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2010;8:235-44; quiz e32. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 266] [Cited by in RCA: 254] [Article Influence: 16.9] [Reference Citation Analysis (0)] |
14. | de Groof AJ, Struyvenberg MR, Fockens KN, van der Putten J, van der Sommen F, Boers TG, Zinger S, Bisschops R, de With PH, Pouw RE, Curvers WL, Schoon EJ, Bergman JJGHM. Deep learning algorithm detection of Barrett's neoplasia with high accuracy during live endoscopic procedures: a pilot study (with video). Gastrointest Endosc. 2020;91:1242-1250. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 66] [Cited by in RCA: 90] [Article Influence: 18.0] [Reference Citation Analysis (0)] |
15. | Struyvenberg MR, de Groof AJ, Fonollà R, van der Sommen F, de With PHN, Schoon EJ, Weusten BLAM, Leggett CL, Kahn A, Trindade AJ, Ganguly EK, Konda VJA, Lightdale CJ, Pleskow DK, Sethi A, Smith MS, Wallace MB, Wolfsen HC, Tearney GJ, Meijer SL, Vieth M, Pouw RE, Curvers WL, Bergman JJ. Prospective development and validation of a volumetric laser endomicroscopy computer algorithm for detection of Barrett's neoplasia. Gastrointest Endosc. 2021;93:871-879. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 7] [Cited by in RCA: 10] [Article Influence: 2.5] [Reference Citation Analysis (0)] |
16. | Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A, Bray F. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144:1941-1953. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3585] [Cited by in RCA: 4908] [Article Influence: 701.1] [Reference Citation Analysis (1)] |
17. | Waki K, Ishihara R, Kato Y, Shoji A, Inoue T, Matsueda K, Miyake M, Shimamoto Y, Fukuda H, Matsuura N, Ono Y, Yao K, Hashimoto S, Terai S, Ohmori M, Tanaka K, Kato M, Shono T, Miyamoto H, Tanaka Y, Tada T. Usefulness of an artificial intelligence system for the detection of esophageal squamous cell carcinoma evaluated with videos simulating overlooking situation. Dig Endosc. 2021;33:1101-1109. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 28] [Cited by in RCA: 24] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
18. | Everson MA, Garcia-Peraza-Herrera L, Wang HP, Lee CT, Chung CS, Hsieh PH, Chen CC, Tseng CH, Hsu MH, Vercauteren T, Ourselin S, Kashin S, Bisschops R, Pech O, Lovat L, Wang WL, Haidry RJ. A clinically interpretable convolutional neural network for the real-time prediction of early squamous cell cancer of the esophagus: comparing diagnostic performance with a panel of expert European and Asian endoscopists. Gastrointest Endosc. 2021;94:273-281. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 16] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
19. | Rosenfeld A, Graham DG, Jevons S, Ariza J, Hagan D, Wilson A, Lovat SJ; BEST2 study group, Sami SS, Ahmad OF, Novelli M, Rodriguez Justo M, Winstanley A, Heifetz EM, Ben-Zecharia M, Noiman U, Fitzgerald RC, Sasieni P, Lovat LB. Development and validation of a risk prediction model to diagnose Barrett's oesophagus (MARK-BE): a case-control machine learning approach. Lancet Digit Health. 2020;2:E37-E48. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 19] [Cited by in RCA: 18] [Article Influence: 3.6] [Reference Citation Analysis (0)] |
20. | Abdelrahim M, Saiko M, Maeda N, Hossain E, Alkandari A, Subramaniam S, Parra-Blanco A, Sanchez-Yague A, Coron E, Repici A, Bhandari P. Development and validation of artificial neural networks model for detection of Barrett's neoplasia: a multicenter pragmatic nonrandomized trial (with video). Gastrointest Endosc. 2023;97:422-434. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 34] [Article Influence: 17.0] [Reference Citation Analysis (0)] |
21. | Struyvenberg MR, de Groof AJ, van der Putten J, van der Sommen F, Baldaque-Silva F, Omae M, Pouw R, Bisschops R, Vieth M, Schoon EJ, Curvers WL, de With PH, Bergman JJ. A computer-assisted algorithm for narrow-band imaging-based tissue characterization in Barrett's esophagus. Gastrointest Endosc. 2021;93:89-98. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 27] [Cited by in RCA: 55] [Article Influence: 13.8] [Reference Citation Analysis (0)] |
22. | Hashimoto R, Requa J, Dao T, Ninh A, Tran E, Mai D, Lugo M, El-Hage Chehade N, Chang KJ, Karnes WE, Samarasena JB. Artificial intelligence using convolutional neural networks for real-time detection of early esophageal neoplasia in Barrett's esophagus (with video). Gastrointest Endosc. 2020;91:1264-1271.e1. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 172] [Cited by in RCA: 145] [Article Influence: 29.0] [Reference Citation Analysis (0)] |
23. | Tokai Y, Yoshio T, Aoyama K, Horie Y, Yoshimizu S, Horiuchi Y, Ishiyama A, Tsuchida T, Hirasawa T, Sakakibara Y, Yamada T, Yamaguchi S, Fujisaki J, Tada T. Application of artificial intelligence using convolutional neural networks in determining the invasion depth of esophageal squamous cell carcinoma. Esophagus. 2020;17:250-256. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 64] [Cited by in RCA: 83] [Article Influence: 16.6] [Reference Citation Analysis (0)] |
24. | Fukuda H, Ishihara R, Kato Y, Matsunaga T, Nishida T, Yamada T, Ogiyama H, Horie M, Kinoshita K, Tada T. Comparison of performances of artificial intelligence versus expert endoscopists for real-time assisted diagnosis of esophageal squamous cell carcinoma (with video). Gastrointest Endosc. 2020;92:848-855. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 73] [Cited by in RCA: 66] [Article Influence: 13.2] [Reference Citation Analysis (0)] |
25. | Li B, Cai SL, Tan WM, Li JC, Yalikong A, Feng XS, Yu HH, Lu PX, Feng Z, Yao LQ, Zhou PH, Yan B, Zhong YS. Comparative study on artificial intelligence systems for detecting early esophageal squamous cell carcinoma between narrow-band and white-light imaging. World J Gastroenterol. 2021;27:281-293. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in CrossRef: 17] [Cited by in RCA: 30] [Article Influence: 7.5] [Reference Citation Analysis (0)] |
26. | Guo L, Xiao X, Wu C, Zeng X, Zhang Y, Du J, Bai S, Xie J, Zhang Z, Li Y, Wang X, Cheung O, Sharma M, Liu J, Hu B. Real-time automated diagnosis of precancerous lesions and early esophageal squamous cell carcinoma using a deep learning model (with videos). Gastrointest Endosc. 2020;91:41-51. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 161] [Cited by in RCA: 149] [Article Influence: 29.8] [Reference Citation Analysis (0)] |
27. | Ohmori M, Ishihara R, Aoyama K, Nakagawa K, Iwagami H, Matsuura N, Shichijo S, Yamamoto K, Nagaike K, Nakahara M, Inoue T, Aoi K, Okada H, Tada T. Endoscopic detection and differentiation of esophageal lesions using a deep neural network. Gastrointest Endosc. 2020;91:301-309.e1. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 112] [Cited by in RCA: 100] [Article Influence: 20.0] [Reference Citation Analysis (0)] |
28. | Goodwin CS. Helicobacter pylori gastritis, peptic ulcer, and gastric cancer: clinical and molecular aspects. Clin Infect Dis. 1997;25:1017-1019. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 52] [Cited by in RCA: 48] [Article Influence: 1.7] [Reference Citation Analysis (0)] |
