BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 21, 2025; 31(39): 110971
Published online Oct 21, 2025. doi: 10.3748/wjg.v31.i39.110971
Artificial intelligence in pancreatitis: A narrative review on advancing precision diagnosis, prognosis, and therapeutic strategies
Xi-Yue Zhang, Meng-Di Hu, Diliare Maimaitijiang, Tao Wang, West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, Natural and Biomimetic Medicine Research Center, Tissue-Orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
Xi-Yue Zhang, Meng-Di Hu, Diliare Maimaitijiang, West China School of Pharmacy, Sichuan University, Chengdu 610041, Sichuan Province, China
Lin Wang, West China School of Nursing, West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
ORCID number: Tao Wang (0000-0002-8955-416X).
Co-first authors: Xi-Yue Zhang and Meng-Di Hu.
Co-corresponding authors: Tao Wang and Lin Wang.
Author contributions: Zhang XY and Hu MD contributed equally to this work and are co-first authors of the manuscript. Wang T and Wang L contributed equally to this work and are co-corresponding authors of the manuscript. Wang T and Wang L conceptualized and designed the study, supervised, and made critical revisions; Zhang XY, Hu MD, and Maimaitijiang D conducted the literature review, created the artwork, performed the analysis and data interpretation, and drafted the original manuscript. All authors prepared the draft and approved the submitted version.
Supported by National Natural Science Foundation of China, No. 82000266; and Natural Science Foundation of Sichuan Province, No. 2025ZNSFSC0700.
Conflict-of-interest statement: The authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Tao Wang, PhD, Associate Professor, West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, Natural and Biomimetic Medicine Research Center, Tissue-Orientated Property of Chinese Medicine Key Laboratory of Sichuan Province, West China School of Medicine, West China Hospital, Sichuan University, No. 2222 Xinchuan Road, Chengdu 610041, Sichuan Province, China. terrywang1126@scu.edu.cn
Received: June 19, 2025
Revised: July 17, 2025
Accepted: September 23, 2025
Published online: October 21, 2025
Processing time: 124 Days and 3 Hours

Abstract

Pancreatitis poses persistent diagnostic and therapeutic challenges due to its heterogeneous clinical presentation, variable disease course, and lack of targeted interventions. Conventional tools, such as serum enzymes, cross-sectional imaging and clinical scoring systems, often exhibit limited sensitivity and prognostic value, especially during early or atypical stages. Moreover, therapeutic development remains slow, with limited progress toward personalized or mechanism-based strategies. These limitations highlight a critical need for integrative data-driven approaches. Artificial intelligence (AI) has emerged as a promising tool to enhance clinical decision-making in pancreatitis. This narrative review synthesizes recent progress in AI applications across three domains. First, AI-enabled diagnostic platforms incorporating radiomics, deep learning-based imaging analysis, and biomarker optimization have improved early detection and differentiation of pancreatic diseases. Second, AI-driven prognostic models now allow real-time severity prediction, complication forecasting, and recurrence risk assessment, some of which have been deployed in hospital information systems for intensive care units and mortality risk triage. Third, AI-assisted drug discovery and network pharmacology, particularly in combination with traditional Chinese medicine, have revealed novel therapeutic opportunities. Despite encouraging developments, challenges remain in data standardization, model transparency and clinical validation. A multidisciplinary strategy integrating omics data, longitudinal monitoring and pharmacological modeling may help bridge current gaps and advance precision medicine in pancreatitis care.

Key Words: Pancreatitis; Artificial intelligence; Machine learning; Deep learning; Diagnosis; Prognosis management; Therapeutics; Network pharmacology; Traditional Chinese medicine

Core Tip: This narrative review summarizes recent advances in artificial intelligence (AI) applications for pancreatitis. It covers AI-enhanced diagnosis through imaging and biomarker analysis, real-time prognostication using machine learning models, and AI-assisted therapeutic innovation, including integration with network pharmacology and traditional Chinese medicine. The review also highlights clinically implemented models and discusses current limitations and future directions for AI deployment in pancreatitis care.



INTRODUCTION

Pancreatitis is an inflammatory disorder of the pancreas. It encompasses a broad clinical spectrum ranging from acute, self-limiting episodes to chronic, progressive forms that lead to irreversible structural damage and functional decline[1,2]. Acute pancreatitis (AP) is among the most common gastrointestinal emergencies worldwide, with rising incidence and significant morbidity and mortality, particularly in severe cases[3]. On the other hand, chronic pancreatitis (CP) poses a substantial burden due to persistent abdominal pain, exocrine and endocrine insufficiency, and increased malignancy risk[4]. The diverse etiologies, overlapping clinical features, and unpredictable disease trajectories make timely diagnosis and optimal management challenging in both acute and chronic forms[5].

Traditional clinical tools, including serum enzyme assays (e.g., amylase, lipase), imaging modalities [contrast-enhanced computed tomography (CT), magnetic resonance imaging (MRI), endoscopic ultrasound (EUS)], and scoring systems such as Ranson, Bedside Index of Severity in Acute Pancreatitis, and Accuracy of Acute Physiology and Chronic Health Evaluation (APACHE) II, have provided foundational frameworks for diagnosis and risk stratification[6-8]. However, these conventional approaches have well-known limitations. Enzyme elevations can be nonspecific (for example, other abdominal issues can raise amylase), and mild or very early pancreatitis can be missed[9]. Imaging may not always distinguish inflammation from other processes, and factors like inter-observer variability or the need for radiation in CT can hinder accuracy[10,11]. While useful, scoring systems are based on a few static parameters and often fail to capture the full complexity of each patient’s condition[12,13]. In short, the sensitivity and specificity of these methods in early or atypical cases are imperfect, and patient subgroups remain hard to identify. Beyond supportive care, therapeutic options remain suboptimal, especially in severe or recurrent pancreatitis, and drug development in this field has been notably slow[14].

Recent advances in artificial intelligence (AI), encompassing machine learning (ML), deep learning (DL), natural language processing, and network pharmacology, offer potential tools to address these limitations[15-17]. AI models can analyze large-scale, heterogeneous datasets, including clinical records, medical images, laboratory values, genomic data, and real-time physiological signals, to identify complex, nonlinear patterns that elude conventional statistical methods[18,19]. For instance, a well-trained model may be able to identify early signals of impending organ failure or highlight imaging features too subtle for the human eye[20-22]. In turn, these insights can enable earlier and more precise diagnosis, better risk prediction for each patient, and even suggest tailored treatment strategies. In essence, AI tools can act as advanced decision-support, augmenting clinical judgment with data-driven intelligence.

This review synthesizes the latest developments in AI applications for both acute and CP, structured around three core domains: (1) Precision diagnostics; (2) Dynamic prognostication; and (3) Integrative therapeutic innovation. We emphasize clinically relevant use cases, cutting-edge modeling techniques, and interdisciplinary approaches, including the incorporation of traditional Chinese medicine (TCM) and omics technologies to provide a comprehensive outlook on the emerging era of intelligent pancreatitis care.

LITERATURE SEARCH STRATEGY

We performed a broad search of Englishlanguage literature published from January 1, 2005 to June 30, 2025 using the following databases: PubMed, Web of Science, and ScienceDirect. The search combined Medical Subject Headings and free-text terms as follows: “Pancreatitis” AND (“artificial intelligence” OR “machine learning” OR “deep learning”) AND (“diagnosis” OR “prognosis” OR “drug discovery” OR “network pharmacology”). Reference lists of seminal articles were also reviewed to capture any additional relevant studies. After removing duplicates, 2066 unique records were screened by title and abstract to exclude non-English publications, conference abstracts, case reports, and studies unrelated to AI applications in pancreatitis. Full texts of 580 remaining articles were then assessed for relevance, yielding 72 final inclusions comprising original research papers and high-quality reviews focused on AI-driven diagnostics, prognostics, or therapeutics in pancreatitis.

In addition, we integrated the key information of 22 representative AI model studies with high accuracy [e.g., area under the curve (AUC) > 0.83] in diagnosis and prognosis into Table 1, including prediction target, patient population, AI method, input data type, performance indicator (e.g., AUC, accuracy), and clinical translation stage. While not a full systematic review, this approach follows essential PRISMA principles[23], such as clear search terms, predefined inclusion criteria, and transparent screening steps, to ensure rigor and reproducibility in our narrative synthesis.

