Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. May 7, 2021; 27(17): 1920-1935
Published online May 7, 2021. doi: 10.3748/wjg.v27.i17.1920
Artificial intelligence applications in inflammatory bowel disease: Emerging technologies and future directions
John Gubatan, Steven Levitte, Akshar Patel, Tatiana Balabanis, Mike T Wei, Sidhartha R Sinha, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Redwood City, CA 94063, United States
ORCID number: John Gubatan (0000-0001-6037-2883); Steven Levitte (0000-0002-3324-5464); Akshar Patel (0000-0002-2524-6061); Tatiana Balabanis (0000-0002-9475-6989); Mike T Wei (0000-0003-4756-9010); Sidhartha R Sinha (0000-0001-5104-6410).
Author contributions: Gubatan J organized and led the literature review; Levitte S, Balabanis T and Patel A performed the primary literature and data extraction; Gubatan J reviewed literature search results and extracted data for inclusion; Gubatan J drafted the manuscript; Wei MT and Sinha SR provided critical review of the manuscript; all authors interpreted the results and contributed to critical review of the manuscript; Gubatan J had full access to the study data and takes responsibility for the integrity of the data and accuracy of the analysis.
Supported by Chan Zuckerberg Biohub Physician Scientist Scholar Award; and National Institutes of Health NIDDK Loan Repayment Program Award, No. GTQR5718.
Conflict-of-interest statement: The authors have no conflicts of interests or financial disclosures relevant to this manuscript.
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: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: John Gubatan, MD, Academic Research, Consultant Physician-Scientist, Postdoctoral Fellow, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, 420 Broadway Street Pavilion D, 2nd Floor, Redwood City, CA 94063, United States. jgubatan@stanford.edu
Received: January 26, 2021
Peer-review started: January 26, 2021
First decision: February 27, 2021
Revised: March 4, 2021
Accepted: April 13, 2021
Article in press: April 13, 2021
Published online: May 7, 2021

Abstract

Inflammatory bowel disease (IBD) is a complex and multifaceted disorder of the gastrointestinal tract that is increasing in incidence worldwide and associated with significant morbidity. The rapid accumulation of large datasets from electronic health records, high-definition multi-omics (including genomics, proteomics, transcriptomics, and metagenomics), and imaging modalities (endoscopy and endomicroscopy) have provided powerful tools to unravel novel mechanistic insights and help address unmet clinical needs in IBD. Although the application of artificial intelligence (AI) methods has facilitated the analysis, integration, and interpretation of large datasets in IBD, significant heterogeneity in AI methods, datasets, and clinical outcomes and the need for unbiased prospective validations studies are current barriers to incorporation of AI into clinical practice. The purpose of this review is to summarize the most recent advances in the application of AI and machine learning technologies in the diagnosis and risk prediction, assessment of disease severity, and prediction of clinical outcomes in patients with IBD.

Key Words: Artificial intelligence, Machine learning, Inflammatory bowel disease, Crohn’s disease, Ulcerative colitis, Clinical outcomes

Core Tip: The application of artificial intelligence (AI) in the field of inflammatory bowel disease (IBD) has grown significantly in the past decade. AI has been used to analyze genomic datasets, construct IBD risk prediction models, and increase IBD diagnosis precision. Machine learning has been used to analyze endoscopic images to improve disease severity grading. AI has enabled the integration of large clinical and laboratory datasets with gene expression profiles to predict clinical outcomes such as therapy response. Future studies will need to validate these findings in independent cohorts and determine whether applying these AI-derived prediction models improves clinical outcomes in IBD.



INTRODUCTION

Inflammatory bowel disease (IBD), which includes Crohn’s disease (CD) and ulcerative colitis (UC), is a chronic inflammatory disorder of the gastrointestinal tract. IBD has emerged as a global disease with increasing incidence worldwide and associated with significant healthcare utilization[1,2]. The pathogenesis of IBD is complex and is thought to involve an interplay between loss of tolerance to commensal gut bacteria, intestinal epithelial barrier dysfunction, and immune dysregulation[3-7]. The diagnosis of IBD is based on a combination of factors including clinical data (e.g., chronicity of gastrointestinal symptoms), laboratory values (elevated inflammatory markers such as C-reactive protein and fecal calprotectin), imaging, endoscopy, and histology (gastrointestinal inflammation with architectural distortion)[8]. Although treatment algorithms based on clinical trials and experience have been developed to inform clinical management in IBD[9], there is significant heterogeneity among patients with IBD with regards to presentation, response to therapy, and long-term clinical outcomes such development of strictures and need for surgery[10,11]. There is a great need for precision medicine strategies to improve diagnostic and therapeutic approaches in IBD.

Precision medicine efforts in IBD have led to more in-depth phenotyping of patients with IBD using large scale databases from clinical trials and cohort studies, deep immunophenotyping using whole genome gene expression datasets, proteomics, transcriptomics, and metagenomics of gut microbiota, and complex predictive models incorporating computer-assisted analysis of endoscopic images and histology[12-14]. This has inevitably led to vast arrays of high dimensional data that pose significant challenges with traditional statistical and computational methods[15]. Technological advances in artificial intelligence (AI) have revolutionized the ability of clinicians and researchers to process, analyze, and interpret high dimensional data and large datasets.

AI is a broad and multidisciplinary field incorporating concepts from computer science, engineering, philosophy, and linguistics aimed at understanding and designing systems that display or mimic human intelligence. The term was first coined in 1965 by McCarthy J[16,17]. Machine learning (ML) is a subdiscipline of AI where computer algorithms apply statistical models to learn associations of predictive power from examples in provided datasets (e.g., Dragon dictation, SPAM, Netflix). ML may be programmed through supervised learning or unsupervised learning. In supervising learning, computer programs are trained to learn associations between inputs and outputs in data through analysis of predefined outputs of interest (by human operator). Once associations have been learned using existing data, supervised ML classifiers could then be used to predict future examples using different datasets. Examples of supervised ML include random forest (RF) and support vector machines (SVM). In unsupervised learning, computer programs learn associations in data without external definitions of associations of interest. This method allows for the identification of previously undiscovered predictors. Deep learning, commonly known as neural networks, includes newer techniques that are based on models with fewer assumptions, rely on multiple layers of representation of the data with successive transformations that amplify aspects of the input which improves discrimination power and thus able to handle more complex data (e.g., Facebook face recognition, credit card fraud)[17]. There has been increased interest in use of AI in IBD in recent years with many prior groups applying ML methods to identify meaningful insights in diagnostics and prediction models in IBD. The purpose of this review is to provide a comprehensive summary of advances in the application of AI and ML technologies in the diagnosis and risk prediction, assessment of disease severity, and prediction of clinical outcomes in patients with IBD.

LITERATURE SEARCH

We performed a literature review using PubMed (MEDLINE) from inception to December 15, 2020 of studies applying AI in IBD. Our search strategy included the following combinations: (((((((((inflammatory bowel disease[Title])) OR (ulcerative colitis[Title])) OR (Crohn's disease[Title])) AND (artificial intelligence[Title])) OR (computer-assisted[Title])) OR (computer-aided[Title])) OR (neural network[Title])) OR (machine learning[Title])) OR (deep learning[Title]). We included studies that used AI in the (1) diagnosis or risk prediction of IBD, (2) assessment of disease severity in IBD, and (3) prediction of therapy response and clinical outcomes in IBD. We excluded reviews, studies with non-human subjects (animal models), or studies that did not provide objective measures of the efficacy of AI applications (e.g., measures of precision, accuracy, area under the curve (AUC), sensitivity, specificity, etc.).

RESULTS

Our search strategy yielded 98 studies evaluating AI in IBD of which 58 studies[18-74] met inclusion criteria and were included in the final review. About 86.2% (50/58) of studies were published within the past 5 years (2015 and later). There were 23 studies[18-39] that focused on IBD diagnosis and risk prediction, 19 studies[40-58] which evaluated disease activity, and 17 studies[45,59-74] which predicted IBD clinical outcomes (response to therapy, colonic neoplasia, post-surgical complications, quality of life, IBD well-being and emotional content). There were 22 studies with combined IBD cohorts (CD and UC), 16 studies with UC patients only, 18 studies with CD only, and 5 pediatric IBD cohorts. The most common AI classifications used were neural networks (convolutional and deep) at 32.7% (19/58 studies), RF at 29.3% (17/58 studies), and SVM at 29.3% (17/58 studies).

AI in diagnosis and risk prediction of IBD

Table 1 summarizes studies included which applied AI in the diagnosis and risk prediction of IBD. There were 17 studies focused on IBD diagnosis, whereas 5 studies focused on predicting risk of IBD. Data modalities included genetic/genomic datasets (n = 16 studies), imaging and endoscopic datasets (n = 4), and protein expression/ proteomics (n = 2 studies). Some groups have used ML to develop IBD risk prediction models based on gene expression datasets. In a cross-sectional study of 180 CD patients, 149 UC patients and 90 healthy controls by Isakov et al[21], RF and SVM used microarray and RNA-seq data sets to classify a list of 16390 genes. Their combined IBD risk prediction model demonstrated an AUC, sensitivity, specificity, and accuracy values of 0.829, 0.577, 0.880, and 0.808, respectively. In another cross-sectional study of 18227 CD patients and 34050 healthy controls, Romagnoni et al[20] used gradient boosted trees and artificial neural networks to analyze gene expression profiles. Using single nucleotide polymorphisms, their final predictive model for CD achieved AUC of 0.80. Likewise, a cross-sectional study of 20 UC patients and 20 healthy controls by Duttagupta et al[33] used SVM to analyze microRNA profiles. Their SVM classifier measurements revealed a predictive score accuracy of 92.8%, specificity of 96.2%, and sensitivity of 89.5% in distinguishing UC patients from normal individuals.