29. | Glover B, Teare J, Patel N. A systematic review of the role of non-magnified endoscopy for the assessment of H. pylori infection. Endosc Int Open. 2020;8:E105-E114. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 12] [Cited by in RCA: 18] [Article Influence: 3.6] [Reference Citation Analysis (0)] |
30. | Shichijo S, Endo Y, Aoyama K, Takeuchi Y, Ozawa T, Takiyama H, Matsuo K, Fujishiro M, Ishihara S, Ishihara R, Tada T. Application of convolutional neural networks for evaluating Helicobacter pylori infection status on the basis of endoscopic images. Scand J Gastroenterol. 2019;54:158-163. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 75] [Cited by in RCA: 65] [Article Influence: 10.8] [Reference Citation Analysis (0)] |
31. | Klein S, Gildenblat J, Ihle MA, Merkelbach-Bruse S, Noh KW, Peifer M, Quaas A, Büttner R. Deep learning for sensitive detection of Helicobacter Pylori in gastric biopsies. BMC Gastroenterol. 2020;20:417. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 13] [Cited by in RCA: 25] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
32. | Yasuda T, Hiroyasu T, Hiwa S, Okada Y, Hayashi S, Nakahata Y, Yasuda Y, Omatsu T, Obora A, Kojima T, Ichikawa H, Yagi N. Potential of automatic diagnosis system with linked color imaging for diagnosis of Helicobacter pylori infection. Dig Endosc. 2020;32:373-381. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 56] [Cited by in RCA: 48] [Article Influence: 9.6] [Reference Citation Analysis (0)] |
33. | Jung MK, Jeon SW, Park SY, Cho CM, Tak WY, Kweon YO, Kim SK, Choi YH, Bae HI. Endoscopic characteristics of gastric adenomas suggesting carcinomatous transformation. Surg Endosc. 2008;22:2705-2711. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 47] [Cited by in RCA: 54] [Article Influence: 3.2] [Reference Citation Analysis (0)] |
34. | Carmack SW, Genta RM, Graham DY, Lauwers GY. Management of gastric polyps: a pathology-based guide for gastroenterologists. Nat Rev Gastroenterol Hepatol. 2009;6:331-341. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 100] [Cited by in RCA: 97] [Article Influence: 6.1] [Reference Citation Analysis (1)] |
35. | He C, Zhang J, Liang Y, Li H. A unified framework harnessing multi-scale feature ensemble and attention mechanism for gastric polyp and protrusion identification in gastroscope imaging. Sci Rep. 2025;15:5734. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
36. | Chen X, Zhang K, Lin S, Dai KF, Yun Y. Single Shot Multibox Detector Automatic Polyp Detection Network Based on Gastrointestinal Endoscopic Images. Comput Math Methods Med. 2021;2021:2144472. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 5] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
37. | Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 75126] [Cited by in RCA: 64880] [Article Influence: 16220.0] [Reference Citation Analysis (177)] |
38. | Colom R, Karama S, Jung RE, Haier RJ. Human intelligence and brain networks. Dialogues Clin Neurosci. 2010;12:489-501. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 154] [Cited by in RCA: 157] [Article Influence: 11.2] [Reference Citation Analysis (0)] |
39. | Li L, Chen Y, Shen Z, Zhang X, Sang J, Ding Y, Yang X, Li J, Chen M, Jin C, Chen C, Yu C. Convolutional neural network for the diagnosis of early gastric cancer based on magnifying narrow band imaging. Gastric Cancer. 2020;23:126-132. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 168] [Cited by in RCA: 140] [Article Influence: 28.0] [Reference Citation Analysis (0)] |
40. | Feng J, Zhang Y, Feng Z, Ma H, Gou Y, Wang P, Feng Y, Wang X, Huang X. A prospective and comparative study on improving the diagnostic accuracy of early gastric cancer based on deep convolutional neural network real-time diagnosis system (with video). Surg Endosc. 2025;39:1874-1884. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
41. | You Y, Liang Y, Chen L, Li Z, Gao B, Wang X, Yuan M, Xue Y, Liu Y, Gao J. Radiomics analysis of dual-energy CT-derived iodine maps for differentiating between T1/2 and T3/4a in gastric cancer: A multicenter study. Eur J Radiol. 2025;186:112054. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
42. | Li Q, Yan S, Yang W, Du Z, Cheng M, Chen R, Shao Q, Tian Y, Sheng M, Peng W, Wu Y. Machine learning models for prediction of lymph node metastasis in patients with gastric cancer: a Chinese single-centre study with external validation in an Asian American population. BMJ Open. 2025;15:e098476. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Reference Citation Analysis (0)] |
43. | Martin DR, Hanson JA, Gullapalli RR, Schultz FA, Sethi A, Clark DP. A Deep Learning Convolutional Neural Network Can Recognize Common Patterns of Injury in Gastric Pathology. Arch Pathol Lab Med. 2020;144:370-378. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 24] [Cited by in RCA: 35] [Article Influence: 5.8] [Reference Citation Analysis (0)] |
44. | Mohan BP, Khan SR, Kassab LL, Ponnada S, Mohy-Ud-Din N, Chandan S, Dulai PS, Kochhar GS. Convolutional neural networks in the computer-aided diagnosis of Helicobacter pylori infection and non-causal comparison to physician endoscopists: a systematic review with meta-analysis. Ann Gastroenterol. 2021;34:20-25. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 8] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
45. | Nakashima H, Kawahira H, Kawachi H, Sakaki N. Endoscopic three-categorical diagnosis of Helicobacter pylori infection using linked color imaging and deep learning: a single-center prospective study (with video). Gastric Cancer. 2020;23:1033-1040. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 27] [Cited by in RCA: 50] [Article Influence: 10.0] [Reference Citation Analysis (0)] |
46. | Yuan XL, Zhou Y, Liu W, Luo Q, Zeng XH, Yi Z, Hu B. Artificial intelligence for diagnosing gastric lesions under white-light endoscopy. Surg Endosc. 2022;36:9444-9453. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 14] [Cited by in RCA: 16] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
47. | Cao C, Wang R, Yu Y, Zhang H, Yu Y, Sun C. Gastric polyp detection in gastroscopic images using deep neural network. PLoS One. 2021;16:e0250632. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 6] [Cited by in RCA: 17] [Article Influence: 4.3] [Reference Citation Analysis (0)] |
48. | Horiuchi Y, Aoyama K, Tokai Y, Hirasawa T, Yoshimizu S, Ishiyama A, Yoshio T, Tsuchida T, Fujisaki J, Tada T. Convolutional Neural Network for Differentiating Gastric Cancer from Gastritis Using Magnified Endoscopy with Narrow Band Imaging. Dig Dis Sci. 2020;65:1355-1363. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 70] [Cited by in RCA: 103] [Article Influence: 20.6] [Reference Citation Analysis (1)] |
49. | Bu F, Shen X, Zhan H, Wang D, Min L, Song Y, Wang S. Efficient Metabolomics Profiling from Plasma Extracellular Vesicles Enables Accurate Diagnosis of Early Gastric Cancer. J Am Chem Soc. 2025;147:8672-8686. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
50. | Wang Y, Liu W, Yu Y, Liu JJ, Jiang L, Xue HD, Lei J, Jin Z, Yu JC. Prediction of the Depth of Tumor Invasion in Gastric Cancer: Potential Role of CT Radiomics. Acad Radiol. 2020;27:1077-1084. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 7] [Cited by in RCA: 28] [Article Influence: 5.6] [Reference Citation Analysis (0)] |
