ArticlePDF AvailableLiterature Review

Artificial intelligence in small intestinal diseases: Application and prospects

Authors:

Abstract and Figures

The small intestine is located in the middle of the gastrointestinal tract, so small intestinal diseases are more difficult to diagnose than other gastrointestinal diseases. However, with the extensive application of artificial intelligence in the field of small intestinal diseases, with its efficient learning capacities and computational power, artificial intelligence plays an important role in the auxiliary diagnosis and prognosis prediction based on the capsule endoscopy and other examination methods, which improves the accuracy of diagnosis and prediction and reduces the workload of doctors. In this review, a comprehensive retrieval was performed on articles published up to October 2020 from PubMed and other databases. Thereby the application status of artificial intelligence in small intestinal diseases was systematically introduced, and the challenges and prospects in this field were also analyzed.
Content may be subject to copyright.
World Journal of
Gastroenterology
ISSN 1007-9327 (print)
ISSN 2219-2840 (online)
World J Gastroenterol 2021 July 7; 27(25): 3693-3950
Published by Baishideng Publishing Group Inc
WJG https://www.wjgnet.com IJuly 7, 2021 Volume 27 Issue 25
World Journal of
Gastroenterology
W J G
Contents Weekly Volume 27 Number 25 July 7, 2021
OPINION REVIEW
Approach to medical therapy in perianal Crohn’s disease
3693
Vasudevan A, Bruining DH, Loftus EV Jr, Faubion W, Ehman EC, Raffals L
REVIEW
Incorporating mucosal-associated invariant T cells into the pathogenesis of chronic liver disease
3705
Czaja AJ
Artificial intelligence in small intestinal diseases: Application and prospects
3734
Yang Y, Li YX, Yao RQ, Du XH, Ren C
Impact of the COVID-19 pandemic on inflammatory bowel disease patients: A review of the current
evidence
3748
Kumric M, Ticinovic Kurir T, Martinovic D, Zivkovic PM, Bozic J
Management of hepatitis B virus infection in patients with inflammatory bowel disease under
immunosuppressive treatment
3762
Axiaris G, Zampeli E, Michopoulos S, Bamias G
MINIREVIEWS
Worldwide management of hepatocellular carcinoma during the COVID-19 pandemic
3780
Inchingolo R, Acquafredda F, Tedeschi M, Laera L, Surico G, Surgo A, Fiorentino A, Spiliopoulos S, de’Angelis N, Memeo
R
Human immune repertoire in hepatitis B virus infection
3790
Zhan Q, Xu JH, Yu YY, Lo KK E, Felicianna, El-Nezami H, Zeng Z
Emerging applications of radiomics in rectal cancer: State of the art and future perspectives
3802
Hou M, Sun JH
Advances in paediatric nonalcoholic fatty liver disease: Role of lipidomics
3815
Di Sessa A, Riccio S, Pirozzi E, Verde M, Passaro AP, Umano GR, Guarino S, Miraglia del Giudice E, Marzuillo P
Autoimmune pancreatitis and pancreatic cancer: Epidemiological aspects and immunological
considerations
3825
Poddighe D
Gut microbiota in obesity
3837
Liu BN, Liu XT, Liang ZH, Wang JH
WJG https://www.wjgnet.com II July 7, 2021 Volume 27 Issue 25
World Journal of Gastroenterology
Contents Weekly Volume 27 Number 25 July 7, 2021
ORIGINAL ARTICLE
Basic Study
Zinc oxide nanoparticles reduce the chemoresistance of gastric cancer by inhibiting autophagy
3851
Miao YH, Mao LP, Cai XJ, Mo XY, Zhu QQ, Yang FT, Wang MH
PPARGC1A rs8192678 G>A polymorphism affects the severity of hepatic histological features and
nonalcoholic steatohepatitis in patients with nonalcoholic fatty liver disease
3863
Zhang RN, Shen F, Pan Q, Cao HX, Chen GY, Fan JG
Retrospective Cohort Study
Does endoscopic intervention prevent subsequent gastrointestinal bleeding in patients with left ventricular
assist devices? A retrospective study
3877
Palchaudhuri S, Dhawan I, Parsikia A, Birati EY, Wald J, Siddique SM, Fisher LR
Retrospective Study
Diverse expression patterns of mucin 2 in colorectal cancer indicates its mechanism related to the intestinal
mucosal barrier
3888
Gan GL, Wu HT, Chen WJ, Li CL, Ye QQ, Zheng YF, Liu J
Clinical characteristics of patients in their forties who underwent surgical resection for colorectal cancer in
Korea
3901
Lee CS, Baek SJ, Kwak JM, Kim J, Kim SH
Observational Study
Effect of gastric microbiota on quadruple Helicobacter pylori eradication therapy containing bismuth
3913
Niu ZY, Li SZ, Shi YY, Xue Y
META-ANALYSIS
Endoscopic submucosal dissection vs endoscopic mucosal resection for colorectal polyps: A meta-analysis
and meta-regression with single arm analysis
3925
Lim XC, Nistala KRY, Ng CH, Lin SY, Tan DJH, Ho KY, Chong CS, Muthiah M
CASE REPORT
Gastric schwannoma treated by endoscopic full-thickness resection and endoscopic purse-string suture: A
case report
3940
Lu ZY, Zhao DY
LETTER TO THE EDITOR
Gastrointestinal cytomegalovirus disease secondary to measles in an immunocompetent infant
3948
Hung CM, Lee PH, Lee HM, Chiu CC
WJG https://www.wjgnet.com III July 7, 2021 Volume 27 Issue 25
World Journal of Gastroenterology
Contents Weekly Volume 27 Number 25 July 7, 2021
ABOUT COVER
Editorial Board Member of World Journal of Gastroenterology, Paola De Nardi, MD, FASCRS, Doctor, Surgeon,
Surgical Oncologist, Division of Gastrointestinal Surgery, IRCCS San Raffaele Scientific Institute, via Olgettina 60,
Milan 20132, Italy. denardi.paola@hsr.it
AIMS AND SCOPE
The primary aim of World Journal of Gastroenterology (WJG, World J Gastroenterol) is to provide scholars and readers
from various fields of gastroenterology and hepatology with a platform to publish high-quality basic and clinical
research articles and communicate their research findings online. WJG mainly publishes articles reporting research
results and findings obtained in the field of gastroenterology and hepatology and covering a wide range of topics
including gastroenterology, hepatology, gastrointestinal endoscopy, gastrointestinal surgery, gastrointestinal
oncology, and pediatric gastroenterology.
INDEXING/ABSTRACTING
The WJG is now indexed in Current Contents®/Clinical Medicine, Science Citation Index Expanded (also known as
SciSearch®), Journal Citation Reports®, Index Medicus, MEDLINE, PubMed, PubMed Central, and Scopus. The 2021
edition of Journal Citation Report® cites the 2020 impact factor (IF) for WJG as 5.742; Journal Citation Indicator: 0.79;
IF without journal self cites: 5.590; 5-year IF: 5.044; Ranking: 28 among 92 journals in gastroenterology and
hepatology; and Quartile category: Q2. The WJG’s CiteScore for 2020 is 6.9 and Scopus CiteScore rank 2020:
Gastroenterology is 19/136.
RESPONSIBLE EDITORS FOR THIS ISSUE
Production Editor: Ying-Yi Yuan; Production Department Director: Xiang Li; Editorial Office Director: Ze-Mao Gong.
NAME OF JOURNAL INSTRUCTIONS TO AUTHORS
World Journal of Gastroenterology https://www.wjgnet.com/bpg/gerinfo/204
ISSN GUIDELINES FOR ETHICS DOCUMENTS
ISSN 1007-9327 (print) ISSN 2219-2840 (online) https://www.wjgnet.com/bpg/GerInfo/287
LAUNCH DATE GUIDELINES FOR NON-NATIVE SPEAKERS OF ENGLISH
October 1, 1995 https://www.wjgnet.com/bpg/gerinfo/240
FREQUENCY PUBLICATION ETHICS
Weekly https://www.wjgnet.com/bpg/GerInfo/288
EDITORS-IN-CHIEF PUBLICATION MISCONDUCT
Andrzej S Tarnawski, Subrata Ghosh https://www.wjgnet.com/bpg/gerinfo/208
EDITORIAL BOARD MEMBERS ARTICLE PROCESSING CHARGE
http://www.wjgnet.com/1007-9327/editorialboard.htm https://www.wjgnet.com/bpg/gerinfo/242
PUBLICATION DATE STEPS FOR SUBMITTING MANUSCRIPTS
July 7, 2021 https://www.wjgnet.com/bpg/GerInfo/239
COPYRIGHT ONLINE SUBMISSION
© 2021 Baishideng Publishing Group Inc https://www.f6publishing.com
© 2021 Baishideng Publishing Group Inc. All rights reserved. 7041 Koll Center Parkway, Suite 160, Pleasanton, CA 94566, USA
E-mail: bpgoffice@wjgnet.com https://www.wjgnet.com
WJG https://www.wjgnet.com 3734 July 7, 2021 Volume 27 Issue 25
World Journal of
Gastroenterology
W J G
Submit a Manuscript: https://www.f6publishing.com World J Gastroenterol 2021 July 7; 27(25): 3734-3747
DOI: 10.3748/wjg.v27.i25.3734 ISSN 1007-9327 (print) ISSN 2219-2840 (online)
REVIEW
Artificial intelligence in small intestinal diseases: Application and
prospects
Yu Yang, Yu-Xuan Li, Ren-Qi Yao, Xiao-Hui Du, Chao Ren
ORCID number: Yu Yang 0000-
0002-0549-8755; Yu-Xuan Li 0000-
0003-2568-6347; Ren-Qi Yao 0000-
0002-3173-4301; Xiao-Hui Du 0000-
0002-8713-1358; Chao Ren 0000-
0003-0655-0209.
Author contributions: Yang Y
searched the literature for recent
advances in the field and wrote the
manuscript; Yang Y, Li YX, Yao RQ
and Du XH edited and revised the
manuscript; Ren C designed the
study; all authors approved the
final version to be published.
Supported by The National Natural
Science Foundation of China, No.
81871317.
Conflict-of-interest statement: The
authors declare that the research
was conducted in the absence of
any commercial or financial
relationships that could be
construed as a potential conflict of
interest.
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
Yu Yang, Yu-Xuan Li, Xiao-Hui Du, Department of General Surgery, Chinese People’s Liberation
Army General Hospital, Beijing 100853, China
Ren-Qi Yao, Chao Ren, Trauma Research Center, The Fourth Medical Center and Medical
Innovation Research Division of the Chinese People‘s Liberation Army General Hospital,
Beijing 100048, China
Ren-Qi Yao, Department of Burn Surgery, Changhai Hospital, Naval Medical University,
Shanghai 200433, China
Corresponding author: Chao Ren, MD, PhD, Trauma Research Center, Fourth Medical Center
and Medical Innovation Research Division of the Chinese People’s Liberation Army General
Hospital, No. 51 Fucheng Road, Beijing 100048, China. rc198@sina.com
Abstract
The small intestine is located in the middle of the gastrointestinal tract, so small
intestinal diseases are more difficult to diagnose than other gastrointestinal
diseases. However, with the extensive application of artificial intelligence in the
field of small intestinal diseases, with its efficient learning capacities and compu-
tational power, artificial intelligence plays an important role in the auxiliary
diagnosis and prognosis prediction based on the capsule endoscopy and other
examination methods, which improves the accuracy of diagnosis and prediction
and reduces the workload of doctors. In this review, a comprehensive retrieval
was performed on articles published up to October 2020 from PubMed and other
databases. Thereby the application status of artificial intelligence in small
intestinal diseases was systematically introduced, and the challenges and pro-
spects in this field were also analyzed.
Key Words: Artificial intelligence; Machine learning; Deep learning; Prognosis prediction;
Small intestinal diseases
©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
Core Tip: Artificial intelligence has been widely used in the management of small
intestinal diseases, which has greatly improved the diagnostic efficiency of capsule
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3735 July 7, 2021 Volume 27 Issue 25
on different terms, provided the
original work is properly cited and
the use is non-commercial. See: htt
p://creativecommons.org/License
s/by-nc/4.0/
Manuscript source: Invited
manuscript
Specialty type: Gastroenterology
and hepatology
Country/Territory of origin: China
Peer-review report’s scientific
quality classification
Grade A (Excellent): 0
Grade B (Very good): B, B
Grade C (Good): C, C
Grade D (Fair): 0
Grade E (Poor): 0
Received: January 25, 2021
Peer-review started: January 25,
2021
First decision: March 29, 2021
Revised: April 9, 2021
Accepted: May 8, 2021
Article in press: May 8, 2021
Published online: July 7, 2021
P-Reviewer: Chen CH, Jiang Y,
Orhan K, Pons-Beltrán V
S-Editor: Zhang H
L-Editor: Filipodia
P-Editor: Liu JH
endoscopy and other examination methods, and at the same time, beneficial
progression has also been obtained in the prognosis prediction of small intestinal
diseases. Although AI still faces risks such as overfitting and black box effects, its
stability and efficiency give it great potential in the management of small intestinal
diseases. This article reviews the current application status of AI in small intestinal
diseases. In addition, challenges and prospects in this field are discussed.