Table 1 Application of high-accuracy artificial intelligence models in the diagnosis and prognosis management of pancreatitis.
Application
Prediction target
AI methodology
Patient cohort
Input data type
Performance metrics
Stage of clinical translation
Ref.
DiagnosisAP early diagnosis and risk stratificationXGBoost model61894Vital signs and lab measurementsAUC: 0.921Experimental[47]
Logistic regression model27528 important radiomics features, lipase levelsAUC: 0.933Experimental[32]
Pancreatic lesions detection and segmentationV-Net model144Pancreatic volume. The split volume ratiosAUC: 0.854Experimental[31]
SegFormer model165EUS imagesAUC: 0.95Validation phase[41]
Pediatric pancreas segmentationPanSegNet84Imaging parameters, biologic sex, age at imaging, clinical diagnosisDice coefficient: Approximately 0.88Validation phase[37]
Non-invasive diagnosis of PDAC and CPDLR model558CEUS imagesTraining cohort AUC: 0.986. Internal validation cohort AUC: 0.978. External validation cohorts 1 AUC: 0.967. External validation cohorts 2 AUC: 0.953Validation phase[39]
491EUS imagesAUC: 0.936Validation phase[40]
Distinguish AIP from PDACSVM-RFE model111251 expert-designed features from 2D and 3D PET/CT imagesAUC: 0.93 ± 0.01Validation phase[44]
Random forest model182431 radiomics features. Two types of CT parameters with dual phase CTAUC: 0.975Validation phase[45]
AP severity early predictionLightGBM model215Radiomics featuresTraining cohort AUC: 0.992. Validation cohort AUC: 0.965. Test cohort AUC: 0.894Hospital IT integration[33,64]
CTA model192Age, gender, BMI, temperature, pulse, systolic blood pressure, WBC, CRP, albumin, calcium, APACHE II and BISAP, Balthazar grade, CTSI, MCTSI, EPIC scoreTraining cohort AUC: 0.853. Validation cohort AUC: 0.833Validation phase[51]
Random forest model648Blood urea nitrogen, serum creatinine, albumin, HDL, LDL, calcium, glucoseTraining cohort AUC: 0.89. Test cohort AUC: 0.96Validation phase[48]
740Age, gender, BMI, comorbidities, serum biochemical index, CTTraining cohort AUC: 0.969. Training cohort AUC: 0.961Validation phase[52]
Prognosis managementMortalityXGBoost model499WBC count, hemoglobin, platelet count, serum creatinine, albuminAUC: 0.881Hospital IT integration[61,64]
ANNs model337Total bilirubin, creatinine, amylase, lipase, LDHAUC: 0.769Experimental[72]
Multiple organ failureANNs model312Age, hematocrit, serum glucose, BUN, serum calciumAUC: 0.96 ± 0.02Experimental[62]
SVM model263HCT, K-time, IL-6, creatinineAUC: 0.840Validation phase[69]
ComplicationsANNs model217Pancreatic necrosis rate, LDH, oxyhemoglobin saturationAUC: 0.859 ± 0.048Experimental[66]
XGBoost model334APACHE II, IAP, PCT rankAUC: 0.9193Experimental[67]
GBDT model1672Age, vasopressors, mechanical ventilation, GCSAUC: 0.985Validation phase[70]
RecurrenceXGBoost model531TG levels, smoking, drinking, ANCAUC: 0.779Experimental[79]
CECT model389Age, etiology, CTSI, hospital stay, pancreatic necrosisAUC: 0.941Experimental[76]
AI-ENHANCED PRECISION DIAGNOSTICS

Accurate diagnosis of pancreatitis (and its specific subtype) is crucial for guiding treatment. However, diagnosis can be difficult; symptoms like abdominal pain, nausea, and vomiting are nonspecific, and routine tests are imperfect[24]. Traditional markers such as serum amylase and lipase can increase in AP, but they can also be elevated for other reasons (e.g., intestinal obstruction or gallbladder disease)[25], and some severe cases of pancreatitis may even present with only mildly elevated enzymes[26]. Imaging modalities such as contrast-enhanced CT, MRI, and EUS provide more direct evidence of pancreatic inflammation and its complications, but they require expert interpretation and are limited by factors such as variable image quality, the need for contrast, or exposure to radiation[27-29]. For example, early AP may not show clear findings on CT, and subtle necrosis or fluid collections can be missed[27]. Even among specialists, diagnostic disagreement is common, especially when differentiating CP from pancreatic cancer[29]. Given these challenges, there is a growing need for tools that can enhance diagnostic precision and reduce subjectivity. AI offers promising solutions across various diagnostic modalities. To contextualize the current landscape, we provide a visual summary of key AI-driven approaches and representative algorithm types in Figure 1 and Table 1.

Figure 1
Figure 1 Schematic overview of high-accuracy artificial intelligence models for the precision diagnosis of pancreatitis and their clinical advantages. Artificial intelligence-enhanced diagnostic tools, encompassing machine learning and deep learning algorithms, integrate multimodal data, ranging from laboratory and biochemical markers to radiologic imaging, endoscopic ultrasound, and computed tomography angiography. Representative models include random forest, XGBoost, SVM-RFE, LightGBM, V-Net, SegFormer, deep learning radiomics, PanSegNet, and others. These approaches have demonstrated utility in improving diagnostic accuracy and efficiency, enabling early severity stratification, differentiating pancreatic neoplasms from chronic pancreatitis, and informing more effective allocation of healthcare resources. The implementation status of each model is indicated by color coding: Experimental (blue), validation phase (orange), or clinically deployed (green). AI: Artificial intelligence; LightGBM: Light gradient boosting machine; CTA: Computed tomography angiography; SVM-RFE: Support vector machine-recursive feature elimination; DLR: Deep learning radiomics.
Medical imaging and radiomics

AI has made rapid inroads into imaging diagnostics. DL models, especially convolutional neural networks (CNNs), excel at processing imaging data[30]. In pancreatitis, CNNs have been trained to recognize visual patterns on CT and MRI scans that correlate with disease severity or presence[31-33]. For example, automated segmentation networks (such as U-Net or V-Net architectures) can precisely outline the pancreas and any necrotic regions on a CT slice (patient cohort: 144; AUC: 0.854)[31]. These segmentations allow quantification of disease burden (e.g., calculating necrosis volume), which can improve early severity grading. Other studies extract radiomic features from scans (e.g., quantitative texture, intensity, and shape descriptors) and feed them into ML classifiers (patient cohort: 275; AUC: 0.933)[32]. One approach uses portal-phase contrast-enhanced CT images of the pancreatic and peripancreatic regions to extract hundreds of radiomic features, then applies a gradient-boosting algorithm or logistic regression to predict who will develop severe AP (SAP) (patient cohort: 215; AUC: 0.992)[33]. These models have shown promising accuracy in small cohorts, suggesting that subtle imaging signals can be harnessed before conventional readout. Importantly, AI-driven imaging can reduce dependence on subjective interpretation[34-36]. For instance, a CNN trained on a large set of MRI images can flag even mild pancreatic inflammation with consistency, acting as a second reader[34]. In practice, such tools could alert clinicians to early pancreatitis changes that might otherwise be overlooked. It is worth noting that most imaging-based AI tools have been trained on adult data. However, anatomical and contrast differences in children, such as smaller organ size, distinct imaging characteristics, and varying etiologies, warrant dedicated models. For example, PanSegNet, a recently developed transformer-augmented nn-UNet version trained exclusively on pediatric MRI, achieves Dice similarity scores of about 0.88 and expert-level performance in acute and chronic pediatric pancreatitis cohorts (patient cohort: 499)[37]. This contrasts with adult models, such as CT-based CNNs, with limited pediatric validation. Consequently, we advocate for age-specific AI pipelines or adaptive learning frameworks that incorporate pediatric imaging and outcome data, supported by rigorous cross-age validation and regulatory approval processes.

Advanced endoscopy and ultrasound

EUS and contrast-enhanced ultrasound are increasingly used for pancreatic evaluation because of their real-time visualization and lack of ionizing radiation (in the case of ultrasound)[38]. However, interpretation of EUS images is notoriously user-dependent, requiring significant expertise. AI is helping to standardize this. DL classifiers trained on thousands of EUS images can learn the visual patterns of CP, pancreatic masses, and other lesions[39-41]. In one reported example, an AI model applied to contrast-enhanced ultrasound images could help distinguish pancreatic ductal adenocarcinoma from benign CP (patient cohort: 558; AUC: 0.986)[39]. With AI assistance, radiologists improved their diagnostic accuracy: Even less-experienced readers achieved performance close to experts when aided by the model[40]. Real-time applications are also in development. Computer-aided detection systems using architectures like SegFormer (a type of visual transformer model) have been built to identify and segment pancreatic lesions on EUS video frames as the endoscope moves (patient cohort: 165; AUC: 0.95)[41]. These systems report high area under the receiver operating characteristic curve for detecting lesions and provide bounding boxes or outlines of suspicious areas. By highlighting abnormalities in real time, such AI tools could reduce missed lesions and decrease the need for repeat procedures.

Autoimmune pancreatitis and other special cases

Some forms of pancreatitis, like autoimmune pancreatitis (AIP), pose particular diagnostic challenges. AIP can mimic pancreatic cancer on imaging, and vice versa[42,43]. ML approaches have been applied here as well. For example, a support vector machine trained on radiomic features from 18-F-fluorodeoxyglucose-positron emission tomography/CT scans could distinguish AIP from pancreatic cancer with accuracy around 85%, outperforming traditional radiologist assessment (patient cohort: 111; AUC: 0.93 ± 0.01)[44]. Similarly, computer analysis of CT texture features has helped identify the characteristic imaging patterns of AIP (patient cohort: 182; AUC: 0.975)[45]. These examples illustrate that AI can pick up on nuanced image traits sometimes imperceptible to the naked eye to improve diagnostic certainty in difficult cases.

Laboratory and biochemical data

In addition to imaging, AI models can enhance diagnosis by integrating laboratory tests. Instead of relying on a single enzyme level, ML can combine dozens of lab values and clinical inputs at once[46-48]. For instance, studies have trained algorithms on admission blood work (amylase, lipase, white cell count, C-reactive protein, calcium, etc.) and demographics to predict whether a patient’s AP will become severe[46]. A notable example used the XGBoost algorithm on a large patient dataset (patient cohort: 61894), which achieved an AUC of about 0.92 for predicting SAP[47]. Other models, including random forests and neural networks, showed similar improved prediction of SAP when combining multiple biomarkers (patient cohort: 648; AUC: 0.89)[48]. AI can also dynamically assess lab trends: For example, temporal patterns in inflammatory markers or vital signs in the first 24 hours could be fed into recurrent neural networks to forecast worsening disease[49]. An interesting application is using unconventional biomarkers: One research group combined quantitative contrast-enhanced ultrasound imaging data with a serum biomarker (heat shock protein 70) and found that an AI classifier could automatically grade the extent of pancreatic necrosis[50]. These approaches demonstrate that AI can fuse biological markers with imaging for more sensitive diagnosis.