Table 1 Artificial intelligence in diagnosis and risk prediction of inflammatory bowel disease.
Ref.
AI classifier vs comparator
IBD type
Study design and sample size
Modality
Outcome
Study results/validation cohort
Mossotto et al[18], 2017Support vector machines (SVM) vs linear discriminantPeds CD/UCProspective cohort, 287 IBD patientsEndoscopic and histologic inflammationDiagnosis of IBDDiagnostic accuracy of 82.7% with an AUC of 0.87 in diagnosing Crohn's disease or ulcerative colitis. Validation cohort included
Wei et al[19], 2013SVM with gradient boosted trees (GBT) vs simple log odds methodCD/UCCross-sectional, 30000 IBD patients, 22000 healthy controlsGenetics, ImmunoChipRisk of IBDThe SVM demonstrated very comparable performance (AUC 0.862 and 0.826 for CD and UC, respectively), whereas GBT showed inferior performance (AUC 0.802 and0.782 for CD and UC, respectively. Validation cohort included
Romagnoni et al[20], 2019Artificial neural networks (ANNs) vs penalized logistic regression (LR), and GBTCD Cross-sectional, 18227 CD patients, 34050 healthy controlsGenetics, ImmunoChipRisk of IBDUsing single nucleotide polymorphisms (SNPs), final predictive model achieved AUC of 0.80. Validation cohort included
Isakov et al[21], 2017Random forest (RF), SVM with svmPoly), extreme gradient boosting vs elastic net regularized generalized linear model (glmnet)CD/UCCross-sectional, 180 CD patients, 149 UC patients, 90 healthy controlsExpression data (microarray and RNA-seq)Risk of IBDThe method was used to classify a list of 16390 genes. Each gene received a score that was used to prioritize it according to its predicted association to IBD. The combined model demonstrated AUC, sensitivity, specificity, and accuracy values of 0.829, 0.577, 0.88, and 0.808, respectively. Validation cohort included
Yuan et al[22], 2017Sequential minimal optimization vs DisGeNET (Version 4.0)CD/UCCross-sectional, 59 CD patients, 26 UC patients, 42 healthy controlsGene Expression datasetsRisk of IBDBy analyzing the gene expression profiles using minimum redundancy maximum relevance and incremental feature selection, 21 genes were obtained that could effectively distinguish samples from IBD and the non-IBD samples. Highest total prediction accuracy was 97.64% using the 1170th feature set. Validation cohort included
Hübenthal et al[23], 2015SVM vs RFCD/UCCross-sectional, 40 CD patients, 36 UC patients, 38 healthy controlsMicroRNAsDiagnosis of IBDMeasured by the AUC the corresponding median holdout-validated accuracy was estimated as ranging from 0.75 to 1.00 and 0.89 to 0.98, respectively. In combination, the corresponding models provide tools for the distinction of CD and UC as well as CD, UC and healthy control with expected classification error rates of 3.1 and 3.3%, respectively. Validation cohort included
Tong et al[24], 2020RF vs convolutional neural network (CNN)CD/UCRetrospective Cohort, 875 CD patients, 5128 UC patientsColonoscopy Endoscopic ImagesDiagnosis of IBDRF sensitivities/specificities of UC/CD were 0.89/0.84, 0.83/0.82, and 0.72/0.77, respectively, while the values for the CNN of CD was 0.90/0.77. The precisions/recalls of UC-CD when employing RF were 0.97/0.97, 0.65/0.53, respectively, and when employing the CNN were 0.99/0.97 and 0.87/0.83, respectively. Validation cohort included
Smolander et al[25], 2019Deep belief networks (DBNs) vs SVMCD/UCCross-sectional, 59 CD patients, 26 UC patients, 42 healthy controlsGene Expression datasetsDiagnosis of IBDUsing DBN only, accuracy for diagnosis of UC was 97.06% and CD was 97.07%. Using both DBN and SVM, accuracy for diagnosis of UC was 97.06% and CD was 97.03%. Validation cohort included
Abbas et al[26], 2019RF vs network-based biomarker discoveryPeds CD/UCCross-sectional, 657 IBD patients, 316 healthy controlsLarge dataset of new-onset pediatric IBD metagenomics biopsy samplesDiagnosis of IBDFor the diagnosis of IBD, highest AUC attained by top Random Forest classifiers was 0.77. No validation cohort included
Khorasani et al[27], 2020SVM vs recently developed feature selection algorithm (robustness-performance tradeoff, RPT)UCCross-sectional, 146 UC patients, 60 healthy controlsGene Expression datasetDiagnosis of IBDOur model perfectly detected all active cases and had an average precision of 0.62 in the inactive cases. Validation cohort included
Rubin et al[28], 2019CITRUS supervised machine learning algorithm. No comparatorCD/UCCross-sectional, 68 IBD patientsPeripheral blood mononuclear cells and intestinal biopsies mass cytometryDiagnosis of IBDAn 8-parameter immune signature distinguished Crohn's disease from ulcerative colitis with an AUC = 0.845 (95%CI: 0.742-0.948). No validation cohort included
Pal et al[29], 2017Naïve Bayes and with a consensus machine learning method vs Critical Assessment of Genome Interpretation (CAGI) 4 methodCDCross-sectional, 64 CD patients, 47 healthy controlsGenotypes from Exome Sequencing DataRisk of IBDThe AUC for predicting risk of Crohn's disease using the SNP model was 0.72. No validation cohort included
Aoki et al[30], 2019Deep CNN. No comparatorCDRetrospective Cohort, 115 IBD patientsWireless capsule endoscopy imagesDiagnosis of IBDThe AUC for the detection of erosions and ulcerations was 0.958 (95%CI: 0.947-0.968). The sensitivity, specificity, and accuracy of the CNN were 88.2% (95%CI: 84.8-91.0), 90.9% (95%CI: 90.3-91.4), and 90.8% (95%CI: 90.2-91.3), respectively. Validation cohort included
Bielecki et al[31], 2012SVM vs human reader (pathologist)CD/UCCross-sectional, 14 CD patients, 13 UC patients, 11 healthy controlsRaman spectroscopic imaging of epithelium cellsDiagnosis of IBDRaman maps of human colon tissue sections were analyzed by utilizing innovative chemometric approaches. Using SVM, it was possible to separate between healthy control patients, patients with Crohn's Disease, and patients with ulcerative colitis with an accuracy of 98.90%. No validation cohort included
Cui et al[32], 2013Recursive SVM vs unsupervised learning strategyCD/UCCross-sectional, 124 IBD patients, 99 healthy controls16S rRNA gene analysisDiagnosis of IBDSelection level of 200 features results in the best leave-one-out cross-validation result (accuracy = 88%, sensitivity = 92%, specificity = 84%). Validation cohort included
Duttagupta et al[33], 2012SVM. No comparatorUCCross-sectional, 20 UC patients, 20 healthy controlsMicroRNAsDiagnosis of IBDSVM classifier measurements revealed a predictive score of 92.8% accuracy, 96.2% specificity and 89.5% sensitivity in distinguishing ulcerative colitis patients from normal individuals. Validation cohort included
Daneshjou et al[34], 2017Naïve bayes, neural networks, random forests vs CAGI methodsCDCross-sectional, 64 ICD patients, 47 healthy controlsExome SequencingDiagnosis of IBDIn CAGI4, 111 exomes were derived from a mix of 64 Crohn’s disease patients. Top performing methods had an AUC of 0.87. Validation cohort included
Geurts et al[35], 2005RF vs SVMCD/UCProspective cohort, 30 CD patients, 30 CD patientsProteomic Mass SpectrometryDiagnosis of IBDRandom forest model to diagnosis IBD had a sensitivity of 81.67%, specificity of 81.17%. Support vector machine model to diagnosis IBD had a sensitivity of 87.92%, specificity of 87.87%. Validation cohort included
Li et al[36], 2020RF vs ANNUCCross-sectional, 193 UC patients, 21 healthy controlsGene Expression ProfilesDiagnosis of IBDThe random forest algorithm was introduced to determine 1 downregulated and 29 upregulated differentially expressed genes contributing highest to ulcerative colitis occurrence. ANN was developed to calculate differentially expressed genes weights to ulcerative colitis. Prediction results agreed with that of an independent data set (AUC = 0.9506/PR-AUC = 0.9747). Validation cohort included
Wingfield et al[37], 2019RF vs SVMCDCross-sectional, 668 CD patientsMetagenomic DataDiagnosis of IBDHighest RPT measure for Crohn’s disease was random forest 0.60 and SVM 0.58. For ulcerative colitis, RPT was random forest 0.70 and SVM 0.48. Validation cohort included
Han et al[38], 2018RF vs LR, CORGCD/UCCross-sectional, 24 CD patients, 59 UC patients, 76 healthy controlsGene Expression ProfilesDiagnosis of IBDThe gene-based feature sets had median AUC on the validation sets ranging from 0.6 to 0.76). Validation cohort included
Wang et al[39], 2019AVADx (Analysis of Variation for Association with Disease) vs two GWAS-based CD evaluation methodsCDCross-sectional, 64 CD patients, 47 healthy controlsWhole Exome or Genome Sequencing DataDiagnosis of IBDAVADx highlighted known CD genes including NOD2and new potential CD genes. AVADx identified 16% (at strict cutoff) of CD patients at 99% precision and 58% of the patients (at default cutoff) with 82% precision in over 3000 individuals from separately sequenced panels. Validation cohort included