Citation: Yang Y, Li YX, Yao RQ, Du XH, Ren C. Artificial intelligence in small intestinal
diseases: Application and prospects. World J Gastroenterol 2021; 27(25): 3734-3747
URL: https://www.wjgnet.com/1007-9327/full/v27/i25/3734.htm
DOI: https://dx.doi.org/10.3748/wjg.v27.i25.3734
INTRODUCTION
The small intestine is located in the middle of the gastrointestinal digestive system,
with a total length of 5 m to 7 m, including the duodenum, jejunum and ileum, and is
the longest organ of the digestive system. Small intestinal diseases (SIDs) mainly
include celiac disease (CD), small intestinal Crohn’s disease (SICD), primary small
intestinal tumor (PSIT), obscure gastrointestinal bleeding and so on. The traditional
examination methods include X-ray barium enterography, computed tomography
(CT), magnetic resonance imaging (MRI), balloon-assisted enteroscopy, deep
enteroscopy and so on. In recent years, the emergence of capsule endoscopy (CE) has
brought a revolutionary breakthrough for the diagnosis of SIDs. However, because of
the special anatomical position of the small intestine (far away from the oral cavity and
anus, overlap and peristalsis), there are still many problems in the diagnosis of SIDs,
such as high technical requirements, low positive rate of diagnosis, inaccurate
qualitative location of the disease, patient intolerance and so on. In addition, the onset
of SIDs is insidious, the specificity of clinical symptoms is low, and the lesion site is
not easy to explore, so the clinical diagnosis of SIDs has always been a difficult
problem. With the emergence of artificial intelligence (AI) and its wide application in
the medical field, it also provides new methods for the whole management process of
SIDs and greatly improves the efficiency of SIDs management.
AI is a concept put forward in the 1950s. It is a frontier cross discipline developed
on the basis of computer science, neuropsychology, philosophy, linguistics, cyber-
netics, information theory and so on[1]. The research fields of AI include expert
system, machine learning (ML), fuzzy logic, natural language processing and so on.
Research methods are also developing continuously, from ML to deep learning and
then to convolutional neural network (CNN), promoting the rapid development of
research in various fields. The research of AI in the medical field is mainly focused on
auxiliary diagnosis. This series of methods of AI has become a hot implementation tool
in the field of medical imaging and digestive endoscope[2,3]. Taking the experiment of
the breast cancer AI detection system established by Google as an example, the
computer-aided diagnosis system based on AI can help doctors reduce the misdia-
gnosis rate of breast cancer by 5.7%[4]. Researchers at Houston Methodist Hospital
also said in their study that they have developed AI software that parses breast X-ray
images 30 times faster than ordinary doctors, with an accuracy of 99%[5]. AI is widely
used in the study of digestive fields such as gastric cancer[6], colorectal cancer[7],
esophageal cancer[8] and so on. AI has also been extensively researched in the field of
SIDs, which will be introduced in this paper.
This study used the keywords of “artificial intelligence” and “small intestine” to
search the relevant literature in the databases of PubMed, Embase, Web of Science and
Cochrane Library up to October 2020. Studies included in our review were required to
meet the following inclusion criteria: (1) full-text paper available in English; and (2)
studies that associated AI with the small intestinal diseases. We excluded descriptive
papers without validation of methods. The application status of AI in SIDs was
summarized, and the challenges and prospects in this field were discussed.
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3736 July 7, 2021 Volume 27 Issue 25
AI IN SMALL INTESTINE ANATOMY
Organ segmentation of the small intestine
With the advent of AI, it is possible to perform computer-assisted organ segmentation
in CT, MRI, endoscopy and other examination methods and has shown good
application potential in the fields like assisted localization of radiotherapy. The
following will introduce the research progress of this aspect in the field of the small
intestine especially the duodenum (Table 1).
CT: Some studies had used the CNN method to automatically segment duodenum
and other abdominal organs from CT images, with clinically acceptable accuracy and
efficiency[9,10]. Tong et al[11] proposed an end-to-end segmentation network for
improving multiorgan segmentation performance using the ML method. The dice
similarity coefficient and average surface distance were quantitatively evaluated, and
the results confirmed this network had good accuracy and timeliness in the anatomical
segmentation of abdominal organs including the duodenum.
MRI: Fu et al[12] conducted a retrospective analysis on 3D MR images of 120 patients
and proposed a CNN model, which has been verified to accurately segment the
abdominal organs including the duodenum and expedite the contouring process for
MRI-guided adaptive radiotherapy. Chen et al[13] also conducted a similar study, and
in their study, the inference process was completed within 1 min, indicating an
obvious advantage of timeliness. The length of the small intestine is an important
factor in the management of patients with short bowel syndrome. Some scholars
designed a special software algorithm to calculate the length of small intestine based
on magnetic resonance enterography images in mice. Compared with the measured
results of anatomical specimens, the mean absolute difference between the two
methods was 1.8 ± 3.8 cm (P = 0.24), and the mean percentage difference was 9.4% ±
6.0%[14].
Endoscopy: In a Japanese study based on GoogLeNet architecture, a CNN diagnostic
program was constructed, using 27335 esophagogastroduodenoscopy (EGD) images
for the training set and using 17081 EGD images for the independent validation set.
The results showed that the CNN has a good effect to classify the anatomical location
of EGD images for stomach and duodenum images, with an area under the curve of
0.99[15]. Igarashi et al[16] used AlexNet (a deep learning framework) to retrospectively
analyze 85246 original images of EGD images in 441 patients with gastric cancer and
developed an anatomical organ classifier. The accuracy rates of the training and
validation sets were 0.993 and 0.965, respectively.
Diagnosis of small intestinal mucosal lesions
With the emergence of CE in 2000[17], it has revolutionized our understanding of
small intestinal mucosa[18-20], enabling doctors to detect small intestinal mucosal
erosion, ulcers, vascular disease, bleeding, polyps, parasite and other lesions more
efficiently. However, reliable and rapid reading of video is still a challenge, but more
and more studies have shown that the combination of AI and CE can greatly improve
the efficiency of our evaluation of small intestinal mucosal lesions; the detection
accuracy was above 90% in most studies[21-27].
Ulcer: Previous studies have confirmed that applying a CNN system of deep learning
to the reading process of CE can reduce the reading time without decreasing the
detection rate of erosion and ulcer lesions[28-32].
Angioectasias and bleeding: Intestinal angioectasias cause more than 8% of all
gastrointestinal bleeding episodes[33]. Different studies have applied ML, CNN and
computer algorithms to the differential diagnosis of intestinal angioectasias and have
achieved high sensitivity and specificity[34-39]. AI is also applied to the direct
examination of intestinal mucosal bleeding by CE, which can directly calculate the
blood content in the digestive tract and infer whether there is active bleeding in the
small intestinal mucosa[40-44].
Protruding lesions: There are a variety of small intestinal mucosal protruding lesions.
CNN can help doctors describe their shape features, help analyze their nature and
distinguish polyps, epithelial tumors, submucosal tumors, etc.[34,45,46].
Villous atrophy: Villous atrophy is a defining symptom of some digestive tract
diseases such as CD. Some scholars combined AI methods with CE for the detection
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3737 July 7, 2021 Volume 27 Issue 25
Table 1 Applications of artificial intelligence in organ segmentation of the small intestine
Ref. Diagnostic method AI technology Training set Validating set Outcomes
Tong et al[11] CT ML 90 images - DSC of duodenum: 69.26%
Kim et al[9] CT CNN 80 images 40 images DSC of duodenum: 0.595
Peng et al[10] CT CNN 43 images - DSC of duodenum: 0.61
Dice coefficient of duodenum:
65.50% ± 8.90%
Fu et al[12] MRI CNN 100 images 20 images
Dice coefficient of bowel: 86.60%
± 2.69%
Chen et al[13] MRI DL 66 images 36 images DSC of duodenum: 0.80
Takiyama et al[15] EGD CNN 27335 images 17081 images AUCs: 0.99
Igarashi et al[16] EGD ML 49174 images 36072 images Accuracy (Ts: 0.993, Vs: 0.965)
AI: Artificial intelligence; AUCs: Area under the curves; CNN: Convolutional neural network; CT: Computed Tomography; DL: Deep learning; DSC: Dice
similarity coefficient; EGD: Esophagogastroduodenoscopy; ML: Machine learning; MRI: Magnetic resonance imaging; Ts: Training set; Vs: Validating set.
and measurement of villous atrophy and successfully mapped the extent of the
diseased small intestine[47].
AI is also used in risk prediction and clinical treatment decisions of small intestinal
mucosal lesions. For example, one study used CNN for the risk prediction of acute
intestinal bleeding[48], and another study applied CNN to risk prediction and
therapeutic tactics selection for duodenal ulcers[49]. Wong et al[50] built a ML model,
based on data from 22854 patients with gastroduodenal ulcer including six clinical
parameters to identify patients at high risk for recurrent ulcer bleeding within 1 year.
Gastrointestinal bleeding is a common complication of left ventricular assist device
treatment. Axelrad et al[51] developed an endoscopic algorithm. Compared with
conventional cohorts, the implementation of the algorithm increased endoscopic
diagnostic efficiency by 68%, treatment efficiency by 113%, the number of procedures
per patient decreased by 27%, the length of hospital stay decreased by 33%, and the
estimated cost decreased by 18%.
In addition, the interference of intestinal contents to CE can also be reduced by AI.
Combined with support vector machine, Bashar et al[52] designed a classifier for
separating useless frames that are highly contaminated by turbid fluids, fecal materials
and/or residual foods. The accuracy of this classifier was more than 80%. Pietri et al
[53] developed a computer algorithm to automatically evaluate the demeanor of small
intestinal bubbles in CE images. The specificity of this algorithm was 95.79%, the
sensitivity was 95.19%, and the calculation time was 0.037 s per frame. It can be used
to reduce the interference of bubbles in CE images. Klein et al[54] created a computed
algorithm based on the pixels in the color bar to score and classify the preparation of
the small intestine for CE, and this automatic scoring method has a concordance rate of
more than 90% with the assessment of clinicians.
AI IN COMMON SMALL INTESTINAL DISEASES
AI in celiac disease
CD is a complex autoimmune disease. Patients who ingest foods containing gluten will
develop an autoimmune response that causes damage to the small intestine. CD is one
of the most common chronic digestive diseases, with a prevalence rate of 1%
worldwide[55]. Duodenal biopsy is the gold standard for diagnosis[56]. Noninvasive
methods such as endoscopy and clinical features analysis are also widely used in
diagnosis, but the diagnostic rate of CD is only 15%–20% through current strategies
[57]. However, with the increasing application of AI in the diagnosis of CD, the
accuracy and efficiency of diagnosis are greatly improved[58] (Table 2).
Previous studies have confirmed that AI-assisted duodenoscope images analysis can
greatly improve the diagnostic efficacy of CD, with the accuracy between 80% and
100% and specificity and sensitivity over 80%[59-61]. In the diagnosis of CD, the
combination of AI and CE is closer, which can improve the accuracy of diagnosis and
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3738 July 7, 2021 Volume 27 Issue 25
Table 2 Applications of artificial intelligence in celiac disease
Ref. Diagnostic
method AI technology Training set Testing set Outcomes
Chetcuti et al[62] CE ML 81 patients - Accuracy: 75.3%
Li et al[63] CE Computer-assisted recognition Ep: 240, Cp: 220 - Accuracy: 93.9%
Vicnesh et al[64] CE Computerized algorithm 21 patients - Accuracy: 89.82%
Zhou et al[65] CE CNN Ep: 6, Cp: 5 Ep: 5, Cp: 5 Accuracy: 100%
Gadermayr et al
[59]
EGD Computer-assisted 290 patients (2835
images)
- Accuracy: 94%-100%
Das et al[67] Mucosal biopsies Computer-assisted Ep: 124, Cp: 137 Ep: 120, Cp:
105
Sen: 90.3%, Spe: 93.5%,
AUCs: 96.2%
Wei et al[66] Mucosal biopsies DL 212 images - Accuracy: 95.3%, AUCs >
0.95
Pastore et al[70] Clinical data Computer-assisted 100 patients - Reliability: 0.813
Tenório et al[60] Clinical data Decision trees, Bayesian inference, k-nearest
neighbor algorithm, support vector machines,
artificial neural networks
178 patients 38 patients Accuracy: 80.0%, Sen:
0.78, Spe: 0.80, AUCs: 0.84
Virta et al[68] Micro-CT Computer-assisted point cloud analysis 81 patients - Accuracy: 100%
Sangineto et al[69] Gene expression in
PBMCs
ML, random forest algorithm Ep: 17, Cp: 20 - Accuracy: 100%
AI: Artificial intelligence; AUCs: Area under the curves; CE: Capsule endoscopy; CNN: Convolutional neural network; Cp: Control group; DL: Deep
learning; EGD: Esophagogastroduodenoscopy; Ep: Experimental group; ML: Machine learning; micro-CT: X-ray microtomography; PBMCs: Peripheral
blood mononuclear cells; Sen: Sensitivity; Spe: Specificity.
significantly save the diagnosis time[62-65]. AI was also used in the analysis of
duodenal mucosa biopsy, which can help with qualitative analysis and play an
important role in quantitative analysis[66,67]. At the same time, the application of AI
with X-ray images[68], peripheral blood mononuclear cells[69] and clinical features[60,
70] in the diagnosis and classification of CD have also achieved progress.