Integrated diagnostic models

Real-world diagnosis often requires synthesizing multiple data streams. Recent AI efforts capitalize on this by creating multimodal models that take in clinical scores, lab tests, and imaging features together[51-53]. For example, a decision tree algorithm was designed to combine standard severity scores (like APACHE II and Bedside Index of Severity in Acute Pancreatitis) with imaging scores (such as the Balthazar grading of CT or early prognostic imaging classification score for necrosis). This hybrid model achieved 100% specificity and 94.8% overall accuracy in early disease severity prediction, far exceeding any single score alone (patient cohort: 192; AUC: 0.853)[51]. Other teams have trained random forests on a full panel of inputs (e.g., patient demographics, serum markers, CT scan features, etc.) to stratify AP risk. These all-in-one classifiers typically outperform traditional univariate rules, offering earlier and more confident identification of patients who need intensive care (patient cohort: 740; AUC: 0.969)[52]. In practice, such AI-driven tools could run in the background of an electronic medical record: As soon as a new lab result or scan report arrives, the model could recalculate the patient’s risk and alert the care team if concerning patterns emerged. By integrating data in this way, AI helps ensure that no critical information is overlooked during the complex diagnostic process.

AI-DRIVEN DYNAMIC PROGNOSTICATION

Once pancreatitis is diagnosed, predicting its course becomes the next priority. Clinical management hinges on anticipating who will do well with standard care and who may rapidly deteriorate. Traditionally, scores like APACHE II or Ranson’s are used to estimate severity and mortality risk[54], but they are relatively crude and static. AI offers a more dynamic and nuanced approach to prognostication by processing longitudinal data and recognizing nonlinear interactions[55-57]. A summary of key AI-driven models and their prognostic applications is illustrated in Figure 2 and Table 1.

Figure 2
Figure 2 Representative artificial intelligence models for prognosis management in pancreatitis. A range of machine learning and neural network-based algorithms have been applied to predict recurrence, assess disease severity and mortality risk, and forecast complications such as acute respiratory distress syndrome, acute kidney injury, and multiple organ failure. Notable models include XGBoost, gradient boosting decision tree, back propagation, support vector machine, and feed-forward neural networks. These models, particularly when integrated into multi-omics platforms, enable individualized prognostication and support precision management strategies in clinical practice. The implementation status of each model is indicated by color coding: Experimental (blue), validation phase (orange), or clinically deployed (green). ARDS: Acute respiratory distress syndrome; AI: Artificial intelligence.
Predicting severity and mortality

SAP can escalate quickly, with organ failure and death rates around 20%-30% in the most critical patients[58]. AI models can help by analyzing early indicators and identifying high-risk individuals. ML algorithms have been trained on cohorts of AP patients to predict outcomes like intensive care unit (ICU) admission or 30-day mortality[59]. In comparative studies, modern models such as XGBoost and gradient boosting decision trees consistently outperformed traditional approaches[60]. For example, an XGBoost model analyzing admission vitals and labs achieved an accuracy of over 90% in forecasting 30-day mortality in SAP patients (patient cohort: 499; AUC: 0.881)[61]. Moreover, an artificial neural network achieved 96% accuracy and 98% specificity in predicting in-hospital mortality (patient cohort: 312; AUC: 0.960 ± 0.020), highlighting the potential of DL in real-time mortality risk assessment[62]. Notably, AI can continuously update risk as new data arrive. Dynamic forecasting models can take time-series inputs (like hourly urine output, blood pressure, or lab trends) to revise severity predictions in real time[63]. This approach enables clinicians to adapt treatment plans in real time. For example, escalating therapy when early warning signs arise or withholding unnecessary interventions if the patient’s trajectory appears favorable. In fact, a recent study integrated LightGBM and XGBoost models into the hospital information system for AP patients, enabling real-time prediction of sepsis, ICU admission, and mortality[64].

Forecasting complications

Beyond overall mortality, predicting specific complications is equally important. Organ failure (respiratory, renal, cardiovascular) is a leading cause of death in SAP[65]. AI systems can warn of impending organ dysfunction by recognizing complex biomarker signatures[66-71]. For example, neural network models have been reported to predict complications such as multiorgan failure with high specificity and sensitivity (patient cohort: 337; AUC: 0.769)[62,72]. ML models, such as artificial neural networks and XGBoost model, have been used to predict acute respiratory distress syndrome in the days following AP onset (patient cohort: 217; AUC: 0.859 ± 0.048)[66] or to identify patients at high risk for acute kidney injury based on their evolving lab data (patient cohort: 334; AUC: 0.9193)[67]. Another serious complication is infected pancreatic necrosis, which demands prompt intervention[68]. Early studies have shown that combining imaging data (e.g., CT necrosis extent) with biochemical markers (like C-reactive protein or procalcitonin) in an AI model improves prediction of infection or abscess formation[51-53]. Venous complications, such as splanchnic vein thrombosis, have also been studied[73]; AI models that incorporate inflammation and coagulation markers along with imaging findings are being developed to identify these risks[74]. In short, any complication that follows certain detectable early signals becomes a target for AI-assisted prediction, allowing preemptive management.

Recurrence prediction

Recurrent AP, defined as multiple episodes of AP in a single patient, poses a risk for progression to CP and long-term pancreatic dysfunction[75]. AI has begun to tackle this by identifying patients prone to relapse. Models have been built using a mix of patient history (such as alcohol use or genetic mutations), initial lab values, and even baseline imaging features[76-79]. For example, an ML classifier applied to first-episode AP data predicted recurrence with high accuracy in both training and validation sets (patient cohort: 389; AUC: 0.941)[76]. Another approach used radiomic analysis of the pancreas on initial CT scans, revealing that subtle texture differences identified by AI could distinguish patients likely to develop recurrent AP[77]. These AI tools enable stratified surveillance and early preventive interventions, such as prioritizing gallstone removal, monitoring alcohol abstinence, or tailoring follow-up schedules for high-risk individuals[78,80,81].

Integrated prognostic platforms

Like diagnostic algorithms, AI-based prognostic systems benefit from the integration of heterogeneous data types, including clinical, imaging, laboratory, and emerging molecular datasets. Some teams have begun to explore “digital phenotyping” platforms that combine genetic, biochemical, and imaging data to construct individualized risk profiles[82]. Though still exploratory in pancreatitis, this integrative approach has shown promise in adjacent fields. For example, in pancreatic cancer, AI has been used to combine radiomic features with immunological markers (e.g., programmed death ligand-1 expression or CD8+ T-cell infiltration) to predict response to immunotherapy[83]. A similar framework could potentially be adapted for pancreatitis, whereby molecular signatures, such as cytokine profiles, oxidative stress markers, or gut microbiome features, may be integrated into prognostic models to stratify patients and guide immunomodulatory or anti-inflammatory treatments. Such multi-modal AI tools may ultimately enable a shift from reactive care to proactive risk interception.

INTEGRATIVE THERAPEUTIC INNOVATION

While diagnostic and prognostic tools help manage pancreatitis, therapeutic innovation is also accelerating with AI[84-88]. Two major directions are emerging: The application of AI to personalize and optimize current treatment strategies (precision therapy), and the use of AI-driven platforms for discovering or repositioning therapeutic agents. Figure 3 summarizes these integrative therapeutic innovations, highlighting the interplay between real-time patient data, AI-based drug discovery, and network pharmacology.

Figure 3
Figure 3 Artificial intelligence-driven strategies for therapeutic innovation in pancreatitis. Artificial intelligence (AI) facilitates precision treatment through clinical decision support systems that integrate multimodal patient data to optimize resuscitation, nutritional support, pain management, and intensive care unit triage. Machine learning algorithms dynamically update risk profiles and stratify patients to guide individualized interventions. AI is also accelerating drug discovery and repositioning by enabling high-throughput screening of chemical libraries, predicting compound-target interactions, and identifying repurposable agents with anti-inflammatory or antifibrotic potential. Additionally, network pharmacology approaches powered by AI are uncovering bioactive compounds and mechanistic targets within traditional Chinese medicine formulations, bridging empirical practices with molecular pharmacology. These integrative frameworks represent a paradigm shift in pancreatitis care, offering data-driven insights into both modern and traditional therapeutic avenues. AI: Artificial intelligence; FDA: Food and Drug Administration; TCM: Traditional Chinese medicine.
Personalized and precision treatment

The goal of precision medicine is to match the right therapy to the right patient[89]. In pancreatitis care, this can mean deciding how aggressively to resuscitate a patient with fluids, when to start enteral nutrition, whether to admit to the ICU, or what kind of pain management to use[90]. AI-driven decision support systems can assist by synthesizing patient-specific information[84-86]. For instance, AI models that continuously update a patient’s risk status could signal whether an AP patient requires urgent transfer to intensive care or invasive monitoring, potentially before any obvious clinical deterioration[84]. ML can also help stratify CP patients; by analyzing patterns in pain scores, imaging, and genetic factors, algorithms might predict which patients will benefit from endoscopic procedures vs those who may need surgery or novel biologic therapies[85]. Although these precision interventions are still in the research phase, early results are encouraging. Several AI models have already demonstrated superior accuracy in outcome prediction[91]. For example, a study reported an XGBoost model using ICU data achieved an area under the receiver operating characteristic curve of about 0.93 for predicting ICU transfer or death, whereas Ranson’s score achieved only approximately 0.74[47]. Such performance gaps suggest that integrating rich clinical data via AI can refine treatment decisions.

AI-accelerated drug discovery

Developing new drugs for pancreatitis has been challenging, partly due to the disease’s complexity. AI is beginning to change the early drug discovery process[87,88]. In other fields (like oncology and infectious diseases), DL frameworks have been used to screen millions of compounds virtually against disease targets, identifying candidates that would take much longer to find manually[89-92]. For pancreatitis, similar strategies could be applied. For instance, AI can analyze large chemical and pharmacological databases to propose existing drugs (or natural compounds) that might inhibit pathways involved in pancreatic inflammation or fibrosis[93]. This drug “repositioning” approach is powerful because it starts with compounds already known to be safe in humans[94]. Early examples include using ML models to predict anti-inflammatory activity of Food and Drug Administration-approved drugs or to match drug-target profiles with the genetic signatures of pancreatitis[95]. While specific pancreatitis drugs are still in preclinical stages[96], the toolbox is available: Techniques like graph neural networks, ensemble learning, and high-throughput virtual screening could soon reveal new candidate therapies. Importantly, these AI-driven predictions would require laboratory validation, but they dramatically speed up the search phase.