A major challenge in IBD diagnosis is the distinction between CD and UC which is based on clinical features such as the distribution of inflammation along the gastrointestinal tract. The misdiagnosis of IBD subtype is not uncommon[74]. Distinguishing between CD and UC is clinically important as IBD subtype informs clinical management. AI has been employed to analyze molecular data to distinguish between CD and UC. In a cross-sectional study of 59 CD patients, 26 UC patients, and 42 healthy controls applying deep belief networks (DBNs) and SVM to gene expression datasets, Smolander et al[25] explored the diagnosis UC from CD. Using DBN only, the accuracy for diagnosis of UC was 97.06% and CD was 97.07%. Using both DBN and SVM, accuracy for diagnosis of UC was 97.06% and CD was 97.03%. In a cross-sectional study of 68 IBD patients using a CITRUS supervised ML algorithm to analyze single cell immunophenotyping of peripheral blood mononuclear cells by mass cytometry, Rubin et al[28] demonstrated that an 8-parameter immune signature distinguished CD from UC with an AUC = 0.845 [95% confidence interval (CI): 0.742-0.948]. ML algorithms have also been applied to analyze large arrays of endoscopic images to differentiate between UC and CD. In a recent retrospective cohort study of 875 CD patients and 5128 UC patients by Tong et al[24] using RF and convolutional neural networks (CNNs) on endoscopic images, the precision of diagnosing UC/CD with RF and CNNs were 0.97/0.65 and 0.99/0.87, respectively. Taken together, these studies suggest that AI classifiers have high performance in diagnosing or predicting risk of IBD but have some variability with type of AI classifier and modality of data (molecular vs endoscopic).

AI in assessment of disease severity in IBD

The assessment of disease activity and grading of severity in IBD could be accomplished using validated clinical symptom scores (e.g., Harvey Bradshaw Index for CD, Mayo Score for UC)[75,76], biomarkers of inflammation (e.g., C-reactive protein, fecal calprotectin)[77,78], endoscopic inflammation indices (e.g., Mayo endoscopic score, simple endoscopic score)[79,80], and histologic scoring systems (e.g., Geboes Score, Robarts Histopathology Index)[81,82]. However, these systems may be subject to recall bias, heterogeneity in patient clinical presentation, and intraobserver and interobserver variability[83]. AI has been applied to these existing systems to improve precision and accuracy of quantifying disease severity in IBD.

Table 2 summarizes studies included which applied AI in the assessment of disease severity in IBD. There were 2 studies that assessed clinical disease activity, 2 studies that assessed disease activity by biomarker (C-reactive protein), 13 studies that focused on endoscopic inflammation, and 3 studies that focused on histologic inflammation. Data modalities included electronic health records (n = 2), molecular datasets (n = 3), endoscopic datasets (n = 11 studies), and histologic datasets via endomicroscopy/ endocytoscopy (n = 2). Using RF to integrate and analyze clinical and laboratory data from publicly available clinical trials (UNITI-1, UNITI-2, and IM-UNITI) data consisting of 401 CD patients, Waljee et al[42] constructed a CD remission prediction model using the week 6 albumin to C-reactive protein ratio with an AUC of 0.76 (95%CI: 0.71-0.82). Reddy et al[44] applied gradient boosting machines to electronic health records and predicted inflammation severity in a retrospective cohort of 3335 CD patients with a very high accuracy (AUC) = 92.82%. In a CNN analysis of colonoscopy images from a retrospective cohort of 841 UC patients by Ozawa et al[55], the CNN-based computer aided diagnostic system showed a high level of performance with AUC of 0.86 and 0.98 to identify Mayo 0 and 0-1, respectively. The performance of the CNN was better for the rectum than for the right side and left side of the colon when identifying Mayo 0 (AUC = 0.92, 0.83, and 0.83, respectively). Likewise, in an ordinal CNN analysis of wireless capsule endoscopy images in a retrospective cohort of 49 CD patients by Barash et al[50], the classification accuracy of the algorithm was 0.91 for grade 1 vs grade 3 ulcers, 0.78 for grade 2 vs grade 3, and 0.624 for grade 1 vs grade 2. The role of AI in grading severity of histologic inflammation in IBD has also been explored. For example, in a retrospective cohort study of 187 UC patients by Maeda et al[46], application of SVM to data derived from endocytoscopy to assess histologic inflammation provided diagnostic sensitivity, specificity, and accuracy of 74% (95%CI: 65-81), 97% (95%CI: 95-99), and 91% (95%CI: 83-95), respectively. These examples highlight the clinical utility, versatility, and performance of AI classifiers in grading the disease activity of IBD patients at the clinical, endoscopic, and histologic level. AI performance may be affected by location of inflammation and may be limited by ability to discriminate between subtle differences.

