Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Jun 24, 2025; 16(6): 104299
Published online Jun 24, 2025. doi: 10.5306/wjco.v16.i6.104299
Comprehensive review of Bayesian network applications in gastrointestinal cancers
Min-Na Zhang, Meng-Ju Xue, Bao-Zhen Zhou, Jing Xu, Hong-Kai Sun, Ji-Han Wang, School of Medicine, Xi'an International University, Xi'an 710077, Shaanxi Province, China
Yang-Yang Wang, School of Physics and Electronic Information, Yan’an University, Yan’an 716000, Shaanxi Province, China
ORCID number: Ji-Han Wang (0000-0003-1925-330X).
Co-first authors: Min-Na Zhang and Meng-Ju Xue.
Co-corresponding authors: Ji-Han Wang and Yang-Yang Wang.
Author contributions: Zhang MN, and Wang YY contributed to conceptualization; Xue MJ and Zhou BZ; validation, Xu J, Wang JH and Wang YY contributed to methodology; Zhang MN, Xue MJ and Sun HK contributed to writing-original draft preparation; Wang JH and Wang YY contributed to writing-review and editing; All the authors have read and approved the final manuscript. Zhang MN proposed and designed the study. Xue MJ prepared the first draft of the manuscript. Both authors have made crucial and indispensable contributions towards the completion of the project and thus qualified as the co-first authors of the paper. Both Wang JH and Wang YY have played important and indispensable roles in the study design, data interpretation and manuscript preparation as the co-corresponding authors.
Supported by Open Funds for Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, No. 2023-KFMS-1.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ji-Han Wang, PhD, Assistant Professor, School of Medicine, Xi'an International University, No. 18 Yudou Road, Yanta District, Xi'an 710077, Shaanxi Province, China. 513837742@qq.com
Received: December 16, 2024
Revised: April 11, 2025
Accepted: May 21, 2025
Published online: June 24, 2025
Processing time: 185 Days and 13.9 Hours

Abstract

Gastrointestinal cancers, including esophageal, gastric, colorectal, liver, gallbladder, cholangiocarcinoma, and pancreatic cancers, pose a significant global health challenge due to their high mortality rates and poor prognosis, particularly when diagnosed at advanced stages. These malignancies, characterized by diverse clinical presentations and etiologies, require innovative approaches for improved management. Bayesian networks (BN) have emerged as a powerful tool in this field, offering the ability to manage uncertainty, integrate heterogeneous data sources, and support clinical decision-making. This review explores the application of BN in addressing critical challenges in gastrointestinal cancers, including the identification of risk factors, early detection, treatment optimization, and prognosis prediction. By integrating genetic predispositions, lifestyle factors, and clinical data, BN hold the potential to enhance survival rates and improve quality of life through personalized treatment strategies. Despite their promise, the widespread adoption of BN is hindered by challenges such as data quality limitations, computational complexities, and the need for greater clinical acceptance. The review concludes with future research directions, emphasizing the development of advanced BN algorithms, the integration of multi-omics data, and strategies to ensure clinical applicability, aiming to fully realize the potential of BN in personalized medicine for gastrointestinal cancers.

Key Words: Gastrointestinal cancers; Bayesian networks; Heterogeneous data integration; Early detection; Risk prediction; Prognosis; Personalized medicine

Core Tip: Bayesian networks (BN) provide a powerful framework for managing the complexity and uncertainty in gastrointestinal cancer research and clinical practice. By integrating diverse data types and modeling causal relationships, BN facilitate personalized treatment strategies. However, challenges such as data availability, computational demands, and clinical adoption must be addressed to fully unlock their potential for improving patient outcomes and quality of life.



INTRODUCTION

Gastrointestinal cancers are among the most prevalent and deadly malignancies worldwide, posing a major public health challenge[1]. These cancers, including but not limited to esophageal, gastric, colorectal, liver, gallbladder, cholangiocarcinoma, and pancreatic cancers, collectively contribute substantially to cancer-related morbidity and mortality[2,3]. Each of these cancers presents unique clinical characteristics and etiological factors, ranging from genetic predispositions and environmental exposures to lifestyle influences such as diet, alcohol consumption, and smoking[4]. Despite advances in cancer biology, diagnostic technologies, and therapeutic interventions, many gastrointestinal cancers continue to exhibit poor outcomes, especially when diagnosed at late stages. This underscores the critical need for improved methods for early detection, risk stratification, and personalized treatment to enhance survival rates and quality of life for affected individuals[5].

Gastrointestinal cancers typically involve complex interactions between genetic, molecular, and environmental factors, making it challenging to identify clear patterns or causal relationships[6]. Moreover, the data generated in cancer research and clinical practice are diverse, spanning epidemiological datasets, clinical observations, imaging studies, and molecular profiles[7]. To address these challenges, computational models have gained prominence as powerful tools for synthesizing and interpreting complex data. Among these models, Bayesian network (BN) has emerged as a particularly promising approach for their ability to handle uncertainty, integrate heterogeneous data, and support decision-making[8]. BN is probabilistic graphical models that represent variables as nodes and their conditional dependencies as directed edges. They provide a framework for modeling causal relationships, enabling researchers and clinicians to infer probabilities and predict outcomes even in the presence of incomplete or uncertain data. By incorporating prior knowledge and learning from data, BN can uncover hidden patterns, facilitate diagnostic decision-making, and improve the accuracy of predictions in complex systems such as cancer biology[9].

In the context of gastrointestinal cancers, BN has been applied to address a wide range of challenges. For example, they have been used to identify risk factors and predict individual cancer risk, integrating genetic predispositions, lifestyle behaviors, and environmental exposures. In diagnosis, BN has shown potential in early detection by combining biomarkers, imaging data, and clinical symptoms[10]. For treatment planning, they offer decision support by weighing the probabilities of different outcomes based on patient-specific data, helping to optimize therapeutic strategies[11]. Additionally, BN has been employed in prognosis, enabling the prediction of survival probabilities and disease progression by analyzing patient-specific characteristics and treatment responses.

Despite their significant promise, the adoption of BN in gastrointestinal oncology is not without challenges. One major limitation is the quality and availability of data, as high-quality, large-scale datasets are often difficult to obtain[12], particularly for rare cancers such as gallbladder and pancreatic cancers. Moreover, the computational complexity of constructing and analyzing large BN can be a barrier, especially when dealing with high-dimensional datasets. The interpretability of the models and their acceptance among clinicians also remain critical issues, as healthcare professionals require intuitive tools that can seamlessly integrate into clinical workflows[13].

This review aims to provide a comprehensive exploration of the applications of BN in gastrointestinal cancers. It begins by introducing the foundational concepts and content of BN, followed by an exploration of their applications across various types of gastrointestinal cancers. It highlights specific use cases such as risk prediction, early diagnosis, treatment decision-making, and survival analysis. The review critically examines challenges and limitations, including data-related issues, computational demands, and the gap between research and clinical implementation, and concludes by identifying promising directions for future research.

FUNDAMENTALS OF BN

BN is a class of probabilistic graphical models that represent a set of variables and their conditional dependencies through a directed acyclic graph (DAG)[14]. These models are uniquely suited for applications in complex systems such as medicine, where uncertainty, interdependence, and incomplete data are prevalent. A BN consists of nodes, representing random variables, and directed edges, denoting the causal or probabilistic relationships between these variables[15]. Each node is associated with a probability distribution that quantifies the likelihood of its states, conditioned on its parent nodes. This structure enables BNs to model joint probability distributions efficiently, facilitating reasoning and inference in uncertain environments[16].

Figure 1 illustrates a classic Asia BN comprising eight random variables: Whether the individual has traveled to Asia (A), has tuberculosis (B), has either tuberculosis or lung cancer (C), has a positive X-ray result (D), smokes (E), has lung cancer (F), has bronchitis (G), and has trouble breathing (H). As shown in the figure, difficulty breathing can result from tuberculosis, lung cancer, bronchitis, a combination of these conditions, or none of them. Recent travel to Asia increases the likelihood of tuberculosis. However, a single chest X-ray cannot differentiate between lung cancer and tuberculosis, nor can it determine the cause of difficulty breathing. It is well-established that smoking is a significant risk factor for both lung cancer and bronchitis. According to the conditional probability distribution, individuals who smoke have a 10% probability of developing lung cancer and a 60% probability of developing bronchitis. This indicates that the likelihood of smoking causing bronchitis is much higher than its likelihood of causing lung cancer. The probability distribution for the DAG in Figure 1 can be expressed as: P(A,B,C,D,E,F,G,H) = P(A)P(E)P(BA)P(FE)P(GE)P(CB,F)P(DC)P(H|C,G).

Figure 1
Figure 1 A typical structure of a Bayesian network. It consists of a directed acyclic graph and the corresponding conditional probability tables. If node C is selected as the target node (yellow node), then the green nodes (B, F, D, H, G) form the Markov blanket of node C, where B and F are parent nodes, D and H are child nodes, and G is a spouse node.

The research on BN s primarily focuses on three aspects: Structure learning, parameter learning, and probabilistic inference. Each of these will be introduced below.

The methodologies of BN structure learning

BN structure learning focuses on constructing a DAG to represent the relationships between variables. This process involves identifying dependency and independence among variables based on data. The learning methods are categorized into DAG-based algorithms, ordering space-based algorithms, and methods for incomplete datasets. Figure 2 illustrates the knowledge context of BN structure learning in a tree-like structure, as well as some representative algorithms.

Figure 2
Figure 2 Classification of algorithms related to Bayesian network structure learning. DAG: Directed acyclic graph; PC: Peter and Clark; ACOG: Ancestral constrained order graph; BDeu: Bayesian Drichlet equivalent uniform; BIC: Bayesian information criterion; MDL: Minimum description length; GS: Greedy search; OBS: Ordering-based search; BiHS: Bidirectional heuristic search; ILP: Integer linear programming; MMHC: Max-Min Hill-Climbing; MMPC: Max-Min Parent and Children; DP: Dynamic programming; AWA*: Anytime Window A* algorithm; MCMC: Markov Chain Monte Carlo; SEM: Structural expectation-maximization.
The methodologies of BN parameter learning

BN parameter learning involves estimating the CPTs that define the relationships between nodes in the network[17]. This process can be categorized into two types: Maximum likelihood estimation (MLE) and Bayesian estimation. MLE is used when the complete data is available, directly calculating probabilities from observed frequencies. Bayesian estimation incorporates prior knowledge into the learning process, making it suitable for handling incomplete or sparse datasets. For incomplete data, the expectation-maximization algorithm is commonly employed to iteratively estimate missing values and refine parameter estimates. Parameter learning is critical for building accurate and reliable BNs, especially in applications like medical diagnosis, where accurate probability estimates directly impact decision-making. Table 1 provides a concise overview of six Bayesian parameter learning algorithms mentioned in the document[18-21].