Network pharmacology and traditional medicines

Network pharmacology, particularly when combined with AI, has emerged as a pivotal approach in decoding the complex mechanisms of TCM and accelerating drug discovery for pancreatitis[97,98]. A growing body of evidence suggests that the multi-component and multi-target nature of TCM is particularly amenable to network-level analysis, offering valuable opportunities for therapeutic innovation in AP and CP[99]. Central to these efforts is the rapid evolution of comprehensive databases that link herbal ingredients to molecular targets and disease phenotypes[100]. Resources such as the Traditional Chinese Medicine Systems Pharmacology Database[101], HERB 2.0[102], BATMAN-TCM 2.0[103], ETCM 2.0[104], and TCMBank[105] collectively provide structured annotations of thousands of herbal compounds, their pharmacokinetic profiles, and predicted or validated interactions with genes and pathways. These are often cross-referenced with disease-specific target databases including GeneCards[106], OMIM[107], TTD[108], and DisGeNET[109], enabling a systematic integration of herbal pharmacology with disease biology. Such synergy between herbal knowledge and disease networks allows researchers to construct multi-layered drug-target-pathway networks, facilitating hypothesis generation and therapeutic repositioning.

This framework has been applied to pancreatitis by identifying shared targets between TCM formulations and genes implicated in inflammatory signaling, oxidative stress, or tissue injury[110-118]. For example, by integrating targets of classic formulas such as Da Chaihu Tang[110] or Xiaochaihu Decoction[111] with AP gene signatures derived from GeneCards and OMIM, researchers have uncovered the modulation of pathways such as mitogen-activated protein kinase, phosphatidylinositol 3-kinase/protein kinase B, and tumor necrosis factor, frequently validated through both protein-protein interaction network analysis and enrichment in Kyoto Encyclopedia of Genes and Genomes or Gene Ontology terms. These computational insights often precede or complement in vivo studies: Compounds like quercetin, kaempferol, or emodin, predicted as central network hubs, have subsequently demonstrated anti-inflammatory or anti-apoptotic effects in preclinical models[112-114]. Further strengthening this approach, transcriptomics and metabolomics data are increasingly incorporated to refine compound selection and mechanism prediction[115-118]. One notable example is the use of integrated network pharmacology and adipose-pancreatic transcriptomic data to elucidate how Chaiqin Chengqi Decoction modulates the nuclear factor-E2-related factor 2/heme oxygenase-1 axis in obesity-associated pancreatitis[110].

ML further enhances this paradigm by enabling high-throughput analysis of TCM prescriptions, prediction of pharmacodynamic activity, and extraction of latent therapeutic patterns[119]. Recent studies have employed supervised learning algorithms such as random forests to model the therapeutic efficacy of hundreds of empirical herbal prescriptions[98], identifying core herbal components, such as Rhubarb (Da Huang), Radix Bupleuri (Chai Hu), and Fructus Aurantii (Zhi Shi), that are most predictive of clinical benefit. These approaches not only confirm traditional usage patterns but also provide quantitative insight into herb-herb synergy and treatment efficacy. Moreover, unsupervised learning and association rule mining can identify novel herb combinations with potential therapeutic relevance, while DL models are increasingly used to screen compound-target interactions and to predict new herb-disease associations based on molecular similarity or knowledge graph embeddings. For example, the TCMBank database integrates three-dimensional chemical structures with AI models to forecast compound functionality and target binding, offering a scalable platform for herbal drug repurposing.

Despite these advances, challenges remain in standardizing herbal data, ensuring batch reproducibility, and experimentally validating predictions. Herbal complexity, variability in preparation, and lack of quantitative pharmacokinetic data hinder AI model calibration and generalizability[120]. Addressing these limitations will require the development of robust ontologies, incorporation of real-world metadata such as geographical origin and processing methods, and integration with experimental pharmacology to verify computational predictions. Nonetheless, the integration of AI with network pharmacology and TCM databases provides a powerful framework for identifying novel therapeutic leads and optimizing personalized treatment strategies in pancreatitis. As these approaches mature, they are likely to reshape how complex diseases like pancreatitis are approached, by combining empirical wisdom with systems-level computation and mechanistic insight.

CHALLENGES AND FUTURE DIRECTIONS

Despite its emerging potential, deploying AI in pancreatitis care faces several key obstacles that must be overcome for safe and effective clinical translation.

Data and methodological robustness

Variability in clinical workflows, imaging protocols, and electronic health record systems across institutions undermines model generalizability. Many AI algorithms have been developed on small, single-center cohorts, heightening the risk of overfitting and limiting their applicability to new patient populations[121,122]. Inconsistent data preprocessing and the absence of standardized benchmarks further hinder cross-study comparisons. Moreover, few tools undergo external or prospective validation, a critical step to confirm real-world performance. To address these gaps, future efforts should prioritize multi-center collaborations, harmonize data collection standards, pre-register development protocols, employ rigorous hold-out test sets, and launch prospective clinical trials.

Transparency, trust, and human-AI integration

Most high-performing AI systems, particularly DL models, operate as “black boxes”, which hampers clinician trust and complicates regulatory approval[15]. The adoption of explainable AI techniques, such as saliency maps, feature-attribution algorithms, and clinically aligned surrogate models, is essential to demystify algorithmic decisions[123]. Even with greater transparency, AI predictions can yield false positives or negatives, each carrying significant clinical consequences[124]. Embedding AI within clinician-in-the-loop frameworks ensures that algorithmic recommendations are reviewed and contextualized by experienced physicians. Notably, meta-analyses show that human-AI hybrid teams outperform either party alone[125]. Interactive platforms (e.g., MedSyn[126]) that support iterative dialogue between clinicians and large language models can refine diagnostic hypotheses, particularly in complex or atypical cases such as AIP or evolving necrosis.

Ethics, equity, and regulatory compliance

Biases in training data, arising from underrepresentation of certain ethnicities, age groups, or comorbidities, can exacerbate health disparities. AI-based medical devices must comply with evolving Food and Drug Administration and European Medicines Agency requirements for transparency[127], continuous-learning oversight, and post-market surveillance. Multi-center data sharing introduces privacy challenges that demand privacy-preserving architectures; federated learning enables decentralized model training, while differential privacy and secure multiparty computation safeguard patient information under the Health Insurance Portability and Accountability Act and General Data Protection Regulation standards[128]. Embedding bias-mitigation measures, such as subgroup performance audits, fairness-aware learning algorithms, and equity dashboards, will be crucial to detect and correct demographic performance gaps before clinical deployment. Notably, while AI-driven network pharmacology approaches integrating TCM compounds have identified promising therapeutic targets, though most of these findings remain at the animal-model or bioinformatic-prediction stage. Prospective clinical trials are required to validate safety and efficacy before real-world implementation.

Five-year roadmap

Looking forward, several concrete milestones are anticipated within the next 5 years. These include: (1) Clinical deployment of real-time AI-powered risk stratification tools for triage in emergency and intensive care settings; (2) Integration of multi-omics profiles (genomic, proteomic, and metabolomic) with clinical and imaging data to refine disease subtyping and therapeutic targeting; (3) Wider adoption of advanced architectures such as transformer-based models (e.g., Vision Transformers[129], ClinicalBERT[130]) to manage sparse or unstructured clinical inputs; and (4) Development of standardized benchmarking frameworks for AI model evaluation, incorporating clinical interpretability, fairness metrics, and real-world impact assessment. Ultimately, realizing the full potential of AI in pancreatitis will require sustained collaboration across disciplines, spanning gastroenterology, computer science, bioethics and health policy, to develop intelligent systems that are not only accurate, but also equitable, interpretable and clinically actionable.

CONCLUSION

AI is rapidly transforming the landscape of pancreatitis care. In diagnostics, ML and DL models are enabling earlier and more accurate identification of pancreatic inflammation using complex inputs such as CT images and biomarker panels. In prognostics, AI allows real-time, individualized risk assessment, helping clinicians anticipate complications, guide ICU triage, and tailor monitoring strategies. In therapeutics, AI-driven methods are accelerating drug discovery and enhancing treatment personalization, especially when coupled with network pharmacology and TCM principles. Recent translational advances illustrate the feasibility of AI deployment in real-world clinical settings. For instance, a 2025 study demonstrated that LightGBM and XGBoost models were successfully integrated into hospital information systems to predict sepsis, ICU admission, and mortality in pancreatitis patients, supporting real-time clinical decision-making. Despite this progress, challenges remain. These include data heterogeneity, model interpretability, regulatory oversight, and the need for external validation in diverse patient populations. Moving forward, successful implementation will require a multidisciplinary strategy that bridges clinical expertise, computational modeling, and ethical design. Moreover, the future of pancreatitis care lies not in replacing clinicians but in augmenting their decision-making with intelligent systems. Human-AI hybrid models, built on reciprocal learning, contextual interpretation, and explainable predictions, may ultimately foster a new paradigm of collaborative, precision-driven medicine.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade B, Grade D

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

Creativity or Innovation: Grade A, Grade B, Grade C, Grade C

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

P-Reviewer: Goel AK, MD, Professor, Senior Researcher, India; Molasy B, MD, PhD, Assistant Professor, Poland; Sinuhaji TRF, Researcher, Indonesia S-Editor: Wang JJ L-Editor: Filipodia P-Editor: Lei YY