Table 2 Artificial Intelligence in assessment of disease severity in inflammatory bowel disease.
Ref.
AI classifier vs comparator
IBD type
Study design and sample size
Modality
Outcomes
Study results/validation cohort
Kumar et al[40], 2012Support vector machines (SVM) vs human observersCDCross-sectional, 50000 images (number of patients not given)Small bowel capsule endoscopyEndoscopic InflammationDatabase of 47 studies including 50000 capsule endoscopy images evaluating severity of small bowel lesions. Method had good precision (> 90% for lesion detection) and recall (> 90%) for lesions of varying severity. Validation cohort included
Biasci et al[41], 2019Logistic regression with an adaptive Elastic-Net penalty. No comparatorCD/UCProspective cohort, 118 IBD patientsTranscriptomics from purified CD8 T cells and/or whole bloodDisease severity, medication escalationA 17-gene qPCR-based classifier stratified patients into two distinct subgroups. IBDhi patients experienced significantly more aggressive disease than IBDlo patients (analogous to IBD2), with earlier need for treatment escalation [HR 2.65 (CD), 3.12 (UC)] and more escalations over time [for multiple escalations within 18 months: sensitivity=72.7% (CD), 100% (UC); negative predictive value = 90.9% (CD), 100% (UC)]. Validation cohort included
Waljee et al[42], 2019RF. No comparatorCDPost-hoc analysis of prospective clinical trials, 401 CD patientsClinical and laboratory data from publicly available clinical trials (UNITI-1, UNITI-2, and IM-UNITI)Crohn's disease remission, C-reactive protein < 5 mg/LA prediction model using the week-6 albumin to C-reactive protein ratio had an AUC of 0.76 [95% confidence interval (CI): 0.71-0.82]. Validation cohort included
Mahapatra et al[43], 2016RF. No comparatorCDCross-sectional, 35 CD patientsAbdominal magnetic resonance imagingSegmentation of diseased colon (intestinal inflammation)Model segmentation accuracy ranged from 82.7% to 92.2%. Validation cohort included
Reddy et al[44], 2019Gradient boosting machines vs logistic regressionCDRetrospective, 3335 CD patientsElectronic medical recordSeverity of intestinal inflammation (by C-reactive protein)Machine-learning-based analytic methods such as gradient boosting machines can predict the inflammation severity with a very high accuracy (AUC) = 92.82%. Validation cohort included
Douglas et al[45], 2018RF. No comparatorPeds CDCross-sectional, 20 CD patients, 20 healthy controlsShotgun metagenomics (MGS), 16S rRNA gene sequencingDisease State (Relapse/Remission)MGS modules significantly classified samples by disease state (accuracy = 68.4%, P = 0.043 and accuracy = 65.8%, P = 0.03, respectively), 16S datasets had a maximum accuracy of 68.4% and P = 0.016 based on strain level for disease state. Validation cohort included
Maeda et al[46], 2019SVM vs human readerUCRetrospective cohort, 187 UC patientsEndocytoscopyHistologic inflammationComputer aided diagnosis (CAD) of histologic inflammation provided diagnostic sensitivity, specificity, and accuracy as follows: 74% (95%CI: 65-81), 97% (95%CI: 95-99), and 91% (95%CI: 83-95), respectively. Its reproducibility was perfect (k = 1). Validation cohort included
Charisis et al[47], 2016SVM vs human readerCDRetrospective cohort, 13 CD patientsWireless capsule endoscopy (WCE) imagesEndoscopic InflammationExperimental results, along with comparison with other related efforts, have shown that the hybrid adaptive filtering [HAF-Differential Lacunarity (DLac) analysis (HAF-DLac)] via SVM approach evidently outperforms them in the field of WCE image analysis for automated lesion detection, providing higher classification results, up to 93.8% (accuracy), 95.2% (sensitivity), 92.4% (specificity) and 92.6% (precision). Validation cohort included
Klang et al[48], 2020Convolutional neural network (CNN) vs human readerCDRetrospective cohort, 49 CD patientsWCE imagesEndoscopic InflammationDataset included 17640 CE images from 49 patients: 7391 images with mucosal ulcers and 10249 images of normal mucosa. For randomly split images results, AUC was 0.99 with accuracies ranging from 95.4% to 96.7%. For individual patient-level experiments, the AUCs were 0.94-0.99. Validation cohort included
Ungaro et al[49], 2021Random survival forest. No comparatorPeds CDRetrospective case-control, 265 peds CD patientsProtein biomarkers using a proximity extension assay (Olink Proteomics)Penetrating and stricturing complicationsA model with 5 protein markers predicted penetrating complications with an AUC of 0.79 (95%CI: 0.76-0.82) compared to 0.69 (95%CI: 0.66-0.72) for serologies and 0.74 (95%CI: 0.71-0.77) for clinical variables. A model with 4 protein markers predicted structuring complications with an AUC of 0.68 (95%CI: 0.65-0.71) compared to 0.62 (95%CI: 0.59-0.65) for serologies and 0.52 (95%CI: 0.50-0.55) for clinical variables. Validation cohort included
Barash et al[50], 2021Ordinal CNN. No comparatorCDRetrospective cohort, 49 CD patientsWCE imagesUlcer Severity GradingThe classification accuracy of the algorithm was 0.91 (95%CI: 0.867-0.954) for grade 1 vs grade 3 ulcers, 0.78 (95%CI: 0.716-0.844) for grade 2 vs grade 3, and 0.624 (95%CI: 0.547-0.701) for grade 1 vs grade 2. Validation cohort included
Lamash et al[51], 2019CNN vs semi-supervised and active learning modelsCDRetrospective cohort, 23 CD patientsMagnetic resonance imagingActive Crohn’s DiseaseCNN exhibited Dice similarity coefficient of 75% ± 18%, 81% ± 8%, and 97% ± 2% for the lumen, wall, and background, respectively. The extracted markers of wall thickness at the location of min radius (P = 0.0013) and the median value of relative contrast enhancement (P = 0.0033) could differentiate active and nonactive disease segments. Other extracted markers could differentiate between segments with strictures and segments without strictures (P < 0.05). Validation cohort included
Takenaka et al[52], 2020Deep neural networks vs human reader (endoscopist)UCProspective cohort, 2012 UC patientsColonoscopy imagesEndoscopic inflammationDeep neural network identified patients with endoscopic remission with 90.1% accuracy (95%CI: 89.2-90.9) and a kappa coefficient of 0.798 (95%CI: 0.780-0.814), using findings reported by endoscopists as the reference standard. Validation cohort included
Bossuyt et al[53], 2020Computer algorithm based on red density (RD) vs blinded central readersUCProspective cohort, 29 UC patients, 6 healthy controlsColonoscopy ImagesEndoscopic and histologic inflammationIn the construction cohort, RD correlated with rhi (r = 0.74, P < 0.0001), Mayo endoscopic subscores (r = 0.76, P < 0.0001) and Endoscopic index of severity scores (r = 0.74, P < 0.0001). The RD sensitivity to change had a standardized effect size of 1.16. in the validation set, RD correlated with rhi (r = 0.65, P = 0.00002). Validation cohort included
Bhambhvani et al[54], 2021CNN vs human reader (endoscopist)UCRetrospective cohort, 777 UC patientsColonoscopy imagesMayo Endoscopic Scores (MES)The final model classified MES 3 disease with an AUC of 0.96, MES 2 disease with an AUC of 0.86, and MES 1 disease with an AUC 0.89. Overall accuracy was 77.2%. Across MES 1, 2, and 3, average specificity was 85.7%, average sensitivity was 72.4%, average PPV was 77.7%, and the average NPV was 87.0%. Validation cohort included
Ozawa et al[55], 2019CNN vs human reader (endoscopist)UCRetrospective cohort, 841 UC patientsColonoscopy imagesMESThe CNN-based CAD system showed a high level of performance with AUC of 0.86 and 0.98 to identify Mayo 0 and 0-1, respectively. The performance of the CNN was better for the rectum than for the right side and left side of the colon when identifying Mayo 0 (AUC = 0.92, 0.83, and 0.83, respectively). Validation cohort included
Bossuyt et al[56], 2021Automated CAD Algorithm vs human readerUCProspective cohort, 48 UC patientsColonoscopy images with confocal laser endomicroscopyHistologic RemissionThe current automated CAD algorithm detects histologic remission with a high performance (sensitivity of 0.79 and specificity of 0.90) compared with the UCEIS (sensitivity of 0.95 and specificity of 0.69) and MES (sensitivity of 0.98 and specificity of 0.61). No validation cohort included
Stidham et al[57], 2019CNN vs human readerUCRetrospective cohort, 3082 UC patientsColonoscopy imagesEndoscopy severityThe CNN was excellent for distinguishing endoscopic remission from moderate-to-severe disease with an AUC of 0.966 (95%CI: 0.967-0.972); a PPV of 0.87 (95%CI: 0.85-0.88) with a sensitivity of 83.0% (95%CI: 80.8-85.4) and specificity of96.0% (95%CI: 95.1-97.1); and NPV of 0.94 (95%CI: 0.93-0.95). No validation cohort included
Gottlieb et al[58], 2021Neural network vs human central readerUCProspective cohort, 249 UC patientsColonoscopy imagesEndoscopy severityThe model's agreement metric was excellent, with a quadratic weighted kappa of 0.844 (95%CI: 0.787-0.901) for endoscopic Mayo Score and 0.855 (95%CI: 0.80-0.91) for UCEIS. No validation cohort included
AI in prediction of therapy response and clinical outcomes in IBD

The armamentarium of therapies in IBD have expanded significantly in recent years with diverse mechanisms of action ranging from biologics that inhibit proinflammatory cytokines (anti-tumor necrosis factor-α, anti-interleukin-12/23) and leukocyte trafficking to the gut (anti-α4β7) to small molecule inhibitors of the JAK-STAT signaling pathway[84-86]. Despite several IBD treatment options available to clinicians, there are no effective biomarkers or tools to predict response to therapy or to guide selection of alternative therapies after a failed response. Likewise, there is also an unmet clinical need to predict long term clinical outcomes in IBD such as colon cancer. To address these challenges, several groups have applied AI and ML algorithms to existing clinical and molecular datasets.

Table 3 summarizes studies included which applied AI in the prediction of therapy response and clinical outcomes in IBD. There were 9 studies that predicted therapy response, 2 studies that predicted presence of extraintestinal manifestations of IBD, 1 study predicting colonic neoplasia, and 1 study predicting post-surgical complications after colectomy. Data modalities included electronic health records (n = 11), molecular datasets (n = 4), and histologic data (n = 1). Waljee et al[59,65] and Popa et al[62] have previously applied RF classifiers to clinical data from electronic health records and laboratory values to predict response to various IBD therapies. In one study using data from a prospective clinical trial consisting of 594 CD patients[59], the AUC for a RF longitudinal model for predicting inpatient hospitalizations in IBD patients prescribed outpatient corticosteroids was 0.85 (95%CI: 0.84-0.85). Using a similar RF approach for predicting remission with thiopurine therapy in a prospective cohort of 55 UC patients yielded an AUC of 0.79[62]. Applying RF to data from a retrospective cohort of 20368 IBD patients with vedolizumab use yielded an AUC of 0.65 (95%CI: 0.53-0.77) for corticosteroid-free vedolizumab remission at week 52 using baseline data and an AUC of 0.75 (95%CI: 0.64-0.86) with data through week 6 of vedolizumab[65]. Molecular datasets have also been used to differentiate between responders and non-responders to various IBD therapies. For example, Morilla et al[66] used a deep neural network classifier to construct a predictive panel of colonic microRNAs for IBD therapies in a retrospective cohort of 47 UC patients. Their panel discriminated responders to steroids from non-responders with 93% accuracy (AUC, 0.91). In addition, three algorithms, based on microRNA levels, identified responders to infliximab vs non-responders (84% accuracy, AUC 0.82) and responders to cyclosporine vs non-responders (80% accuracy, AUC 0.79). A more recent prospective cohort study of 55 UC patients by Popa et al[62] integrated clinical, laboratory, and endoscopic (Mayo scores) datasets using a neural network classifier to predict disease activity after one year of anti-tumor necrosis factor therapy in patients with UC. This classifier achieved an AUC of 0.92 for predicting the disease activity at one year on the test set and an AUC of 1.00 on the validation set. These studies suggest that AI classifiers may play a role in predicting clinical outcomes and response to specific therapies in patients with IBD. However, future clinical trials are needed to compare the efficacy of AI applications in IBD clinical management vs standard of care before incorporation into real life clinical practice.