Table 1 Six Bayesian network parameter learning algorithms.
Algorithms
For incomplete datasets
Basic principle
Advantages & disadvantages
Ref.
Maximum likelihood estimateNoEstimates parameters by maximizing the likelihood function based on observed dataFast convergence; no prior knowledge used, leading to slow convergence[18]
Bayesian methodNoUses a prior distribution (often Dirichlet) and updates it with observed data to obtain a posterior distributionIncorporates prior knowledge; computationally intensive[19]
Expectation-maximizationYesEstimates parameters by iteratively applying expectation (E) and maximization (M) steps to handle missing dataEffective with missing data; can converge to local optima[20]
Robust bayesian estimateYesEstimates parameters using probability intervals to represent the ranges of conditional probabilities without assumptionsDoes not require assumptions about missing data; interval width indicates reliability of estimation[12]
Monte-Carlo methodYesUses random sampling (e.g., Gibbs sampling) to estimate the expectation of the joint probability distributionFlexible and can handle complex models; computationally expensive and convergence can be slow[21]
The methodologies of BN inference

Inference is a critical capability of BNs. It involves calculating the probabilities of unobserved variables given the observed evidence, making BNs powerful tools for prediction and decision support. For instance, in cancer diagnosis, if a BN includes nodes for patient symptoms, test results, and possible cancer types, it can estimate the likelihood of each cancer type given observed symptoms and test results. Table 2 summarizes the commonly used algorithms in the field of BN inference[22-25]. The choice of these inference algorithms typically depends on the complexity of the problem, the size of the network, and the availability of computational resources.

Table 2 Some methodologies of Bayesian network inference.
Algorithm
Network type
Complexity
Accuracy
Advantages
Ref.
Variable eliminationSingle, multi-connected networksExponential in the number of variables in factorizationExactSimple, easy to use[22]
Junction treeSingle, multi-connected networksExponential in the size of the largest cliqueExactFastest method, suitable for sparse networks[22]
Differential methodSingle, multi-connected networksProportional to the complexity of differentiation operationsExactCan solve multiple problems simultaneously[23]
Stochastic samplingSingle, multi-connected networksInversely proportional to the probability of evidence variablesApproximateSimple, widely applicable, and generally effective[24]
Loopy belief propagationSingle, multi-connected networksExponential in the number of loops in the networkApproximatePerforms well when the algorithm converges[25]
Tools for BN

Several software tools and algorithms facilitate the development and application of BN in medicine. These tools simplify the creation of BNs, making them accessible to researchers without extensive computational expertise, as shown in Table 3[26-32].

Table 3 Some popular Bayesian network software tools.
Tools
Language
Description
Links
Bnlearn[26]RPython package for causal discovery by learning the graphical structure of Bayesian networkshttp://www.bnlearn.com/
BNT[27]MATLABBayes net toolbox for Matlabhttps://github.com/bayesnet/bnt
GOBNILPCLearning Bayesian network structure with integer programminghttps://www.cs.york.ac.uk/aig/sw/gobnilp/
BnstructRBnstruct is an R package which learns Bayesian networks from data with missing valueshttps://cran.r-project.org/web/packages/bnstruct
BmmaloneC++This project implements a number of algorithms for learning Bayesian network structures using state space search techniques.https://github.com/bmmalone/urlearning-cpp
Causal-Learner[28]MATLABA toolbox for causal structure and Markov blanket learninghttps://github.com/z-dragonl/Causal-Learner
CausalFS[29]C/C++An open-source package of causal feature selection and causal (Bayesian network) structure learninghttps://github.com/kuiy/CausalFS
Weka[30]JavaWeka contains a collection of visualization tools and algorithms for data analysis and predictive modeling, together with graphical user interfaces for easy access to these functionshttps://git.cms.waikato.ac.nz/weka/weka
BeneCAn exact Bayesian network structure learning software based on dynamic programminghttps://github.com/tomisilander/bene
Causal-learnPythonCausal discovery in Python. It also includes (conditional) independence tests and score functionshttps://github.com/py-why/causal-learn
pyCausalFSPythonAn open-source package of causal feature selection and causal (Bayesian network) structure learninghttps://github.com/kuiy/pyCausalFS
CausalExplorer[31]MATLABA MATLAB library of computational causal discovery and variable selection algorithmshttps://github.com/mensxmachina/CausalExplorer
PgmpyPythonPython library for learning (structure and parameter), inference (probabilistic and causal), and simulations in Bayesian networkshttps://github.com/pgmpy/pgmpy
TetradJavaIt provides algorithms the capability to discover causal models, search for models of latent structurehttps://github.com/cmu-phil/tetrad
Causal discovery toolboxPythonThe causal discovery toolbox is a package for causal inference in graphshttps://github.com/FenTechSolutions/CausalDiscoveryToolbox
DoWhy[32]PythonDoWhy is a Python library for causal inference that supports explicit modeling and testing of causal assumptionshttps://github.com/py-why/dowhy
APPLICATIONS OF BN IN GASTROINTESTINAL CANCERS

BN has been effectively applied to address various challenges in gastrointestinal cancers. Their abilities to integrate diverse datasets, model uncertainty, and uncover causal relationships make them invaluable tools for improving early diagnosis, risk prediction, treatment planning, and prognosis. This section provides a detailed analysis of their applications in specific gastrointestinal cancers, including esophageal, gastric, colorectal, liver, gallbladder, and pancreatic cancers.

Esophageal cancer

Esophageal cancer is a highly aggressive malignancy that is often diagnosed at advanced stages, highlighting the urgent need for effective tools to enable early detection and personalized management[33]. BNs have emerged as valuable tools in addressing these challenges, demonstrating significant potential in various areas of esophageal cancer research and clinical practice.

One notable study developed machine learning models, including BN, to predict the five-year survival of esophageal cancer patients[34]. By identifying key predictors, this approach contributed to personalized treatment strategies and improved clinical outcomes. Similarly, another study utilized BN to synthesize data from 114 studies, creating a personalized risk prediction model for adenocarcinoma in Barrett's esophagus. By integrating critical risk factors, the model achieved high predictive accuracy, paving the way for more tailored surveillance strategies and enhancing early intervention[35]. Additionally, a serum-based biomarker panel for the early detection of esophageal adenocarcinoma was developed using a BN-based rule-learning predictive model. Leveraging proteomics data, this approach demonstrated high accuracy and area under curve (AUC), showcasing its potential for noninvasive disease classification and early diagnosis[36]. The integration of BN models into clinical workflows may enhance early detection and risk stratification in esophageal cancer, potentially allowing for more timely interventions and tailored surveillance in high-risk individuals.

Overall, these studies underscore the versatility and effectiveness of BN in advancing esophageal cancer research. From survival prediction and risk assessment to treatment optimization and biomarker development, BN offers powerful tools to enhance early detection, refine surveillance strategies, and improve patient care outcomes.

Gastric cancer

Gastric cancer remains a leading cause of cancer-related mortality worldwide, with significant variability in incidence and outcomes across populations. The application of BN has demonstrated great utility in risk prediction, diagnosis, prognosis, and treatment planning for gastric cancer. By integrating epidemiological factors, patient clinical data, and molecular profiles, BN models help identify high-risk populations, enabling targeted screening and early detection strategies. For instance, BN models using patient data from the public Surveillance, Epidemiology and End Results database and hospital cohorts identified key predictors such as age, T-stage, tumor size, and tumor location for distant metastasis in early-stage gastric cancer[37]. Similarly, models based on lncRNA expression profiles effectively stratified patients into risk groups and provided robust prognostic predictions with high AUC values[37]. In terms of treatment planning and survival prediction, BN models have been instrumental in handling high-dimensional clinical and molecular data. A two-slice temporal BN improved prediction accuracy for survival outcomes[38], while Bayesian meta-analysis identified nivolumab as the optimal therapy for metastatic gastric cancer without peritoneal metastasis, balancing efficacy and safety[39]. Additionally, Bayesian approaches integrating gene expression data successfully revealed clinically relevant pathways linked to molecular subtypes and immunotherapy response[40,41]. BN applications highlight key prognostic and predictive factors, supporting more accurate metastasis prediction and individualized treatment strategies, which could improve patient outcomes and resource allocation in clinical settings.

Collectively, the studies summarized in Table 4 demonstrate the versatility of BNs in advancing gastric cancer research, from risk prediction and molecular subtype identification to personalized treatment strategies and procedure outcome prediction[42-47]. This integrative approach holds significant promise for improving clinical decision-making and patient outcomes in gastric cancer management.

Table 4 Summary of Bayesian network applications in gastric cancer research.
Data type
Bayesian network algorithm
Key findings
Ref.
Patient data from the public SEER database; patient data from a hospital cohortNaïve Bayesian modelThe Bayesian network model identified key risk factors, including age, T-stage, N-stage, tumor size, grade, and tumor location, contributing to the prediction of distant metastasis in stage T1 gastric cancer[37]
LncRNA expression profiles from 375 STAD samples in the TCGA databaseBayesian Lasso-logistic regressionThe Bayesian-based approach identified seven lncRNAs, effectively stratified STAD patients by risk, and demonstrated robust prognostic prediction accuracy with AUC values above 0.69 for 1-, 3-, and 5-year survival[37]
Survival and censorship data from 760 gastric cancer patientsA two-slice temporal Bayesian network modelThe Bayesian network improved prediction accuracy, reduced bias, and aligned with classical methods while handling high-dimensional data effectively[38]
Data from seven randomized clinical trials involving 2655 metastatic gastric cancer patientsBayesian fixed-effect network meta-analysis modelThe Bayesian analysis identified nivolumab as the optimal choice for OS in mGC patients without peritoneal metastases, providing the best balance of efficacy and safety[39]
Gene expression data from TCGA gastric cancer and metastatic gastric cancer immunotherapy clinical trial datasetsBayesian semi-nonnegative matrix trifactorization methodThe Bayesian method identified clinically relevant pathways associated with molecular subtypes and immunotherapy response, enabling patient stratification and prognosis prediction in independent validation datasets[40]
LncRNA-miRNA-disease association data, including known associations related to gastric cancerA Naïve Bayesian Classifier was integrated into a CFNBCCFNBC demonstrated reliable prediction performance (AUC of 0.8576) and successfully identified potential lncRNA-disease associations for gastric cancer in case studies[42]
Clinical data from 339 gastric cancer patientsBNNThe BNN outperformed the ANN in predicting survival of gastric cancer patients, with higher sensitivity, specificity, prediction accuracy, and AUCs[43]
Data from 245 gastric endoscopic submucosal dissectionsNaïve Bayesian modelThe Bayesian model demonstrated good discriminative power in predicting ESD outcomes, with naïve Bayesian models presenting AUCs of approximately 80% in the derivation cohort and at least 74% in cross-validation for both outcomes[44]
Data from the structural domain characteristics of the p42.3 protein moleculeBayesian network modelThe study identified the most likely acting pathway for p42.3 in gastric cancer as "S100A11" - RAGE - P38 - MAPK - Microtubule-associated protein - spindle protein - centromere protein - cell proliferation" through Bayesian probability optimizing calculation, which was subsequently validated by biological experiments[45]
Genome-wide gene expression profilesCategorical Bayesian networksThe BN approach outperformed benchmark methods and successfully identified disease-specific changes in gene regulation that differentiate cancer types, improving prediction[46]
Gene expression profile data from gastric cancer patientsA Bayesian Network was constructed using 18 genes selected by multiple logistic regressionThe constructed Bayesian Network was very similar to the network from GeneMANIA, indicating the effectiveness of the Bayesian approach in modeling the relationships among genes associated with gastric cancer subtypes[47]
Colorectal cancer

Colorectal cancer (CRC) is one of the most extensively studied gastrointestinal cancers in the context of BNs. The application of BN in CRC research spans over two decades, with early studies illustrating their utility in predicting nodal metastasis through tumor biomarkers[48]. For instance, a Bayesian neural network with automatic relevance determination identified tumor matrilysin as a key predictor of nodal metastasis, providing valuable insights into its potential causal role. This study highlighted the predictive power of Bayesian approaches in analyzing clinical-pathological data[48].