51. | Shang H, Feng T, Han D, Liang F, Zhao B, Xu L, Cao Z. Deep learning and radiomics for gastric cancer serosal invasion: automated segmentation and multi-machine learning from two centers. J Cancer Res Clin Oncol. 2025;151:60. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Reference Citation Analysis (0)] |
52. | Kang D, Jeon HJ, Kim JH, Oh SI, Seong YS, Jang JY, Kim JW, Kim JS, Nam SJ, Bang CS, Choi HS. Enhancing Lymph Node Metastasis Risk Prediction in Early Gastric Cancer Through the Integration of Endoscopic Images and Real-World Data in a Multimodal AI Model. Cancers (Basel). 2025;17:869. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
53. | Chen G, Xie Y, Yang B, Tan J, Zhong G, Zhong L, Zhou S, Han F. Artificial intelligence model for perigastric blood vessel recognition during laparoscopic radical gastrectomy with D2 lymphadenectomy in locally advanced gastric cancer. BJS Open. 2024;9:zrae158. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Reference Citation Analysis (0)] |
54. | Zhang W, Fang M, Dong D, Wang X, Ke X, Zhang L, Hu C, Guo L, Guan X, Zhou J, Shan X, Tian J. Development and validation of a CT-based radiomic nomogram for preoperative prediction of early recurrence in advanced gastric cancer. Radiother Oncol. 2020;145:13-20. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 38] [Cited by in RCA: 106] [Article Influence: 21.2] [Reference Citation Analysis (0)] |
55. | Dong D, Fang MJ, Tang L, Shan XH, Gao JB, Giganti F, Wang RP, Chen X, Wang XX, Palumbo D, Fu J, Li WC, Li J, Zhong LZ, De Cobelli F, Ji JF, Liu ZY, Tian J. Deep learning radiomic nomogram can predict the number of lymph node metastasis in locally advanced gastric cancer: an international multicenter study. Ann Oncol. 2020;31:912-920. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 90] [Cited by in RCA: 270] [Article Influence: 54.0] [Reference Citation Analysis (1)] |
56. | Huang P, Li S, Deng Z, Hu F, Jin D, Xiong S, Fan B. Machine-Learning Based Computed Tomography Radiomics Nomgram For Predicting Perineural Invasion In Gastric Cancer. Curr Med Imaging. 2025;21:e15734056323323. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
57. | Podolsky DK. Inflammatory bowel disease. N Engl J Med. 2002;347:417-429. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2693] [Cited by in RCA: 2748] [Article Influence: 119.5] [Reference Citation Analysis (2)] |
58. | Jackson BD, Con D, Gorelik A, Liew D, Knowles S, De Cruz P. Examination of the relationship between disease activity and patient-reported outcome measures in an inflammatory bowel disease cohort. Intern Med J. 2018;48:1234-1241. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 7] [Cited by in RCA: 11] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
59. | Denson LA, Curran M, McGovern DPB, Koltun WA, Duerr RH, Kim SC, Sartor RB, Sylvester FA, Abraham C, de Zoeten EF, Siegel CA, Burns RM, Dobes AM, Shtraizent N, Honig G, Heller CA, Hurtado-Lorenzo A, Cho JH. Challenges in IBD Research: Precision Medicine. Inflamm Bowel Dis. 2019;25:S31-S39. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 49] [Cited by in RCA: 73] [Article Influence: 12.2] [Reference Citation Analysis (0)] |
60. | Kim TW, Park SK, Chun J, Kim S, Choi CH, Kang SB, Bang KB, Kim TO, Seo GS, Cha JM, Jung Y, Kim HG, Im JP, Ahn KS, Lee CK, Kim HJ, Kim S, Park DI. Early Progression Prediction in Korean Crohn's Disease Using a Korean-Specific PrediXcan Model. Int J Mol Sci. 2025;26:2910. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
61. | Udristoiu AL, Stefanescu D, Gruionu G, Gruionu LG, Iacob AV, Karstensen JG, Vilman P, Saftoiu A. Deep Learning Algorithm for the Confirmation of Mucosal Healing in Crohn's Disease, Based on Confocal Laser Endomicroscopy Images. J Gastrointestin Liver Dis. 2021;30:59-65. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 14] [Article Influence: 3.5] [Reference Citation Analysis (0)] |
62. | Ferreira JPS, de Mascarenhas Saraiva MJDQEC, Afonso JPL, Ribeiro TFC, Cardoso HMC, Ribeiro Andrade AP, de Mascarenhas Saraiva MNG, Parente MPL, Natal Jorge R, Lopes SIO, de Macedo GMG. Identification of Ulcers and Erosions by the Novel Pillcam™ Crohn's Capsule Using a Convolutional Neural Network: A Multicentre Pilot Study. J Crohns Colitis. 2022;16:169-172. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 11] [Cited by in RCA: 35] [Article Influence: 11.7] [Reference Citation Analysis (1)] |
63. | Ahmed M. Gastrointestinal neuroendocrine tumors in 2020. World J Gastrointest Oncol. 2020;12:791-807. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in CrossRef: 153] [Cited by in RCA: 138] [Article Influence: 27.6] [Reference Citation Analysis (11)] |
64. | Xu Z, Wang L, Dai S, Chen M, Li F, Sun J, Luo F. Epidemiologic Trends of and Factors Associated With Overall Survival for Patients With Gastroenteropancreatic Neuroendocrine Tumors in the United States. JAMA Netw Open. 2021;4:e2124750. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 50] [Cited by in RCA: 150] [Article Influence: 37.5] [Reference Citation Analysis (0)] |
65. | Blazevic A, Starmans MPA, Brabander T, Dwarkasing RS, van Gils RAH, Hofland J, Franssen GJH, Feelders RA, Niessen WJ, Klein S, de Herder WW. Predicting symptomatic mesenteric mass in small intestinal neuroendocrine tumors using radiomics. Endocr Relat Cancer. 2021;28:529-539. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Article Influence: 0.5] [Reference Citation Analysis (0)] |
66. | Li X, Liang D, Meng J, Zhou J, Chen Z, Huang S, Lu B, Qiu Y, Baker ME, Ye Z, Cao Q, Wang M, Yuan C, Chen Z, Feng S, Zhang Y, Iacucci M, Ghosh S, Rieder F, Sun C, Chen M, Li Z, Mao R, Huang B, Feng ST. Development and Validation of a Novel Computed-Tomography Enterography Radiomic Approach for Characterization of Intestinal Fibrosis in Crohn's Disease. Gastroenterology. 2021;160:2303-2316.e11. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 79] [Cited by in RCA: 79] [Article Influence: 19.8] [Reference Citation Analysis (0)] |
67. | Majtner T, Brodersen JB, Herp J, Kjeldsen J, Halling ML, Jensen MD. A deep learning framework for autonomous detection and classification of Crohn's disease lesions in the small bowel and colon with capsule endoscopy. Endosc Int Open. 2021;9:E1361-E1370. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 14] [Article Influence: 3.5] [Reference Citation Analysis (0)] |
68. | Klang E, Grinman A, Soffer S, Margalit Yehuda R, Barzilay O, Amitai MM, Konen E, Ben-Horin S, Eliakim R, Barash Y, Kopylov U. Automated Detection of Crohn's Disease Intestinal Strictures on Capsule Endoscopy Images Using Deep Neural Networks. J Crohns Colitis. 2021;15:749-756. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 18] [Cited by in RCA: 56] [Article Influence: 14.0] [Reference Citation Analysis (0)] |
69. | Konikoff T, Goren I, Yalon M, Tamir S, Avni-Biron I, Yanai H, Dotan I, Ollech JE. Machine learning for selecting patients with Crohn's disease for abdominopelvic computed tomography in the emergency department. Dig Liver Dis. 2021;53:1559-1564. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 3] [Article Influence: 0.8] [Reference Citation Analysis (0)] |