AI in small intestinal Crohn’s disease
Crohn’s disease is a chronic nonspecific inflammatory bowel disease that affects the
entire gastrointestinal tract, in which 30% of patients are confined to the small
intestine, commonly known as small intestinal Crohn‘s disease[71]. SICD most often
involves the distal ileum as well as the jejunum and the digestive tract above and has a
higher incidence of intestinal strictures than colonic Crohn‘s disease[72,73]. The
application of AI in the management of SICD is comprehensive, including diagnosis,
risk prediction, extra-intestinal manifestation (EIM) prediction and so on (Table 3).
Diagnosis: Lamash et al[74,75] used CNN to analyze MRI images and construct an
assessment model for SICD. Their model could effectively distinguish active and
inactive inflammatory segments, distinguish segments with strictures and segments
without strictures and could be used to measure the length of intestinal strictures.
Parfеnov et al[76] used a software diagnostic algorithm to analyze the CE images of 25
SICD patients, preliminarily confirming that CE could be used to diagnose early SICD
with intestinal mucosal inflammation. Klang et al[77] performed automatic analysis of
CE images of 49 SICD patients using a CNN method, achieving diagnostic accuracy of
more than 95% and significantly reducing reading time. Yang et al[78] also attempted
to combine CNN with a microultrasound system for early diagnosis of SICD in mice
and achieved good effectiveness in the identification of early inflammation.
Risk prediction of SICD: Taylor et al[79] used ML classifiers (elastic network and
random forest) to classify small intestine inflammation in asymptomatic first-degree
relatives of patients with SICD. They found that genetic variants associated with SICD,
family history and fecal calprotectin together identified individuals with presymp-
tomatic intestinal inflammation who are therefore at risk for SICD. Shen et al[80]
developed a web-based SICD hazard stratification tool. Predicting high-risk
populations for SICD based on altered bowel habit, abdominal pain, white blood cell
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3739 July 7, 2021 Volume 27 Issue 25
Table 3 Applications of artificial intelligence in small intestinal Crohn’s disease
Ref. Diagnostic method AI technology Training set Testing set Outcomes
Yang et al[78] Microultrasound CNN 43 mice - AUCs: 0.8831
Shen et al[80] Clinical data Computerized
algorithm
Ep1: 61, Cp1: 78 Ep2:42, Cp2: 57;
Ep3:84, Cp3: 495
AUCs: 0.92
Bottigliengo et al
[81]
Clinical data BMLTs (NB, BN,
BART)
152 patients - AUCs without genetic variables (NB: 0.71,
BN: 0.50, BART: 0.76), AUCs with genetic
variables (NB: 0.75, BN: 0.67, BART: 0.78)
Taylor et al[79] Clinical data ML (elastic net and
random forest)
480 first-degree
relatives
- AUCs (elastic net): 0.80, AUCs (random
forest): 0.87
Menti et al[82] Clinical data BMLTs 152 patients - Accuracy without genetic variables: 82%,
accuracy with genetic variables: 89%
Klang et al[77] CE DL 49 patients
(17640 images)
- AUCs: 0.94-0.99, accuracy: 95.4%-96.7%
Parfеnov et al[76] CE Computerized
algorithm
25 patients - 44% patients confirmed only with the help
of AI
Lamash et al[74,
75]
MRI CNN 15 patients 8 patients Dice coefficients: 75%-97%
AI: Artificial intelligence; AUCs: Area under the curves; BART: Bayes additive return trees; BMLTs: Bayesian machine learning techniques; BN: Bayesian
network; CE: Capsule endoscopy; DL: Deep learning; CNN: Convolutional neural network; Cp: Control group; Ep: Experimental group; ML: Machine
learning; MRI: Magnetic resonance imaging; NB: Naive Bayes.
count, albumin and platelet count abnormalities allowed clinicians to identify
potential SICD earlier.
Risk prediction of EIMs: AI is controversial in the evaluation of the EIMs of SICD. In
the study of Bottigliengo et al[81], based on Bayesian machine learning technology
evaluation combined with genetic factors to predict the occurrence of EIMs in Crohn’s
disease, it has no advantage over traditional statistical tools. Whereas Menti et al[82]
used Bayesian machine learning technology to predict the risk of occurrence of EIMs
in Crohn’s disease, and the prediction accuracy was 82% when considering only
clinical factor and 89% combined with genetic factors, which was outperforming other
prediction techniques.
AI in primary small intestinal tumor
The incidence of PSIT is about 5% of gastrointestinal tumors and 0.2% of all kinds of
tumors[83,84]. The main site of PSIT is the duodenum, followed by the jejunum and
ileum[85]. There are a variety of pathological types of malignant PSIT. Adenocar-
cinoma is the most common pathological type, up to 40%, followed by neuroendocrine
tumors (25%), malignant lymphomas (10%-15%) and malignant stromal tumors (9%)
[86]. PSIT lacks specific manifestations in the early stage, and they are faced with many
problems in the clinic, such as difficult diagnosis, high misdiagnosis rate, nonstandard
treatment and so on[87]. AI has been applied in the field of auxiliary diagnosis and
prognostic analysis of PSIT, and has an important impact on the management
(Table 4).
Diagnosis: Inoue et al[88] used CNN to analyze EGD images for the diagnosis of
superficial nonampullary duodenal epithelial tumors. The overall diagnosis accuracy
of CNN was 94.7%, including 94% for adenomas and 100% for high-grade dysplasias,
and it only took 12-31 s for analysis. The method of support vector machine was
applied to the automatic analysis of CE images, which greatly improved the accuracy
and efficiency of diagnosis[89-92]. In addition, Barbosa et al[93] used the CNN to
automatically analyze CE images for the diagnosis of PSIT, which also had high
sensitivity and specificity, reaching 98.7% and 96.6%, respectively.
Risk stratification and prognosis prediction: In different studies, ML was used to
analyze the pathological tissue samples, plasma protein multibiomarker and miRNA
markers of patients with small intestinal neuroendocrine tumors[94-96]. Their studies
provided some new and effective methods for early diagnosis, treatment strategy
selection, prognosis prediction and recurrence risk prediction of small intestinal
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3740 July 7, 2021 Volume 27 Issue 25
Table 4 Applications of artificial intelligence in primary small intestinal tumor
Ref. Diagnostic method AI technology Training set Testing set Outcomes
Inoue et al[88] EGD CNN 531 images 1080 images Accuracy: 94.7%-100%
Liu et al[90] CE SVM 89 patients - Sen: 97.8%, Spe: 96.7%
Vieira et al[89,91] CE SVM 29 patients (936
images)
- This SVM outperforms others by more than
5%
Barbosa et al[93] CE CNN Ep: 104, Cp: 100 Ep: 92, Cp: 100 Sen: 98.7%, Spe: 96.6%
Panarelli et al[94] MicroRNA sequencing ML 84 samples - Accuracy (Ts: 98.5%, Vs: 94.4%)
Drozdov et al[95] Gene expression profiling ML 73 samples - Differentiated from normal cells (Sen: 100%,
Spe: 92%), metastases prediction (Sen:
100%, Spe: 100%)
Kjellman et al[96] Plasma protein
multibiomarker
Random forestmodel Ep:135, Cp: 143 - AUCs: 0.97
Yan et al[97] CT Random forestmodel 213 patients - AUCs: 0.943
AI: Artificial intelligence; AUCs: Area under the curves; CE: Capsule endoscopy; CNN: Convolutional neural network; CT: Computed tomography; Cp:
Control group; EGD: esophagogastroduodenoscopy; Ep: Experimental group; ML: Machine learning; SVM: Support vector machine; Sen: Sensitivity; Spe:
Specificity; Ts: Training set; Vs: Validating set.
neuroendocrine tumors. In the study of Yan et al[97], random forest models were
performed to evaluate the correlation of risk stratification for small intestinal stromal
tumors. Their study suggested multidetector CT texture analysis may become an
important comprehensive tool for preoperative risk stratification of small intestinal
stromal tumors.
AI in other small intestinal diseases
Small intestinal obstruction: Cheng et al[98,99] used CNN to analyze abdominal
radiographs to assist in the diagnosis of small intestinal obstruction (SIO). The
sensitivity and specificity of the CNN diagnostic system were 83.8% and 68.1%,
respectively, based on the training set of 2210 abdominal radiographs. When the
training set was expanded to 7768 abdominal radiographs, the diagnostic sensitivity
and specificity were increased to 91.4% and 91.9%, respectively. Their study suggests
that the accuracy of detection of SIO by CNN improves significantly with the
increasing number of training radiographs. Lucas et al[100] explored the development
of an ML tool for SIO detection based on CT images. They evaluated the accuracy of
eye tracking in image centerline annotation of the small intestine as the first step in the
development of an ML tool for SIO. Their results showed that the eye tracking-based
annotation was accurate and precise enough for application in ML-based small
intestinal centerline annotation.
Small intestinal motor dysfunction: Small bowel intestinal dysfunction (SIMD) can
occur during the development of many diseases, so the evaluation of small intestinal
motor function is an important means for the auxiliary diagnosis and severity
evaluation of these diseases. AI is widely used to evaluate small intestinal motor
function through CE images. Using intraintestinal manometry as the gold standard for
the diagnosis of SIMD, Malagelada et al[101] proved that an ML model was reliable to
evaluate CE images for the diagnosis of SIMD. Applying this model, they found that
29% of patients with functional intestinal disorders had SIMD, significantly higher
than that of the healthy population (3%), confirming the pathophysiological changes in
the intestine of functional intestinal disorders[102]. Furthermore, a classification
method for classifying functional intestinal disorders according to small intestinal
motor function was also proposed[103].
De Iorio et al[104] also demonstrated that ML can reliably detect reduced intestinal
muscle activity and motion by CE images through the method of injecting intestinal
muscle inhibition (glucagon) into healthy subjects. Moreover, the study of Seguí et al
[105] suggested that CNN was also reliable in the description and classification of
small intestine motor characteristics with the classification accuracy reaching 96%.
Malagelada et al[106] also used ML to analyze CE and abdominal MRI images of
patients with cystic fibrosis and confirmed that the delay in small bowel and colonic
transit times in patients with cystic fibrosis is associated with known endocrine
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3741 July 7, 2021 Volume 27 Issue 25
dysfunction and with SIMD.
Small intestinal ischemia-reperfusion injury: Intraoperative evaluation of intestinal
viability in patients with acute intestinal ischemia is a critical factor for surgical
decision making. In the pig jejunum experiment, Strand-Amundsen et al[107]
attempted to apply ML to the analysis of multivariate time-series of bioimpedance
sensor data to analyze intestinal viability after intestinal ischemia-reperfusion. The
results suggested that the measurement should be made before the onset of
reperfusion, and the prediction effect was better when the measurement was repeated
continuously during ischemia and reperfusion. The detection accuracy of irreversible
damage may be close to 100%.
Enteropathies associated with undernutrition: There is a significant histopathological
overlap in duodenal biopsies of enteropathies associated with undernutrition such as
environmental enteropathy and CD. Syed et al[108] used CNN to establish a histopath-
ological analysis model, which can effectively distinguish environmental enteropathy,
CD and normal intestinal mucosa. The detection accuracy was 93.4%, and the false-
negative rate was 2.4%.