Over the years, BN models have significantly broadened their scope, playing a pivotal role in early detection and risk stratification of CRC[49]. These models integrate diverse data types, including stool-based biomarkers (e.g., gut microbiota profiles), genetic testing, and clinical information, to predict the likelihood of advanced adenomas or CRC during routine screenings. Moreover, BNs have been utilized to incorporate lifestyle factors such as diet, smoking, and physical activity, along with genetic predispositions, enabling the identification of modifiable risk factors and the stratification of individuals into distinct risk categories based on their susceptibility to CRC.

Beyond early detection and risk assessment, BN have become indispensable in the treatment and prognosis of CRC. These models are employed to predict chemotherapy response, recurrence risk, and long-term survival outcomes by integrating tumor-specific genomic, proteomic, and clinical data[50]. For example, Bayesian belief networks and Bayesian additive regression trees have shown superior performance in survival prediction and patient stratification[51,52]. Table 5 provides a comprehensive summary of BN applications in CRC research over the past two decades, showcasing the transformative potential of Bayesian methodologies in advancing personalized medicine and supporting evidence-based clinical decision-making[48,50,52-78]. The use of BN in CRC enables comprehensive risk modeling, integrating modifiable and genetic factors. These tools could guide personalized screening intervals and optimize treatment decisions, improving overall care efficiency.

Table 5 Summary of Bayesian network applications in colorectal cancer research.
Data type
Bayesian network algorithm
Key findings
Ref.
Observational data on CRC, including risk factors such as alcohol consumption, smoking, diabetes, and hypertensionStructure learning algorithms combined with expert knowledge to construct BN modelThe BN model effectively segmented populations into risk subgroups and identified modifiable risk factors with significant predictive influence on CRC risk[53]
Simulation models of CRC progression and natural history, including parameters for risk factors and disease progressionBayesian calibration using Hamiltonian Monte Carlo-based algorithms integrated with ANN emulatorsThe Bayesian framework successfully calibrated CRC simulation models, accurately predicting outcomes within confidence intervals, and reduced computational complexity, enabling efficient uncertainty quantification and improved policy analysis for CRC[54]
Genetic and expression data from 275 normal colon and 276 CRC samples in the SYSCOL cohortBayesian network modelBN revealed tumor-specific (transposable elements) TE-eQTLs that influence the expression of cancer driver genes, demonstrating TEs' role in activating oncogenic pathways and providing insights into tumor-specific regulatory mechanisms[55]
Clinical data of 1253 CRC patients under 50 years of age from the Yonsei Cancer Center, encompassing 93 clinical featuresBayesian network-based synthesizing modelThe BN-based model generated a synthetic population of 5005 individuals with no significant statistical differences from the original data. Training predictive models with synthetic data improved performance, especially for small datasets[56]
Plasma concentrations of heavy metals (As, Cd, Cr, Hg, Pb) and tumor tissue NGS data from CRC patientsBKMRBKMR analysis revealed that Pb, As, and Cd were significant contributors to increased mutation rates, particularly indels. Mutational signatures showed strong correlations with heavy metal exposure, and shifts in the mutational landscape were observed between high and low exposure groups[57]
CRC-associated loci from genome-wide association studies (GWAS) and multi-omics datasetsiRIGS, a Bayesian approachThe iRIGS identified 105 high-confidence risk genes, including CEBPB, which promotes CRC cell proliferation through oncogenic pathways such as MAPK, PI3K-Akt, and Ras signaling[58]
Epidemiological data related to gut microbiome and CRC riskMultivariate Mendelian randomization analysis based on Bayesian modelNine bacteria were identified with a robust causal relationship to CRC development, including Streptococcus thermophilus, Bacteroides ovatus, and others[59]
Clinicopathologic, immune, microbial, and genomic variables from 815 stage II-III CRC patientsBARTThe BART risk model identified seven stable survival predictors and successfully stratified patients into low, intermediate, and high-risk groups with statistically significant survival differences[52]
CRC patients with poorly differentiated and moderately differentiated tumors, analyzed through fecal microbiotaRDP classifier Bayesian algorithmThe study identified distinct GM associated with poorly differentiated CRC, including high abundance of Bifidobacterium and other bacteria[60]
Colon cancer (microsatellite stable/instable stage III) samples analyzed through multi-omics data (gene expression, DNA methylation, copy number variation)IntOMICS, an integrative framework based on Bayesian networksIntOMICS successfully integrated multi-omics data and biological prior knowledge to uncover regulatory networks, revealing deeper insights into genetic information flow and identifying potential predictive biomarkers for stage III colon cancer[61]
Rectal cancer clinical data from 705 patients who underwent radical resectionTree-augmented naïve Bayes algorithmThe BN model, incorporating factors like age, CEA, CA19-9, CA125, differentiation status, T stage, N stage, KRAS mutation, and postoperative chemotherapy, showed higher accuracy (AUC = 80.11%) in predicting 3-year OS compared to a nomogram (AUC = 74.23%)[62]
Time series transcriptomic data from normal and tumor cells of colorectal tissueDBNsThe DBN-based classifier achieved high classification accuracy, revealing significant differences in gene regulatory networks between normal and tumor cells in CRC, particularly in the neighborhoods of oncogenes and cancer tissue markers[63]
Gene expression profiles of COAD tumor samples from TCGA and normal colon tissues from GTExBayesian network modelThe BN analysis identified 14 upregulated DEGs significantly correlated with tumor stages, and Cox regression highlighted tumor stage, STMN4, and FAM135B dysregulation as independent prognostic factors for COAD survival outcomes[64]
Clinical data of colon cancer patients, including 18 prognostic biomarkers and three clinical featuresBayesian binary classifiers, including a Bayesian bimodal neural network and a single modal BNN classifierThe Bayesian bimodal neural network achieved the best results in terms of AUC (0.8083), macro F1-score (0.7300), and concordance index (0.7238), demonstrating superior robustness compared to non-Bayesian models and the Bayesian single modal classifier[65]
Normal mucosa samples from 100 colon cancer patients and 50 healthy donors, including genetic variants, DNA methylation markers, and gene expression dataBayesian network modelThe BN analysis revealed that most combinations showed the canonical pathway where methylation markers cause gene expression variation (60.1%), with 33.9% showing non-causal relationships, and 6% indicating gene expression causes variation in methylation markers[66]
Genetic data from 55105 CRC cases and 65079 controls, along with an independent cohort of 101987 individuals including 1699 CRC casesLDpred, a Bayesian approachThe LDpred-derived polygenic risk score showed the highest discriminatory accuracy for CRC risk prediction, identifying 30% of individuals without a family history at similar risk to those with a family history, suggesting the potential for earlier screening[67]
Fecal microbiome samples from 45 rectal cancer patients before preoperative CCRTBayesian network modelThe BN analysis identified Duodenibacillus massiliensis as linked with an improved complete response rate after preoperative CCRT, suggesting its potential as a predictive biomarker[68]
Gene expression data from primary colon cancer and CLM samplesFast and FFBNFFBN successfully constructed gene regulatory networks for colon cancer and colon to liver metastasis, revealing unique molecular mechanisms for CLM and shared similarities with primary liver and colon cancers[69]
Gut microbiota data related to CRCBayesian networks combined with IDA (Intervention calculus when the DAG is absent)Four species-Fusobacterium, Citrobacter, Microbacterium, and Slackia-were identified as having non-null lower bounds of causal effects on CRC, supporting the role of specific microbial communities in CRC progression[70]
CRC metastasis-related transcription factors (RNA and protein levels)Bayesian network modelThe BN analysis identified LMO7 and ARL8A as potential clinical biomarkers for CRC metastasis[71]
Gene expression data from 153 colon cancer samples and 19 normal control samples (from TCGA project)BRPCAThe approach identified 7 molecular subtypes of colon cancer with 44 feature genes, offering a finer classification compared to previous studies[72]
Protein-protein interaction network data for CRCDynamic Bayesian networkThe study identified biomarkers with high accuracy and F1-scores, with Alpha-2-HS-glycoprotein identified as a dominant hub gene in CRC[73]
Gene expression data from LS174T cell lines, normal and adenoma samples, and CRC-related samplesNaive Bayesian networkThe BN model demonstrated accurate and reproducible prediction results for normal, adenoma, CRC, and related test samples, with high prediction accuracies[74]
Gene expression data related to Wnt signaling pathway in human CRCStatic Bayesian networkThe biologically inspired Bayesian models, which include epigenetic modifications, improved prediction accuracy for CRC, revealing a significant difference in the activation state of the β-catenin transcription complex between tumorous and normal samples[75]
Registry data of patients with colon cancer from the Department of Defense Automated Central Tumor Registryml-BBNsThe ml-BBNs demonstrated high accuracy in predicting recurrence and mortality in colon cancer, with AUCs ranging from 0.85 to 0.90, and positive predictive values for recurrence and mortality between 78% and 84%; the model identified which high-risk patients benefit from adjuvant therapy, with the largest benefit for elderly patients with high T-stage tumors[50]
Somatic mutation data from 906 stage II/III CRC from the VICTOR clinical trialBayesian network modelThe BN analysis revealed significant associations between microsatellite instability, chromosomal instability, and specific mutations (TP53, KRAS, BRAF, PIK3CA, NRAS), and proposed a new molecular classification for CRC with improved prognostic capabilities, particularly for disease-free survival in certain groups[76]
Population-based data from the SEER registry, including 146248 records of colon cancer patientsml-BBNThe ml-BBN model accurately estimated OS with an AUC of 0.85, identifying significant prognostic factors such as age, race, tumor histology, and AJCC staging, and demonstrating improved survival predictions compared to existing models[77]
Clinical data from 53 patients with colon carcinomatosis, including 31 clinical-pathological, treatment-related, and outcome variablesStep-wise ml-BBNThe BBN model identified three predictors of OS: Performance status, Peritoneal Cancer Index, and the ability to undergo CRS +/- HIPEC. The model achieved an AUC of 0.71, with positive and negative predictive values of 63.3% and 68.3%, respectively, and demonstrated strong classification for OS predictions[51]
Clinical data from 278 CRC patients undergoing SLN mappingA probabilistic Bayesian network modelThe BN model predicted FN SLN mapping with an (AUC of 0.84-0.86, achieving positive and negative predictive values of 83% and 97%, respectively. The number of SLN (< 3) and tumor-replaced nodes independently predicted FN SLN[78]
Gene expression data from cDNA arrays and clinical-pathological data of 494 CRC patients, focused on nodal metastasis predictionA Bayesian neural network with automatic relevance determinationTumor matrilysin was identified as a key predictor of nodal metastasis, with the Bayesian model achieving strong predictive performance, suggesting potential causality between matrilysin expression and nodal metastasis[48]
Liver cancer

Liver cancer, primarily hepatocellular carcinoma (HCC), presents unique challenges due to its association with underlying liver diseases like hepatitis and cirrhosis. BN has been utilized to model the progression from chronic liver disease to cancer by incorporating clinical, serological, and genetic data, offering insights into various aspects of diagnosis, prognosis, and treatment. By integrating diverse data types such as radiomics features, genetic variations, gene expression profiles, and clinical data, BN can model complex relationships and identify key factors influencing disease progression. For instance, a logistic sparsity-based feature selection model optimized using Bayesian optimization has been shown to significantly improve classification performance for HCC, especially under limited training data conditions[79]. Additionally, BNs are used to predict survival outcomes by incorporating clinical factors such as tumor size, liver function, and recurrence patterns. For example, in HCC patients after hepatectomy, a tree-augmented naïve Bayes algorithm identified portal vein tumor thrombosis as the most significant predictor of survival time, highlighting the model's potential in clinical decision-making[80]. These models also enable the discovery of significant biomarkers for early diagnosis, such as the identification of gene polymorphisms and miRNA-mRNA pairs that play crucial roles in HCC susceptibility and progression[81,82]. In diagnostic settings, BNs have been utilized to classify regions of the liver as malignant or benign based on functional computed tomography (CT) perfusion data, aiding in the differentiation of cancerous tissues from healthy ones[83]. BN models that incorporate imaging, clinical, and genomic data may support earlier diagnosis and personalized prognostic evaluations in HCC, aiding in treatment planning and surveillance for patients with chronic liver disease.