References
1.  Kalivarathan J, Yadav K, Bataller W, Brigle NW, Kanak MA.   Etiopathogenesis and pathophysiology of chronic pancreatitis. In: Transplantation, Bioengineering, and Regeneration of the Endocrine Pancreas. Netherlands: Elsevier, 2020; 5-32.  [PubMed]  [DOI]  [Full Text]
2.  Ashraf H, Colombo JP, Marcucci V, Rhoton J, Olowoyo O. A Clinical Overview of Acute and Chronic Pancreatitis: The Medical and Surgical Management. Cureus. 2021;13:e19764.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
3.  Lee PJ, Papachristou GI. New insights into acute pancreatitis. Nat Rev Gastroenterol Hepatol. 2019;16:479-496.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 218]  [Cited by in RCA: 532]  [Article Influence: 88.7]  [Reference Citation Analysis (0)]
4.  Pham A, Forsmark C. Chronic pancreatitis: review and update of etiology, risk factors, and management. F1000Res. 2018;7:F1000 Faculty Rev-F1000 Faculty 607.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 50]  [Cited by in RCA: 72]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
5.  Libby P, Smith R, Rubin EJ, Glassberg MK, Farkouh ME, Rosenson RS. Inflammation unites diverse acute and chronic diseases. Eur J Clin Invest. 2024;54:e14280.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 13]  [Reference Citation Analysis (0)]
6.  Chan KS, Shelat VG. Diagnosis, severity stratification and management of adult acute pancreatitis-current evidence and controversies. World J Gastrointest Surg. 2022;14:1179-1197.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 24]  [Cited by in RCA: 24]  [Article Influence: 8.0]  [Reference Citation Analysis (5)]
7.  Hussain S, Mubeen I, Ullah N, Shah SSUD, Khan BA, Zahoor M, Ullah R, Khan FA, Sultan MA. Modern Diagnostic Imaging Technique Applications and Risk Factors in the Medical Field: A Review. Biomed Res Int. 2022;2022:5164970.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 166]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
8.  Yang L, Liu J, Xing Y, Du L, Chen J, Liu X, Hao J. Comparison of BISAP, Ranson, MCTSI, and APACHE II in Predicting Severity and Prognoses of Hyperlipidemic Acute Pancreatitis in Chinese Patients. Gastroenterol Res Pract. 2016;2016:1834256.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
9.  Rajca B, Guda NM.   Elevated Amylase and Lipase: Physiology Including Non-pancreatitis related Elevations. In: Dua K, Shaker R. Pancreas and Biliary Disease. Cham: Springer, 2016.  [PubMed]  [DOI]  [Full Text]
10.  Quinn L, Tryposkiadis K, Deeks J, De Vet HCW, Mallett S, Mokkink LB, Takwoingi Y, Taylor-Phillips S, Sitch A. Interobserver variability studies in diagnostic imaging: a methodological systematic review. Br J Radiol. 2023;96:20220972.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
11.  McErlean A, Panicek DM, Zabor EC, Moskowitz CS, Bitar R, Motzer RJ, Hricak H, Ginsberg MS. Intra- and interobserver variability in CT measurements in oncology. Radiology. 2013;269:451-459.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 38]  [Reference Citation Analysis (0)]
12.  Shi Z, Wang S, Yue L, Zhang Y, Adhikari BK, Xue S, Zuo W, Li X. Dual-core mutual learning between scoring systems and clinical features for ICU mortality prediction. Inf Sci. 2023;637:118984.  [PubMed]  [DOI]  [Full Text]
13.  Desai N, Gross J. Scoring systems in the critically ill: uses, cautions, and future directions. BJA Educ. 2019;19:212-218.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 25]  [Cited by in RCA: 31]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
14.  Beij A, Verdonk RC, van Santvoort HC, de-Madaria E, Voermans RP. Acute Pancreatitis: An Update of Evidence-Based Management and Recent Trends in Treatment Strategies. United European Gastroenterol J. 2025;13:97-106.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
15.  Maletz S, Balagurunathan Y, Murphy K, Folio L, Chima R, Zaheer A, Vadvala H. AI-powered innovations in pancreatitis imaging: a comprehensive literature synthesis. Abdom Radiol (NY). 2025;50:438-452.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
16.  Kröner PT, Engels MM, Glicksberg BS, Johnson KW, Mzaik O, van Hooft JE, Wallace MB, El-Serag HB, Krittanawong C. Artificial intelligence in gastroenterology: A state-of-the-art review. World J Gastroenterol. 2021;27:6794-6824.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 28]  [Cited by in RCA: 88]  [Article Influence: 22.0]  [Reference Citation Analysis (7)]
17.  Zhang D, Wu C, Yang Z, Yin H, Liu Y, Li W, Huang H, Jin Z. The application of artificial intelligence in EUS. Endosc Ultrasound. 2024;13:65-75.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 13]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
18.  Acosta JN, Falcone GJ, Rajpurkar P, Topol EJ. Multimodal biomedical AI. Nat Med. 2022;28:1773-1784.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 399]  [Article Influence: 133.0]  [Reference Citation Analysis (0)]
19.  Billot B, Magdamo C, Cheng Y, Arnold SE, Das S, Iglesias JE. Robust machine learning segmentation for large-scale analysis of heterogeneous clinical brain MRI datasets. Proc Natl Acad Sci U S A. 2023;120:e2216399120.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 94]  [Reference Citation Analysis (0)]
20.  da Silva DB, Schmidt D, da Costa CA, da Rosa Righi R, Eskofier B. DeepSigns: A predictive model based on Deep Learning for the early detection of patient health deterioration. Expert Syst Appl. 2021;165:113905.  [PubMed]  [DOI]  [Full Text]
21.  Thiele D, Rodseth R, Friedland R, Berger F, Mathew C, Maslo C, Moll V, Leithner C, Storm C, Krannich A, Nee J. Machine Learning Models for the Early Real-Time Prediction of Deterioration in Intensive Care Units-A Novel Approach to the Early Identification of High-Risk Patients. J Clin Med. 2025;14:350.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
22.  Ma X, Wang M, Lin S, Zhang Y, Zhang Y, Ouyang W, Liu X. Knowledge and data-driven prediction of organ failure in critical care patients. Health Inf Sci Syst. 2023;11:7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
23.  Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, Chou R, Glanville J, Grimshaw JM, Hróbjartsson A, Lalu MM, Li T, Loder EW, Mayo-Wilson E, McDonald S, McGuinness LA, Stewart LA, Thomas J, Tricco AC, Welch VA, Whiting P, Moher D. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 44932]  [Cited by in RCA: 42834]  [Article Influence: 10708.5]  [Reference Citation Analysis (2)]
24.  Chinese Pancreatic Surgery Association; Chinese Society of Surgery;  Chinese Medical Association. [Guidelines for diagnosis and treatment of acute pancreatitis in China (2021)]. Zhonghua Wai Ke Za Zhi. 2021;59:578-587.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 22]  [Reference Citation Analysis (0)]
25.  Hu JX, Zhao CF, Wang SL, Tu XY, Huang WB, Chen JN, Xie Y, Chen CR. Acute pancreatitis: A review of diagnosis, severity prediction and prognosis assessment from imaging technology, scoring system and artificial intelligence. World J Gastroenterol. 2023;29:5268-5291.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 35]  [Reference Citation Analysis (4)]
26.  Kamil Faiz FZ, Mehrabian S, Saad M, Aisenberg GM. Prognostic value of serum lipase levels in patients with small bowel obstruction. Proc (Bayl Univ Med Cent). 2018;31:276-279.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
27.  Lee DW, Cho CM. Predicting Severity of Acute Pancreatitis. Medicina (Kaunas). 2022;58:787.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 32]  [Reference Citation Analysis (0)]
28.  Du C, Chai NL, Linghu EQ, Li HK, Sun LH, Jiang L, Wang XD, Tang P, Yang J. Comparison of endoscopic ultrasound, computed tomography and magnetic resonance imaging in assessment of detailed structures of pancreatic cystic neoplasms. World J Gastroenterol. 2017;23:3184-3192.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 36]  [Cited by in RCA: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
29.  Sun C, Li Z, Shi Z, Li G. Current diagnosis and treatment of acute pancreatitis in China: a real-world, multicenter study. BMC Gastroenterol. 2021;21:210.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
30.  Yasaka K, Akai H, Kunimatsu A, Kiryu S, Abe O. Deep learning with convolutional neural network in radiology. Jpn J Radiol. 2018;36:257-272.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 150]  [Cited by in RCA: 221]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
31.  Lu CX, Zhou J, Feng YC, Meng SJ, Guo XL, Su WS, Ngo T, Hsu TH, Lin P, Huang J, Liu ST, Palacio MLB, Change WL, Qin G, Hu YQ, Zhan LH. Artificial intelligence models assisting physicians in quantifying pancreatic necrosis in acute pancreatitis. Quant Imaging Med Surg. 2025;15:135-148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
32.  Bette S, Canalini L, Feitelson LM, Woźnicki P, Risch F, Huber A, Decker JA, Tehlan K, Becker J, Wollny C, Scheurig-Münkler C, Wendler T, Schwarz F, Kroencke T. Radiomics-Based Machine Learning Model for Diagnosis of Acute Pancreatitis Using Computed Tomography. Diagnostics (Basel). 2024;14:718.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
33.  Zhao Y, Wei J, Xiao B, Wang L, Jiang X, Zhu Y, He W. Early prediction of acute pancreatitis severity based on changes in pancreatic and peripancreatic computed tomography radiomics nomogram. Quant Imaging Med Surg. 2023;13:1927-1936.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
34.  Gao X, Wang X. Performance of deep learning for differentiating pancreatic diseases on contrast-enhanced magnetic resonance imaging: A preliminary study. Diagn Interv Imaging. 2020;101:91-100.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 32]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