CHALLANGES AND PROSPECTS
At present, in order to promote the application of AI in the field of SIDs, we still need
to solve some problems and challenges: (1) Insufficiency of training sample size. The
incidence of most SIDs is not high. For example, the incidence of PSIT is much lower
than that of gastric tumors and colorectal tumors, which affects the amount of data in
the training set. Measurement errors are easy to occur when the sample size is small
[109], and as suggested by the continuity study of Cheng et al[98,99], expanding the
sample size of the training set can significantly improve the inspection accuracy of AI
model; (2) Lack of prospective data. Most of the studies used retrospective data, which
have been artificially screened before AI model training and lack prospective studies;
(3) Single source of data. Most of the training sets and verification sets used in the
study come from single-center data, so it is still necessary to further improve the
repeatability and stability of the model through multicenter data. As in Alzheimer‘s
disease research, each single center should be encouraged to share data in anticipation
of establishing large-scale and open databases[110]; (4) The interpretation of the
results. Due to the inevitable problems of AI, like overfitting of training set data[111]
and the “black box” characteristic of the algorithm[112], the accuracy and
interpretation of the AI model are inconsistent, which may have a negative impact on
clinical application[113]. More intensive basic research and extensive verification are
needed to improve this deficiency; (5) Ethical and legal issues. Can we trust the results
of AI? Once the AI diagnosis and treatment prediction fails, it will give rise to a series
of social, ethical and legal problems[114,115]. It is necessary to combine human
supervision with AI tools more reliably; and (6) AI has been widely researched in
various fields. We should attach importance to learning experience from different
research fields and try to carry out related research in the field of SIDs, so as to
promote the continuous progress of AI research in the field of SIDs.
CONCLUSION
The advantages of AI in the diagnosis and prognosis analysis of SIDs have been
increasingly recognized, and the high accuracy and efficiency of AI detection greatly
reduce the workload of doctors. Although there are still various challenges in the
application of AI, the potential of AI in improving the management efficiency of
diseases cannot be ignored. Clinicians should work together with experts in various
fields to promote the development of AI in SIDs.
REFERENCES
Hamet P, Tremblay J. Artificial intelligence in medicine. Metabolism 2017; 69S: S36-S40 [PMID:
28126242 DOI: 10.1016/j.metabol.2017.01.011]
1
Lee JG, Jun S, Cho YW, Lee H, Kim GB, Seo JB, Kim N. Deep Learning in Medical Imaging:
General Overview. Korean J Radiol 2017; 18: 570-584 [PMID: 28670152 DOI:
2
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3742 July 7, 2021 Volume 27 Issue 25
10.3348/kjr.2017.18.4.570]
He YS, Su JR, Li Z, Zuo XL, Li YQ. Application of artificial intelligence in gastrointestinal
endoscopy. J Dig Dis 2019; 20: 623-630 [PMID: 31639272 DOI: 10.1111/1751-2980.12827]
3
McKinney SM, Sieniek M, Godbole V, Godwin J, Antropova N, Ashrafian H, Back T, Chesus M,
Corrado GS, Darzi A, Etemadi M, Garcia-Vicente F, Gilbert FJ, Halling-Brown M, Hassabis D,
Jansen S, Karthikesalingam A, Kelly CJ, King D, Ledsam JR, Melnick D, Mostofi H, Peng L,
Reicher JJ, Romera-Paredes B, Sidebottom R, Suleyman M, Tse D, Young KC, De Fauw J, Shetty S.
International evaluation of an AI system for breast cancer screening. Nature 2020; 577: 89-94
[PMID: 31894144 DOI: 10.1038/s41586-019-1799-6]
4
Bozkurt S, Gimenez F, Burnside ES, Gulkesen KH, Rubin DL. Using automatically extracted
information from mammography reports for decision-support. J Biomed Inform 2016; 62: 224-231
[PMID: 27388877 DOI: 10.1016/j.jbi.2016.07.001]
5
Niu PH, Zhao LL, Wu HL, Zhao DB, Chen YT. Artificial intelligence in gastric cancer: Application
and future perspectives. World J Gastroenterol 2020; 26: 5408-5419 [PMID: 33024393 DOI:
10.3748/wjg.v26.i36.5408]
6
Wang KW, Dong M. Potential applications of artificial intelligence in colorectal polyps and cancer:
Recent advances and prospects. World J Gastroenterol 2020; 26: 5090-5100 [PMID: 32982111 DOI:
10.3748/wjg.v26.i34.5090]
7
Zhang YH, Guo LJ, Yuan XL, Hu B. Artificial intelligence-assisted esophageal cancer
management: Now and future. World J Gastroenterol 2020; 26: 5256-5271 [PMID: 32994686 DOI:
10.3748/wjg.v26.i35.5256]
8
Kim H, Jung J, Kim J, Cho B, Kwak J, Jang JY, Lee SW, Lee JG, Yoon SM. Abdominal multi-
organ auto-segmentation using 3D-patch-based deep convolutional neural network. Sci Rep 2020;
10: 6204 [PMID: 32277135 DOI: 10.1038/s41598-020-63285-0]
9
Peng Z, Fang X, Yan P, Shan H, Liu T, Pei X, Wang G, Liu B, Kalra MK, Xu XG. A method of
rapid quantification of patient-specific organ doses for CT using deep-learning-based multi-organ
segmentation and GPU-accelerated Monte Carlo dose computing. Med Phys 2020; 47: 2526-2536
[PMID: 32155670 DOI: 10.1002/mp.14131]
10
Tong N, Gou S, Niu T, Yang S, Sheng K. Self-paced DenseNet with boundary constraint for
automated multi-organ segmentation on abdominal CT images. Phys Med Biol 2020; 65: 135011
[PMID: 32657281 DOI: 10.1088/1361-6560/ab9b57]
11
Fu Y, Mazur TR, Wu X, Liu S, Chang X, Lu Y, Li HH, Kim H, Roach MC, Henke L, Yang D. A
novel MRI segmentation method using CNN-based correction network for MRI-guided adaptive
radiotherapy. Med Phys 2018; 45: 5129-5137 [PMID: 30269345 DOI: 10.1002/mp.13221]
12
Chen Y, Ruan D, Xiao J, Wang L, Sun B, Saouaf R, Yang W, Li D, Fan Z. Fully automated
multiorgan segmentation in abdominal magnetic resonance imaging with deep neural networks. Med
Phys 2020; 47: 4971-4982 [PMID: 32748401 DOI: 10.1002/mp.14429]
13
Wilson NA, Park HS, Lee KS, Barron LK, Warner BW. A Novel Approach to Calculating Small
Intestine Length Based on Magnetic Resonance Enterography. J Am Coll Surg 2017; 225: 266-273.
e1 [PMID: 28445795 DOI: 10.1016/j.jamcollsurg.2017.04.009]
14
Takiyama H, Ozawa T, Ishihara S, Fujishiro M, Shichijo S, Nomura S, Miura M, Tada T.
Automatic anatomical classification of esophagogastroduodenoscopy images using deep
convolutional neural networks. Sci Rep 2018; 8: 7497 [PMID: 29760397 DOI:
10.1038/s41598-018-25842-6]
15
Igarashi S, Sasaki Y, Mikami T, Sakuraba H, Fukuda S. Anatomical classification of upper
gastrointestinal organs under various image capture conditions using AlexNet. Comput Biol Med
2020; 124: 103950 [PMID: 32798923 DOI: 10.1016/j.compbiomed.2020.103950]
16
Iddan G, Meron G, Glukhovsky A, Swain P. Wireless capsule endoscopy. Nature 2000; 405: 417
[PMID: 10839527 DOI: 10.1038/35013140]
17
Kopylov U, Seidman EG. Diagnostic modalities for the evaluation of small bowel disorders. Curr
Opin Gastroenterol 2015; 31: 111-117 [PMID: 25635667 DOI: 10.1097/MOG.0000000000000159]
18
Eliakim R. Video capsule endoscopy of the small bowel. Curr Opin Gastroenterol 2008; 24: 159-
163 [PMID: 18301265 DOI: 10.1097/MOG.0b013e3282f3d946]
19
Kopylov U, Seidman EG. Clinical applications of small bowel capsule endoscopy. Clin Exp
Gastroenterol 2013; 6: 129-137 [PMID: 23983481 DOI: 10.2147/CEG.S48005]
20
Oh DJ, Kim KS, Lim YJ. A New Active Locomotion Capsule Endoscopy under Magnetic Control
and Automated Reading Program. Clin Endosc 2020; 53: 395-401 [PMID: 32746536 DOI:
10.5946/ce.2020.127]
21
Beg S, Wronska E, Araujo I, González Suárez B, Ivanova E, Fedorov E, Aabakken L, Seitz U, Rey
JF, Saurin JC, Tari R, Card T, Ragunath K. Use of rapid reading software to reduce capsule
endoscopy reading times while maintaining accuracy. Gastrointest Endosc 2020; 91: 1322-1327
[PMID: 31981645 DOI: 10.1016/j.gie.2020.01.026]
22
Pérez-Cuadrado-Robles E, Pinho R, Gonzalez B, Mão de Ferro S, Chagas C, Esteban Delgado P,
Carretero C, Figueiredo P, Rosa B, García Lledó J, Nogales Ó, Ponte A, Andrade P, Juanmartiñena-
Fernández JF, San-Juan-Acosta M, Lopes S, Prieto-Frías C, Egea-Valenzuela J, Caballero N,
Valdivieso-Cortazar E, Cardoso H, Gálvez C, Almeida N, Borque Barrera P, Gómez-Rodríguez BJ,
Sánchez Ceballos F, Bernardes C, Alonso P, Argüelles-Arias F, Mascarenhas Saraiva M, Pérez-
Cuadrado-Martínez E. Small Bowel Enteroscopy - A Joint Clinical Guideline from the Spanish and
Portuguese Small Bowel Study Groups. GE Port J Gastroenterol 2020; 27: 324-335 [PMID:
23
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3743 July 7, 2021 Volume 27 Issue 25
32999905 DOI: 10.1159/000507375]
Gomes C, Pinho R, Ponte A, Rodrigues A, Sousa M, Silva JC, Afecto E, Carvalho J. Evaluation of
the sensitivity of the Express View function in the Mirocam® capsule endoscopy software. Scand J
Gastroenterol 2020; 55: 371-375 [PMID: 32150486 DOI: 10.1080/00365521.2020.1734650]
24
Oumrani S, Histace A, Abou Ali E, Pietri O, Becq A, Houist G, Nion-Larmurier I, Camus M,
Florent C, Dray X. Multi-criterion, automated, high-performance, rapid tool for assessing mucosal
visualization quality of still images in small bowel capsule endoscopy. Endosc Int Open 2019; 7:
E944-E948 [PMID: 31367673 DOI: 10.1055/a-0918-5883]
25
Ding Z, Shi H, Zhang H, Meng L, Fan M, Han C, Zhang K, Ming F, Xie X, Liu H, Liu J, Lin R,
Hou X. Gastroenterologist-Level Identification of Small-Bowel Diseases and Normal Variants by
Capsule Endoscopy Using a Deep-Learning Model. Gastroenterology 2019; 157: 1044-1054. e5
[PMID: 31251929 DOI: 10.1053/j.gastro.2019.06.025]
26
Karargyris A, Bourbakis N. Detection of small bowel polyps and ulcers in wireless capsule
endoscopy videos. IEEE Trans Biomed Eng 2011; 58: 2777-2786 [PMID: 21592915 DOI:
10.1109/TBME.2011.2155064]
27
Hwang Y, Lee HH, Park C, Tama BA, Kim JS, Cheung DY, Chung WC, Cho YS, Lee KM, Choi
MG, Lee S, Lee BI. Improved classification and localization approach to small bowel capsule
endoscopy using convolutional neural network. Dig Endosc 2020 [PMID: 32640059 DOI:
10.1111/den.13787]
28
Otani K, Nakada A, Kurose Y, Niikura R, Yamada A, Aoki T, Nakanishi H, Doyama H, Hasatani
K, Sumiyoshi T, Kitsuregawa M, Harada T, Koike K. Automatic detection of different types of
small-bowel lesions on capsule endoscopy images using a newly developed deep convolutional
neural network. Endoscopy 2020; 52: 786-791 [PMID: 32557474 DOI: 10.1055/a-1167-8157]
29
Aoki T, Yamada A, Aoyama K, Saito H, Fujisawa G, Odawara N, Kondo R, Tsuboi A, Ishibashi R,
Nakada A, Niikura R, Fujishiro M, Oka S, Ishihara S, Matsuda T, Nakahori M, Tanaka S, Koike K,
Tada T. Clinical usefulness of a deep learning-based system as the first screening on small-bowel
capsule endoscopy reading. Dig Endosc 2020; 32: 585-591 [PMID: 31441972 DOI:
10.1111/den.13517]
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 [PMID: 30670179 DOI: 10.1016/j.gie.2018.10.027]
31
Fan S, Xu L, Fan Y, Wei K, Li L. Computer-aided detection of small intestinal ulcer and erosion in
wireless capsule endoscopy images. Phys Med Biol 2018; 63: 165001 [PMID: 30033931 DOI:
10.1088/1361-6560/aad51c]
32
Fan GW, Chen TH, Lin WP, Su MY, Sung CM, Hsu CM, Chi CT. Angiodysplasia and bleeding in
the small intestine treated by balloon-assisted enteroscopy. J Dig Dis 2013; 14: 113-116 [PMID:
23216888 DOI: 10.1111/1751-2980.12021]
33
Aoki T, Yamada A, Kato Y, Saito H, Tsuboi A, Nakada A, Niikura R, Fujishiro M, Oka S, Ishihara
S, Matsuda T, Nakahori M, Tanaka S, Koike K, Tada T. Automatic detection of various
abnormalities in capsule endoscopy videos by a deep learning-based system: a multicenter study.