A summary of BN applications in liver cancer research is provided in Table 6[79-89], which highlights the versatility of BN in handling diverse data sources, from radiomics and genetic data to clinical records and functional imaging, underscoring their potential to improve diagnosis, prognosis, and treatment strategies in liver cancer. Overall, BNs offer a comprehensive framework for integrating and analyzing heterogeneous data in liver cancer research, facilitating more accurate predictions and personalized treatment strategies.

Table 6 Summary of Bayesian network applications in liver cancer research.
Data type
Bayesian network algorithm
Key findings
Ref.
Radiomics featuresA logistic sparsity-based feature selection model optimized using Bayesian optimizationThe Bayesian optimization-based feature selection model significantly improved classification performance for HCC and other focal liver lesions, especially under limited training data conditions[79]
Simulated concentration time curves for DCE-MRI and in vivo patient data with hepatic tumor lesionsBNNThe BNN provided more accurate parameter estimates compared to NLLS fitting and effectively identified uncertainties, particularly under high noise levels and out-of-distribution data, improving robustness for clinical applications[84]
Genetic variation data from 33 meta-analytic studies on 45 polymorphisms across 35 genes related to HCCBFDPFourteen gene polymorphisms, including CCND1, CTLA4, EGF, IL6, IL12A, KIF1B, MDM2, MICA, miR-499, MTHFR, PNPLA3, STAT4, TM6SF2, and XPD genes, were identified as significant biomarkers for HCC susceptibility[81]
Gene expression profiles of liver tissue samples from two microarray platforms analyzed for HCCAn empirical Bayesian methodThree genes were identified as specific biomarkers for HCC diagnosis, achieving an AUC of 0.931[85]
Single-cell multiomics data, including RNA-seq, Reduced Representation Bisulfite Sequencing, and copy number variation estimatesBayesian network modelsBest-fitted BN models identified 295 genes and provided novel insights into the mechanistic relationships of human lymphocyte antigen class I genes in HCC[86]
miRNA and mRNA expression data from 39 HCC patients and 25 liver cirrhosis patientsA flexible Bayesian two-step integrative methodThe study identified 66 significant miRNA-mRNA pairs, including molecules previously recognized as potential biomarkers in liver cancer[82]
Multi-omics data, including genome (mutation and copy number), transcriptome, proteome, and phosphoproteome from HCC samplesA Bayesian network mixture modelThe study identified three main HCC subtypes with distinct molecular characteristics, some associated with survival independent of clinical stage. Cluster-specific networks revealed connections between genotypes and molecular phenotypes[87]
Electronic medical records from 10060 primary liver cancer patients, including TCM symptoms, signs, tongue diagnosis, and pulse diagnosis informationBayesian network modelThe Bayesian network model achieved a classification accuracy of 85.84% for syndrome diagnosis in primary liver cancer, demonstrating its effectiveness in mining nonlinear relationships in clinical data and providing reliable support for TCM-based syndrome differentiation and treatment in liver cancer[88]
Clinical data of HCC patients, including recurrence outcomes (early, late, or no recurrence)Bayesian network-based modelThe Bayesian network model effectively distinguished between early, late, and no recurrence, significantly outperforming benchmark techniques in accuracy, precision, recall, and F-measures. It addressed the challenge of insufficient early-stage information by integrating latent variables, offering robust and reliable predictions validated across datasets, with potential implications for improving HCC recurrence management in clinical practice[89]
Dataset of 299 HCC patients after hepatectomy, including factors like preoperative AFP level, liver function grade, tumor size, and postoperative treatmentTree-augmented naïve Bayes algorithmThe Bayesian network model identified PVTT as the most significant predictor of survival time for HCC patients after hepatectomy. The model also highlighted the preoperative AFP level and postoperative performance of TACE as independent survival factors[80]
Functional CT perfusion data of hepatic regions, including measurements from malignant and benign liver tissues, acquired over 590 seconds using repeated scansA Bayesian semiparametric modelThe model facilitated the clustering of liver regions based on their CT profiles, which can be used to predict and classify regions as malignant or benign, aiding in the discrimination of cancerous tissue from healthy tissue in diagnostic settings[83]
Gallbladder cancer

Gallbladder cancer, although rare, is an aggressive malignancy that is often diagnosed incidentally during procedures for gallstone disease. Early detection and accurate prognosis prediction remain critical for improving outcomes. Recent studies have demonstrated the BN applications in enhancing diagnostic precision, survival predictions, and decision-making support for gallbladder cancer management.

For early detection, BN models have been particularly instrumental in identifying neoplastic gallbladder polyps. Using preoperative ultrasound features, these models accurately identified polyps larger than 10 mm with neoplastic potential, surpassing current clinical guidelines with an accuracy of over 81% in both training and testing sets[15,90]. Such findings provide refined surgical criteria, aiding clinicians in determining the necessity of surgical interventions.

For prognosis prediction, BN models have been developed to identify key survival factors and support treatment decisions. One study utilizing clinical data from 362 patients with gallbladder adenocarcinoma employed a tree-augmented naïve Bayesian algorithm to classify survival outcomes. The results emphasized that R0 resection significantly improves survival, even in advanced stages such as T4 and N2, suggesting that surgery remains a viable option for these patients[91]. Similarly, another study using data from 366 patients constructed a BN model with BayesiaLab software, identifying surgical type and tumor-node-metastasis stages as the most significant prognostic factors for gallbladder cancer[92].

Furthermore, BN models have been applied to optimize lymph node (LN) assessment in gallbladder adenocarcinoma. A study analyzing data from 1268 patients revealed that harvesting a minimum of seven LNs, with an optimal range of 7 to 10, maximized survival outcomes for patients undergoing curative resection[93]. This finding underscores the importance of adequate lymphadenectomy in surgical management.

To further enhance survival prediction, BN models have outperformed traditional methods such as nomograms. By integrating imaging findings, such as gallbladder wall thickening, with patient risk factors like age, gender, and gallstone history, BN models have demonstrated superior accuracy in differentiating benign from malignant conditions and predicting survival following curative-intent resections[94]. In addition, BN-based survival models have provided decision support for advanced gallbladder carcinoma, showing that adjuvant chemoradiotherapy may significantly improve survival in node-positive patients[95]. BN-based predictions support surgical decision-making and prognosis estimation, offering clinicians a more objective basis for managing complex cases and potentially expanding curative treatment options for advanced-stage patients.

In summary, BNs have shown significant promise in the management of gallbladder cancer, offering robust tools for early detection, prognosis prediction, and treatment optimization. Their ability to integrate diverse clinical, imaging, and surgical data allows for improved accuracy over traditional methods, ultimately guiding clinicians in delivering personalized and timely interventions for better patient outcomes.

Cholangiocarcinoma

BN models have emerged as valuable tools for prognostic and predictive modeling in cholangiocarcinoma, offering insights into complex clinical decision-making scenarios. A study utilizing clinical data from 531 patients with intrahepatic cholangiocarcinoma (ICC) developed and validated a BN prediction model for microvascular invasion (MVI). The model, constructed using five preoperative risk factors-obstructive jaundice, prognostic nutritional index, carbohydrate antigen 19-9 (CA19-9), tumor size, and major vascular invasion-achieved AUC values of 78.92% and 83.01% in the training and testing sets, respectively, demonstrating its effectiveness in preoperative MVI prediction[96]. Similarly, another study leveraged clinical and pathological data from 516 ICC patients to construct BN models for predicting 1-year survival after curative resection. The naïve Bayesian model, based on seven independent prognostic factors, demonstrated the highest predictive performance with an AUC of 79.5%, outperforming nomogram-based models in survival prognostication[97]. These findings underscore the potential of BN models in enhancing personalized care and improving predictive accuracy in cholangiocarcinoma management. BN models help predict key outcomes like MVI and survival, offering noninvasive, data-driven tools to inform surgical planning and postoperative care, thus enhancing precision in clinical decision-making.

Pancreatic cancer

Pancreatic cancer is one of the deadliest malignancies, often diagnosed at advanced stages due to the lack of early symptoms. To address this challenge, multiple studies have employed BNs to improve diagnostic, prognostic, and treatment strategies for pancreatic cancer.

Using DNA methylation and transcriptome data from the TCGA database, researchers identified 14 specifically methylated genes that served as the foundation for a BN-based prognostic prediction model in pancreatic adenocarcinoma. This model achieved an impressive predictive performance (AUC = 0.937), showcasing its potential to refine subtype classification and guide personalized treatment strategies[98]. Similarly, a study using small RNA sequencing data from blood small extracellular vesicle miRNAs applied a BN model to analyze causal relationships. This revealed miR-95-3p as linked to pancreatic ductal adenocarcinoma (PDAC) and miR-26b-5p to chronic pancreatitis (CP), providing valuable insights into miRNA-mediated disease mechanisms and aiding differentiation between PDAC and CP[99].

In the realm of survival analysis, researchers developed a BN model using clinical data to predict survival outcomes for potentially resectable pancreatic cancer. The model achieved AUCs of 0.7 for pre-operative predictions and 0.8 for post-operative updates, offering clinicians valuable tools for tailored patient management[100]. Furthermore, another study utilized delta radiomic features (DRFs) extracted from longitudinal non-contrast CT images of pancreatic cancer patients to predict treatment response. A Bayesian-regularization-neural-network model achieved a cross-validated AUC of 0.94, demonstrating the utility of DRFs in early response prediction[101].