Table 3 Artificial intelligence in prediction of therapy response and clinical outcomes in inflammatory bowel disease.
Ref.
AI classifier vs comparator
IBD type
Study design and sample size
Modality
Outcomes
Study results/validation cohort
Waljee et al[59], 2018Random forest (RF). No comparatorCD/UCPost-hoc analysis of prospective clinical trial, 594 CD patientsVeteran’s Health Administration Electronic Health Record (EHR)Outpatient corticosteroids prescribed for IBD and inpatient hospitalizations associated with a diagnosis of IBDAUC for the RF longitudinal model was 0.85 [95% confidence interval (CI): 0.84–0.85]. AUC for the RF longitudinal model using previous hospitalization or steroid use was 0.87 (95%CI: 0.87-0.88). Validation cohort included
Uttam et al[60], 2019Support vector machines (SVM) vs nanoscale nuclear architecture mapping (NanoNAM)CD/UCProspective cohort, 103 IBD patients3-dimensional NanoNAM of normal-appearing rectal biopsiesColonic neoplasiaNanoNAM detects colonic neoplasia with an AUC of 0.87 ± 0.04, sensitivity of 0.81 ± 0.09, and specificity of 0.82 ± 0.07 in the independent validation set. Validation cohort included
Waljee et al[61], 2017RF. No comparatorCD/UCRetrospective cohort, 1080 IBD patientsEHR, lab valuesRemission and clinical outcomes with thiopurinesAUC for algorithm-predicted remission in the validation set was 0.79 vs 0.49 for 6-TGN. The mean number of clinical events per year in patients with sustained algorithm-predicted remission (APR) was 1.08 vs 3.95 in those that did not have sustained APR (P < 1 × 10-5). Validation cohort included
Popa et al[62], 2020Neural network model. No comparatorUCProspective cohort, 55 UC patientsClinical and biological parameters and the endoscopic Mayo scoreDisease activity after one year of anti-TNF treatmentThe classifier achieved an excellent performance predicting the disease activity at one year with an accuracy of 90% and AUC 0.92 on the test set and an accuracy of 100% and an AUC of 1 on the validation set. Validation cohort included
Douglas et al[45], 2018RF. No comparatorPeds CDCross-sectional, 20 CD patients, 20 healthy controlsShotgun metagenomics (MGS), 16S rRNA gene sequencingResponse to induction therapy16S genera were again the top dataset (accuracy = 77.8%; P = 0.008) for predicting response to therapy. MGS strain (P = 0.029), genus (P = 0.013), and KEGG pathway (P = 0.018) datasets could also classify patients according to therapy response with accuracy = 72.2% for all three. Validation cohort included
Waljee et al[63], 2010RF vs boosted trees, RuleFitCD/UCCross-sectional, 774 IBD patientsEHR, lab values (thiopurine metabolites)Response to thiopurine therapyA RF algorithm using laboratory values and patient age differentiated clinical response from nonresponse in the model validation data set with an AUC of 0.856 (95%CI: 0.793-0.919). Validation cohort included
Menti et al[64], 2016Naïve bayes vs Bayesian additive regression trees vs Bayesian networksCD/UCRetrospective cohort, 152 CD patientsGenomic DNA, genetic polymorphismPresence of extra-intestinal manifestations in IBD patientsBayesian networks achieved accuracy of 82% when considering only clinical factors and 89% when considering also genetic information, outperforming the other techniques. Validation cohort included
Waljee et al[65], 2017RF vs baseline regression modelCD/UCRetrospective cohort, 20368 IBD patientsEHR, lab valuesCorticosteroid-free biologic remission with vedolizumabThe AUC for corticosteroid-free biologic remission at week 52 using baseline data was only 0.65 (95%CI: 0.53-0.77), but was 0.75 (95%CI: 0.64-0.86) with data through week 6 of vedolizumab. Validation cohort included
Morilla et al[66], 2019Deep neural networks. No comparatorUCRetrospective cohort, 47 UC patientsColonic microrna profilesResponses to therapyA deep neural network-based classifier identified 9 microRNAs plus 5 clinical factors, routinely recorded at time of hospital admission, that were associated with responses of patients to treatment. This panel discriminated responders to steroids from non-responders with 93% accuracy (AUC, 0.91). Three algorithms, based on microRNA levels, identified responders to infliximab vs non-responders (84% accuracy, AUC 0.82) and responders to cyclosporine vs non-responders (80% accuracy, AUC 0.79). Validation cohort included
Wang et al[67], 2020Back-propagation neural network (BPNN), SVM vs logistic regressionCD Cross-sectional, 446 CD patientsEHRMedication nonadherence to maintenance therapyThe average classification accuracy and AUC of the three models were 85.9% and 0.912 for BPNN, and 87.7% and 0.930 for SVM, respectively. Validation cohort included
Bottigliengo et al[68], 2019Bayesian machine learning techniques (BMLTs) vs logistic regressionCD/UCRetrospective cohort, 142 IBD patientsEHR, genetic polymorphismsPresence of extra-intestinal manifestations in IBD patientsBMLTs had an AUC of 0.50 for classifying the presence of extra-intestinal manifestations. Validation cohort included
Ghoshal et al[69], 2020Nonlinear artificial neural network (ANN) vs multivariate linear PCAUCProspective cohort, 263 UC patientsEHRResponses to therapyThe multilayer perceptron neural network was trained by back-propagation algorithm (10 networks retained out of 16 tested). The classification accuracy rate was 73% in correctly classifying response to medical treatment in UC patients. No validation cohort included
Sofo et al[70], 2020SVM leave-one-out cross-validation. No comparatorUCRetrospective cohort, 32 UC patientsEHRPost-surgical complications after colectomyEvaluating only preoperative features, machine learning algorithms were able to predict minor postoperative complications with a high strike rate (84.3%), high sensitivity (87.5%) and high specificity (83.3%) during the testing phase. Validation cohort included
Kang et al[71], 2017ANN vs logistic regressionUCCross-sectional, 24 UC patientsGene expression profilesResponse to anti-TNFBalanced accuracy in cross validation test for predicting response to anti-TNF therapy in ulcerative colitis patient was 82%. Validation cohort included
Babic et al[72], 1997CART vs back propagation neural network (BPNN)CD/UCCross-sectional, 200 IBD patientsEHRQuality of lifeBest reached classification accuracy did not exceed 80% in any case. Other classifiers namely, K-nearest-neighbor, learning vector quantization and BPNN confirmed that outcome. Validation cohort included
Dong et al[73], 2019RF, SVM, ANN vs logistic regressionCDRetrospective cohort, 239 CD patientsEHR, laboratory testsCrohn's related surgeryThe results revealed that RF predictive model performed better than LR model in terms of accuracy (93.11% vs 91.15%), precision (53.42% vs 44.81%), F1 score (0.6016 vs 0.5763), TN rate (95.08% vs 92.00%), and the AUC (0.8926 vs 0.8809). The AUCs were excellent at 0.9864 in RF,0.9538 in LR, 0.8809 in DT, 0.9497 in SVM, and 0.9059 in ANN, respectively. Validation cohort included
Lerrigo et al[74], 2019Latent Dirichlet allocation, unsupervised machine learning algorithm. No comparatorCD/UCRetrospective cohort, 28623 IBD patientsOnline posts from the Crohn’s and colitis foundation community forumImpact of online community forums on well-being and their emotional content10702 (20.8%) posts were identified expressing: gratitude (40%), anxiety/fear (20.8%), empathy (18.2%), anger/frustration (13.4%), hope (13.2%), happiness (10.0%), sadness/depression (5.8%), shame/guilt (2.5%), and/or loneliness (2.5%). A common subtheme was the importance of fostering social support. No validation cohort included

Finally, AI algorithms have been previously applied to enhance the detection of colonic polyps[87] and distinguish among subtypes of neoplastic colorectal lesions[88] in the general population. Although patients with IBD who have extensive colitis have a significantly greater risk of colorectal cancer compared to the general population[89,90], there have been limited studies applying AI technologies to improve colorectal cancer surveillance or develop prediction risk models in patients with IBD. Most studies evaluating polyp detection have excluded IBD patients[91-93]. Our literature search yielded only one study applying AI for the detection of colonic neoplasia in IBD. Uttam et al[60] employed support SVM to analyze 3-dimensional nanoscale nuclear architecture mapping (NanoNAM) of normal-appearing rectal biopsies in a prospective cohort of 103 IBD patients. In their study, NanoNAM detected colonic neoplasia with an AUC of 0.87 ± 0.04, sensitivity of 0.81 ± 0.09, and specificity of 0.82 ± 0.07 in the independent validation set. Further studies should focus on determining the clinical utility of incorporating AI methods to enhance standard of cancer surveillance in patients with IBD such as chromoendoscopy[94] and to develop predictive models for risks of colorectal malignancy in IBD patient populations.

CONCLUSION

In conclusion, our literature review has revealed that the applications of AI in IBD have significantly increased in recent years. Our review also highlighted that various AI classifiers may be applied to analyze and integrate large datasets ranging from clinical data from electronic health records, molecular data including gene expression and protein-based studies to a wide array of datasets consisting of endoscopic and histologic images. The application of AI has the potential to improve the accuracy and precision of predicting risk and diagnosis of IBD, assessing disease severity, and predicting outcomes with various IBD therapies. Currently, the application of AI methods in IBD has been limited to the research setting and has not yet been adopted in real life clinical practice. Furthermore, studies applying AI in the context of colorectal cancer surveillance or prediction in IBD are much needed. Given the current status of the field of AI in IBD, future directions should include: (1) Prospective validation of AI applications in IBD in independent cohorts as there is a risk of bias from internal training cohorts and potential limitations with generalizability; (2) Standardization of AI methods and comparative studies evaluating effect of heterogeneity from using different types of datasets on outcomes of interest; (3) Randomized controlled trials to determine whether application of AI in the clinical management of IBD improves clinical outcomes and could be translated into clinical practice; and (4) Randomized controlled trials to determine whether application of AI leads to greater clinical efficacy and cost-effectiveness compared to standard of care in IBD.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Rath T, Schmidt PT S-Editor: Gao CC L-Editor: A P-Editor: Ma YJ