35.  Khalifa M, Albadawy M. AI in diagnostic imaging: Revolutionising accuracy and efficiency. Comput Methods Programs Biomed Update. 2024;5:100146.  [PubMed]  [DOI]  [Full Text]
36.  Pinto-Coelho L. How Artificial Intelligence Is Shaping Medical Imaging Technology: A Survey of Innovations and Applications. Bioengineering (Basel). 2023;10:1435.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 113]  [Reference Citation Analysis (0)]
37.  Keles E, Yazol M, Durak G, Hong Z, Aktas HE, Zhang Z, Peng L, Susladkar O, Guzelyel N, Boyunaga OL, Yazici C, Lowe M, Uc A, Bagci U. Pediatric pancreas segmentation from MRI scans with deep learning. Pancreatology. 2025;25:648-657.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
38.  Dong Y, D'Onofrio M, Hocke M, Jenssen C, Potthoff A, Atkinson N, Ignee A, Dietrich CF. Autoimmune pancreatitis: Imaging features. Endosc Ultrasound. 2018;7:196-203.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 56]  [Cited by in RCA: 48]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
39.  Tong T, Gu J, Xu D, Song L, Zhao Q, Cheng F, Yuan Z, Tian S, Yang X, Tian J, Wang K, Jiang T. Deep learning radiomics based on contrast-enhanced ultrasound images for assisted diagnosis of pancreatic ductal adenocarcinoma and chronic pancreatitis. BMC Med. 2022;20:74.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 45]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
40.  Gu J, Pan J, Hu J, Dai L, Zhang K, Wang B, He M, Zhao Q, Jiang T. Prospective assessment of pancreatic ductal adenocarcinoma diagnosis from endoscopic ultrasonography images with the assistance of deep learning. Cancer. 2023;129:2214-2223.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
41.  Konikoff T, Loebl N, Benson AA, Green O, Sandler H, Gingold-Belfer R, Levi Z, Perl L, Dotan I, Shamah S. Enhancing detection of various pancreatic lesions on endoscopic ultrasound through artificial intelligence: a basis for computer-aided detection systems. J Gastroenterol Hepatol. 2025;40:235-240.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
42.  Udriștoiu AL, Cazacu IM, Gruionu LG, Gruionu G, Iacob AV, Burtea DE, Ungureanu BS, Costache MI, Constantin A, Popescu CF, Udriștoiu Ș, Săftoiu A. Real-time computer-aided diagnosis of focal pancreatic masses from endoscopic ultrasound imaging based on a hybrid convolutional and long short-term memory neural network model. PLoS One. 2021;16:e0251701.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 26]  [Cited by in RCA: 39]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
43.  Hammami M, Noomen F, Toumi O, Harzallah O, Mahmoudi A, Kallel W, Zouari K, Hamdi A. Autoimmune pancreatitis mimicking pancreatic cancer. N Am J Med Sci. 2011;3:520-523.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
44.  Zhang Y, Cheng C, Liu Z, Wang L, Pan G, Sun G, Chang Y, Zuo C, Yang X. Radiomics analysis for the differentiation of autoimmune pancreatitis and pancreatic ductal adenocarcinoma in (18) F-FDG PET/CT. Med Phys. 2019;46:4520-4530.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 59]  [Cited by in RCA: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
45.  Park S, Chu LC, Hruban RH, Vogelstein B, Kinzler KW, Yuille AL, Fouladi DF, Shayesteh S, Ghandili S, Wolfgang CL, Burkhart R, He J, Fishman EK, Kawamoto S. Differentiating autoimmune pancreatitis from pancreatic ductal adenocarcinoma with CT radiomics features. Diagn Interv Imaging. 2020;101:555-564.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 40]  [Cited by in RCA: 76]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
46.  Wang L, Qi X, Tian F, Li H, Zhao H, Lv J, Zhou X, Ba D, Song W, Ma X. Diagnostic value of hematological parameters in acute pancreatitis. Ann Palliat Med. 2020;9:2716-2722.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
47.  Thapa R, Iqbal Z, Garikipati A, Siefkas A, Hoffman J, Mao Q, Das R. Early prediction of severe acute pancreatitis using machine learning. Pancreatology. 2022;22:43-50.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
48.  Hong W, Lu Y, Zhou X, Jin S, Pan J, Lin Q, Yang S, Basharat Z, Zippi M, Goyal H. Usefulness of Random Forest Algorithm in Predicting Severe Acute Pancreatitis. Front Cell Infect Microbiol. 2022;12:893294.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
49.  Baek Y, Han K, Jeon E, Yoo HY. Prediction of Nursing Need Proxies Using Vital Signs and Biomarkers Data: Application of Deep Learning Models. J Clin Nurs. 2025;34:4121-4131.  [PubMed]  [DOI]  [Full Text]
50.  Kielaite-Gulla A, Samuilis A, Raisutis R, Dzemyda G, Strupas K. The Concept of AI-Based Algorithm: Analysis of CEUS Images and HSPs for Identification of Early Parenchymal Changes in Severe Acute Pancreatitis. Informatica. 2021;32:305-319.  [PubMed]  [DOI]  [Full Text]
51.  Choi HW, Park HJ, Choi SY, Do JH, Yoon NY, Ko A, Lee ES. Early Prediction of the Severity of Acute Pancreatitis Using Radiologic and Clinical Scoring Systems With Classification Tree Analysis. AJR Am J Roentgenol. 2018;211:1035-1043.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
52.  Luo Z, Shi J, Fang Y, Pei S, Lu Y, Zhang R, Ye X, Wang W, Li M, Li X, Zhang M, Xiang G, Pan Z, Zheng X. Development and evaluation of machine learning models and nomogram for the prediction of severe acute pancreatitis. J Gastroenterol Hepatol. 2023;38:468-475.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
53.  Chen Z, Lin Z, Lin Z, Zhang Q, Zhang H, Li H, Chang Q, Sun J, Li F. The applications of CT with artificial intelligence in the prognostic model of idiopathic pulmonary fibrosis. Ther Adv Respir Dis. 2024;18:17534666241282538.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
54.  Harshit Kumar A, Singh Griwan M. A comparison of APACHE II, BISAP, Ranson's score and modified CTSI in predicting the severity of acute pancreatitis based on the 2012 revised Atlanta Classification. Gastroenterol Rep (Oxf). 2018;6:127-131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 55]  [Cited by in RCA: 109]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
55.  Di MY, Liu H, Yang ZY, Bonis PA, Tang JL, Lau J. Prediction Models of Mortality in Acute Pancreatitis in Adults: A Systematic Review. Ann Intern Med. 2016;165:482-490.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 79]  [Cited by in RCA: 72]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
56.  Stam WT, Goedknegt LK, Ingwersen EW, Schoonmade LJ, Bruns ERJ, Daams F. The prediction of surgical complications using artificial intelligence in patients undergoing major abdominal surgery: A systematic review. Surgery. 2022;171:1014-1021.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
57.  Mumuni A, Mumuni F. Automated data processing and feature engineering for deep learning and big data applications: A survey. J Inf Intell. 2025;3:113-153.  [PubMed]  [DOI]  [Full Text]
58.  Tenner S, Vege SS, Sheth SG, Sauer B, Yang A, Conwell DL, Yadlapati RH, Gardner TB. American College of Gastroenterology Guidelines: Management of Acute Pancreatitis. Am J Gastroenterol. 2024;119:419-437.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 64]  [Cited by in RCA: 114]  [Article Influence: 114.0]  [Reference Citation Analysis (0)]
59.  Zhou Y, Ge YT, Shi XL, Wu KY, Chen WW, Ding YB, Xiao WM, Wang D, Lu GT, Hu LH. Machine learning predictive models for acute pancreatitis: A systematic review. Int J Med Inform. 2022;157:104641.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 50]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
60.  Montomoli J, Romeo L, Moccia S, Bernardini M, Migliorelli L, Berardini D, Donati A, Carsetti A, Bocci MG, Wendel Garcia PD, Fumeaux T, Guerci P, Schüpbach RA, Ince C, Frontoni E, Hilty MP; RISC-19-ICU Investigators. Machine learning using the extreme gradient boosting (XGBoost) algorithm predicts 5-day delta of SOFA score at ICU admission in COVID-19 patients. J Intensive Med. 2021;1:110-116.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
61.  Li X, Tian Y, Li S, Wu H, Wang T. Interpretable prediction of 30-day mortality in patients with acute pancreatitis based on machine learning and SHAP. BMC Med Inform Decis Mak. 2024;24:328.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
62.  Hong WD, Chen XR, Jin SQ, Huang QK, Zhu QH, Pan JY. Use of an artificial neural network to predict persistent organ failure in patients with acute pancreatitis. Clinics (Sao Paulo). 2013;68:27-31.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 23]  [Cited by in RCA: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
63.  Ghassemi M, Pimentel MA, Naumann T, Brennan T, Clifton DA, Szolovits P, Feng M. A Multivariate Timeseries Modeling Approach to Severity of Illness Assessment and Forecasting in ICU with Sparse, Heterogeneous Clinical Data. Proc AAAI Conf Artif Intell. 2015;2015:446-453.  [PubMed]  [DOI]
64.  Chang CH, Chen CJ, Ma YS, Shen YT, Sung MI, Hsu CC, Lin HJ, Chen ZC, Huang CC, Liu CF. Real-time artificial intelligence predicts adverse outcomes in acute pancreatitis in the emergency department: Comparison with clinical decision rule. Acad Emerg Med. 2024;31:149-155.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
65.  Li J, Gao J, Huang M, Fu X, Fu B. Risk Factors for Death in Patients with Severe Acute Pancreatitis in Guizhou Province, China. Gastroenterol Res Pract. 2024;2024:8236616.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
66.  Fei Y, Gao K, Li WQ. Prediction and evaluation of the severity of acute respiratory distress syndrome following severe acute pancreatitis using an artificial neural network algorithm model. HPB (Oxford). 2019;21:891-897.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 31]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