Gastrointest Endosc 2021; 93: 165-173. e1 [PMID: 32417297 DOI: 10.1016/j.gie.2020.04.080]
34
Leenhardt R, Li C, Le Mouel JP, Rahmi G, Saurin JC, Cholet F, Boureille A, Amiot X, Delvaux M,
Duburque C, Leandri C, Gérard R, Lecleire S, Mesli F, Nion-Larmurier I, Romain O, Sacher-
Huvelin S, Simon-Shane C, Vanbiervliet G, Marteau P, Histace A, Dray X. CAD-CAP: a 25,000-
image database serving the development of artificial intelligence for capsule endoscopy. Endosc Int
Open 2020; 8: E415-E420 [PMID: 32118115 DOI: 10.1055/a-1035-9088]
35
Vezakis IA, Toumpaniaris P, Polydorou AA, Koutsouris D. A Novel Real-time Automatic
Angioectasia Detection Method in Wireless Capsule Endoscopy Video Feed. Annu Int Conf IEEE
Eng Med Biol Soc 2019; 2019: 4072-4075 [PMID: 31946766 DOI: 10.1109/EMBC.2019.8857445]
36
Tsuboi A, Oka S, Aoyama K, Saito H, Aoki T, Yamada A, Matsuda T, Fujishiro M, Ishihara S,
Nakahori M, Koike K, Tanaka S, Tada T. Artificial intelligence using a convolutional neural
network for automatic detection of small-bowel angioectasia in capsule endoscopy images. Dig
Endosc 2020; 32: 382-390 [PMID: 31392767 DOI: 10.1111/den.13507]
37
Vieira PM, Silva CP, Costa D, Vaz IF, Rolanda C, Lima CS. Automatic Segmentation and
Detection of Small Bowel Angioectasias in WCE Images. Ann Biomed Eng 2019; 47: 1446-1462
[PMID: 30919139 DOI: 10.1007/s10439-019-02248-7]
38
Leenhardt R, Vasseur P, Li C, Saurin JC, Rahmi G, Cholet F, Becq A, Marteau P, Histace A, Dray
X; CAD-CAP Database Working Group. A neural network algorithm for detection of GI angiectasia
during small-bowel capsule endoscopy. Gastrointest Endosc 2019; 89: 189-194 [PMID: 30017868
DOI: 10.1016/j.gie.2018.06.036]
39
Aoki T, Yamada A, Kato Y, Saito H, Tsuboi A, Nakada A, Niikura R, Fujishiro M, Oka S, Ishihara
S, Matsuda T, Nakahori M, Tanaka S, Koike K, Tada T. Automatic detection of blood content in
capsule endoscopy images based on a deep convolutional neural network. J Gastroenterol Hepatol
2020; 35: 1196-1200 [PMID: 31758717 DOI: 10.1111/jgh.14941]
40
Arieira C, Monteiro S, Dias de Castro F, Boal Carvalho P, Rosa B, Moreira MJ, Cotter J. Capsule
endoscopy: Is the software TOP 100 a reliable tool in suspected small bowel bleeding? Dig Liver Dis
2019; 51: 1661-1664 [PMID: 31281069 DOI: 10.1016/j.dld.2019.06.008]
41
Xiao Jia, Meng MQ. A deep convolutional neural network for bleeding detection in Wireless 42
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3744 July 7, 2021 Volume 27 Issue 25
Capsule Endoscopy images. Annu Int Conf IEEE Eng Med Biol Soc 2016; 2016: 639-642 [PMID:
28268409 DOI: 10.1109/EMBC.2016.7590783]
Han S, Fahed J, Cave DR. Suspected Blood Indicator to Identify Active Gastrointestinal Bleeding:
A Prospective Validation. Gastroenterology Res 2018; 11: 106-111 [PMID: 29707077 DOI:
10.14740/gr949w]
43
Liu DY, Gan T, Rao NN, Xing YW, Zheng J, Li S, Luo CS, Zhou ZJ, Wan YL. Identification of
lesion images from gastrointestinal endoscope based on feature extraction of combinational methods
with and without learning process. Med Image Anal 2016; 32: 281-294 [PMID: 27236223 DOI:
10.1016/j.media.2016.04.007]
44
Saito H, Aoki T, Aoyama K, Kato Y, Tsuboi A, Yamada A, Fujishiro M, Oka S, Ishihara S,
Matsuda T, Nakahori M, Tanaka S, Koike K, Tada T. Automatic detection and classification of
protruding lesions in wireless capsule endoscopy images based on a deep convolutional neural
network. Gastrointest Endosc 2020; 92: 144-151. e1 [PMID: 32084410 DOI:
10.1016/j.gie.2020.01.054]
45
Li B, Meng MQ, Xu L. A comparative study of shape features for polyp detection in wireless
capsule endoscopy images. Annu Int Conf IEEE Eng Med Biol Soc 2009; 2009: 3731-3734 [PMID:
19965014 DOI: 10.1109/IEMBS.2009.5334875]
46
Ciaccio EJ, Bhagat G, Lewis SK, Green PH. Extraction and processing of videocapsule data to
detect and measure the presence of villous atrophy in celiac disease patients. Comput Biol Med 2016;
78: 97-106 [PMID: 27673492 DOI: 10.1016/j.compbiomed.2016.09.009]
47
Das A, Wong RC. Prediction of outcome in acute lower gastrointestinal hemorrhage: role of
artificial neural network. Eur J Gastroenterol Hepatol 2007; 19: 1064-1069 [PMID: 17998830 DOI:
10.1097/MEG.0b013e3282f198f7]
48
Nemytin IuV, Petrov VP, Zuev VK, Osipov VV, Esin SV, Baryshev SS. [The use of artificial
neuronal networks in the treatment of peptic ulcer]. Voen Med Zh 2000; 321: 40-44, 96 [PMID:
10929514]
49
Wong GL, Ma AJ, Deng H, Ching JY, Wong VW, Tse YK, Yip TC, Lau LH, Liu HH, Leung CM,
Tsang SW, Chan CW, Lau JY, Yuen PC, Chan FK. Machine learning model to predict recurrent
ulcer bleeding in patients with history of idiopathic gastroduodenal ulcer bleeding. Aliment
Pharmacol Ther 2019; 49: 912-918 [PMID: 30761584 DOI: 10.1111/apt.15145]
50
Axelrad JE, Faye AS, Pinsino A, Thanataveerat A, Cagliostro B, Pineda MFT, Ross K, Te-Frey RT,
Effner L, Garan AR, Topkara VK, Takayama H, Takeda K, Naka Y, Ramirez I, Garcia-Carrasquillo
R, Colombo PC, Gonda T, Yuzefpolskaya M. Endoscopic Algorithm for Management of
Gastrointestinal Bleeding in Patients With Continuous Flow LVADs: A Prospective Validation
Study. J Card Fail 2020; 26: 324-332 [PMID: 31794863 DOI: 10.1016/j.cardfail.2019.11.027]
51
Bashar MK, Kitasaka T, Suenaga Y, Mekada Y, Mori K. Automatic detection of informative frames
from wireless capsule endoscopy images. Med Image Anal 2010; 14: 449-470 [PMID: 20137998
DOI: 10.1016/j.media.2009.12.001]
52
Pietri O, Rezgui G, Histace A, Camus M, Nion-Larmurier I, Li C, Becq A, Ali EA, Romain O,
Chaput U, Marteau P, Florent C, Dray X. Development and validation of an automated algorithm to
evaluate the abundance of bubbles in small bowel capsule endoscopy. Endosc Int Open 2018; 6:
E462-E469 [PMID: 29616238 DOI: 10.1055/a-0573-1044]
53
Klein A, Gizbar M, Bourke MJ, Ahlenstiel G. Validated computed cleansing score for video capsule
endoscopy. Dig Endosc 2016; 28: 564-569 [PMID: 26716407 DOI: 10.1111/den.12599]
54
Ludvigsson JF, Card TR, Kaukinen K, Bai J, Zingone F, Sanders DS, Murray JA. Screening for
celiac disease in the general population and in high-risk groups. United European Gastroenterol J
2015; 3: 106-120 [PMID: 25922671 DOI: 10.1177/2050640614561668]
55
Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease: time for a
standardized report scheme for pathologists. Eur J Gastroenterol Hepatol 1999; 11: 1185-1194
[PMID: 10524652 DOI: 10.1097/00042737-199910000-00019]
56
Molder A, Balaban DV, Jinga M, Molder CC. Current Evidence on Computer-Aided Diagnosis of
Celiac Disease: Systematic Review. Front Pharmacol 2020; 11: 341 [PMID: 32372947 DOI:
10.3389/fphar.2020.00341]
57
Gadermayr M, Wimmer G, Kogler H, Vécsei A, Merhof D, Uhl A. Automated classification of
celiac disease during upper endoscopy: Status quo and quo vadis. Comput Biol Med 2018; 102: 221-
226 [PMID: 29739614 DOI: 10.1016/j.compbiomed.2018.04.020]
58
Gadermayr M, Kogler H, Karla M, Merhof D, Uhl A, Vécsei A. Computer-aided texture analysis
combined with experts’ knowledge: Improving endoscopic celiac disease diagnosis. World J
Gastroenterol 2016; 22: 7124-7134 [PMID: 27610022 DOI: 10.3748/wjg.v22.i31.7124]
59
Tenório JM, Hummel AD, Cohrs FM, Sdepanian VL, Pisa IT, de Fátima Marin H. Artificial
intelligence techniques applied to the development of a decision-support system for diagnosing
celiac disease. Int J Med Inform 2011; 80: 793-802 [PMID: 21917512 DOI:
10.1016/j.ijmedinf.2011.08.001]
60
Wimmer G, Vécsei A, Häfner M, Uhl A. Fisher encoding of convolutional neural network features
for endoscopic image classification. J Med Imaging (Bellingham) 2018; 5: 034504 [PMID:
30840751 DOI: 10.1117/1.JMI.5.3.034504]
61
Chetcuti Zammit S, Bull LA, Sanders DS, Galvin J, Dervilis N, Sidhu R, Worden K. Towards the
Probabilistic Analysis of Small Bowel Capsule Endoscopy Features to Predict Severity of Duodenal
Histology in Patients with Villous Atrophy. J Med Syst 2020; 44: 195 [PMID: 33005996 DOI:
62
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3745 July 7, 2021 Volume 27 Issue 25
10.1007/s10916-020-01657-9]
Li BNN, Wang X, Wang R, Zhou T, Gao R, Ciaccio EJ, Green PH. Celiac Disease Detection from
Videocapsule Endoscopy Images Using Strip Principal Component Analysis. IEEE/ACM Trans
Comput Biol Bioinform 2019; PP [PMID: 31751282 DOI: 10.1109/TCBB.2019.2953701]
63
Vicnesh J, Wei JKE, Ciaccio EJ, Oh SL, Bhagat G, Lewis SK, Green PH, Acharya UR. Automated
diagnosis of celiac disease by video capsule endoscopy using DAISY Descriptors. J Med Syst 2019;
43: 157 [PMID: 31028562 DOI: 10.1007/s10916-019-1285-6]
64
Zhou T, Han G, Li BN, Lin Z, Ciaccio EJ, Green PH, Qin J. Quantitative analysis of patients with
celiac disease by video capsule endoscopy: A deep learning method. Comput Biol Med 2017; 85: 1-6
[PMID: 28412572 DOI: 10.1016/j.compbiomed.2017.03.031]
65
Wei JW, Wei JW, Jackson CR, Ren B, Suriawinata AA, Hassanpour S. Automated Detection of
Celiac Disease on Duodenal Biopsy Slides: A Deep Learning Approach. J Pathol Inform 2019; 10: 7
[PMID: 30984467 DOI: 10.4103/jpi.jpi_87_18]
66
Das P, Gahlot GP, Singh A, Baloda V, Rawat R, Verma AK, Khanna G, Roy M, George A, Nalwa
A, Ramteke P, Yadav R, Ahuja V, Sreenivas V, Gupta SD, Makharia GK. Quantitative histology-
based classification system for assessment of the intestinal mucosal histological changes in patients
with celiac disease. Intest Res 2019; 17: 387-397 [PMID: 30996219 DOI: 10.5217/ir.2018.00167]
67
Virta J, Hannula M, Tamminen I, Lindfors K, Kaukinen K, Popp A, Taavela J, Saavalainen P,
Hiltunen P, Hyttinen J, Kurppa K. X-ray microtomography is a novel method for accurate evaluation
of small-bowel mucosal morphology and surface area. Sci Rep 2020; 10: 13164 [PMID: 32753621
DOI: 10.1038/s41598-020-69487-w]
68
Sangineto M, Graziano G, D’Amore S, Salvia R, Palasciano G, Sabbà C, Vacca M, Cariello M.