To enhance diagnostic accuracy, several BN-based models have been developed. A Bayesian combination model integrating a random forest classifier and a convolutional neural network used CT image data and demographic information to classify four common types of pancreatic cysts, achieving a classification accuracy of 83.6%[102]. Similarly, an automated differential diagnosis system for pancreatic cystic tumors utilized digital pathology data, distinguishing between benign serous cystadenoma and potentially malignant mucinous cystadenoma. Among the classifiers tested, the Bayesian model demonstrated superior performance based on morphological features, emphasizing its diagnostic value[103].

Innovative approaches have also integrated external knowledge into BN frameworks. A study combined PubMed knowledge with Electronic Health Records (EHR) to develop a weighted Bayesian Network Inference (BNI) model for pancreatic cancer prediction. By incorporating risk factor weights derived from PubMed abstracts, the weighted BNI model demonstrated superior accuracy compared to conventional BNI and other classifiers, underscoring its potential for clinical applications[104]. Another study applied a BN model to hospital admission data, including type of admission, procedure, primary diagnosis, and Charlson co-morbidity index, to predict the probability of in-hospital death. This analysis highlighted the role of co-morbidities and admission details in influencing survival outcomes, showcasing the BN model’s ability to evaluate multiple hypotheses and measure variable impact[105]. These models may facilitate early intervention and guide therapy selection in high-risk pancreatic cancer patients.

Collectively, these studies illustrate the versatility and potential of BN in improving the diagnosis, prognosis, and treatment of pancreatic cancer. By integrating diverse data types and leveraging advanced algorithms, BN-based approaches are paving the way for more precise and personalized healthcare solutions in pancreatic cancer management.

Other related studies

BN applications in other (or rare) gastrointestinal cancer research: A recent study applied BN methodology to develop a computational tool, PROMETheus, for preoperative risk stratification in gastrointestinal stromal tumors (GISTs). Using clinicopathological data from 160 real-world GIST cases, the BN model integrated key parameters such as tumor size, location, biopsy-derived mitotic count, and tumor response to therapy. The model exhibited excellent diagnostic performance, with posterior predictive checks confirming its accuracy. PROMETheus, an intuitive tool based on this BN model, provides dynamic predictions of mitotic count and risk assessment, enhancing preoperative decision-making for GIST patients and offering a foundation for personalized therapeutic strategies[106]. Moreover, study on malignant peritoneal mesothelioma, a rare gastrointestinal cancer, utilized clinicopathological data from 154 patients treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy to establish a BN model. The model incorporated ten prognostic factors identified through survival analysis, with its structure refined based on clinical expertise and previous research. Demonstrating an accuracy of 72.7% and an AUC of 0.74, the BN model showed strong potential as a clinical decision support tool for predicting patient survival[107]. Similarly, another study on pseudomyxoma peritonei leveraged clinicopathological data from 453 patients undergoing similar treatments to establish a BN model. Incorporating seven independent prognostic factors, including gender, previous operation history, histological grading, lymphatic metastasis, peritoneal cancer index, completeness of cytoreduction, and splenectomy. Internal validation demonstrated the BN model's predictive accuracy of 70.3% and an AUC of 73.5%[108]. These findings underscore the versatility and efficacy of BN approaches in addressing the complexities of diverse gastrointestinal cancers.

BN applications in multi-cancer research for gastrointestinal cancers: BN models have been applied in multi-cancer research to uncover critical insights into gastrointestinal cancers. In liver and pancreatic cancers, BN models were used to integrate dose volume histograms and clinical factors, such as tumor location, patient age, and planning target volume size. This approach achieved an AUC of 83.56% and identified tumor location as the most critical factor in selecting between radiotherapy plans using TrueBeam or MRIdian[109]. In another study focusing on gastric and CRCs analyzed within a pan-cancer series, BN were employed to examine gene expression data and survival outcomes. The analysis highlighted key prognostic markers, such as TYMS and RGS1, and revealed gene interactions related to cell cycle, apoptosis, and metabolism[110]. These findings provided valuable insights into survival prediction and the molecular mechanisms underlying gastrointestinal cancers, showcasing the significant utility of BNs in multi-cancer research.

CHALLENGES IN BN APPLICATIONS

BN holds considerable promise in advancing gastrointestinal cancer research and clinical practice, yet their implementation faces several significant challenges. These challenges span issues related to data, computational methodologies, and clinical translation. A detailed analysis is presented below, incorporating both general challenges and those specific to gastrointestinal cancers.

Data-related challenges

Data-related challenges are prominent in cancer studies, particularly in gastrointestinal cancers which heterogeneity and missing data complicate model construction. For esophageal and gastric cancers, early-stage data are scarce due to late diagnoses, hindering the development of reliable early detection models[111]. Multi-omics integration. CRC, while benefiting from large-scale data from screening programs, faces challenges in generalization due to population variations like genetic diversity and dietary differences. Liver cancer is further complicated by associated comorbidities such as hepatitis and cirrhosis, which introduce additional variability. The rarity of gallbladder and pancreatic cancers limits the availability of high-quality datasets, increasing the risk of overfitting in models. Additionally, imbalanced data representation, where advanced-stage cases dominate, skew predictions and hinder early detection efforts, particularly for pancreatic and liver cancers. Integrating data from multiple centers also presents difficulties due to differences in data collection protocols and standards. Expanding early screening programs, creating dedicated biobanks, and leveraging natural language processing to extract structured data from electronic medical records can help enrich early-stage datasets. Standardizing data collection, promoting data sharing, and employing techniques like transfer learning and synthetic data generation can effectively address data scarcity, heterogeneity, and imbalance.

Computational and methodological challenges

BN faces significant computational and methodological challenges, particularly in handling large, complex datasets. Multi-omics integration in cancers like colorectal and gastric cancers, which generate high-dimensional data, requires substantial computational resources for training BN[87,112]. Furthermore, dynamic models used for cancers like liver cancer, which track progression or treatment response over time, are computationally intensive. Rare cancers like gallbladder and pancreatic cancers face the risk of overfitting due to limited data, as smaller datasets may fail to capture essential relationships. Balancing model complexity is crucial-simpler models may not capture necessary nuances, while more complex models risk overfitting[113]. Techniques like cross-validation and Bayesian regularization can help, but they may not fully address the limitations posed by small datasets. Dimensionality reduction, regularization, and scalable computing frameworks help manage high-dimensional data and prevent overfitting in complex or rare cancer models.

Clinical translation challenges

The clinical translation of BN faces challenges related to interpretability and acceptance among clinicians. While BN has an intuitive structure, their outputs, such as probabilistic risk scores or survival probabilities, may not align with traditional clinical decision-making, especially in cancers like esophageal and pancreatic cancer[114]. Clinicians often prefer simple, actionable outputs, and models lacking clear visualization or an understandable explanation may face resistance. Validation for real-world implementation is also crucial but challenging. Regional differences in patient characteristics and risk factors complicate external validation in cancers like esophageal and gastric cancer. Liver cancer’s dynamic models require longitudinal datasets, which are resource-intensive to obtain. For rare cancers, such as gallbladder and pancreatic cancers, the limited availability of validation cohorts reduces confidence in the robustness of the models. Enhancing model interpretability, aligning outputs with clinical workflows, and validating across diverse populations are key to improving clinical acceptance and real-world utility.

FUTURE DIRECTIONS

Addressing the challenges in applying BN to gastrointestinal cancers requires a forward-looking approach. Future advancements span algorithmic innovations, technological integration, personalized medicine, and collaborative efforts.

Advances in BN algorithms

BN-based methodologies are evolving to accommodate the complexities of cancer research. Dynamic BN is promising for modeling disease progression and treatment response over time[115]. For example, these models could track biomarker changes in liver or pancreatic cancer, providing real-time insights into disease dynamics. Additionally, developing scalable algorithms optimized for high-dimensional data is critical for reducing computational burdens, allowing for faster training and real-time inference. This is especially important for cancers with large multi-omics datasets, such as colorectal and gastric cancer. Developing scalable algorithms optimized for high-dimensional datasets will enable faster training and real-time inference, crucial for integrating extensive genomic, transcriptomic, and epigenetic data from cancers such as gastric and CRC.

Integration with emerging technologies

Emerging technologies are significantly enhancing the utility and scope of BN in cancer research. The integration of multi-omics data, including genomic, proteomic, and metabolomic information, offers a more comprehensive understanding of cancer biology[64], particularly for complex cancers like pancreatic and liver cancer. Moreover, artificial intelligence (AI) has emerged as techniques, such as deep learning, complement BN by identifying latent patterns and improving feature selection. For example, AI-powered preprocessing can refine inputs for Bayesian models in esophageal and gastric cancer studies. Furthermore, BN can be combined with data from wearable devices (e.g., continuous glucose monitors, activity trackers) and EHRs using real-time data fusion approaches to enable continuous risk monitoring and early alert systems in outpatient settings.

Personalized medicine and precision oncology

BN are well-suited to advancing personalized medicine and precision oncology. By incorporating individual patient data, these networks can identify the most effective treatment pathways, such as recommending targeted therapies for colorectal or gastric cancer patients based on their genetic profiles. Furthermore, dynamic Bayesian models can monitor treatment responses over time, enabling timely adjustments to therapeutic strategies. This capability is particularly valuable for cancers with aggressive progression, such as pancreatic cancer, where rapid adaptation of treatment plans can significantly impact patient outcomes. By advancing algorithmic capabilities, integrating with emerging technologies, and fostering collaborative ecosystems, BN can transform the diagnosis, treatment, and management of gastrointestinal cancers. These future directions provide a roadmap for overcoming current limitations and realizing the full potential of this promising technology.

CONCLUSION

BNs are powerful tools in gastrointestinal cancer research, excelling in integrating diverse data, identifying causal relationships, and managing uncertainty. They have shown significant potential in risk prediction, early diagnosis, treatment optimization, and outcome forecasting. However, challenges remain, such as data scarcity, particularly in early detection, computational complexity as network size increases, and barriers to clinical implementation. To address these challenges, future efforts should focus on advancing algorithms to handle dynamic and temporal data, integrating BNs with multi-omics and AI approaches, and promoting collaborative data sharing to improve data availability. Developing user-friendly tools and aligning BN models with clinical workflows will further support their real-world adoption. As precision oncology continues to evolve, BNs hold great promise in enabling personalized treatment strategies and dynamic monitoring of therapeutic responses, ultimately transforming gastrointestinal cancer care and advancing personalized medicine to improve patient outcomes.