67.  Qu C, Gao L, Yu XQ, Wei M, Fang GQ, He J, Cao LX, Ke L, Tong ZH, Li WQ. Machine Learning Models of Acute Kidney Injury Prediction in Acute Pancreatitis Patients. Gastroenterol Res Pract. 2020;2020:3431290.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
68.  Lan L, Guo Q, Zhang Z, Zhao W, Yang X, Lu H, Zhou Z, Zhou X. Classification of Infected Necrotizing Pancreatitis for Surgery Within or Beyond 4 Weeks Using Machine Learning. Front Bioeng Biotechnol. 2020;8:541.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 20]  [Cited by in RCA: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
69.  Qiu Q, Nian YJ, Guo Y, Tang L, Lu N, Wen LZ, Wang B, Chen DF, Liu KJ. Development and validation of three machine-learning models for predicting multiple organ failure in moderately severe and severe acute pancreatitis. BMC Gastroenterol. 2019;19:118.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 23]  [Cited by in RCA: 43]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
70.  Liu F, Yao J, Liu C, Shou S. Construction and validation of machine learning models for sepsis prediction in patients with acute pancreatitis. BMC Surg. 2023;23:267.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
71.  Song B, Liu P, Fu K, Liu C. Developing a predictive model for septic shock risk in acute pancreatitis patients using interpretable machine learning algorithms. Digit Health. 2025;11:20552076251346361.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
72.  Ding N, Guo C, Li C, Zhou Y, Chai X. An Artificial Neural Networks Model for Early Predicting In-Hospital Mortality in Acute Pancreatitis in MIMIC-III. Biomed Res Int. 2021;2021:6638919.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 31]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
73.  Chiasakul T, Lam BD, McNichol M, Robertson W, Rosovsky RP, Lake L, Vlachos IS, Adamski A, Reyes N, Abe K, Zwicker JI, Patell R. Artificial intelligence in the prediction of venous thromboembolism: A systematic review and pooled analysis. Eur J Haematol. 2023;111:951-962.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
74.  Lehmann ALCF, Alfieri DF, de Araújo MCM, Trevisani ER, Nagao MR, Pesente FS, Gelinski JR, de Freitas LB, Flauzino T, Lehmann MF, Lozovoy MAB, Breganó JW, Simão ANC, Maes M, Reiche EMV. Immune-inflammatory, coagulation, adhesion, and imaging biomarkers combined in machine learning models improve the prediction of death 1 year after ischemic stroke. Clin Exp Med. 2022;22:111-123.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
75.  Trikudanathan G, Yazici C, Evans Phillips A, Forsmark CE. Diagnosis and Management of Acute Pancreatitis. Gastroenterology. 2024;167:673-688.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 40]  [Article Influence: 40.0]  [Reference Citation Analysis (1)]
76.  Chen Y, Chen TW, Wu CQ, Lin Q, Hu R, Xie CL, Zuo HD, Wu JL, Mu QW, Fu QS, Yang GQ, Zhang XM. Radiomics model of contrast-enhanced computed tomography for predicting the recurrence of acute pancreatitis. Eur Radiol. 2019;29:4408-4417.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 64]  [Cited by in RCA: 67]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
77.  Tang L, Ma L, Chen Y, Hu Y, Chen X, Huang X, Liu N. Radiomics analysis of contrast-enhanced T1W MRI: predicting the recurrence of acute pancreatitis. Sci Rep. 2023;13:2762.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
78.  Gardner TB, Adler DG, Forsmark CE, Sauer BG, Taylor JR, Whitcomb DC. ACG Clinical Guideline: Chronic Pancreatitis. Am J Gastroenterol. 2020;115:322-339.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 118]  [Cited by in RCA: 212]  [Article Influence: 42.4]  [Reference Citation Analysis (0)]
79.  Ren W, Zou K, Chen Y, Huang S, Luo B, Jiang J, Zhang W, Shi X, Shi L, Zhong X, Lü M, Tang X. Application of a Machine Learning Predictive Model for Recurrent Acute Pancreatitis. J Clin Gastroenterol. 2024;58:923-930.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
80.  Thierens N, Verdonk RC, Löhr JM, van Santvoort HC, Bouwense SA, van Hooft JE. Chronic pancreatitis. Lancet. 2025;404:2605-2618.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 14]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
81.  Whitcomb DC; North American Pancreatitis Study Group. Pancreatitis: TIGAR-O Version 2 Risk/Etiology Checklist With Topic Reviews, Updates, and Use Primers. Clin Transl Gastroenterol. 2019;10:e00027.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 45]  [Cited by in RCA: 72]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
82.  Cui C, Yang H, Wang Y, Zhao S, Asad Z, Coburn LA, Wilson KT, Landman BA, Huo Y. Deep multimodal fusion of image and non-image data in disease diagnosis and prognosis: a review. Prog Biomed Eng (Bristol). 2023;5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 39]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
83.  Yu G, Zhang Z, Eresen A, Hou Q, Amirrad F, Webster S, Nauli S, Yaghmai V, Zhang Z. Predicting and Monitoring Immune Checkpoint Inhibitor Therapy Using Artificial Intelligence in Pancreatic Cancer. Int J Mol Sci. 2024;25:12038.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
84.  De Micco F, Di Palma G, Ferorelli D, De Benedictis A, Tomassini L, Tambone V, Cingolani M, Scendoni R. Artificial intelligence in healthcare: transforming patient safety with intelligent systems-A systematic review. Front Med (Lausanne). 2024;11:1522554.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
85.  Gorris M, Hoogenboom SA, Wallace MB, van Hooft JE. Artificial intelligence for the management of pancreatic diseases. Dig Endosc. 2021;33:231-241.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 21]  [Cited by in RCA: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (1)]
86.  Kaissis G, Ziegelmayer S, Lohöfer F, Steiger K, Algül H, Muckenhuber A, Yen HY, Rummeny E, Friess H, Schmid R, Weichert W, Siveke JT, Braren R. A machine learning algorithm predicts molecular subtypes in pancreatic ductal adenocarcinoma with differential response to gemcitabine-based versus FOLFIRINOX chemotherapy. PLoS One. 2019;14:e0218642.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 43]  [Cited by in RCA: 44]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
87.  Paul D, Sanap G, Shenoy S, Kalyane D, Kalia K, Tekade RK. Artificial intelligence in drug discovery and development. Drug Discov Today. 2021;26:80-93.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 363]  [Cited by in RCA: 543]  [Article Influence: 135.8]  [Reference Citation Analysis (0)]
88.  Zhang K, Yang X, Wang Y, Yu Y, Huang N, Li G, Li X, Wu JC, Yang S. Artificial intelligence in drug development. Nat Med. 2025;31:45-59.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 57]  [Article Influence: 57.0]  [Reference Citation Analysis (1)]
89.  Akhoon N. Precision Medicine: A New Paradigm in Therapeutics. Int J Prev Med. 2021;12:12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 23]  [Reference Citation Analysis (0)]
90.  van den Berg FF, Boermeester MA. Update on the management of acute pancreatitis. Curr Opin Crit Care. 2023;29:145-151.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 23]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
91.  Hassan AM, Rajesh A, Asaad M, Nelson JA, Coert JH, Mehrara BJ, Butler CE. Artificial Intelligence and Machine Learning in Prediction of Surgical Complications: Current State, Applications, and Implications. Am Surg. 2023;89:25-30.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 37]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
92.  Zhang P, Wang X, Cen X, Zhang Q, Fu Y, Mei Y, Wang X, Wang R, Wang J, Ouyang H, Liang T, Xia H, Han X, Guo G. A deep learning framework for in silico screening of anticancer drugs at the single-cell level. Natl Sci Rev. 2025;12:nwae451.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
93.  Wu D, Gao W, Li X, Tian C, Jiao N, Fang S, Xiao J, Xu Z, Zhu L, Zhang G, Zhu R. Dr AFC: drug repositioning through anti-fibrosis characteristic. Brief Bioinform. 2021;22:bbaa115.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
94.  Saranraj K, Kiran PU. Drug repurposing: Clinical practices and regulatory pathways. Perspect Clin Res. 2025;16:61-68.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
95.  Agarwal V, Haldhar R, Hirad AH, Ahmed B, Han SB, Gupta A, Raj V, Lee S. Repurposing FDA-approved drugs as NLRP3 inhibitors against inflammatory diseases: machine learning and molecular simulation approaches. J Biomol Struct Dyn. 2025;43:4327-4339.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
96.  Kambhampati S, Park W, Habtezion A. Pharmacologic therapy for acute pancreatitis. World J Gastroenterol. 2014;20:16868-16880.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 46]  [Cited by in RCA: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (1)]
97.  Li S, Huang Y, Liu L, Zhang F, Ao H, Luo Y. Mechanism of Magnolia Volatile Oil in the Treatment of Acute Pancreatitis Based on GC-MS, Network Pharmacology, and Molecular Docking. Evid Based Complement Alternat Med. 2023;2023:3503888.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
98.  Lu WW, Chen X, Ni JL, Cai WJ, Zhu SL, Fei AH, Wang XS. Study on the medication rule of traditional Chinese medicine in the treatment of acute pancreatitis based on machine learning technology. Ann Palliat Med. 2021;10:10616-10625.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
99.  Li X, Yan Z, Cao X, Chen X, Guan Z, Tang S, Fan J, Duan L, Xu X, Zhang H. Dachaihu Decoction alleviates chronic pancreatitis by regulating MAPK signaling pathway: Insights from network pharmacology and experimental validation. J Ethnopharmacol. 2025;337:118833.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