Identification of peculiar gene expression profile in peripheral blood mononuclear cells (PBMC) of
celiac patients on gluten free diet. PLoS One 2018; 13: e0197915 [PMID: 29795662 DOI:
10.1371/journal.pone.0197915]
69
Pastore RL, Murray JA, Coffman FD, Mitrofanova A, Srinivasan S. Physician Review of a Celiac
Disease Risk Estimation and Decision-Making Expert System. J Am Coll Nutr 2019; 38: 722-728
[PMID: 31063433 DOI: 10.1080/07315724.2019.1608477]
70
Feuerstein JD, Cheifetz AS. Crohn Disease: Epidemiology, Diagnosis, and Management. Mayo
Clin Proc 2017; 92: 1088-1103 [PMID: 28601423 DOI: 10.1016/j.mayocp.2017.04.010]
71
Nóbrega VG, Silva INN, Brito BS, Silva J, Silva MCMD, Santana GO. The onset of clinical
manifestations in inflammatory bowel disease patients. Arq Gastroenterol 2018; 55: 290-295
[PMID: 30540094 DOI: 10.1590/S0004-2803.201800000-73]
72
Lazarev M, Huang C, Bitton A, Cho JH, Duerr RH, McGovern DP, Proctor DD, Regueiro M, Rioux
JD, Schumm PP, Taylor KD, Silverberg MS, Steinhart AH, Hutfless S, Brant SR. Relationship
between proximal Crohn’s disease location and disease behavior and surgery: a cross-sectional study
of the IBD Genetics Consortium. Am J Gastroenterol 2013; 108: 106-112 [PMID: 23229423 DOI:
10.1038/ajg.2012.389]
73
Lamash Y, Kurugol S, Warfield SK. Semi-Automated Extraction of Crohns Disease MR Imaging
Markers using a 3D Residual CNN with Distance Prior. Deep Learn Med Image Anal Multimodal
Learn Clin Decis Support (2018) 2018; 11045: 218-226 [PMID: 30450491 DOI:
10.1007/978-3-030-00889-5_25]
74
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 [PMID: 30353957 DOI: 10.1002/jmri.26330]
75
Parfеnov АI, Аkopova АО, Shcherbakov PL, Мikcheeva ОМ. Role of video capsulе endoscopy in
the diagnostic algorithm of small bowel Crohn’s disease. Ter Arkh 2019; 91: 37-42 [PMID:
31094474 DOI: 10.26442/00403660.2019.04.000079]
76
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 [PMID: 31743689 DOI:
10.1016/j.gie.2019.11.012]
77
Yang S, Lemke C, Cox BF, Newton IP, Nathke I, Cochran S. A Learning-Based Microultrasound
System for the Detection of Inflammation of the Gastrointestinal Tract. IEEE Trans Med Imaging
2021; 40: 38-47 [PMID: 32881684 DOI: 10.1109/TMI.2020.3021560]
78
Taylor KM, Hanscombe KB, Prescott NJ, Iniesta R, Traylor M, Taylor NS, Fong S, Powell N,
Irving PM, Anderson SH, Mathew CG, Lewis CM, Sanderson JD. Genetic and Inflammatory
Biomarkers Classify Small Intestine Inflammation in Asymptomatic First-degree Relatives of
Patients With Crohn’s Disease. Clin Gastroenterol Hepatol 2020; 18: 908-916. e13 [PMID:
31202982 DOI: 10.1016/j.cgh.2019.05.061]
79
Shen EX, Lord A, Doecke JD, Hanigan K, Irwin J, Cheng RKY, Radford-Smith G. A validated risk
stratification tool for detecting high-risk small bowel Crohn’s disease. Aliment Pharmacol Ther
2020; 51: 281-290 [PMID: 31769537 DOI: 10.1111/apt.15550]
80
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 [PMID: 31212952 DOI: 10.3390/jcm8060865]
81
Menti E, Lanera C, Lorenzoni G, Giachino DF, Marchi M, Gregori D, Berchialla P; Piedmont 82
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3746 July 7, 2021 Volume 27 Issue 25
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 [PMID: 28269885]
Mellouki I, Jellali K, Ibrahimi A. [Tumors of the small bowel: about 27 cases]. Pan Afr Med J 2018;
30: 13 [PMID: 30167041 DOI: 10.11604/pamj.2018.30.13.5407]
83
Sarosiek T, Stelmaszuk M. [Small intestine neoplasms]. Pol Merkur Lekarski 2018; 44: 45-48
[PMID: 29498365]
84
Tran TB, Qadan M, Dua MM, Norton JA, Poultsides GA, Visser BC. Prognostic relevance of
lymph node ratio and total lymph node count for small bowel adenocarcinoma. Surgery 2015; 158:
486-493 [PMID: 26013988 DOI: 10.1016/j.surg.2015.03.048]
85
Zhang Y, Zulfiqar M, Bluth MH, Bhalla A, Beydoun R. Molecular Diagnostics in the Neoplasms of
Small Intestine and Appendix: 2018 Update. Clin Lab Med 2018; 38: 343-355 [PMID: 29776634
DOI: 10.1016/j.cll.2018.03.002]
86
Zhao Z, Guan X, Chen Y, Wang X. [Progression of diagnosis and treatment in primary malignant
small bowel tumor]. Zhonghua Wei Chang Wai Ke Za Zhi 2017; 20: 117-120 [PMID: 28105627]
87
Inoue S, Shichijo S, Aoyama K, Kono M, Fukuda H, Shimamoto Y, Nakagawa K, Ohmori M,
Iwagami H, Matsuno K, Iwatsubo T, Nakahira H, Matsuura N, Maekawa A, Kanesaka T, Yamamoto
S, Takeuchi Y, Higashino K, Uedo N, Ishihara R, Tada T. Application of Convolutional Neural
Networks for Detection of Superficial Nonampullary Duodenal Epithelial Tumors in
Esophagogastroduodenoscopic Images. Clin Transl Gastroenterol 2020; 11: e00154 [PMID:
32352719 DOI: 10.14309/ctg.0000000000000154]
88
Vieira PM, Ramos J, Lima CS. Automatic detection of small bowel tumors in endoscopic capsule
images by ROI selection based on discarded lightness information. Annu Int Conf IEEE Eng Med
Biol Soc 2015; 2015: 3025-3028 [PMID: 26736929 DOI: 10.1109/EMBC.2015.7319029]
89
Liu G, Yan G, Kuang S, Wang Y. Detection of small bowel tumor based on multi-scale curvelet
analysis and fractal technology in capsule endoscopy. Comput Biol Med 2016; 70: 131-138 [PMID:
26829705 DOI: 10.1016/j.compbiomed.2016.01.021]
90
Vieira PM, Freitas NR, Valente J, Vaz IF, Rolanda C, Lima CS. Automatic detection of small bowel
tumors in wireless capsule endoscopy images using ensemble learning. Med Phys 2020; 47: 52-63
[PMID: 31299096 DOI: 10.1002/mp.13709]
91
Li BP, Meng MQ. Comparison of several texture features for tumor detection in CE images. J Med
Syst 2012; 36: 2463-2469 [PMID: 21523427 DOI: 10.1007/s10916-011-9713-2]
92
Barbosa DJ, Ramos J, Lima CS. Detection of small bowel tumors in capsule endoscopy frames
using texture analysis based on the discrete wavelet transform. Annu Int Conf IEEE Eng Med Biol
Soc 2008; 2008: 3012-3015 [PMID: 19163340 DOI: 10.1109/IEMBS.2008.4649837]
93
Panarelli N, Tyryshkin K, Wong JJM, Majewski A, Yang X, Scognamiglio T, Kim MK, Bogardus
K, Tuschl T, Chen YT, Renwick N. Evaluating gastroenteropancreatic neuroendocrine tumors
through microRNA sequencing. Endocr Relat Cancer 2019; 26: 47-57 [PMID: 30021866 DOI:
10.1530/ERC-18-0244]
94
Drozdov I, Kidd M, Nadler B, Camp RL, Mane SM, Hauso O, Gustafsson BI, Modlin IM.
Predicting neuroendocrine tumor (carcinoid) neoplasia using gene expression profiling and
supervised machine learning. Cancer 2009; 115: 1638-1650 [PMID: 19197975 DOI:
10.1002/cncr.24180]
95
Kjellman M, Knigge U, Welin S, Thiis-Evensen E, Gronbæk H, Schalin-Jäntti C, Sorbye H,
Joergensen MT, Johanson V, Metso S, Waldum H, Søreide JA, Ebeling T, Lindberg F, Landerholm
K, Wallin G, Salem F, Schneider MDP, Belusa R. A plasma protein biomarker strategy for detection
of small intestinal neuroendocrine tumors. Neuroendocrinology 2020 [PMID: 32721955 DOI:
10.1159/000510483]
96
Yan J, Zhao X, Han S, Wang T, Miao F. Evaluation of Clinical Plus Imaging Features and
Multidetector Computed Tomography Texture Analysis in Preoperative Risk Grade Prediction of
Small Bowel Gastrointestinal Stromal Tumors. J Comput Assist Tomogr 2018; 42: 714-720 [PMID:
30015796 DOI: 10.1097/RCT.0000000000000756]
97
Cheng PM, Tejura TK, Tran KN, Whang G. Detection of high-grade small bowel obstruction on
conventional radiography with convolutional neural networks. Abdom Radiol (NY) 2018; 43: 1120-
1127 [PMID: 28828625 DOI: 10.1007/s00261-017-1294-1]
98
Cheng PM, Tran KN, Whang G, Tejura TK. Refining Convolutional Neural Network Detection of
Small-Bowel Obstruction in Conventional Radiography. AJR Am J Roentgenol 2019; 212: 342-350
[PMID: 30476452 DOI: 10.2214/AJR.18.20362]
99
Lucas A, Wang K, Santillan C, Hsiao A, Sirlin CB, Murphy PM. Image Annotation by Eye
Tracking: Accuracy and Precision of Centerlines of Obstructed Small-Bowel Segments Placed Using
Eye Trackers. J Digit Imaging 2019; 32: 855-864 [PMID: 31144146 DOI:
10.1007/s10278-018-0169-5]
100
Malagelada C, De Iorio F, Azpiroz F, Accarino A, Segui S, Radeva P, Malagelada JR. New insight
into intestinal motor function via noninvasive endoluminal image analysis. Gastroenterology 2008;
135: 1155-1162 [PMID: 18691579 DOI: 10.1053/j.gastro.2008.06.084]
101
Malagelada C, De Lorio F, Seguí S, Mendez S, Drozdzal M, Vitria J, Radeva P, Santos J, Accarino
A, Malagelada JR, Azpiroz F. Functional gut disorders or disordered gut function?
Neurogastroenterol Motil 2012; 24: 223-228, e104 [PMID: 22129212 DOI:
10.1111/j.1365-2982.2011.01823.x]
102
Yang Y et al. Artificial intelligence in small intestinal diseases
WJG https://www.wjgnet.com 3747 July 7, 2021 Volume 27 Issue 25
Malagelada C, Drozdzal M, Seguí S, Mendez S, Vitrià J, Radeva P, Santos J, Accarino A,
Malagelada JR, Azpiroz F. Classification of functional bowel disorders by objective physiological
criteria based on endoluminal image analysis. Am J Physiol Gastrointest Liver Physiol 2015; 309:
G413-G419 [PMID: 26251472 DOI: 10.1152/ajpgi.00193.2015]
103
de Iorio F, Malagelada C, Azpiroz F, Maluenda M, Violanti C, Igual L, Vitrià J, Malagelada JR.