References
1.  Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, Panaccione R, Ghosh S, Wu JCY, Chan FKL, Sung JJY, Kaplan GG. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2017;390:2769-2778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2677]  [Cited by in F6Publishing: 3169]  [Article Influence: 452.7]  [Reference Citation Analysis (0)]
2.  Click B, Ramos Rivers C, Koutroubakis IE, Babichenko D, Anderson AM, Hashash JG, Dunn MA, Schwartz M, Swoger J, Baidoo L, Barrie A 3rd, Regueiro M, Binion DG. Demographic and Clinical Predictors of High Healthcare Use in Patients with Inflammatory Bowel Disease. Inflamm Bowel Dis. 2016;22:1442-1449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
3.  Ananthakrishnan AN, Bernstein CN, Iliopoulos D, Macpherson A, Neurath MF, Ali RAR, Vavricka SR, Fiocchi C. Environmental triggers in IBD: a review of progress and evidence. Nat Rev Gastroenterol Hepatol. 2018;15:39-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 374]  [Cited by in F6Publishing: 485]  [Article Influence: 80.8]  [Reference Citation Analysis (0)]
4.  de Souza HSP, Fiocchi C, Iliopoulos D. The IBD interactome: an integrated view of aetiology, pathogenesis and therapy. Nat Rev Gastroenterol Hepatol. 2017;14:739-749.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 258]  [Article Influence: 36.9]  [Reference Citation Analysis (0)]
5.  Shouval DS, Rufo PA. The Role of Environmental Factors in the Pathogenesis of Inflammatory Bowel Diseases: A Review. JAMA Pediatr. 2017;171:999-1005.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 102]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
6.  van der Sloot KWJ, Amini M, Peters V, Dijkstra G, Alizadeh BZ. Inflammatory Bowel Diseases: Review of Known Environmental Protective and Risk Factors Involved. Inflamm Bowel Dis. 2017;23:1499-1509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 60]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
7.  Danese S, Sans M, Fiocchi C. Inflammatory bowel disease: the role of environmental factors. Autoimmun Rev. 2004;3:394-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 258]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
8.  Tontini GE, Vecchi M, Pastorelli L, Neurath MF, Neumann H. Differential diagnosis in inflammatory bowel disease colitis: state of the art and future perspectives. World J Gastroenterol. 2015;21:21-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 120]  [Cited by in F6Publishing: 120]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
9.  Panaccione R, Rutgeerts P, Sandborn WJ, Feagan B, Schreiber S, Ghosh S. Review article: treatment algorithms to maximize remission and minimize corticosteroid dependence in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2008;28:674-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 46]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
10.  Bettenworth D, Lopez R, Hindryckx P, Levesque BG, Rieder F. Heterogeneity in endoscopic treatment of Crohn's disease-associated strictures: An international inflammatory bowel disease specialist survey. J Gastroenterol. 2016;51:939-948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 24]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
11.  Ananthakrishnan AN, Shi HY, Tang W, Law CC, Sung JJ, Chan FK, Ng SC. Systematic Review and Meta-analysis: Phenotype and Clinical Outcomes of Older-onset Inflammatory Bowel Disease. J Crohns Colitis. 2016;10:1224-1236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 95]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
12.  Anderson AJ, Click B, Ramos-Rivers C, Babichenko D, Koutroubakis IE, Hartman DJ, Hashash JG, Schwartz M, Swoger J, Barrie AM 3rd, Dunn MA, Regueiro M, Binion DG. Development of an Inflammatory Bowel Disease Research Registry Derived from Observational Electronic Health Record Data for Comprehensive Clinical Phenotyping. Dig Dis Sci. 2016;61:3236-3245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
13.  Weersma RK, Xavier RJ;  IBD Multi Omics Consortium; Vermeire S, Barrett JC. Multiomics Analyses to Deliver the Most Effective Treatment to Every Patient With Inflammatory Bowel Disease. Gastroenterology. 2018;155:e1-e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
14.  Syed S, Stidham RW. Potential for Standardization and Automation for Pathology and Endoscopy in Inflammatory Bowel Disease. Inflamm Bowel Dis. 2020;26:1490-1497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
15.  Johnstone IM, Titterington DM. Statistical challenges of high-dimensional data. Philos Trans A Math Phys Eng Sci. 2009;367:4237-4253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 122]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
16.  Wang W, Siau K. Artificial intelligence, machine learning, automation, robotics, future of work and future of humanity: a review and research agenda. J Database Manag. 2019;30:61-79.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Panch T, Szolovits P, Atun R. Artificial intelligence, machine learning and health systems. J Glob Health. 2018;8:020303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 133]  [Article Influence: 26.6]  [Reference Citation Analysis (0)]
18.  Mossotto E, Ashton JJ, Coelho T, Beattie RM, MacArthur BD, Ennis S. Classification of Paediatric Inflammatory Bowel Disease using Machine Learning. Sci Rep. 2017;7:2427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 83]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
19.  Wei Z, Wang W, Bradfield J, Li J, Cardinale C, Frackelton E, Kim C, Mentch F, Van Steen K, Visscher PM, Baldassano RN, Hakonarson H;  International IBD Genetics Consortium. Large sample size, wide variant spectrum, and advanced machine-learning technique boost risk prediction for inflammatory bowel disease. Am J Hum Genet. 2013;92:1008-1012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 121]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
20.  Romagnoni A, Jégou S, Van Steen K, Wainrib G, Hugot JP;  International Inflammatory Bowel Disease Genetics Consortium (IIBDGC). Comparative performances of machine learning methods for classifying Crohn Disease patients using genome-wide genotyping data. Sci Rep. 2019;9:10351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
21.  Isakov O, Dotan I, Ben-Shachar S. Machine Learning-Based Gene Prioritization Identifies Novel Candidate Risk Genes for Inflammatory Bowel Disease. Inflamm Bowel Dis. 2017;23:1516-1523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
22.  Yuan F, Zhang YH, Kong XY, Cai YD. Identification of Candidate Genes Related to Inflammatory Bowel Disease Using Minimum Redundancy Maximum Relevance, Incremental Feature Selection, and the Shortest-Path Approach. Biomed Res Int. 2017;2017:5741948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
23.  Hübenthal M, Hemmrich-Stanisak G, Degenhardt F, Szymczak S, Du Z, Elsharawy A, Keller A, Schreiber S, Franke A. Sparse Modeling Reveals miRNA Signatures for Diagnostics of Inflammatory Bowel Disease. PLoS One. 2015;10:e0140155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
24.  Tong Y, Lu K, Yang Y, Li J, Lin Y, Wu D, Yang A, Li Y, Yu S, Qian J. Can natural language processing help differentiate inflammatory intestinal diseases in China? BMC Med Inform Decis Mak. 2020;20:248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
25.  Smolander J, Dehmer M, Emmert-Streib F. Comparing deep belief networks with support vector machines for classifying gene expression data from complex disorders. FEBS Open Bio. 2019;9:1232-1248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
26.  Abbas M, Matta J, Le T, Bensmail H, Obafemi-Ajayi T, Honavar V, El-Manzalawy Y. Biomarker discovery in inflammatory bowel diseases using network-based feature selection. PLoS One. 2019;14:e0225382.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
27.  Khorasani HM, Usefi H, Peña-Castillo L. Detecting ulcerative colitis from colon samples using efficient feature selection and machine learning. Sci Rep. 2020;10:13744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (1)]
28.  Rubin SJS, Bai L, Haileselassie Y, Garay G, Yun C, Becker L, Streett SE, Sinha SR, Habtezion A. Mass cytometry reveals systemic and local immune signatures that distinguish inflammatory bowel diseases. Nat Commun. 2019;10:2686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 59]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
29.  Pal LR, Kundu K, Yin Y, Moult J. CAGI4 Crohn's exome challenge: Marker SNP vs exome variant models for assigning risk of Crohn disease. Hum Mutat. 2017;38:1225-1234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
30.  Aoki T, Yamada A, Aoyama K, Saito H, Tsuboi A, Nakada A, Niikura R, Fujishiro M, Oka S, Ishihara S, Matsuda T, Tanaka S, Koike K, Tada T. Automatic detection of erosions and ulcerations in wireless capsule endoscopy images based on a deep convolutional neural network. Gastrointest Endosc 2019; 89: 357-363. e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 145]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
31.  Bielecki C, Bocklitz TW, Schmitt M, Krafft C, Marquardt C, Gharbi A, Knösel T, Stallmach A, Popp J. Classification of inflammatory bowel diseases by means of Raman spectroscopic imaging of epithelium cells. J Biomed Opt. 2012;17:076030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
32.  Cui H, Zhang X. Alignment-free supervised classification of metagenomes by recursive SVM. BMC Genomics. 2013;14:641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
33.  Duttagupta R, DiRienzo S, Jiang R, Bowers J, Gollub J, Kao J, Kearney K, Rudolph D, Dawany NB, Showe MK, Stamato T, Getts RC, Jones KW. Genome-wide maps of circulating miRNA biomarkers for ulcerative colitis. PLoS One. 2012;7:e31241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 86]  [Article Influence: 7.2]  [Reference Citation Analysis (1)]