Footnotes

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

Peer-review model: Single blind

Specialty type: Oncology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade D

Novelty: Grade A, Grade B, Grade D

Creativity or Innovation: Grade A, Grade A, Grade D

Scientific Significance: Grade A, Grade A, Grade D

P-Reviewer: Liu HR; Su YH S-Editor: Li L L-Editor: A P-Editor: Zhao YQ

References
1.  Xie Y, Shi L, He X, Luo Y. Gastrointestinal cancers in China, the USA, and Europe. Gastroenterol Rep (Oxf). 2021;9:91-104.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 70]  [Cited by in RCA: 122]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
2.  Arnold M, Abnet CC, Neale RE, Vignat J, Giovannucci EL, McGlynn KA, Bray F. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology. 2020;159:335-349.e15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 857]  [Cited by in RCA: 1192]  [Article Influence: 238.4]  [Reference Citation Analysis (0)]
3.  Sonkin D, Thomas A, Teicher BA. Cancer treatments: Past, present, and future. Cancer Genet. 2024;286-287:18-24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 133]  [Article Influence: 133.0]  [Reference Citation Analysis (0)]
4.  Huang J, Lucero-Prisno DE 3rd, Zhang L, Xu W, Wong SH, Ng SC, Wong MCS. Updated epidemiology of gastrointestinal cancers in East Asia. Nat Rev Gastroenterol Hepatol. 2023;20:271-287.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 114]  [Reference Citation Analysis (0)]
5.  Lu L, Mullins CS, Schafmayer C, Zeißig S, Linnebacher M. A global assessment of recent trends in gastrointestinal cancer and lifestyle-associated risk factors. Cancer Commun (Lond). 2021;41:1137-1151.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 184]  [Cited by in RCA: 176]  [Article Influence: 44.0]  [Reference Citation Analysis (0)]
6.  Alsina M, Arrazubi V, Diez M, Tabernero J. Current developments in gastric cancer: from molecular profiling to treatment strategy. Nat Rev Gastroenterol Hepatol. 2023;20:155-170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 173]  [Article Influence: 86.5]  [Reference Citation Analysis (1)]
7.  Kim JH, Zang DY, Jang HJ, Kim HS. A Bayesian network meta-analysis on systemic therapy for previously treated gastric cancer. Crit Rev Oncol Hematol. 2021;167:103505.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
8.  Park S, Nam CM, Kim SG, Mun JE, Rha SY, Chung HC. Comparative efficacy and tolerability of third-line treatments for advanced gastric cancer: A systematic review with Bayesian network meta-analysis. Eur J Cancer. 2021;144:49-60.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
9.  Scutari M, Denis J.   Bayesian Networks. 2nd ed. New York: Imprint Chapman and Hall/CRC, 2021.  [PubMed]  [DOI]  [Full Text]
10.  Becker AK, Dörr M, Felix SB, Frost F, Grabe HJ, Lerch MM, Nauck M, Völker U, Völzke H, Kaderali L. From heterogeneous healthcare data to disease-specific biomarker networks: A hierarchical Bayesian network approach. PLoS Comput Biol. 2021;17:e1008735.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
11.  Fahd F, Veitch B, Khan F. Risk assessment of Arctic aquatic species using ecotoxicological biomarkers and Bayesian network. Mar Pollut Bull. 2020;156:111212.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
12.  Ji Z, Xia Q, Meng G.   A Review of Parameter Learning Methods in Bayesian Network. In: Huang DS, Han K, editors. Advanced Intelligent Computing Theories and Applications. ICIC 2015. Lecture Notes in Computer Science. Cham: Springer, 2015.  [PubMed]  [DOI]  [Full Text]
13.  Kyrimi E, Neves MR, McLachlan S, Neil M, Marsh W, Fenton N. Medical idioms for clinical Bayesian network development. J Biomed Inform. 2020;108:103495.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
14.  Kitson NK, Constantinou AC, Guo Z, Liu Y, Chobtham K. A survey of Bayesian Network structure learning. Artif Intell Rev. 2023;56:8721-8814.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
15.  Li Q, Dou M, Zhang J, Jia P, Wang X, Lei D, Li J, Yang W, Yang R, Yang C, Zhang X, Hao Q, Geng X, Zhang Y, Liu Y, Guo Z, Yao C, Cai Z, Si S, Geng Z, Zhang D. A Bayesian network model to predict neoplastic risk for patients with gallbladder polyps larger than 10 mm based on preoperative ultrasound features. Surg Endosc. 2023;37:5453-5463.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
16.  Vaniš M, Lokaj Z, Šrotýř M. A Novel Algorithm for Merging Bayesian Networks. Symmetry. 2023;15:1461.  [PubMed]  [DOI]  [Full Text]
17.  Ru X, Gao X, Wang Y, Liu X. Bayesian network parameter learning using constraint-based data extension method. Appl Intell. 2023;53:9958-9977.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
18.  Wang D, Amriljaharadak A, Xiao Y. Dynamic Knowledge Inference Based on Bayesian Network Learning. Math Probl Eng. 2020;2020:1-9.  [PubMed]  [DOI]  [Full Text]
19.  Ru X, Gao X, Wang Z, Wang Y, Liu X. Bayesian network parameter learning using fuzzy constraints. Neurocomputing. 2023;544:126239.  [PubMed]  [DOI]  [Full Text]
20.  Zhang SZ, Zhang ZN, Yang NH, Zhang JY, Wang XK.   An improved EM algorithm for Bayesian networks parameter learning. Proceedings of 2004 International Conference on Machine Learning and Cybernetics (IEEE Cat. No.04EX826); 2004 Aug 26-29; Shanghai, China. IEEE, 2005: 1503-1508.  [PubMed]  [DOI]  [Full Text]
21.  Niu D, Shi H, Wu DD. Short-term load forecasting using bayesian neural networks learned by Hybrid Monte Carlo algorithm. Appl Soft Comput. 2012;12:1822-1827.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 86]  [Cited by in RCA: 66]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
22.  Lauritzen SL, Spiegelhalter DJ. Local Computations with Probabilities on Graphical Structures and Their Application to Expert Systems. J R Stat Soc B. 1988;50:157-194.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 269]  [Cited by in RCA: 100]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
23.  Larrañaga P, Karshenas H, Bielza C, Santana R. A review on evolutionary algorithms in Bayesian network learning and inference tasks. Inform Sci. 2013;233:109-125.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 97]  [Cited by in RCA: 97]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
24.  Yuan C, Druzdzel MJ. Importance sampling algorithms for Bayesian networks: Principles and performance. Math Comput Modell. 2006;43:1189-1207.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 47]  [Cited by in RCA: 47]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
25.  Salmerón A, Rumí R, Langseth H, Nielsen TD, Madsen AL. A Review of Inference Algorithms for Hybrid Bayesian Networks. J Artif Intell Res. 2018;62:799-828.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
26.  Bhimagavni NK, Adilakshmi T.   Structure Learning of Bayesian Network from the Data. In: Kumar A, Ghinea G, Merugu S, editors. Proceedings of the 2nd International Conference on Cognitive and Intelligent Computing. ICCIC 2022. Cognitive Science and Technology. Singapore: Springer, 2023.  [PubMed]  [DOI]  [Full Text]
27.  Burigana L. Bayesian networks and knowledge structures in cognitive assessment: Remarks on basic comparable aspects. J Math Psychol. 2024;123:102875.  [PubMed]  [DOI]  [Full Text]
28.  Ling Z, Yu K, Zhang Y, Liu L, Li J. Causal learner: A toolbox for causal structure and Markov blanket learning. Pattern Recognit Lett. 2022;163:92-95.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
29.  Yu K, Guo X, Liu L, Li J, Wang H, Ling Z, Wu X. Causality-based Feature Selection. ACM Comput Surv. 2021;53:1-36.  [PubMed]  [DOI]  [Full Text]
30.  Alpan K, Ilgi GS.   Classification of Diabetes Dataset with Data Mining Techniques by Using WEKA Approach. 2020 4th International Symposium on Multidisciplinary Studies and Innovative Technologies (ISMSIT); 2020 Oct 22-24; Istanbul, Turkey. IEEE, 2020 1-7.  [PubMed]  [DOI]  [Full Text]
31.  Hasan U, Hossain E, Gani MO.   A Survey on Causal Discovery Methods for I.I.D. and Time Series Data. 2024 Preprint. Available from: arxiv:15027.  [PubMed]  [DOI]  [Full Text]
32.  Sharma A, Kiciman E.   DoWhy: An End-to-End Library for Causal Inference. 2020. 2020 Preprint. Available from: arxiv:04216.  [PubMed]  [DOI]  [Full Text]
33.  Rizzo A. Bayesian analysis supports the role of neoadjuvant chemoradiation followed by surgery for resectable locoregional esophageal cancer. Thorac Cancer. 2022;13:3098.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
34.  Nopour R. Prediction of five-year survival among esophageal cancer patients using machine learning. Heliyon. 2023;9:e22654.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
35.  Bradley A, Sami S, N G H, Macleod A, Prasanth M, Zafar M, Hemadasa N, Neagle G, Rosindell I, Apollos J. A predictive Bayesian network that risk stratifies patients undergoing Barrett's surveillance for personalized risk of developing malignancy. PLoS One. 2020;15:e0240620.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
36.  Zaidi AH, Gopalakrishnan V, Kasi PM, Zeng X, Malhotra U, Balasubramanian J, Visweswaran S, Sun M, Flint MS, Davison JM, Hood BL, Conrads TP, Bergman JJ, Bigbee WL, Jobe BA. Evaluation of a 4-protein serum biomarker panel-biglycan, annexin-A6, myeloperoxidase, and protein S100-A9 (B-AMP)-for the detection of esophageal adenocarcinoma. Cancer. 2014;120:3902-3913.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 33]  [Cited by in RCA: 35]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
37.  Liao T, Lu Y, Li W, Wang K, Zhang Y, Luo Z, Ju Y, Ouyang M. Construction and validation of a glycolysis-related lncRNA signature for prognosis prediction in Stomach Adenocarcinoma. Front Genet. 2022;13:794621.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
38.  Sheidaei A, Foroushani AR, Gohari K, Zeraati H. A novel dynamic Bayesian network approach for data mining and survival data analysis. BMC Med Inform Decis Mak. 2022;22:251.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
39.  Pan WT, Zhou SN, Pan MX, Luo QY, Zhang L, Yang DJ, Qiu M. Role of Systemic Treatment for Advanced/Metastatic Gastric Carcinoma in the Third-Line Setting: A Bayesian Network Analysis. Front Oncol. 2020;10:513.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
40.  Park S, Kar N, Cheong JH, Hwang TH. Bayesian semi-nonnegative matrix tri-factorization to identify pathways associated with cancer phenotypes. Pac Symp Biocomput. 2020;25:427-438.  [PubMed]  [DOI]  [Full Text]
41.  Ou L, Liu H, Peng C, Zou Y, Jia J, Li H, Feng Z, Zhang G, Yao M. Helicobacter pylori infection facilitates cell migration and potentially impact clinical outcomes in gastric cancer. Heliyon. 2024;10:e37046.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 17]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
42.  Yu J, Xuan Z, Feng X, Zou Q, Wang L. A novel collaborative filtering model for LncRNA-disease association prediction based on the Naïve Bayesian classifier. BMC Bioinformatics. 2019;20:396.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 40]  [Cited by in RCA: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
43.  Korhani Kangi A, Bahrampour A. Predicting the Survival of Gastric Cancer Patients Using Artificial and Bayesian Neural Networks. Asian Pac J Cancer Prev. 2018;19:487-490.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