Intestinal motor activity, endoluminal motion and transit. Neurogastroenterol Motil 2009; 21: 1264-
e119 [PMID: 19614865 DOI: 10.1111/j.1365-2982.2009.01363.x]
104
Seguí S, Drozdzal M, Pascual G, Radeva P, Malagelada C, Azpiroz F, Vitrià J. Generic feature
learning for wireless capsule endoscopy analysis. Comput Biol Med 2016; 79: 163-172 [PMID:
27810622 DOI: 10.1016/j.compbiomed.2016.10.011]
105
Malagelada C, Bendezú RA, Seguí S, Vitrià J, Merino X, Nieto A, Sihuay D, Accarino A, Molero
X, Azpiroz F. Motor dysfunction of the gut in cystic fibrosis. Neurogastroenterol Motil 2020; 32:
e13883 [PMID: 32475007 DOI: 10.1111/nmo.13883]
106
Strand-Amundsen RJ, Tronstad C, Reims HM, Reinholt FP, Høgetveit JO, Tønnessen TI. Machine
learning for intraoperative prediction of viability in ischemic small intestine. Physiol Meas 2018; 39:
105011 [PMID: 30207981 DOI: 10.1088/1361-6579/aae0ea]
107
Syed S, Al-Boni M, Khan MN, Sadiq K, Iqbal NT, Moskaluk CA, Kelly P, Amadi B, Ali SA, Moore
SR, Brown DE. Assessment of Machine Learning Detection of Environmental Enteropathy and
Celiac Disease in Children. JAMA Netw Open 2019; 2: e195822 [PMID: 31199451 DOI:
10.1001/jamanetworkopen.2019.5822]
108
Loken E, Gelman A. Measurement error and the replication crisis. Science 2017; 355: 584-585
[PMID: 28183939 DOI: 10.1126/science.aal3618]
109
Jack CR Jr, Bernstein MA, Fox NC, Thompson P, Alexander G, Harvey D, Borowski B, Britson
PJ, L Whitwell J, Ward C, Dale AM, Felmlee JP, Gunter JL, Hill DL, Killiany R, Schuff N, Fox-
Bosetti S, Lin C, Studholme C, DeCarli CS, Krueger G, Ward HA, Metzger GJ, Scott KT, Mallozzi
R, Blezek D, Levy J, Debbins JP, Fleisher AS, Albert M, Green R, Bartzokis G, Glover G, Mugler J,
Weiner MW. The Alzheimer’s Disease Neuroimaging Initiative (ADNI): MRI methods. J Magn
Reson Imaging 2008; 27: 685-691 [PMID: 18302232 DOI: 10.1002/jmri.21049]
110
Chartrand G, Cheng PM, Vorontsov E, Drozdzal M, Turcotte S, Pal CJ, Kadoury S, Tang A. Deep
Learning: A Primer for Radiologists. Radiographics 2017; 37: 2113-2131 [PMID: 29131760 DOI:
10.1148/rg.2017170077]
111
Lawrence DR, Palacios-González C, Harris J. Artificial Intelligence. Camb Q Healthc Ethics 2016;
25: 250-261 [PMID: 26957450 DOI: 10.1017/S0963180115000559]
112
Cabitza F, Rasoini R, Gensini GF. Unintended Consequences of Machine Learning in Medicine.
JAMA 2017; 318: 517-518 [PMID: 28727867 DOI: 10.1001/jama.2017.7797]
113
Yu KH, Beam AL, Kohane IS. Artificial intelligence in healthcare. Nat Biomed Eng 2018; 2: 719-
731 [PMID: 31015651 DOI: 10.1038/s41551-018-0305-z]
114
O’Sullivan S, Nevejans N, Allen C, Blyth A, Leonard S, Pagallo U, Holzinger K, Holzinger A,
Sajid MI, Ashrafian H. Legal, regulatory, and ethical frameworks for development of standards in
artificial intelligence (AI) and autonomous robotic surgery. Int J Med Robot 2019; 15: e1968 [PMID:
30397993 DOI: 10.1002/rcs.1968]
115
Published by Baishideng Publishing Group Inc
7041 Koll Center Parkway, Suite 160, Pleasanton, CA 94566, USA
Telephone: +1-925-3991568
E-mail: bpgoffice@wjgnet.com
Help Desk: https://www.f6publishing.com/helpdesk
https://www.wjgnet.com
© 2021 Baishideng Publishing Group Inc. All rights reserved.
... The clinical utility of segmenting the bowel includes several oncologic applications, like pre-radiation planning before pelvic radiation therapy. Other uses, like the detection and classification of inflammatory bowel disease, mucosal lesions, or bowel obstruction, have also been explored [23]. ...
Article
Full-text available
Artificial intelligence (AI) has been a topic of substantial interest for radiologists in recent years. Although many of the first clinical applications were in the neuro, cardiothoracic, and breast imaging subspecialties, the number of investigated and real-world applications of body imaging has been increasing, with more than 30 FDA-approved algorithms now available for applications in the abdomen and pelvis. In this manuscript, we explore some of the fundamentals of artificial intelligence and machine learning, review major functions that AI algorithms may perform, introduce current and potential future applications of AI in abdominal imaging, provide a basic understanding of the pathways by which AI algorithms can receive FDA approval, and explore some of the challenges with the implementation of AI in clinical practice.
... Beyond observer bias, patch testing also demands valuable human resources, a challenge that can be particularly pronounced in large clinics. To address similar challenges in medical settings, researchers have proposed the use of deep learning models [7][8][9]. However, the application of deep learning to automate the detection of skin reactions in patch testing remains largely unexplored. ...
Article
Full-text available
Epicutaneous patch testing is a well-established diagnostic method for identifying substances that may cause Allergic Contact Dermatitis (ACD), a common skin condition caused by exposure to environmental allergens. While the patch test remains the gold standard for identifying allergens, it is prone to observer bias and consumes valuable human resources. Deep learning models can be employed to address this challenge. In this study, we collected a dataset of 1579 multi-modal skin images from 200 patients using the Antera 3D® camera. We then investigated the feasibility of using a deep learning classifier for automating the identification of the allergens causing ACD. We propose a deep learning approach that utilizes a context-retaining pre-processing technique to improve the accuracy of the classifier. In addition, we find promise in the combination of the color image and false-color map of hemoglobin concentration to improve diagnostic accuracy. Our results showed that this approach can potentially achieve more than 86% recall and 94% specificity in identifying skin reactions, and contribute to faster and more accurate diagnosis while reducing clinician workload.
... [92][93][94][95][96][97][98][99][100][101]. ...
Chapter
Full-text available
Autoimmune diseases (AD) have emerged as a pandemic in our modern societies, especially after the World War II. In part I, we have reviewed five main diseases and shed light on different aspects from introducing the concept of autoimmunity, the description of the disease's pathogenesis and the diagnosis, the role of antibod-ies as markers for the prediction of the disease, the link between the gut and brain through what is known as the gut-brain axis, and the relationship of this axis in GI autoimmune diseases. In this chapter, we review the role of antibodies as markers for the prediction of the disease, artificial intelligence in GI autoimmune diseases, the nutritional role and implications in the five GI autoimmune diseases, and finally the treatment of those diseases.
... Based on the result of this study and the existing literature, we established that a CC prediction model enrolled the weighted features including antibiotic significantly reduce the workload and cost in molecular typing while achieved a comparable accuracy comparing to MLST method. Artificial intelligence is prominent in the field of medical diagnosis with extensive application nowadays [22]. Recently, artificial intelligence is being increasingly applied into molecular typing of clinical isolates. ...
Article
Full-text available
Background The clinical significance of group B streptococcus (GBS) was different among different clonal complexes (CCs), accurate strain typing of GBS would facilitate clinical prognostic evaluation, epidemiological investigation and infection control. The aim of this study was to construct a practical and facile CCs prediction model for S. agalactiae. Methods A total of 325 non-duplicated GBS strains were collected from clinical samples in Xinhua Hospital, Shanghai, China. Multilocus sequence typing (MLST) method was used for molecular classification, the results were analyzed to derive CCs by Bionumeric 8.0 software. Antibiotic susceptibility test was performed using Vitek-2 Compact system combined with K-B method. Multiplex PCR method was used for serotype identification. A total of 45 virulence genes associated with adhesion, invasion, immune evasion were detected by PCR method and electrophoresis. Three types of features, including antibiotic susceptibility (A), serotypes (S) and virulence genes (V) tests, and XGBoost algorithm was established to develop multi-class CCs identification models. The performance of proposed models was evaluated by the receiver operating characteristic curve (ROC). Results The 325 GBS were divided into 47 STs, and then calculated into 7 major CCs, including CC1, CC10, CC12, CC17, CC19, CC23, CC24. A total of 18 features in three kinds of tests (A, S, V) were significantly different from each CC. The model based on all the features (S&A&V) performed best with AUC 0.9536. The model based on serotype and antibiotic resistance (S&A) only enrolled 5 weighed features, performed well in predicting CCs with mean AUC 0.9212, and had no statistical difference in predicting CC10, CC12, CC17, CC19, CC23 and CC24 when compared with S&A&V model (all p > 0.05). Conclusions The S&A model requires least parameters while maintaining a high accuracy and predictive power of CCs prediction. The established model could be used as a promising tool to classify the GBS molecular types, and suggests a substantive improvement in clinical application and epidemiology surveillance in GBS phenotyping.
... As one proceeds from the structure to the content of relevant studies, as we documented, the most popular topics are tumor type identification and grade, tumor detection, 5-year survival, cell segmentation, disease progression, disease recurrence and Ki-67 scoring. In a recent review, Yang et al. showed similar applications of AI with satisfactory prediction accuracy in the diagnosis, risk stratification and prognosis of small intestinal tumors [59]. Interestingly, this review shares 3 studies with the review in hand [14,21,33], which is not surprising given the rarity of small intestinal tumors and the major share of NENs among them. ...
Article
Full-text available
Neuroendocrine neoplasms (NENs) and tumors (NETs) are rare neoplasms that may affect any part of the gastrointestinal system. In this scoping review, we attempt to map existing evidence on the role of artificial intelligence, machine learning and deep learning in the diagnosis and management of NENs of the gastrointestinal system. After implementation of inclusion and exclusion criteria, we retrieved 44 studies with 53 outcome analyses. We then classified the papers according to the type of studied NET (26 Pan-NETs, 59.1%; 3 metastatic liver NETs (6.8%), 2 small intestinal NETs, 4.5%; colorectal, rectal, non-specified gastroenteropancreatic and non-specified gastrointestinal NETs had from 1 study each, 2.3%). The most frequently used AI algorithms were Supporting Vector Classification/Machine (14 analyses, 29.8%), Convolutional Neural Network and Random Forest (10 analyses each, 21.3%), Random Forest (9 analyses, 19.1%), Logistic Regression (8 analyses, 17.0%), and Decision Tree (6 analyses, 12.8%). There was high heterogeneity on the description of the prediction model, structure of datasets, and performance metrics, whereas the majority of studies did not report any external validation set. Future studies should aim at incorporating a uniform structure in accordance with existing guidelines for purposes of reproducibility and research quality, which are prerequisites for integration into clinical practice.
... With enhanced CT, heterogeneous enhancement is typical of small bowel neoplasms, especially SBA and gastrointestinal stromal tumors but detection of SBA at an early stage remains difficult [12]. Recently, some authors combine the conventional modalities with artificiel intelligence to improve the diagnosis of small bowel pathologies both inflammatory and tumors [13]. The treatment of SBA is surgical based on R0 resection of the tumor with lymphe node dissection. ...
Article
Full-text available
Introduction Small bowel adenocarcinoma is a rare but increasing disease. It poses both a diagnostic and therapeutic challange. Small bowel adenocarcinoma is a rare cause of small bowel obstruction. We present the case of a patient admitted to our emergency department for a bowel obstruction due to a mass of the jejunum and whose anatomopathological diagnosis was adenocarcinoma. Patient and method It is a 62-year-old woman with unparticular history, admitted to the emergency of visceral surgery of Ibn Rochd University Hospital for subocclusive syndrome evolving for one year, with early postprandial vomiting becoming stenosing two months ago. The abdominal CT scan showed thickening jejunal wall of 46 mm with upstream distension. She underwent a segmental bowel resection of 50 cm of small bowel with 3 cm stenotic mass located at 40 cm from the duodenojejunal angle. The pathophysiology revealed an invasive liberkhunian adenocarcinoma. The postoperative follow-up was simple, feeding allowed at D4 with discharge allowed at D6 and functional improvement at the time of the control performed three months after the intervention. Discussion Small bowel adenocarcinoma is rare and represents only 1–3% of all gastrointestinal cancers. The incidence of SBA is 24 to 66 times lower than that of colorectal cancer (CRC). Due to its non-specific clinical manifestation and less accessible location, SBA is diagnosed at an advanced stage, and often at specimen analysis. The treatment is resection and the overall survival is increased when diagnostic is early made. Conclusion Small bowel adenocarcinoma is a rare but increasing cause of gastrointestinal malignancy, being both a diagnostic and therapeutic challenge. In front of the occlusive syndrome of small bowel appearance, adenocarcinoma must be ruled out.