70. | Con D, van Langenberg DR, Vasudevan A. Deep learning vs conventional learning algorithms for clinical prediction in Crohn's disease: A proof-of-concept study. World J Gastroenterol. 2021;27:6476-6488. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in CrossRef: 16] [Cited by in RCA: 17] [Article Influence: 4.3] [Reference Citation Analysis (2)] |
71. | Ungaro RC, Hu L, Ji J, Nayar S, Kugathasan S, Denson LA, Hyams J, Dubinsky MC, Sands BE, Cho JH. Machine learning identifies novel blood protein predictors of penetrating and stricturing complications in newly diagnosed paediatric Crohn's disease. Aliment Pharmacol Ther. 2021;53:281-290. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 28] [Cited by in RCA: 27] [Article Influence: 6.8] [Reference Citation Analysis (0)] |
72. | Stidham RW, Liu Y, Enchakalody B, Van T, Krishnamurthy V, Su GL, Zhu J, Waljee AK. The Use of Readily Available Longitudinal Data to Predict the Likelihood of Surgery in Crohn Disease. Inflamm Bowel Dis. 2021;27:1328-1334. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 7] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
73. | Kjellman M, Knigge U, Welin S, Thiis-Evensen E, Gronbaek H, Schalin-Jäntti C, Sorbye H, Joergensen MT, Johanson V, Metso S, Waldum H, Søreide JA, Ebeling T, Lindberg F, Landerholm K, Wallin G, Salem F, Schneider MDP, Belusa R. A Plasma Protein Biomarker Strategy for Detection of Small Intestinal Neuroendocrine Tumors. Neuroendocrinology. 2021;111:840-849. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 5] [Cited by in RCA: 8] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
74. | Clift AK, Mahon H, Khan G, Boardman-Pretty F, Worker A, Marchini E, Buendia O, Fish P, Khan MS. Identifying patients with undiagnosed small intestinal neuroendocrine tumours in primary care using statistical and machine learning: model development and validation study. Br J Cancer. 2024;131:305-311. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
75. | Ungaro R, Mehandru S, Allen PB, Peyrin-Biroulet L, Colombel JF. Ulcerative colitis. Lancet. 2017;389:1756-1770. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2199] [Cited by in RCA: 2497] [Article Influence: 312.1] [Reference Citation Analysis (2)] |
76. | Baumgart DC, Le Berre C. Newer Biologic and Small-Molecule Therapies for Inflammatory Bowel Disease. N Engl J Med. 2021;385:1302-1315. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 57] [Cited by in RCA: 217] [Article Influence: 54.3] [Reference Citation Analysis (0)] |
77. | Pariente B, Cosnes J, Danese S, Sandborn WJ, Lewin M, Fletcher JG, Chowers Y, D'Haens G, Feagan BG, Hibi T, Hommes DW, Irvine EJ, Kamm MA, Loftus EV Jr, Louis E, Michetti P, Munkholm P, Oresland T, Panés J, Peyrin-Biroulet L, Reinisch W, Sands BE, Schoelmerich J, Schreiber S, Tilg H, Travis S, van Assche G, Vecchi M, Mary JY, Colombel JF, Lémann M. Development of the Crohn's disease digestive damage score, the Lémann score. Inflamm Bowel Dis. 2011;17:1415-1422. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 412] [Cited by in RCA: 464] [Article Influence: 33.1] [Reference Citation Analysis (1)] |
78. | Sturm A, Maaser C, Calabrese E, Annese V, Fiorino G, Kucharzik T, Vavricka SR, Verstockt B, van Rheenen P, Tolan D, Taylor SA, Rimola J, Rieder F, Limdi JK, Laghi A, Krustiņš E, Kotze PG, Kopylov U, Katsanos K, Halligan S, Gordon H, González Lama Y, Ellul P, Eliakim R, Castiglione F, Burisch J, Borralho Nunes P, Bettenworth D, Baumgart DC, Stoker J; European Crohn’s and Colitis Organisation [ECCO] and the European Society of Gastrointestinal and Abdominal Radiology [ESGAR]. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 2: IBD scores and general principles and technical aspects. J Crohns Colitis. 2019;13:273-284. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 319] [Cited by in RCA: 304] [Article Influence: 50.7] [Reference Citation Analysis (1)] |
79. | Maaser C, Sturm A, Vavricka SR, Kucharzik T, Fiorino G, Annese V, Calabrese E, Baumgart DC, Bettenworth D, Borralho Nunes P, Burisch J, Castiglione F, Eliakim R, Ellul P, González-Lama Y, Gordon H, Halligan S, Katsanos K, Kopylov U, Kotze PG, Krustinš E, Laghi A, Limdi JK, Rieder F, Rimola J, Taylor SA, Tolan D, van Rheenen P, Verstockt B, Stoker J; European Crohn’s and Colitis Organisation [ECCO] and the European Society of Gastrointestinal and Abdominal Radiology [ESGAR]. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial diagnosis, monitoring of known IBD, detection of complications. J Crohns Colitis. 2019;13:144-164. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1242] [Cited by in RCA: 1180] [Article Influence: 196.7] [Reference Citation Analysis (0)] |
80. | Mohammed Vashist N, Samaan M, Mosli MH, Parker CE, MacDonald JK, Nelson SA, Zou GY, Feagan BG, Khanna R, Jairath V. Endoscopic scoring indices for evaluation of disease activity in ulcerative colitis. Cochrane Database Syst Rev. 2018;1:CD011450. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 36] [Cited by in RCA: 89] [Article Influence: 12.7] [Reference Citation Analysis (0)] |
81. | Wang L, Chen L, Wang X, Liu K, Li T, Yu Y, Han J, Xing S, Xu J, Tian D, Seidler U, Xiao F. Development of a Convolutional Neural Network-Based Colonoscopy Image Assessment Model for Differentiating Crohn's Disease and Ulcerative Colitis. Front Med (Lausanne). 2022;9:789862. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 7] [Reference Citation Analysis (0)] |
82. | Higuchi N, Hiraga H, Sasaki Y, Hiraga N, Igarashi S, Hasui K, Ogasawara K, Maeda T, Murai Y, Tatsuta T, Kikuchi H, Chinda D, Mikami T, Matsuzaka M, Sakuraba H, Fukuda S. Automated evaluation of colon capsule endoscopic severity of ulcerative colitis using ResNet50. PLoS One. 2022;17:e0269728. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 14] [Article Influence: 4.7] [Reference Citation Analysis (0)] |
83. | Bhambhvani HP, Zamora A. Deep learning enabled classification of Mayo endoscopic subscore in patients with ulcerative colitis. Eur J Gastroenterol Hepatol. 2021;33:645-649. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 16] [Cited by in RCA: 34] [Article Influence: 8.5] [Reference Citation Analysis (0)] |
84. | Gschwantler M, Kriwanek S, Langner E, Göritzer B, Schrutka-Kölbl C, Brownstone E, Feichtinger H, Weiss W. High-grade dysplasia and invasive carcinoma in colorectal adenomas: a multivariate analysis of the impact of adenoma and patient characteristics. Eur J Gastroenterol Hepatol. 2002;14:183-188. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 115] [Cited by in RCA: 101] [Article Influence: 4.4] [Reference Citation Analysis (0)] |
85. | Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in colorectal cancer incidence and mortality. Gut. 2017;66:683-691. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3058] [Cited by in RCA: 3310] [Article Influence: 413.8] [Reference Citation Analysis (3)] |
86. | Leufkens AM, van Oijen MG, Vleggaar FP, Siersema PD. Factors influencing the miss rate of polyps in a back-to-back colonoscopy study. Endoscopy. 2012;44:470-475. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 205] [Cited by in RCA: 202] [Article Influence: 15.5] [Reference Citation Analysis (0)] |