44.  Libânio D, Dinis-Ribeiro M, Pimentel-Nunes P, Dias CC, Rodrigues PP. Predicting outcomes of gastric endoscopic submucosal dissection using a Bayesian approach: a step for individualized risk assessment. Endosc Int Open. 2017;5:E563-E572.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
45.  Liu X, Hao Y, Fan T, Nan K. Application of intelligent algorithm in the optimization of novel protein regulatory pathway: Mechanism of action of gastric carcinoma protein p42.3. J Cancer Res Ther. 2016;12:650-656.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
46.  Balov N. A categorical network approach for discovering differentially expressed regulations in cancer. BMC Med Genomics. 2013;6 Suppl 3:S1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
47.  Hiroyuki I, Hiroki S, Kazuhiko A, Toshimasa Y.   Classification of Gastric Cancer Subtypes using ICA, MLR and Bayesian Network. In: Lehmann CH, Ammenwerth E, Nøhr C, editors. Studies in Health Technology and Informatics. Amsterdam, The Netherlands: IOS Press, 2013.  [PubMed]  [DOI]  [Full Text]
48.  Kurokawa S, Arimura Y, Yamamoto H, Adachi Y, Endo T, Sato T, Suga T, Hosokawa M, Shinomura Y, Imai K. Tumour matrilysin expression predicts metastatic potential of stage I (pT1) colon and rectal cancers. Gut. 2005;54:1751-1758.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 26]  [Cited by in RCA: 28]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
49.  Osong B, Masciocchi C, Damiani A, Bermejo I, Meldolesi E, Chiloiro G, Berbee M, Lee SH, Dekker A, Valentini V, Gerard JP, Rödel C, Bujko K, van de Velde C, Folkesson J, Sainato A, Glynne-Jones R, Ngan S, Brændengen M, Sebag-Montefiore D, van Soest J. Bayesian network structure for predicting local tumor recurrence in rectal cancer patients treated with neoadjuvant chemoradiation followed by surgery. Phys Imaging Radiat Oncol. 2022;22:1-7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
50.  Steele SR, Bilchik A, Johnson EK, Nissan A, Peoples GE, Eberhardt JS, Kalina P, Petersen B, Brücher B, Protic M, Avital I, Stojadinovic A. Time-dependent estimates of recurrence and survival in colon cancer: clinical decision support system tool development for adjuvant therapy and oncological outcome assessment. Am Surg. 2014;80:441-453.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
51.  Stojadinovic A, Nissan A, Eberhardt J, Chua TC, Pelz JO, Esquivel J. Development of a Bayesian Belief Network Model for personalized prognostic risk assessment in colon carcinomatosis. Am Surg. 2011;77:221-230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 24]  [Cited by in RCA: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
52.  Zhao M, Lau MC, Haruki K, Väyrynen JP, Gurjao C, Väyrynen SA, Dias Costa A, Borowsky J, Fujiyoshi K, Arima K, Hamada T, Lennerz JK, Fuchs CS, Nishihara R, Chan AT, Ng K, Zhang X, Meyerhardt JA, Song M, Wang M, Giannakis M, Nowak JA, Yu KH, Ugai T, Ogino S. Bayesian risk prediction model for colorectal cancer mortality through integration of clinicopathologic and genomic data. NPJ Precis Oncol. 2023;7:57.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
53.  Corrales D, Santos-Lozano A, López-Ortiz S, Lucia A, Insua DR. Colorectal cancer risk mapping through Bayesian networks. Comput Methods Programs Biomed. 2024;257:108407.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
54.  Pineda-Antunez C, Seguin C, van Duuren LA, Knudsen AB, Davidi B, Nascimento de Lima P, Rutter C, Kuntz KM, Lansdorp-Vogelaar I, Collier N, Ozik J, Alarid-Escudero F. Emulator-Based Bayesian Calibration of the CISNET Colorectal Cancer Models. Med Decis Making. 2024;44:543-553.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
55.  Lykoskoufis NMR, Planet E, Ongen H, Trono D, Dermitzakis ET. Transposable elements mediate genetic effects altering the expression of nearby genes in colorectal cancer. Nat Commun. 2024;15:749.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Reference Citation Analysis (0)]
56.  Kim H, Jang WS, Sim WS, Kim HS, Choi JE, Baek ES, Park YR, Shin SJ. Synthetic Data Improve Survival Status Prediction Models in Early-Onset Colorectal Cancer. JCO Clin Cancer Inform. 2024;8:e2300201.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
57.  Liu M, Hong Y, Duan X, Zhou Q, Chen J, Liu S, Su J, Han L, Zhang J, Niu B. Unveiling the metal mutation nexus: Exploring the genomic impacts of heavy metal exposure in lung adenocarcinoma and colorectal cancer. J Hazard Mater. 2024;461:132590.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
58.  Zhang M, Wang X, Yang N, Zhu X, Lu Z, Cai Y, Li B, Zhu Y, Li X, Wei Y, Zhang S, Tian J, Miao X. Prioritization of risk genes in colorectal cancer by integrative analysis of multi-omics data and gene networks. Sci China Life Sci. 2024;67:132-148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Reference Citation Analysis (0)]
59.  Li H, Sheng D, Jin C, Zhao G, Zhang L. Identifying and ranking causal microbial biomarkers for colorectal cancer at different cancer subsites and stages: a Mendelian randomization study. Front Oncol. 2023;13:1224705.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
60.  Qi Z, Zhibo Z, Jing Z, Zhanbo Q, Shugao H, Weili J, Jiang L, Shuwen H. Prediction model of poorly differentiated colorectal cancer (CRC) based on gut bacteria. BMC Microbiol. 2022;22:312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
61.  Pačínková A, Popovici V. Correction: Using empirical biological knowledge to infer regulatory networks from multi-omics data. BMC Bioinformatics. 2022;23:376.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
62.  Li R, Zhang C, Du K, Dan H, Ding R, Cai Z, Duan L, Xie Z, Zheng G, Wu H, Ren G, Dou X, Feng F, Zheng J. Analysis of Prognostic Factors of Rectal Cancer and Construction of a Prognostic Prediction Model Based on Bayesian Network. Front Public Health. 2022;10:842970.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
63.  Suter P, Kuipers J, Beerenwinkel N. Discovering gene regulatory networks of multiple phenotypic groups using dynamic Bayesian networks. Brief Bioinform. 2022;23.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Reference Citation Analysis (0)]
64.  Wang Y, Gao X, Ru X, Sun P, Wang J. Identification of gene signatures for COAD using feature selection and Bayesian network approaches. Sci Rep. 2022;12:8761.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
65.  Hsu TC, Lin C. Training with Small Medical Data: Robust Bayesian Neural Networks for Colon Cancer Overall Survival Prediction. Annu Int Conf IEEE Eng Med Biol Soc. 2021;2021:2030-2033.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
66.  Díez-Villanueva A, Jordà M, Carreras-Torres R, Alonso H, Cordero D, Guinó E, Sanjuan X, Santos C, Salazar R, Sanz-Pamplona R, Moreno V. Identifying causal models between genetically regulated methylation patterns and gene expression in healthy colon tissue. Clin Epigenetics. 2021;13:162.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
67.  Thomas M, Sakoda LC, Hoffmeister M, Rosenthal EA, Lee JK, van Duijnhoven FJB, Platz EA, Wu AH, Dampier CH, de la Chapelle A, Wolk A, Joshi AD, Burnett-Hartman A, Gsur A, Lindblom A, Castells A, Win AK, Namjou B, Van Guelpen B, Tangen CM, He Q, Li CI, Schafmayer C, Joshu CE, Ulrich CM, Bishop DT, Buchanan DD, Schaid D, Drew DA, Muller DC, Duggan D, Crosslin DR, Albanes D, Giovannucci EL, Larson E, Qu F, Mentch F, Giles GG, Hakonarson H, Hampel H, Stanaway IB, Figueiredo JC, Huyghe JR, Minnier J, Chang-Claude J, Hampe J, Harley JB, Visvanathan K, Curtis KR, Offit K, Li L, Le Marchand L, Vodickova L, Gunter MJ, Jenkins MA, Slattery ML, Lemire M, Woods MO, Song M, Murphy N, Lindor NM, Dikilitas O, Pharoah PDP, Campbell PT, Newcomb PA, Milne RL, MacInnis RJ, Castellví-Bel S, Ogino S, Berndt SI, Bézieau S, Thibodeau SN, Gallinger SJ, Zaidi SH, Harrison TA, Keku TO, Hudson TJ, Vymetalkova V, Moreno V, Martín V, Arndt V, Wei WQ, Chung W, Su YR, Hayes RB, White E, Vodicka P, Casey G, Gruber SB, Schoen RE, Chan AT, Potter JD, Brenner H, Jarvik GP, Corley DA, Peters U, Hsu L. Genome-wide Modeling of Polygenic Risk Score in Colorectal Cancer Risk. Am J Hum Genet. 2020;107:432-444.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 137]  [Cited by in RCA: 132]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
68.  Jang BS, Chang JH, Chie EK, Kim K, Park JW, Kim MJ, Song EJ, Nam YD, Kang SW, Jeong SY, Kim HJ. Gut Microbiome Composition Is Associated with a Pathologic Response After Preoperative Chemoradiation in Patients with Rectal Cancer. Int J Radiat Oncol Biol Phys. 2020;107:736-746.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 39]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
69.  Liu E, Li J, Kinnebrew GH, Zhang P, Zhang Y, Cheng L, Li L. A Fast and Furious Bayesian Network and Its Application of Identifying Colon Cancer to Liver Metastasis Gene Regulatory Networks. IEEE/ACM Trans Comput Biol Bioinform. 2021;18:1325-1335.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
70.  Kharrat N, Assidi M, Abu-Elmagd M, Pushparaj PN, Alkhaldy A, Arfaoui L, Naseer MI, El Omri A, Messaoudi S, Buhmeida A, Rebai A. Data mining analysis of human gut microbiota links Fusobacterium spp. with colorectal cancer onset. Bioinformation. 2019;15:372-379.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 11]  [Cited by in RCA: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
71.  Zhou B, Guo R. Genomic and regulatory characteristics of significant transcription factors in colorectal cancer metastasis. Sci Rep. 2018;8:17836.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
72.  Wang WH, Xie TY, Xie GL, Ren ZL, Li JM. An Integrated Approach for Identifying Molecular Subtypes in Human Colon Cancer Using Gene Expression Data. Genes (Basel). 2018;9:397.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
73.  Akutekwe A, Seker H, Yang S. In silico discovery of significant pathways in colorectal cancer metastasis using a two-stage optimisation approach. IET Syst Biol. 2015;9:294-302.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
74.  Sinha S. Reproducibility of parameter learning with missing observations in naive Wnt Bayesian network trained on colorectal cancer samples and doxycycline-treated cell lines. Mol Biosyst. 2015;11:1802-1819.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
75.  Sinha S. Integration of prior biological knowledge and epigenetic information enhances the prediction accuracy of the Bayesian Wnt pathway. Integr Biol (Camb). 2014;6:1034-1048.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11]  [Cited by in RCA: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
76.  Domingo E, Ramamoorthy R, Oukrif D, Rosmarin D, Presz M, Wang H, Pulker H, Lockstone H, Hveem T, Cranston T, Danielsen H, Novelli M, Davidson B, Xu ZZ, Molloy P, Johnstone E, Holmes C, Midgley R, Kerr D, Sieber O, Tomlinson I. Use of multivariate analysis to suggest a new molecular classification of colorectal cancer. J Pathol. 2013;229:441-448.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 60]  [Cited by in RCA: 63]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