Chapter
In the early 1900s, multiple significant studies showed high incidences of cancer. During this period, study with infectious agents produced only modest results which looked irrelevant to people. Then, in the 1980s, groundbreaking evidence that a number of viruses can cause cancer in people began to emerge. Machine learning and deep learning techniques have been widely employed in cancer detection and classification that include support vector machines (SVMs), artificial neural networks (ANNs), and conventional neural networks (CNNs). The recurrence of cancer is also an important issue that needs to be predicted with significant accuracy. This chapter reviews current state-of-the-art of ANNs model in the prediction of cancer recurrence.
Chapter
Computational intelligence (CI) is one of the most popular and advanced fields that mimics human intelligence and transforms numerous aspects of the healthcare industry. For the past two decades, CI has grown extensively and reached a new height in clinical research on cancer. The subfields of artificial intelligence, like deep learning and machine learning, are also applied to tackle various scientific problems. Cancer is one of the most aggressive and dreaded diseases, which every year costs more than 10 million lives globally, and it is expected that these tolls may reach 16.4 million by 2040. Hence, meticulous prognosis and early diagnosis of this disease are very significant to increase the chance of patient survival. In recent years, breakthroughs in various AI-based stratagems for precision oncology, digital pathology, and next-generation sequencing (NSG) are driving the need for new prognostic and predictive assays that facilitate the stratification and selection of patients for treatment. This chapter briefly includes the applications of CI and its subfields in cancer biomarkers and therapeutic target prediction. However, we explore how these applications have assisted in solving biomedical problems with unprecedented accuracy and deep learning. We also demonstrated how these data could be run on natural and mathematical algorithms to derive a classification of cancer genomes, identify potential biomarkers, therapeutic targets, drugs, and draw scientifically sound conclusions based on them. By providing a fresh viewpoint on how computational tools might aid enhance cancer detection, this chapter can help make significant advances in the medical disciplines and scientific community.
Article
Full-text available
Gastric cancer is the fourth leading cause of cancer-related mortality across the globe, with a 5-year survival rate of less than 40%. In recent years, several applications of artificial intelligence (AI) have emerged in the gastric cancer field based on its efficient computational power and learning capacities, such as image-based diagnosis and prognosis prediction. AI-assisted diagnosis includes pathology, endoscopy, and computerized tomography, while researchers in the prognosis circle focus on recurrence, metastasis, and survival prediction. In this review, a comprehensive literature search was performed on articles published up to April 2020 from the databases of PubMed, Embase, Web of Science, and the Cochrane Library. Thereby the current status of AI-applications was systematically summarized in gastric cancer. Moreover, future directions that target this field were also analyzed to overcome the risk of overfitting AI models and enhance their accuracy as well as the applicability in clinical practice.
Article
Full-text available
Small bowel capsule endoscopy (SBCE) can be complementary to histological assessment of celiac disease (CD) and serology negative villous atrophy (SNVA). Determining the severity of disease on SBCE using statistical machine learning methods can be useful in the follow up of patients. SBCE can play an additional role in differentiating between CD and SNVA. De-identified SBCEs of patients with CD and SNVA were included. Probabilistic analysis of features on SBCE were used to predict severity of duodenal histology and to distinguish between CD and SNVA. Patients with higher Marsh scores were more likely to have a positive SBCE and a continuous distribution of macroscopic features of disease than those with lower Marsh scores. The same pattern was also true for patients with CD when compared to patients with SNVA. The validation accuracy when predicting the severity of Marsh scores and when distinguishing between CD and SNVA was 69.1% in both cases. When the proportions of each SBCE class group within the dataset were included in the classification model, to distinguish between the two pathologies, the validation accuracy increased to 75.3%. The findings of this work suggest that by using features of CD and SNVA on SBCE, predictions can be made of the type of pathology and the severity of disease.
Article
Full-text available
Esophageal cancer poses diagnostic, therapeutic and economic burdens in high-risk regions. Artificial intelligence (AI) has been developed for diagnosis and outcome prediction using various features, including clinicopathologic, radiologic, and genetic variables, which can achieve inspiring results. One of the most recent tasks of AI is to use state-of-the-art deep learning technique to detect both early esophageal squamous cell carcinoma and esophageal adenocarcinoma in Barrett's esophagus. In this review, we aim to provide a comprehensive overview of the ways in which AI may help physicians diagnose advanced cancer and make clinical decisions based on predicted outcomes, and combine the endoscopic images to detect precancerous lesions or early cancer. Pertinent studies conducted in recent two years have surged in numbers, with large datasets and external validation from multi-centers, and have partly achieved intriguing results of expert's performance of AI in real time. Improved pre-trained computer-aided diagnosis algorithms in the future studies with larger training and external validation datasets, aiming at real-time video processing, are imperative to produce a diagnostic efficacy similar to or even superior to experienced endoscopists. Meanwhile, supervised randomized controlled trials in real clinical practice are highly essential for a solid conclusion, which meets patient-centered satisfaction. Notably, ethical and legal issues regarding the black-box nature of computer algorithms should be addressed, for both clinicians and regulators.
Article
Full-text available
Since the advent of artificial intelligence (AI) technology, it has been constantly studied and has achieved rapid development. The AI assistant system is expected to improve the quality of automatic polyp detection and classification. It could also help prevent endoscopists from missing polyps and make an accurate optical diagnosis. These functions provided by AI could result in a higher adenoma detection rate and decrease the cost of polypectomy for hyperplastic polyps. In addition, AI has good performance in the staging, diagnosis, and segmentation of colorectal cancer. This article provides an overview of recent research focusing on the application of AI in colorectal polyps and cancer and highlights the advances achieved. © The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
Article
Full-text available
The often poorly orientated small-bowel mucosal biopsies taken for the diagnostics of celiac disease and other intestinal disorders are prone to misinterpretation. Furthermore, conventional histopathology has suboptimal sensitivity for early histopathological changes observed in short-term challenge studies. X-ray microtomography (micro-CT) is a promising new method for accurate imaging of human-derived biological samples. Here, we report that micro-CT could be utilized to create virtual reconstructions of endoscopically obtained intestinal biopsies. The formed digital 3D images enabled selection of always optimal cutting angles for accurate measurement of the mucosal damage and revealed diagnostic lesions in cases interpreted as normal with conventional histomorphometry. We also demonstrate that computer-assisted point cloud analysis can be used to calculate biologically meaningful surface areas of the biopsies in different stages of mucosal damage with excellent replicability and correlation with other disease parameters. We expect the improved diagnostic accuracy and capability to measure the surface areas to provide a powerful tool for the diagnostics of intestinal diseases and for future clinical and pharmaceutical trials.
Article
Full-text available
Purpose: Segmentation of multiple organs-at-risk (OARs) is essential for MR-only radiation therapy treatment planning and MR-guided adaptive radiotherapy of abdominal cancers. Current practice requires manual delineation that is labor-intensive, time-consuming, and prone to intra- and inter-observer variations. We developed a deep learning (DL) technique for fully automated segmentation of multiple OARs on clinical abdominal MR images with high accuracy, reliability, and efficiency. Methods: We developed Automated deep Learning-based Abdominal Multi-Organ segmentation (ALAMO) technique based on 2D U-net and a densely connected network structure with tailored design in data augmentation and training procedures such as deep connection, auxiliary supervision, and multi-view. The model takes in multi-slice MR images and generates the output of segmentation results. 3.0-Tesla T1 VIBE (Volumetric Interpolated Breath-hold Examination) images of 102 subjects were used in our study and split into 66 for training, 16 for validation, and 20 for testing. Ten OARs were studied, including the liver, spleen, pancreas, left/right kidneys, stomach, duodenum, small intestine, spinal cord, and vertebral bodies. An experienced radiologist manually labeled each OAR, followed by reediting, if necessary, by a senior radiologist, to create the ground-truth. The performance was measured using volume overlapping and surface distance. Results: The ALAMO technique generated segmentation labels in good agreement with the manual results. Specifically, among the 10 OARs, 9 achieved high Dice Similarity Coefficients (DSCs) in the range of 0.87-0.96, except for the duodenum with a DSC of 0.80. The inference completed within one minute for a 3D volume of 320x288x180. Overall, the ALAMO model matched the state-of-the-art techniques in performance. Conclusion: The proposed ALAMO technique allows for fully automated abdominal MR segmentation with high accuracy and practical memory and computation time demands.
Article
Full-text available
Capsule endoscopy (CE) is the first-line diagnostic modality for detecting small bowel lesions. CE is non-invasive and does not require sedation, but its movements cannot be controlled, it requires a long time for interpretation, and it has lower image quality compared to wired endoscopy. With the rapid advancement of technology, several methods to solve these problems have been developed. This article describes the ongoing developments regarding external CE locomotion using magnetic force, artificial intelligence-based interpretation, and image-enhancing technologies with the CE system.
Article
Full-text available
Background: Small intestinal neuroendocrine tumors (SI-NETs) are difficult to diagnose in the early stage of disease. Current blood biomarkers such as chromogranin A (CgA) and 5-hydroxyindolacetic acid (5-HIAA) have low sensitivity and specificity. This is a first pre-planned interim analysis (NORDIC non-interventional, exploratory, EXPLAIN study (NCT02630654)). Its objective is to investigate if a plasma protein multi-biomarker strategy can improve diagnostic accuracy in SI-NETs. Methods: At time of diagnosis, prior any disease specific treatment was initiated, blood was collected from patients with advanced SI-NETs and 92 putative cancer-related plasma proteins from 135 patients were analyzed and compared with the results of age and gender matched controls (n=143), using multiplex proximity extension assay and machine learning techniques. Results: Using a random forest model including 12 top ranked plasma proteins in patients with SI-NETs, the multi-biomarker strategy showed sensitivity (SEN) and specificity (SPE) of 89% and 91%, respectively, with negative predictive value (NPV) and positive predictive value (PPV) of 90% and 91%, respectively, to identify patients with regional or metastatic disease with an area under the receiver operator characteristic curve (AUROC) of 99%. In thirty patients with normal CgA concentrations the model provided diagnostic SPE of 98%, a SEN of 56%, and NPV 90%, PPV of 90%, and AUROC 97%, regardless of proton pump inhibitor intake. Conclusion: This interim analysis demonstrate that a multi-biomarker/machine learning strategy improve diagnostic accuracy of patients with SI-NET at the time of diagnosis, especially in patients with normal CgA levels. The results indicate that this multi-biomarker strategy can be useful for early detection of SI-NETs at presentation and conceivably detect recurrence after radical primary resection.
Article
Inflammation of the gastrointestinal (GI) tract accompanies several diseases, including Crohn’s disease. Currently, video capsule endoscopy and deep bowel enteroscopy are the main means for direct visualisation of the bowel surface. However, the use of optical imaging limits visualisation to the luminal surface only, which makes early-stage diagnosis difficult. In this study, we propose a learning enabled microultrasound (μUS) system that aims to classify inflamed and non-inflamed bowel tissues. μUS images of the caecum, small bowel and colon were obtained from mice treated with agents to induce inflammation. Those images were then used to train three deep learning networks and to provide a ground truth of inflammation status. The classification accuracy was evaluated using 10-fold evaluation and additional B-scan images. Our deep learning approach allowed robust differentiation between healthy tissue and tissue with early signs of inflammation that is not detectable by current endoscopic methods or by human inspection of the μUS images. The methods may be a foundation for future early GI disease diagnosis and enhanced management with computer-aided imaging.
Article
Background Machine learning has led to several endoscopic studies about the automated localization of digestive lesions and prediction of cancer invasion depth. Training and validation dataset collection are required for a disease in each digestive organ under a similar image capture condition; this is the first step in system development. This data cleansing task in data collection causes a great burden among experienced endoscopists. Thus, this study classified upper gastrointestinal (GI) organ images obtained via routine esophagogastroduodenoscopy (EGD) into precise anatomical categories using AlexNet. Method In total, 85,246 raw upper GI endoscopic images from 441 patients with gastric cancer were collected retrospectively. The images were manually classified into 14 categories: 0) white-light (WL) stomach with indigo carmine (IC); 1) WL esophagus with iodine; 2) narrow-band (NB) esophagus; 3) NB stomach with IC; 4) NB stomach; 5) WL duodenum; 6) WL esophagus; 7) WL stomach; 8) NB oral–pharynx–larynx; 9) WL oral–pharynx–larynx; 10) WL scaling paper; 11) specimens; 12) WL muscle fibers during endoscopic submucosal dissection (ESD); and 13) others. AlexNet is a deep learning framework and was trained using 49,174 datasets and validated using 36,072 independent datasets. Results The accuracy rates of the training and validation dataset were 0.993 and 0.965, respectively. Conclusions A simple anatomical organ classifier using AlexNet was developed and found to be effective in data cleansing task for collection of EGD images. Moreover, it could be useful to both expert and non-expert endoscopists as well as engineers in retrospectively assessing upper GI images.