87. | Rabeneck L, Souchek J, El-Serag HB. Survival of colorectal cancer patients hospitalized in the Veterans Affairs Health Care System. Am J Gastroenterol. 2003;98:1186-1192. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 66] [Cited by in RCA: 70] [Article Influence: 3.2] [Reference Citation Analysis (0)] |
88. | Ozawa T, Ishihara S, Fujishiro M, Kumagai Y, Shichijo S, Tada T. Automated endoscopic detection and classification of colorectal polyps using convolutional neural networks. Therap Adv Gastroenterol. 2020;13:1756284820910659. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 105] [Cited by in RCA: 76] [Article Influence: 15.2] [Reference Citation Analysis (0)] |
89. | Song EM, Park B, Ha CA, Hwang SW, Park SH, Yang DH, Ye BD, Myung SJ, Yang SK, Kim N, Byeon JS. Endoscopic diagnosis and treatment planning for colorectal polyps using a deep-learning model. Sci Rep. 2020;10:30. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 65] [Cited by in RCA: 64] [Article Influence: 12.8] [Reference Citation Analysis (0)] |
90. | Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RGS, Barzi A, Jemal A. Colorectal cancer statistics, 2017. CA Cancer J Clin. 2017;67:177-193. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2526] [Cited by in RCA: 2912] [Article Influence: 364.0] [Reference Citation Analysis (3)] |
91. | Knudsen AB, Zauber AG, Rutter CM, Naber SK, Doria-Rose VP, Pabiniak C, Johanson C, Fischer SE, Lansdorp-Vogelaar I, Kuntz KM. Estimation of Benefits, Burden, and Harms of Colorectal Cancer Screening Strategies: Modeling Study for the US Preventive Services Task Force. JAMA. 2016;315:2595-2609. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 366] [Cited by in RCA: 378] [Article Influence: 42.0] [Reference Citation Analysis (0)] |
92. | Simon K. Colorectal cancer development and advances in screening. Clin Interv Aging. 2016;11:967-976. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 245] [Cited by in RCA: 428] [Article Influence: 47.6] [Reference Citation Analysis (0)] |
93. | Sharara AI, El Reda ZD, Harb AH, Abou Fadel CG, Sarkis FS, Chalhoub JM, Abou Mrad R. The burden of bowel preparations in patients undergoing elective colonoscopy. United European Gastroenterol J. 2016;4:314-318. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 36] [Cited by in RCA: 33] [Article Influence: 3.7] [Reference Citation Analysis (0)] |
94. | Ribeiro MS, Wallace MB. Endoscopic Treatment of Early Cancer of the Colon. Gastroenterol Hepatol (N Y). 2015;11:445-452. [PubMed] |
95. | Zhang ZY, Gao W, Luo QF, Yin XW, Basnet S, Dai ZL, Ge HY. A nomogram improves AJCC stages for colorectal cancers by introducing CEA, modified lymph node ratio and negative lymph node count. Sci Rep. 2016;6:39028. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 12] [Cited by in RCA: 20] [Article Influence: 2.2] [Reference Citation Analysis (0)] |
96. | Schneider JL, Layefsky E, Udaltsova N, Levin TR, Corley DA. Validation of an Algorithm to Identify Patients at Risk for Colorectal Cancer Based on Laboratory Test and Demographic Data in Diverse, Community-Based Population. Clin Gastroenterol Hepatol. 2020;18:2734-2741.e6. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 12] [Cited by in RCA: 15] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
97. | Ito N, Kawahira H, Nakashima H, Uesato M, Miyauchi H, Matsubara H. Endoscopic Diagnostic Support System for cT1b Colorectal Cancer Using Deep Learning. Oncology. 2019;96:44-50. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 76] [Cited by in RCA: 60] [Article Influence: 8.6] [Reference Citation Analysis (0)] |
98. | Jiang D, Liao J, Duan H, Wu Q, Owen G, Shu C, Chen L, He Y, Wu Z, He D, Zhang W, Wang Z. A machine learning-based prognostic predictor for stage III colon cancer. Sci Rep. 2020;10:10333. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 25] [Cited by in RCA: 29] [Article Influence: 5.8] [Reference Citation Analysis (0)] |
99. | Sutton RT, Zai Ane OR, Goebel R, Baumgart DC. Artificial intelligence enabled automated diagnosis and grading of ulcerative colitis endoscopy images. Sci Rep. 2022;12:2748. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 52] [Cited by in RCA: 48] [Article Influence: 16.0] [Reference Citation Analysis (0)] |
100. | Lo B, Liu Z, Bendtsen F, Igel C, Vind I, Burisch J. High Accuracy in Classifying Endoscopic Severity in Ulcerative Colitis Using Convolutional Neural Network. Am J Gastroenterol. 2022;117:1648-1654. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 17] [Reference Citation Analysis (0)] |
101. | Ruan G, Qi J, Cheng Y, Liu R, Zhang B, Zhi M, Chen J, Xiao F, Shen X, Fan L, Li Q, Li N, Qiu Z, Xiao Z, Xu F, Lv L, Chen M, Ying S, Chen L, Tian Y, Li G, Zhang Z, He M, Qiao L, Zhang Z, Chen D, Cao Q, Nian Y, Wei Y. Development and Validation of a Deep Neural Network for Accurate Identification of Endoscopic Images From Patients With Ulcerative Colitis and Crohn's Disease. Front Med (Lausanne). 2022;9:854677. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 21] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
102. | Gutierrez Becker B, Arcadu F, Thalhammer A, Gamez Serna C, Feehan O, Drawnel F, Oh YS, Prunotto M. Training and deploying a deep learning model for endoscopic severity grading in ulcerative colitis using multicenter clinical trial data. Ther Adv Gastrointest Endosc. 2021;14:2631774521990623. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 9] [Cited by in RCA: 28] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
103. | Bossuyt P, Nakase H, Vermeire S, de Hertogh G, Eelbode T, Ferrante M, Hasegawa T, Willekens H, Ikemoto Y, Makino T, Bisschops R. Automatic, computer-aided determination of endoscopic and histological inflammation in patients with mild to moderate ulcerative colitis based on red density. Gut. 2020;69:1778-1786. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 49] [Cited by in RCA: 84] [Article Influence: 16.8] [Reference Citation Analysis (0)] |
104. | Iacucci M, Cannatelli R, Parigi TL, Nardone OM, Tontini GE, Labarile N, Buda A, Rimondi A, Bazarova A, Bisschops R, Del Amor R, Meseguer P, Naranjo V, Ghosh S, Grisan E; PICaSSO group. A virtual chromoendoscopy artificial intelligence system to detect endoscopic and histologic activity/remission and predict clinical outcomes in ulcerative colitis. Endoscopy. 2023;55:332-341. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 37] [Cited by in RCA: 38] [Article Influence: 19.0] [Reference Citation Analysis (0)] |
105. | Huang TY, Zhan SQ, Chen PJ, Yang CW, Lu HH. Accurate diagnosis of endoscopic mucosal healing in ulcerative colitis using deep learning and machine learning. J Chin Med Assoc. 2021;84:678-681. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 5] [Cited by in RCA: 19] [Article Influence: 4.8] [Reference Citation Analysis (0)] |
106. | Popa IV, Burlacu A, Mihai C, Prelipcean CC. A Machine Learning Model Accurately Predicts Ulcerative Colitis Activity at One Year in Patients Treated with Anti-Tumour Necrosis Factor α Agents. Medicina (Kaunas). 2020;56:628. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 6] [Cited by in RCA: 13] [Article Influence: 2.6] [Reference Citation Analysis (0)] |