34.  Daneshjou R, Wang Y, Bromberg Y, Bovo S, Martelli PL, Babbi G, Lena PD, Casadio R, Edwards M, Gifford D, Jones DT, Sundaram L, Bhat RR, Li X, Pal LR, Kundu K, Yin Y, Moult J, Jiang Y, Pejaver V, Pagel KA, Li B, Mooney SD, Radivojac P, Shah S, Carraro M, Gasparini A, Leonardi E, Giollo M, Ferrari C, Tosatto SCE, Bachar E, Azaria JR, Ofran Y, Unger R, Niroula A, Vihinen M, Chang B, Wang MH, Franke A, Petersen BS, Pirooznia M, Zandi P, McCombie R, Potash JB, Altman RB, Klein TE, Hoskins RA, Repo S, Brenner SE, Morgan AA. Working toward precision medicine: Predicting phenotypes from exomes in the Critical Assessment of Genome Interpretation (CAGI) challenges. Hum Mutat. 2017;38:1182-1192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 33]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
35.  Geurts P, Fillet M, de Seny D, Meuwis MA, Malaise M, Merville MP, Wehenkel L. Proteomic mass spectra classification using decision tree based ensemble methods. Bioinformatics. 2005;21:3138-3145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 99]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
36.  Li H, Lai L, Shen J. Development of a susceptibility gene based novel predictive model for the diagnosis of ulcerative colitis using random forest and artificial neural network. Aging (Albany NY). 2020;12:20471-20482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
37.  Wingfield B, Coleman S, McGinnity TM, Bjourson AJ. Robust Microbial Markers for Non-Invasive Inflammatory Bowel Disease Identification. IEEE/ACM Trans Comput Biol Bioinform. 2019;16:2078-2088.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
38.  Han L, Maciejewski M, Brockel C, Gordon W, Snapper SB, Korzenik JR, Afzelius L, Altman RB. A probabilistic pathway score (PROPS) for classification with applications to inflammatory bowel disease. Bioinformatics. 2018;34:985-993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 19]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
39.  Wang Y, Miller M, Astrakhan Y, Petersen BS, Schreiber S, Franke A, Bromberg Y. Identifying Crohn's disease signal from variome analysis. Genome Med. 2019;11:59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
40.  Kumar R, Zhao Q, Seshamani S, Mullin G, Hager G, Dassopoulos T. Assessment of Crohn's disease lesions in wireless capsule endoscopy images. IEEE Trans Biomed Eng. 2012;59:355-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
41.  Biasci D, Lee JC, Noor NM, Pombal DR, Hou M, Lewis N, Ahmad T, Hart A, Parkes M, McKinney EF, Lyons PA, Smith KGC. A blood-based prognostic biomarker in IBD. Gut. 2019;68:1386-1395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 117]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
42.  Waljee AK, Wallace BI, Cohen-Mekelburg S, Liu Y, Liu B, Sauder K, Stidham RW, Zhu J, Higgins PDR. Development and Validation of Machine Learning Models in Prediction of Remission in Patients With Moderate to Severe Crohn Disease. JAMA Netw Open. 2019;2:e193721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 52]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
43.  Mahapatra D, Vos FM, Buhmann JM. Active learning based segmentation of Crohns disease from abdominal MRI. Comput Methods Programs Biomed. 2016;128:75-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
44.  Reddy BK, Delen D, Agrawal RK. Predicting and explaining inflammation in Crohn's disease patients using predictive analytics methods and electronic medical record data. Health Informatics J. 2019;25:1201-1218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
45.  Douglas GM, Hansen R, Jones CMA, Dunn KA, Comeau AM, Bielawski JP, Tayler R, El-Omar EM, Russell RK, Hold GL, Langille MGI, Van Limbergen J. Multi-omics differentially classify disease state and treatment outcome in pediatric Crohn's disease. Microbiome. 2018;6:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 74]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
46.  Maeda Y, Kudo SE, Mori Y, Misawa M, Ogata N, Sasanuma S, Wakamura K, Oda M, Mori K, Ohtsuka K. Fully automated diagnostic system with artificial intelligence using endocytoscopy to identify the presence of histologic inflammation associated with ulcerative colitis (with video). Gastrointest Endosc. 2019;89:408-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 130]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
47.  Charisis VS, Hadjileontiadis LJ. Potential of hybrid adaptive filtering in inflammatory lesion detection from capsule endoscopy images. World J Gastroenterol. 2016;22:8641-8657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 26]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
48.  Klang E, Barash Y, Margalit RY, Soffer S, Shimon O, Albshesh A, Ben-Horin S, Amitai MM, Eliakim R, Kopylov U. Deep learning algorithms for automated detection of Crohn's disease ulcers by video capsule endoscopy. Gastrointest Endosc 2020; 91: 606-613. e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 117]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
49.  Ungaro RC, Hu L, Ji J, Nayar S, Kugathasan S, Denson LA, Hyams J, Dubinsky MC, Sands BE, Cho JH. Machine learning identifies novel blood protein predictors of penetrating and stricturing complications in newly diagnosed paediatric Crohn's disease. Aliment Pharmacol Ther. 2021;53:281-290.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Barash Y, Azaria L, Soffer S, Margalit Yehuda R, Shlomi O, Ben-Horin S, Eliakim R, Klang E, Kopylov U. Ulcer severity grading in video capsule images of patients with Crohn's disease: an ordinal neural network solution. Gastrointest Endosc. 2021;93:187-192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 45]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
51.  Lamash Y, Kurugol S, Freiman M, Perez-Rossello JM, Callahan MJ, Bousvaros A, Warfield SK. Curved planar reformatting and convolutional neural network-based segmentation of the small bowel for visualization and quantitative assessment of pediatric Crohn's disease from MRI. J Magn Reson Imaging. 2019;49:1565-1576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
52.  Takenaka K, Ohtsuka K, Fujii T, Negi M, Suzuki K, Shimizu H, Oshima S, Akiyama S, Motobayashi M, Nagahori M, Saito E, Matsuoka K, Watanabe M. Development and Validation of a Deep Neural Network for Accurate Evaluation of Endoscopic Images From Patients With Ulcerative Colitis. Gastroenterology. 2020;158:2150-2157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 128]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
53.  Bossuyt P, Nakase H, Vermeire S, de Hertogh G, Eelbode T, Ferrante M, Hasegawa T, Willekens H, Ikemoto Y, Makino T, Bisschops R. Automatic, computer-aided determination of endoscopic and histological inflammation in patients with mild to moderate ulcerative colitis based on red density. Gut. 2020;69:1778-1786.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 59]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
54.  Bhambhvani HP, Zamora A. Deep learning enabled classification of Mayo endoscopic subscore in patients with ulcerative colitis. Eur J Gastroenterol Hepatol. 2021;33:645-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
55.  Ozawa T, Ishihara S, Fujishiro M, Saito H, Kumagai Y, Shichijo S, Aoyama K, Tada T. Novel computer-assisted diagnosis system for endoscopic disease activity in patients with ulcerative colitis. Gastrointest Endosc 2019; 89: 416-421. e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 107]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
56.  Bossuyt P, De Hertogh G, Eelbode T, Vermeire S, Bisschops R. Computer-Aided Diagnosis With Monochromatic Light Endoscopy for Scoring Histologic Remission in Ulcerative Colitis. Gastroenterology. 2021;160:23-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
57.  Stidham RW, Liu W, Bishu S, Rice MD, Higgins PDR, Zhu J, Nallamothu BK, Waljee AK. Performance of a Deep Learning Model vs Human Reviewers in Grading Endoscopic Disease Severity of Patients With Ulcerative Colitis. JAMA Netw Open. 2019;2:e193963.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 112]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
58.  Gottlieb K, Requa J, Karnes W, Chandra Gudivada R, Shen J, Rael E, Arora V, Dao T, Ninh A, McGill J. Central Reading of Ulcerative Colitis Clinical Trial Videos Using Neural Networks. Gastroenterology 2021; 160: 710-719. e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 56]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
59.  Waljee AK, Liu B, Sauder K, Zhu J, Govani SM, Stidham RW, Higgins PDR. Predicting Corticosteroid-Free Biologic Remission with Vedolizumab in Crohn's Disease. Inflamm Bowel Dis. 2018;24:1185-1192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
60.  Uttam S, Hashash JG, LaFace J, Binion D, Regueiro M, Hartman DJ, Brand RE, Liu Y. Three-Dimensional Nanoscale Nuclear Architecture Mapping of Rectal Biopsies Detects Colorectal Neoplasia in Patients with Inflammatory Bowel Disease. Cancer Prev Res (Phila). 2019;12:527-538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
61.  Waljee AK, Sauder K, Patel A, Segar S, Liu B, Zhang Y, Zhu J, Stidham RW, Balis U, Higgins PDR. Machine Learning Algorithms for Objective Remission and Clinical Outcomes with Thiopurines. J Crohns Colitis. 2017;11:801-810.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 52]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
62.  Popa IV, Burlacu A, Mihai C, Prelipcean CC. A Machine Learning Model Accurately Predicts Ulcerative Colitis Activity at One Year in Patients Treated with Anti-Tumour Necrosis Factor α Agents. Medicina (Kaunas). 2020;56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
63.  Waljee AK, Joyce JC, Wang S, Saxena A, Hart M, Zhu J, Higgins PD. Algorithms outperform metabolite tests in predicting response of patients with inflammatory bowel disease to thiopurines. Clin Gastroenterol Hepatol. 2010;8:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 61]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