77.  Stojadinovic A, Bilchik A, Smith D, Eberhardt JS, Ward EB, Nissan A, Johnson EK, Protic M, Peoples GE, Avital I, Steele SR. Clinical decision support and individualized prediction of survival in colon cancer: bayesian belief network model. Ann Surg Oncol. 2013;20:161-174.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 38]  [Cited by in RCA: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
78.  Nissan A, Protic M, Bilchik A, Eberhardt J, Peoples GE, Stojadinovic A. Predictive model of outcome of targeted nodal assessment in colorectal cancer. Ann Surg. 2010;251:265-274.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
79.  Tang VH, Duong STM, Nguyen CDT, Huynh TM, Duc VT, Phan C, Le H, Bui T, Truong SQH. Wavelet radiomics features from multiphase CT images for screening hepatocellular carcinoma: analysis and comparison. Sci Rep. 2023;13:19559.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
80.  Cai ZQ, Si SB, Chen C, Zhao Y, Ma YY, Wang L, Geng ZM. Analysis of prognostic factors for survival after hepatectomy for hepatocellular carcinoma based on a bayesian network. PLoS One. 2015;10:e0120805.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
81.  Penha Mesquita A, Victor Oliveira Monteiro A, Luiz Araújo Bentes Leal A, Dos Santos Pessoa L, de Siqueira Amorim Júnior J, Rogério Souza Monteiro J, Andrade de Sousa A, Fernando Pereira Vasconcelos D, Carolina Alves de Oliveira A, Leão Pereira A, Rodolfo Pereira da Silva F. Gene variations related to the hepatocellular carcinoma: Results from a field synopsis and Bayesian revaluation. Gene. 2023;869:147392.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
82.  Denis M, Varghese RS, Barefoot ME, Tadesse MG, Ressom HW. A Bayesian two-step integrative procedure incorporating prior knowledge for the identification of miRNA-mRNAs involved in hepatocellular carcinoma. Annu Int Conf IEEE Eng Med Biol Soc. 2022;2022:81-86.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
83.  Fronczyk KM, Guindani M, Hobbs BP, Ng CS, Vannucci M. A Bayesian Nonparametric Approach for Functional Data Classification with Application to Hepatic Tissue Characterization. Cancer Inform. 2015;14:151-162.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
84.  Dejene EM, Brenner W, Makowski MR, Kolbitsch C. Unified Bayesian network for uncertainty quantification of physiological parameters in dynamic contrast enhanced (DCE) MRI of the liver. Phys Med Biol. 2023;68.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
85.  Gholizadeh M, Mazlooman SR, Hadizadeh M, Drozdzik M, Eslami S. Detection of key mRNAs in liver tissue of hepatocellular carcinoma patients based on machine learning and bioinformatics analysis. MethodsX. 2023;10:102021.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
86.  Jihad M, Yet İ. Multiomics Integration at Single-Cell Resolution Using Bayesian Networks: A Case Study in Hepatocellular Carcinoma. OMICS. 2023;27:24-33.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
87.  Suter P, Dazert E, Kuipers J, Ng CKY, Boldanova T, Hall MN, Heim MH, Beerenwinkel N. Multi-omics subtyping of hepatocellular carcinoma patients using a Bayesian network mixture model. PLoS Comput Biol. 2022;18:e1009767.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
88.  Ding L, Zhang XY, Wu DY, Liu ML. Application of an extreme learning machine network with particle swarm optimization in syndrome classification of primary liver cancer. J Integr Med. 2021;19:395-407.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
89.  Xu D, Sheng JQ, Hu PJ, Huang TS, Lee WC. Predicting hepatocellular carcinoma recurrences: A data-driven multiclass classification method incorporating latent variables. J Biomed Inform. 2019;96:103237.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
90.  Li Q, Zhang J, Cai Z, Jia P, Wang X, Geng X, Zhang Y, Lei D, Li J, Yang W, Yang R, Zhang X, Yang C, Yao C, Hao Q, Liu Y, Guo Z, Si S, Geng Z, Zhang D. A Bayesian network prediction model for gallbladder polyps with malignant potential based on preoperative ultrasound. Surg Endosc. 2023;37:518-527.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
91.  Cong LL, Cai ZQ, Guo P, Chen C, Liu DC, Li WZ, Wang L, Zhao Y, Si SB, Geng ZM. Decision of surgical approach for advanced gallbladder adenocarcinoma based on a Bayesian network. J Surg Oncol. 2017;116:1123-1131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
92.  Cai ZQ, Guo P, Si SB, Geng ZM, Chen C, Cong LL. Analysis of prognostic factors for survival after surgery for gallbladder cancer based on a Bayesian network. Sci Rep. 2017;7:293.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
93.  Zhang R, Wu YH, Cai ZQ, Xue F, Zhang D, Chen C, Li Q, Fu JL, Tang ZH, Si SB, Geng ZM. Optimal number of harvested lymph nodes for curatively resected gallbladder adenocarcinoma based on a Bayesian network model. J Surg Oncol. 2020;122:1409-1417.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
94.  Haddawy P, Kahn CE Jr, Butarbutar M. A Bayesian network model for radiological diagnosis and procedure selection: work-up of suspected gallbladder disease. Med Phys. 1994;21:1185-1192.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 16]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
95.  Geng ZM, Cai ZQ, Zhang Z, Tang ZH, Xue F, Chen C, Zhang D, Li Q, Zhang R, Li WZ, Wang L, Si SB. Estimating survival benefit of adjuvant therapy based on a Bayesian network prediction model in curatively resected advanced gallbladder adenocarcinoma. World J Gastroenterol. 2019;25:5655-5666.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 16]  [Cited by in RCA: 18]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
96.  Li Q, Zhang J, Cai Z, Chen C, Wu H, Qiu Y, Song T, Mao X, He Y, Cheng Z, Zhai W, Li J, Si S, Zhang D, Geng Z, Tang Z. A Bayesian Network Prediction Model for Microvascular Invasion in Patients with Intrahepatic Cholangiocarcinoma: A Multi-institutional Study. World J Surg. 2023;47:773-784.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
97.  Chen C, Wu YH, Zhang JW, Qiu YH, Wu H, Li Q, Song TQ, He Y, Mao XH, Zhai WL, Cheng ZJ, Li JD, Si SB, Cai ZQ, Geng ZM, Tang ZH. [A prognostic model of intrahepatic cholangiocarcinoma after curative intent resection based on Bayesian network]. Zhonghua Wai Ke Za Zhi. 2021;59:265-271.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
98.  Li X, Zhang X, Lin X, Cai L, Wang Y, Chang Z. Classification and Prognosis Analysis of Pancreatic Cancer Based on DNA Methylation Profile and Clinical Information. Genes (Basel). 2022;13:1913.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
99.  Guo S, Qin H, Liu K, Wang H, Bai S, Liu S, Shao Z, Zhang Y, Song B, Xu X, Shen J, Zeng P, Shi X, Chen H, Gao S, Xu J, Pan Y, Xiong L, Li F, Zhang D, Jiao X, Jin G. Blood small extracellular vesicles derived miRNAs to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Clin Transl Med. 2021;11:e520.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
100.  Bradley A, Van der Meer R, McKay CJ. A prognostic Bayesian network that makes personalized predictions of poor prognostic outcome post resection of pancreatic ductal adenocarcinoma. PLoS One. 2019;14:e0222270.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
101.  Nasief H, Zheng C, Schott D, Hall W, Tsai S, Erickson B, Allen Li X. A machine learning based delta-radiomics process for early prediction of treatment response of pancreatic cancer. NPJ Precis Oncol. 2019;3:25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 75]  [Cited by in RCA: 97]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
102.  Dmitriev K, Kaufman AE, Javed AA, Hruban RH, Fishman EK, Lennon AM, Saltz JH. Classification of Pancreatic Cysts in Computed Tomography Images Using a Random Forest and Convolutional Neural Network Ensemble. Med Image Comput Comput Assist Interv. 2017;10435:150-158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 24]  [Cited by in RCA: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
103.  Song JW, Lee JH, Choi JH, Chun SJ. Automatic differential diagnosis of pancreatic serous and mucinous cystadenomas based on morphological features. Comput Biol Med. 2013;43:1-15.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
104.  Zhao D, Weng C. Combining PubMed knowledge and EHR data to develop a weighted bayesian network for pancreatic cancer prediction. J Biomed Inform. 2011;44:859-868.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 104]  [Cited by in RCA: 69]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
105.  Alvaro-Meca A, Gil-Prieto R, Gil de Miguel A. In-hospital death caused by pancreatic cancer in Spain: application with a bayesian network. Int J Biomed Sci. 2011;7:125-130.  [PubMed]  [DOI]
106.  Renne SL, Cammelli M, Santori I, Tassan-Mangina M, Samà L, Ruspi L, Sicoli F, Colombo P, Terracciano LM, Quagliuolo V, Cananzi FCM. True Mitotic Count Prediction in Gastrointestinal Stromal Tumors: Bayesian Network Model and PROMETheus (Preoperative Mitosis Estimator Tool) Application Development. J Med Internet Res. 2024;26:e50023.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
107.  Su YD, Zhao X, Ma R, Fu YB, Yang ZR, Wu HL, Yu Y, Yang R, Liang XL, Du XM, Chen Y, Li Y. Establishment of a Bayesian network model to predict the survival of malignant peritoneal mesothelioma patients after cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy. Int J Hyperthermia. 2023;40:2223374.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
108.  Zhao X, Li X, Lin Y, Ma R, Zhang Y, Xu D, Li Y. Survival prediction by Bayesian network modeling for pseudomyxoma peritonei after cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy. Cancer Med. 2023;12:2637-2645.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
109.  Kim DY, Jang BS, Kim E, Chie EK. Integrating Deep Learning-Based Dose Distribution Prediction with Bayesian Networks for Decision Support in Radiotherapy for Upper Gastrointestinal Cancer. Cancer Res Treat. 2025;57:186-197.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
110.  Carreras J, Nakamura N, Hamoudi R. Artificial Intelligence Analysis of Gene Expression Predicted the Overall Survival of Mantle Cell Lymphoma and a Large Pan-Cancer Series. Healthcare (Basel). 2022;10:155.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 25]  [Cited by in RCA: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
111.  Yan S, Fu X, Yang Y, Jia L, Liang J, Li Y, Yan L, Zhou Y, Zhou X, Li S, Mao X. Artificial intelligence in early screening for esophageal squamous cell carcinoma. Best Pract Res Cl Ga. 2025;102004.  [PubMed]  [DOI]  [Full Text]
112.  Ruiz-Perez D, Lugo-Martinez J, Bourguignon N, Mathee K, Lerner B, Bar-Joseph Z, Narasimhan G. Dynamic Bayesian Networks for Integrating Multi-omics Time Series Microbiome Data. mSystems. 2021;6:e01105-20.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
113.  Briscoe E, Feldman J. Conceptual complexity and the bias/variance tradeoff. Cognition. 2011;118:2-16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 55]  [Cited by in RCA: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
114.  Demirbaga U, Kaur N, Aujla GS. Uncovering hidden and complex relations of pandemic dynamics using an AI driven system. Sci Rep. 2024;14:15433.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
115.  Zhang T, Ma Y, Xiao X, Lin Y, Zhang X, Yin F, Li X. Dynamic Bayesian network in infectious diseases surveillance: a simulation study. Sci Rep. 2019;9:10376.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]