107. | Takenaka K, Ohtsuka K, Fujii T, Negi M, Suzuki K, Shimizu H, Oshima S, Akiyama S, Motobayashi M, Nagahori M, Saito E, Matsuoka K, Watanabe M. Development and Validation of a Deep Neural Network for Accurate Evaluation of Endoscopic Images From Patients With Ulcerative Colitis. Gastroenterology. 2020;158:2150-2157. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 205] [Cited by in RCA: 181] [Article Influence: 36.2] [Reference Citation Analysis (0)] |
108. | Maeda Y, Kudo SE, Ogata N, Misawa M, Iacucci M, Homma M, Nemoto T, Takishima K, Mochida K, Miyachi H, Baba T, Mori K, Ohtsuka K, Mori Y. Evaluation in real-time use of artificial intelligence during colonoscopy to predict relapse of ulcerative colitis: a prospective study. Gastrointest Endosc. 2022;95:747-756.e2. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 7] [Cited by in RCA: 45] [Article Influence: 15.0] [Reference Citation Analysis (0)] |
109. | Wang W, Tian J, Zhang C, Luo Y, Wang X, Li J. An improved deep learning approach and its applications on colonic polyp images detection. BMC Med Imaging. 2020;20:83. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 17] [Cited by in RCA: 34] [Article Influence: 6.8] [Reference Citation Analysis (0)] |
110. | Jin EH, Lee D, Bae JH, Kang HY, Kwak MS, Seo JY, Yang JI, Yang SY, Lim SH, Yim JY, Lim JH, Chung GE, Chung SJ, Choi JM, Han YM, Kang SJ, Lee J, Chan Kim H, Kim JS. Improved Accuracy in Optical Diagnosis of Colorectal Polyps Using Convolutional Neural Networks with Visual Explanations. Gastroenterology. 2020;158:2169-2179.e8. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 59] [Cited by in RCA: 91] [Article Influence: 18.2] [Reference Citation Analysis (0)] |
111. | Sakamoto T, Nakashima H, Nakamura K, Nagahama R, Saito Y. Performance of Computer-Aided Detection and Diagnosis of Colorectal Polyps Compares to That of Experienced Endoscopists. Dig Dis Sci. 2022;67:3976-3983. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 19] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
112. | Zachariah R, Samarasena J, Luba D, Duh E, Dao T, Requa J, Ninh A, Karnes W. Prediction of Polyp Pathology Using Convolutional Neural Networks Achieves "Resect and Discard" Thresholds. Am J Gastroenterol. 2020;115:138-144. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 52] [Cited by in RCA: 84] [Article Influence: 16.8] [Reference Citation Analysis (0)] |
113. | Wickstrøm K, Kampffmeyer M, Jenssen R. Uncertainty and interpretability in convolutional neural networks for semantic segmentation of colorectal polyps. Med Image Anal. 2020;60:101619. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 52] [Cited by in RCA: 64] [Article Influence: 10.7] [Reference Citation Analysis (0)] |
114. | Su JR, Li Z, Shao XJ, Ji CR, Ji R, Zhou RC, Li GC, Liu GQ, He YS, Zuo XL, Li YQ. Impact of a real-time automatic quality control system on colorectal polyp and adenoma detection: a prospective randomized controlled study (with videos). Gastrointest Endosc. 2020;91:415-424.e4. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 153] [Cited by in RCA: 209] [Article Influence: 41.8] [Reference Citation Analysis (0)] |
115. | Luo H, Zhao Q, Wei W, Zheng L, Yi S, Li G, Wang W, Sheng H, Pu H, Mo H, Zuo Z, Liu Z, Li C, Xie C, Zeng Z, Li W, Hao X, Liu Y, Cao S, Liu W, Gibson S, Zhang K, Xu G, Xu RH. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci Transl Med. 2020;12:eaax7533. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 186] [Cited by in RCA: 282] [Article Influence: 56.4] [Reference Citation Analysis (0)] |
116. | Arabameri A, Asemani D, Teymourpour P. Detection of Colorectal Carcinoma Based on Microbiota Analysis Using Generalized Regression Neural Networks and Nonlinear Feature Selection. IEEE/ACM Trans Comput Biol Bioinform. 2020;17:547-557. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 5] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
117. | Zeng Y, Xu S, Chapman WC Jr, Li S, Alipour Z, Abdelal H, Chatterjee D, Mutch M, Zhu Q. Real-time colorectal cancer diagnosis using PR-OCT with deep learning. Theranostics. 2020;10:2587-2596. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 36] [Cited by in RCA: 35] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
118. | Wang D, Xu J, Zhang Z, Li S, Zhang X, Zhou Y, Zhang X, Lu Y. Evaluation of Rectal Cancer Circumferential Resection Margin Using Faster Region-Based Convolutional Neural Network in High-Resolution Magnetic Resonance Images. Dis Colon Rectum. 2020;63:143-151. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 14] [Cited by in RCA: 26] [Article Influence: 5.2] [Reference Citation Analysis (0)] |
119. | Yang C, Jiang ZK, Liu LH, Zeng MS. Pre-treatment ADC image-based random forest classifier for identifying resistant rectal adenocarcinoma to neoadjuvant chemoradiotherapy. Int J Colorectal Dis. 2020;35:101-107. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 15] [Cited by in RCA: 21] [Article Influence: 4.2] [Reference Citation Analysis (0)] |
120. | Fu J, Zhong X, Li N, Van Dams R, Lewis J, Sung K, Raldow AC, Jin J, Qi XS. Deep learning-based radiomic features for improving neoadjuvant chemoradiation response prediction in locally advanced rectal cancer. Phys Med Biol. 2020;65:075001. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 22] [Cited by in RCA: 42] [Article Influence: 8.4] [Reference Citation Analysis (0)] |
121. | Xu Y, Ju L, Tong J, Zhou CM, Yang JJ. Machine Learning Algorithms for Predicting the Recurrence of Stage IV Colorectal Cancer After Tumor Resection. Sci Rep. 2020;10:2519. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 61] [Cited by in RCA: 51] [Article Influence: 10.2] [Reference Citation Analysis (0)] |
122. | Zhao X, Xie P, Wang M, Li W, Pickhardt PJ, Xia W, Xiong F, Zhang R, Xie Y, Jian J, Bai H, Ni C, Gu J, Yu T, Tang Y, Gao X, Meng X. Deep learning-based fully automated detection and segmentation of lymph nodes on multiparametric-mri for rectal cancer: A multicentre study. EBioMedicine. 2020;56:102780. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 56] [Cited by in RCA: 51] [Article Influence: 10.2] [Reference Citation Analysis (0)] |
123. | Reichling C, Taieb J, Derangere V, Klopfenstein Q, Le Malicot K, Gornet JM, Becheur H, Fein F, Cojocarasu O, Kaminsky MC, Lagasse JP, Luet D, Nguyen S, Etienne PL, Gasmi M, Vanoli A, Perrier H, Puig PL, Emile JF, Lepage C, Ghiringhelli F. Artificial intelligence-guided tissue analysis combined with immune infiltrate assessment predicts stage III colon cancer outcomes in PETACC08 study. Gut. 2020;69:681-690. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 55] [Cited by in RCA: 84] [Article Influence: 16.8] [Reference Citation Analysis (0)] |
124. | Skrede OJ, De Raedt S, Kleppe A, Hveem TS, Liestøl K, Maddison J, Askautrud HA, Pradhan M, Nesheim JA, Albregtsen F, Farstad IN, Domingo E, Church DN, Nesbakken A, Shepherd NA, Tomlinson I, Kerr R, Novelli M, Kerr DJ, Danielsen HE. Deep learning for prediction of colorectal cancer outcome: a discovery and validation study. Lancet. 2020;395:350-360. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 332] [Cited by in RCA: 368] [Article Influence: 73.6] [Reference Citation Analysis (0)] |