100.  Xue R, Fang Z, Zhang M, Yi Z, Wen C, Shi T. TCMID: Traditional Chinese Medicine integrative database for herb molecular mechanism analysis. Nucleic Acids Res. 2013;41:D1089-D1095.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 300]  [Cited by in RCA: 398]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
101.  Ru J, Li P, Wang J, Zhou W, Li B, Huang C, Li P, Guo Z, Tao W, Yang Y, Xu X, Li Y, Wang Y, Yang L. TCMSP: a database of systems pharmacology for drug discovery from herbal medicines. J Cheminform. 2014;6:13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1623]  [Cited by in RCA: 3211]  [Article Influence: 291.9]  [Reference Citation Analysis (0)]
102.  Gao K, Liu L, Lei S, Li Z, Huo P, Wang Z, Dong L, Deng W, Bu D, Zeng X, Li C, Zhao Y, Zhang W, Wang W, Wu Y. HERB 2.0: an updated database integrating clinical and experimental evidence for traditional Chinese medicine. Nucleic Acids Res. 2025;53:D1404-D1414.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
103.  Kong X, Liu C, Zhang Z, Cheng M, Mei Z, Li X, Liu P, Diao L, Ma Y, Jiang P, Kong X, Nie S, Guo Y, Wang Z, Zhang X, Wang Y, Tang L, Guo S, Liu Z, Li D. BATMAN-TCM 2.0: an enhanced integrative database for known and predicted interactions between traditional Chinese medicine ingredients and target proteins. Nucleic Acids Res. 2024;52:D1110-D1120.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 55]  [Cited by in RCA: 64]  [Article Influence: 64.0]  [Reference Citation Analysis (0)]
104.  Zhang Y, Li X, Shi Y, Chen T, Xu Z, Wang P, Yu M, Chen W, Li B, Jing Z, Jiang H, Fu L, Gao W, Jiang Y, Du X, Gong Z, Zhu W, Yang H, Xu H. ETCM v2.0: An update with comprehensive resource and rich annotations for traditional Chinese medicine. Acta Pharm Sin B. 2023;13:2559-2571.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 52]  [Reference Citation Analysis (0)]
105.  Lv Q, Chen G, He H, Yang Z, Zhao L, Zhang K, Chen CY. TCMBank-the largest TCM database provides deep learning-based Chinese-Western medicine exclusion prediction. Signal Transduct Target Ther. 2023;8:127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 39]  [Reference Citation Analysis (0)]
106.  Stelzer G, Rosen N, Plaschkes I, Zimmerman S, Twik M, Fishilevich S, Stein TI, Nudel R, Lieder I, Mazor Y, Kaplan S, Dahary D, Warshawsky D, Guan-Golan Y, Kohn A, Rappaport N, Safran M, Lancet D. The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Curr Protoc Bioinformatics. 2016;54:1.30.1-1.30.33.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1289]  [Cited by in RCA: 3002]  [Article Influence: 333.6]  [Reference Citation Analysis (0)]
107.  Amberger JS, Hamosh A. Searching Online Mendelian Inheritance in Man (OMIM): A Knowledgebase of Human Genes and Genetic Phenotypes. Curr Protoc Bioinformatics. 2017;58:1.2.1-1.2.12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 200]  [Cited by in RCA: 428]  [Article Influence: 53.5]  [Reference Citation Analysis (0)]
108.  Zhou Y, Zhang Y, Zhao D, Yu X, Shen X, Zhou Y, Wang S, Qiu Y, Chen Y, Zhu F. TTD: Therapeutic Target Database describing target druggability information. Nucleic Acids Res. 2024;52:D1465-D1477.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 273]  [Article Influence: 273.0]  [Reference Citation Analysis (0)]
109.  Piñero J, Saüch J, Sanz F, Furlong LI. The DisGeNET cytoscape app: Exploring and visualizing disease genomics data. Comput Struct Biotechnol J. 2021;19:2960-2967.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 225]  [Cited by in RCA: 335]  [Article Influence: 83.8]  [Reference Citation Analysis (0)]
110.  Yang X, Yao L, Yuan M, Zhang X, Jakubowska MA, Ferdek PE, Dai L, Yang J, Jin T, Deng L, Fu X, Du D, Liu T, Criddle DN, Sutton R, Huang W, Xia Q. Transcriptomics and Network Pharmacology Reveal the Protective Effect of Chaiqin Chengqi Decoction on Obesity-Related Alcohol-Induced Acute Pancreatitis via Oxidative Stress and PI3K/Akt Signaling Pathway. Front Pharmacol. 2022;13:896523.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
111.  Zhu K, Zhang M, Long J, Zhang S, Luo H. Elucidating the Mechanism of Action of Salvia miltiorrhiza for the Treatment of Acute Pancreatitis Based on Network Pharmacology and Molecular Docking Technology. Comput Math Methods Med. 2021;2021:8323661.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
112.  Aggarwal D, Chaudhary M, Mandotra SK, Tuli HS, Chauhan R, Joshi NC, Kaur D, Dufossé L, Chauhan A. Anti-inflammatory potential of quercetin: From chemistry and mechanistic insight to nanoformulations. Curr Res Pharmacol Drug Discov. 2025;8:100217.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
113.  Carvalho KM, Morais TC, de Melo TS, de Castro Brito GA, de Andrade GM, Rao VS, Santos FA. The natural flavonoid quercetin ameliorates cerulein-induced acute pancreatitis in mice. Biol Pharm Bull. 2010;33:1534-1539.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 42]  [Cited by in RCA: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
114.  Yang Q, Luo Y, Ge P, Lan B, Liu J, Wen H, Cao Y, Sun Z, Zhang G, Yuan H, Zhang L, Chen H. Emodin Ameliorates Severe Acute Pancreatitis-Associated Acute Lung Injury in Rats by Modulating Exosome-Specific miRNA Expression Profiles. Int J Nanomedicine. 2023;18:6743-6761.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 16]  [Reference Citation Analysis (0)]
115.  Zhang J, Li H, Wang W, Li H. Assessing the anti-inflammatory effects of quercetin using network pharmacology and in vitro experiments. Exp Ther Med. 2022;23:301.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
116.  Bastin A, Teimouri M, Faramarz S, Shabani M, Doustimotlagh AH, Sadeghi A. In vitro and Molecular Docking Analysis of Quercetin as an Anti-inflammatory and Antioxidant. Curr Pharm Des. 2023;29:883-891.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
117.  Aney KJ, Jeong WJ, Vallejo AF, Burdziak C, Chen E, Wang A, Koak P, Wise K, Jensen K, Pe'er D, Dougan SK, Martelotto L, Nissim S. Novel Approach for Pancreas Transcriptomics Reveals the Cellular Landscape in Homeostasis and Acute Pancreatitis. Gastroenterology. 2024;166:1100-1113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 15]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
118.  Xu H, Zhang L, Kang H, Zhang J, Liu J, Liu S. Serum Metabonomics of Mild Acute Pancreatitis. J Clin Lab Anal. 2016;30:990-998.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 12]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
119.  Cheng N, Chen Y, Gao W, Liu J, Huang Q, Yan C, Huang X, Ding C. An Improved Deep Learning Model: S-TextBLCNN for Traditional Chinese Medicine Formula Classification. Front Genet. 2021;12:807825.  [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)]
120.  Ma S, Liu J, Li W, Liu Y, Hui X, Qu P, Jiang Z, Li J, Wang J. Machine learning in TCM with natural products and molecules: current status and future perspectives. Chin Med. 2023;18:43.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 17]  [Reference Citation Analysis (0)]
121.  Cho JH, Kim TN, Chung HH, Kim KH. Comparison of scoring systems in predicting the severity of acute pancreatitis. World J Gastroenterol. 2015;21:2387-2394.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 95]  [Cited by in RCA: 116]  [Article Influence: 11.6]  [Reference Citation Analysis (1)]
122.  Hamesch K, Hollenbach M, Guilabert L, Lahmer T, Koch A. Practical management of severe acute pancreatitis. Eur J Intern Med. 2025;133:1-13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
123.  López Gordo S, Ramirez-Maldonado E, Fernandez-Planas MT, Bombuy E, Memba R, Jorba R. AI and Machine Learning for Precision Medicine in Acute Pancreatitis: A Narrative Review. Medicina (Kaunas). 2025;61:629.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
124.  Ucdal M, Bakhshandehpour A, Durak MB, Balaban Y, Kekilli M, Simsek C. Evaluating the Role of Artificial Intelligence in Making Clinical Decisions for Treating Acute Pancreatitis. J Clin Med. 2025;14:4347.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
125.  Podină N, Gheorghe EC, Constantin A, Cazacu I, Croitoru V, Gheorghe C, Balaban DV, Jinga M, Țieranu CG, Săftoiu A. Artificial Intelligence in Pancreatic Imaging: A Systematic Review. United European Gastroenterol J. 2025;13:55-77.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
126.  Sayin B, Schlicht IB, Hong NV, Allievi S, Staiano J, Minervini P, Passerini A.   MedSyn: Enhancing Diagnostics with Human-AI Collaboration. 2025 Preprint. Available from: arXiv:2506.14774v1.  [PubMed]  [DOI]  [Full Text]
127.  Han GR, Goncharov A, Eryilmaz M, Ye S, Palanisamy B, Ghosh R, Lisi F, Rogers E, Guzman D, Yigci D, Tasoglu S, Di Carlo D, Goda K, McKendry RA, Ozcan A. Machine learning in point-of-care testing: innovations, challenges, and opportunities. Nat Commun. 2025;16:3165.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 17]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
128.  Alwhbi IA, Zou CC, Alharbi RN. Encrypted Network Traffic Analysis and Classification Utilizing Machine Learning. Sensors (Basel). 2024;24:3509.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
129.  Noguchi A, Numata Y, Sugawara T, Miura H, Konno K, Adachi Y, Yamaguchi R, Ishida M, Kokumai T, Douchi D, Miura T, Ariake K, Nakayama S, Maeda S, Ohtsuka H, Mizuma M, Nakagawa K, Morikawa H, Akatsuka J, Maeda I, Unno M, Yamamoto Y, Furukawa T. Deep learning predicts the 1-year prognosis of pancreatic cancer patients using positive peritoneal washing cytology. Sci Rep. 2024;14:17059.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
130.  Banerjee I, Davis MA, Vey BL, Mazaheri S, Khan F, Zavaletta V, Gerard R, Gichoya JW, Patel B. Natural Language Processing Model for Identifying Critical Findings-A Multi-Institutional Study. J Digit Imaging. 2023;36:105-113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]