64.  Menti E, Lanera C, Lorenzoni G, Giachino DF, Marchi M, Gregori D, Berchialla P;  Piedmont Study Group on the Genetics of IBD. Bayesian Machine Learning Techniques for revealing complex interactions among genetic and clinical factors in association with extra-intestinal Manifestations in IBD patients. AMIA Annu Symp Proc. 2016;2016:884-893.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Waljee AK, Lipson R, Wiitala WL, Zhang Y, Liu B, Zhu J, Wallace B, Govani SM, Stidham RW, Hayward R, Higgins PDR. Predicting Hospitalization and Outpatient Corticosteroid Use in Inflammatory Bowel Disease Patients Using Machine Learning. Inflamm Bowel Dis. 2017;24:45-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 66]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
66.  Morilla I, Uzzan M, Laharie D, Cazals-Hatem D, Denost Q, Daniel F, Belleannee G, Bouhnik Y, Wainrib G, Panis Y, Ogier-Denis E, Treton X. Colonic MicroRNA Profiles, Identified by a Deep Learning Algorithm, That Predict Responses to Therapy of Patients With Acute Severe Ulcerative Colitis. Clin Gastroenterol Hepatol. 2019;17:905-913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (1)]
67.  Wang L, Fan R, Zhang C, Hong L, Zhang T, Chen Y, Liu K, Wang Z, Zhong J. Applying Machine Learning Models to Predict Medication Nonadherence in Crohn's Disease Maintenance Therapy. Patient Prefer Adherence. 2020;14:917-926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
68.  Bottigliengo D, Berchialla P, Lanera C, Azzolina D, Lorenzoni G, Martinato M, Giachino D, Baldi I, Gregori D. The Role of Genetic Factors in Characterizing Extra-Intestinal Manifestations in Crohn's Disease Patients: Are Bayesian Machine Learning Methods Improving Outcome Predictions? J Clin Med. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
69.  Ghoshal UC, Rai S, Kulkarni A, Gupta A. Prediction of outcome of treatment of acute severe ulcerative colitis using principal component analysis and artificial intelligence. JGH Open. 2020;4:889-897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
70.  Sofo L, Caprino P, Schena CA, Sacchetti F, Potenza AE, Ciociola A. New perspectives in the prediction of postoperative complications for high-risk ulcerative colitis patients: machine learning preliminary approach. Eur Rev Med Pharmacol Sci. 2020;24:12781-12787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
71.  Kang T, Ding W, Zhang L, Ziemek D, Zarringhalam K. A biological network-based regularized artificial neural network model for robust phenotype prediction from gene expression data. BMC Bioinformatics. 2017;18:565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
72.  Babic A, Ster B, Pavesic N, Wigertz O. Machine learning for the quality of life in inflammatory bowel disease. Stud Health Technol Inform. 1997;43 Pt B:661-665.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Dong Y, Xu L, Fan Y, Xiang P, Gao X, Chen Y, Zhang W, Ge Q. A novel surgical predictive model for Chinese Crohn's disease patients. Medicine (Baltimore). 2019;98:e17510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
74.  Lerrigo R, Coffey JT, Kravitz JL, Jadhav P, Nikfarjam A, Shah NH, Jurafsky D, Sinha SR. The Emotional Toll of Inflammatory Bowel Disease: Using Machine Learning to Analyze Online Community Forum Discourse. Crohn's & Colitis 360. 2019;1:otz011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
75.  Papadakis KA, Tabibzadeh S. Diagnosis and misdiagnosis of inflammatory bowel disease. Gastrointest Endosc Clin N Am. 2002;12:433-449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
76.  Best WR. Predicting the Crohn's disease activity index from the Harvey-Bradshaw Index. Inflamm Bowel Dis. 2006;12:304-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 194]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
77.  Lewis JD, Chuai S, Nessel L, Lichtenstein GR, Aberra FN, Ellenberg JH. Use of the noninvasive components of the Mayo score to assess clinical response in ulcerative colitis. Inflamm Bowel Dis. 2008;14:1660-1666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 512]  [Cited by in F6Publishing: 589]  [Article Influence: 36.8]  [Reference Citation Analysis (0)]
78.  Jones J, Loftus EV Jr, Panaccione R, Chen LS, Peterson S, McConnell J, Baudhuin L, Hanson K, Feagan BG, Harmsen SW, Zinsmeister AR, Helou E, Sandborn WJ. Relationships between disease activity and serum and fecal biomarkers in patients with Crohn's disease. Clin Gastroenterol Hepatol. 2008;6:1218-1224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 295]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
79.  Schoepfer AM, Beglinger C, Straumann A, Safroneeva E, Romero Y, Armstrong D, Schmidt C, Trummler M, Pittet V, Vavricka SR. Fecal calprotectin more accurately reflects endoscopic activity of ulcerative colitis than the Lichtiger Index, C-reactive protein, platelets, hemoglobin, and blood leukocytes. Inflamm Bowel Dis. 2013;19:332-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 207]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
80.  Xie T, Zhang T, Ding C, Dai X, Li Y, Guo Z, Wei Y, Gong J, Zhu W, Li J. Ulcerative Colitis Endoscopic Index of Severity (UCEIS) vs Mayo Endoscopic Score (MES) in guiding the need for colectomy in patients with acute severe colitis. Gastroenterol Rep (Oxf). 2018;6:38-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 61]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
81.  Daperno M, D'Haens G, Van Assche G, Baert F, Bulois P, Maunoury V, Sostegni R, Rocca R, Pera A, Gevers A, Mary JY, Colombel JF, Rutgeerts P. Development and validation of a new, simplified endoscopic activity score for Crohn's disease: the SES-CD. Gastrointest Endosc. 2004;60:505-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 999]  [Cited by in F6Publishing: 1089]  [Article Influence: 54.5]  [Reference Citation Analysis (0)]
82.  Novak G, Parker CE, Pai RK, MacDonald JK, Feagan BG, Sandborn WJ, D'Haens G, Jairath V, Khanna R. Histologic scoring indices for evaluation of disease activity in Crohn's disease. Cochrane Database Syst Rev. 2017;7:CD012351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
83.  Mosli MH, Feagan BG, Sandborn WJ, Dʼhaens G, Behling C, Kaplan K, Driman DK, Shackelton LM, Baker KA, Macdonald JK, Vandervoort MK, Geboes K, Levesque BG. Histologic evaluation of ulcerative colitis: a systematic review of disease activity indices. Inflamm Bowel Dis. 2014;20:564-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 87]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
84.  Falvey JD, Hoskin T, Meijer B, Ashcroft A, Walmsley R, Day AS, Gearry RB. Disease activity assessment in IBD: clinical indices and biomarkers fail to predict endoscopic remission. Inflamm Bowel Dis. 2015;21:824-831.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 110]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
85.  Neurath MF. Current and emerging therapeutic targets for IBD. Nat Rev Gastroenterol Hepatol. 2017;14:269-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 315]  [Cited by in F6Publishing: 386]  [Article Influence: 55.1]  [Reference Citation Analysis (0)]
86.  Weisshof R, El Jurdi K, Zmeter N, Rubin DT. Emerging Therapies for Inflammatory Bowel Disease. Adv Ther. 2018;35:1746-1762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 54]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
87.  Luo Y, Zhang Y, Liu M, Lai Y, Liu P, Wang Z, Xing T, Huang Y, Li Y, Li A, Wang Y, Luo X, Liu S, Han Z. Artificial Intelligence-Assisted Colonoscopy for Detection of Colon Polyps: a Prospective, Randomized Cohort Study. J Gastrointest Surg. 2020;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 43]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
88.  Selaru FM, Xu Y, Yin J, Zou T, Liu TC, Mori Y, Abraham JM, Sato F, Wang S, Twigg C, Olaru A, Shustova V, Leytin A, Hytiroglou P, Shibata D, Harpaz N, Meltzer SJ. Artificial neural networks distinguish among subtypes of neoplastic colorectal lesions. Gastroenterology. 2002;122:606-613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 103]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
89.  Gillen CD, Walmsley RS, Prior P, Andrews HA, Allan RN. Ulcerative colitis and Crohn's disease: a comparison of the colorectal cancer risk in extensive colitis. Gut. 1994;35:1590-1592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 375]  [Cited by in F6Publishing: 390]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
90.  Bernstein CN, Blanchard JF, Kliewer E, Wajda A. Cancer risk in patients with inflammatory bowel disease: a population-based study. Cancer. 2001;91:854-862.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
91.  Misawa M, Kudo SE, Mori Y, Cho T, Kataoka S, Yamauchi A, Ogawa Y, Maeda Y, Takeda K, Ichimasa K, Nakamura H, Yagawa Y, Toyoshima N, Ogata N, Kudo T, Hisayuki T, Hayashi T, Wakamura K, Baba T, Ishida F, Itoh H, Roth H, Oda M, Mori K. Artificial Intelligence-Assisted Polyp Detection for Colonoscopy: Initial Experience. Gastroenterology 2018; 154: 2027-2029. e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 229]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
92.  Wang P, Berzin TM, Glissen Brown JR, Bharadwaj S, Becq A, Xiao X, Liu P, Li L, Song Y, Zhang D, Li Y, Xu G, Tu M, Liu X. Real-time automatic detection system increases colonoscopic polyp and adenoma detection rates: a prospective randomised controlled study. Gut. 2019;68:1813-1819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 398]  [Cited by in F6Publishing: 450]  [Article Influence: 90.0]  [Reference Citation Analysis (0)]
93.  Wang P, Liu X, Berzin TM, Glissen Brown JR, Liu P, Zhou C, Lei L, Li L, Guo Z, Lei S, Xiong F, Wang H, Song Y, Pan Y, Zhou G. Effect of a deep-learning computer-aided detection system on adenoma detection during colonoscopy (CADe-DB trial): a double-blind randomised study. Lancet Gastroenterol Hepatol. 2020;5:343-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 241]  [Article Influence: 60.3]  [Reference Citation Analysis (0)]
94.  Ogawa R, Nishikawa J, Hideura E, Goto A, Koto Y, Ito S, Unno M, Yamaoka Y, Kawasato R, Hashimoto S, Okamoto T, Ogihara H, Hamamoto Y, Sakaida I. Objective Assessment of the Utility of Chromoendoscopy with a Support Vector Machine. J Gastrointest Cancer. 2019;50:386-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]