125. | Väyrynen JP, Lau MC, Haruki K, Väyrynen SA, Dias Costa A, Borowsky J, Zhao M, Fujiyoshi K, Arima K, Twombly TS, Kishikawa J, Gu S, Aminmozaffari S, Shi S, Baba Y, Akimoto N, Ugai T, Da Silva A, Song M, Wu K, Chan AT, Nishihara R, Fuchs CS, Meyerhardt JA, Giannakis M, Ogino S, Nowak JA. Prognostic Significance of Immune Cell Populations Identified by Machine Learning in Colorectal Cancer Using Routine Hematoxylin and Eosin-Stained Sections. Clin Cancer Res. 2020;26:4326-4338. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 33] [Cited by in RCA: 49] [Article Influence: 9.8] [Reference Citation Analysis (0)] |
126. | Xu SS, Chang C, Su C, Phu PQ. Classification of Liver Diseases Based on Ultrasound Image Texture Features. Appl Sci. 2019;9:342. [RCA] [DOI] [Full Text] [Cited by in Crossref: 14] [Cited by in RCA: 16] [Article Influence: 2.7] [Reference Citation Analysis (0)] |
127. | Pisani P, Parkin DM, Bray F, Ferlay J. Estimates of the worldwide mortality from 25 cancers in 1990. Int J Cancer. 1999;83:18-29. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 30] [Reference Citation Analysis (0)] |
128. | Smith A, Baumgartner K, Bositis C. Cirrhosis: Diagnosis and Management. Am Fam Physician. 2019;100:759-770. [PubMed] |
129. | Intraobserver and interobserver variations in liver biopsy interpretation in patients with chronic hepatitis C. The French METAVIR Cooperative Study Group. Hepatology. 1994;20:15-20. [PubMed] |
130. | Barbois S, Arvieux C, Leroy V, Reche F, Stürm N, Borel AL. Benefit-risk of intraoperative liver biopsy during bariatric surgery: review and perspectives. Surg Obes Relat Dis. 2017;13:1780-1786. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 21] [Cited by in RCA: 25] [Article Influence: 3.1] [Reference Citation Analysis (0)] |
131. | Brattain LJ, Telfer BA, Dhyani M, Grajo JR, Samir AE. Machine learning for medical ultrasound: status, methods, and future opportunities. Abdom Radiol (NY). 2018;43:786-799. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 157] [Cited by in RCA: 135] [Article Influence: 19.3] [Reference Citation Analysis (0)] |
132. | Park BK, Kim CK, Choi HY, Lee HM, Jeon SS, Seo SI, Han DH. Limitation for performing ultrasound-guided radiofrequency ablation of small renal masses. Eur J Radiol. 2010;75:248-252. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 18] [Cited by in RCA: 18] [Article Influence: 1.1] [Reference Citation Analysis (0)] |
133. | 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: 15] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
134. | Agrawal S, Umapathy S, Dhiman RK. Minimal hepatic encephalopathy impairs quality of life. J Clin Exp Hepatol. 2015;5:S42-S48. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 55] [Cited by in RCA: 60] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
135. | Zhang G, Cheng Y, Shen W, Liu B, Huang L, Xie S. The short-term effect of liver transplantation on the low-frequency fluctuation of brain activity in cirrhotic patients with and without overt hepatic encephalopathy. Brain Imaging Behav. 2017;11:1849-1861. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 12] [Article Influence: 1.7] [Reference Citation Analysis (0)] |
136. | Zhang G, Li Y, Zhang X, Huang L, Cheng Y, Shen W. Identifying Mild Hepatic Encephalopathy Based on Multi-Layer Modular Algorithm and Machine Learning. Front Neurosci. 2020;14:627062. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 4] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
137. | Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7-30. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 12667] [Cited by in RCA: 15334] [Article Influence: 3066.8] [Reference Citation Analysis (4)] |
138. | Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, Washington KM, Carneiro F, Cree IA; WHO Classification of Tumours Editorial Board. The 2019 WHO classification of tumours of the digestive system. Histopathology. 2020;76:182-188. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2554] [Cited by in RCA: 2457] [Article Influence: 491.4] [Reference Citation Analysis (3)] |
139. | Petrowsky H, Fritsch R, Guckenberger M, De Oliveira ML, Dutkowski P, Clavien PA. Modern therapeutic approaches for the treatment of malignant liver tumours. Nat Rev Gastroenterol Hepatol. 2020;17:755-772. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 69] [Cited by in RCA: 152] [Article Influence: 30.4] [Reference Citation Analysis (0)] |
140. | Ayuso C, Rimola J, Vilana R, Burrel M, Darnell A, García-Criado Á, Bianchi L, Belmonte E, Caparroz C, Barrufet M, Bruix J, Brú C. Corrigendum to "Diagnosis and staging of hepatocellular carcinoma (HCC): Current guidelines" [Eur. J. Radiol. 101 (2018) 72-81]. Eur J Radiol. 2019;112:229. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 6] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
141. | Cheng N, Ren Y, Zhou J, Zhang Y, Wang D, Zhang X, Chen B, Liu F, Lv J, Cao Q, Chen S, Du H, Hui D, Weng Z, Liang Q, Su B, Tang L, Han L, Chen J, Shao C. Deep Learning-Based Classification of Hepatocellular Nodular Lesions on Whole-Slide Histopathologic Images. Gastroenterology. 2022;162:1948-1961.e7. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 23] [Cited by in RCA: 58] [Article Influence: 19.3] [Reference Citation Analysis (0)] |
142. | Qu H, Zhang S, Guo M, Miao Y, Han Y, Ju R, Cui X, Li Y. Deep Learning Model for Predicting Proliferative Hepatocellular Carcinoma Using Dynamic Contrast-Enhanced MRI: Implications for Early Recurrence Prediction Following Radical Resection. Acad Radiol. 2024;31:4445-4455. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 1] [Reference Citation Analysis (0)] |
143. | Xu Z, An C, Shi F, Ren H, Li Y, Chen S, Dou J, Wang Y, Yan S, Lu J, Chen H. Automatic prediction of hepatic arterial infusion chemotherapy response in advanced hepatocellular carcinoma with deep learning radiomic nomogram. Eur Radiol. 2023;33:9038-9051. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 11] [Reference Citation Analysis (0)] |
144. | Hua Y, Sun Z, Xiao Y, Li H, Ma X, Luo X, Tan W, Xie Z, Zhang Z, Tang C, Zhuang H, Xu W, Zhu H, Chen Y, Shang C. Pretreatment CT-based machine learning radiomics model predicts response in unresectable hepatocellular carcinoma treated with lenvatinib plus PD-1 inhibitors and interventional therapy. J Immunother Cancer. 2024;12:e008953. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 15] [Reference Citation Analysis (0)] |
145. | Han JW, Lee SK, Kwon JH, Nam SW, Yang H, Bae SH, Kim JH, Nam H, Kim CW, Lee HL, Kim HY, Lee SW, Lee A, Chang UI, Song DS, Kim SH, Song MJ, Sung PS, Choi JY, Yoon SK, Jang JW. A Machine Learning Algorithm Facilitates Prognosis Prediction and Treatment Selection for Barcelona Clinic Liver Cancer Stage C Hepatocellular Carcinoma. Clin Cancer Res. 2024;30:2812-2821. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Reference Citation Analysis (0)] |
146. | Rhyou SY, Yoo JC. Aggregated micropatch-based deep learning neural network for ultrasonic diagnosis of cirrhosis. Artif Intell Med. 2023;139:102541. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
147. | Luetkens JA, Nowak S, Mesropyan N, Block W, Praktiknjo M, Chang J, Bauckhage C, Sifa R, Sprinkart AM, Faron A, Attenberger U. Deep learning supports the differentiation of alcoholic and other-than-alcoholic cirrhosis based on MRI. Sci Rep. 2022;12:8297. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 18] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
148. | Chang D, Truong E, Mena EA, Pacheco F, Wong M, Guindi M, Todo TT, Noureddin N, Ayoub W, Yang JD, Kim IK, Kohli A, Alkhouri N, Harrison S, Noureddin M. Machine learning models are superior to noninvasive tests in identifying clinically significant stages of NAFLD and NAFLD-related cirrhosis. Hepatology. 2023;77:546-557. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 42] [Article Influence: 21.0] [Reference Citation Analysis (0)] |
149. | Mazumder NR, Enchakalody B, Zhang P, Su GL. Using Artificial Intelligence to Predict Cirrhosis From Computed Tomography Scans. Clin Transl Gastroenterol. 2023;14:e00616. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 4] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
150. | Guo C, Liu Z, Fan H, Wang H, Zhang X, Zhao S, Li Y, Han X, Wang T, Chen X, Zhang T. Machine-learning-based plasma metabolomic profiles for predicting long-term complications of cirrhosis. Hepatology. 2025;81:168-180. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 9] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
151. | Calvo Córdoba A, García Cena CE, Montoliu C. Automatic Video-Oculography System for Detection of Minimal Hepatic Encephalopathy Using Machine Learning Tools. Sensors (Basel). 2023;23:8073. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
152. | Sparacia G, Colelli G, Parla G, Mamone G, Maruzzelli L, Lo Re V, Avorio F, Miraglia R, Pichiecchio A. Brain Magnetic Resonance Imaging Radiomic Signature and Machine Learning Model Prediction of Hepatic Encephalopathy in Adult Cirrhotic Patients. Life (Basel). 2025;15:346. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
153. | Chen QF, Zou TX, Yang ZT, Chen HJ. Identification of patients with and without minimal hepatic encephalopathy based on gray matter volumetry using a support vector machine learning algorithm. Sci Rep. 2020;10:2490. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 8] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
154. | Liu DJ, Jia LX, Zeng FX, Zeng WX, Qin GG, Peng QF, Tan Q, Zeng H, Ou ZY, Kun LZ, Zhao JB, Chen WG. Machine learning prediction of hepatic encephalopathy for long-term survival after transjugular intrahepatic portosystemic shunt in acute variceal bleeding. World J Gastroenterol. 2025;31:100401. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 1] [Reference Citation Analysis (3)] |
155. | Zhong BY, Wang WS, Shen J, Du H, Zhang S, Li WC, Yin Y, Yang J, Ni CF, Zhu XL. Single-Centre Retrospective Training Cohort Using Artificial Intelligence for Prognostic Prediction of Encephalopathy, Mortality, and Liver Dysfunction after Early TIPS Creation. Cardiovasc Intervent Radiol. 2021;44:1597-1608. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3] [Cited by in RCA: 13] [Article Influence: 3.3] [Reference Citation Analysis (0)] |
156. | Gao R, Zhao S, Aishanjiang K, Cai H, Wei T, Zhang Y, Liu Z, Zhou J, Han B, Wang J, Ding H, Liu Y, Xu X, Yu Z, Gu J. Deep learning for differential diagnosis of malignant hepatic tumors based on multi-phase contrast-enhanced CT and clinical data. J Hematol Oncol. 2021;14:154. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 85] [Cited by in RCA: 80] [Article Influence: 20.0] [Reference Citation Analysis (0)] |
157. | Xu Y, Zhou C, He X, Song R, Liu Y, Zhang H, Wang Y, Fan Q, Chen W, Wu J, Wang J, Guo D. Deep learning-assisted LI-RADS grading and distinguishing hepatocellular carcinoma (HCC) from non-HCC based on multiphase CT: a two-center study. Eur Radiol. 2023;33:8879-8888. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
158. | Li M, Fan Y, You H, Li C, Luo M, Zhou J, Li A, Zhang L, Yu X, Deng W, Zhou J, Zhang D, Zhang Z, Chen H, Xiao Y, Huang B, Wang J. Dual-Energy CT Deep Learning Radiomics to Predict Macrotrabecular-Massive Hepatocellular Carcinoma. Radiology. 2023;308:e230255. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 25] [Cited by in RCA: 32] [Article Influence: 16.0] [Reference Citation Analysis (0)] |
159. | Ma Y, Gong Y, Qiu Q, Ma C, Yu S. Research on multi-model imaging machine learning for distinguishing early hepatocellular carcinoma. BMC Cancer. 2024;24:363. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Reference Citation Analysis (0)] |
160. | An C, Wei R, Liu W, Fu Y, Gong X, Li C, Yao W, Zuo M, Li W, Li Y, Wu F, Liu K, Yan D, Wu P, Han J. Machine learning-based decision support model for selecting intra-arterial therapies for unresectable hepatocellular carcinoma: A national real-world evidence-based study. Br J Cancer. 2024;131:832-842. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
161. | Cao S, Yu S, Huang L, Seery S, Xia Y, Zhao Y, Si Z, Zhang X, Zhu J, Lang R, Kou J, Zhang H, Wei L, Zhou G, Sun L, Wang L, Li T, He Q, Zhu Z. Deep learning for hepatocellular carcinoma recurrence before and after liver transplantation: a multicenter cohort study. Sci Rep. 2025;15:7730. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 2] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
162. | Altaf A, Mustafa A, Dar A, Nazer R, Riyaz S, Rana A, Bhatti ABH. Artificial intelligence-based model for the recurrence of hepatocellular carcinoma after liver transplantation. Surgery. 2024;176:1500-1506. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
163. | Dong B, Zhang H, Duan Y, Yao S, Chen Y, Zhang C. Development of a machine learning-based model to predict prognosis of alpha-fetoprotein-positive hepatocellular carcinoma. J Transl Med. 2024;22:455. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 11] [Reference Citation Analysis (0)] |
164. | Yan M, Zhang X, Zhang B, Geng Z, Xie C, Yang W, Zhang S, Qi Z, Lin T, Ke Q, Li X, Wang S, Quan X. Deep learning nomogram based on Gd-EOB-DTPA MRI for predicting early recurrence in hepatocellular carcinoma after hepatectomy. Eur Radiol. 2023;33:4949-4961. [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 (0)] |
165. | Krois J, Garcia Cantu A, Chaurasia A, Patil R, Chaudhari PK, Gaudin R, Gehrung S, Schwendicke F. Generalizability of deep learning models for dental image analysis. Sci Rep. 2021;11:6102. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 33] [Cited by in RCA: 37] [Article Influence: 9.3] [Reference Citation Analysis (0)] |
166. | Lin H, Li R, Liu Z, Chen J, Yang Y, Chen H, Lin Z, Lai W, Long E, Wu X, Lin D, Zhu Y, Chen C, Wu D, Yu T, Cao Q, Li X, Li J, Li W, Wang J, Yang M, Hu H, Zhang L, Yu Y, Chen X, Hu J, Zhu K, Jiang S, Huang Y, Tan G, Huang J, Lin X, Zhang X, Luo L, Liu Y, Liu X, Cheng B, Zheng D, Wu M, Chen W, Liu Y. Diagnostic Efficacy and Therapeutic Decision-making Capacity of an Artificial Intelligence Platform for Childhood Cataracts in Eye Clinics: A Multicentre Randomized Controlled Trial. EClinicalMedicine. 2019;9:52-59. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 96] [Cited by in RCA: 112] [Article Influence: 18.7] [Reference Citation Analysis (0)] |