BPG is committed to discovery and dissemination of knowledge
Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Nov 14, 2025; 31(42): 111291
Published online Nov 14, 2025. doi: 10.3748/wjg.v31.i42.111291
Could artificial intelligence-powered colonoscopies change the future of colorectal cancer screening?
Mircea Mănuc, Cătălin-Andrei Duței, Teodora-Ecaterina Mănuc, Andreea-Elena Chifulescu, Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, Bucharest 050474, Romania
Florin Andrei Grama, Department of Surgery, “Carol Davila” University of Medicine and Pharmacy, Bucharest 050474, Romania
ORCID number: Mircea Mănuc (0000-0003-1142-5859); Cătălin-Andrei Duței (0000-0002-8834-4718); Teodora-Ecaterina Mănuc (0000-0002-0589-4571); Andreea-Elena Chifulescu (0000-0001-7942-5375); Florin Andrei Grama (0000-0001-5728-2860).
Co-corresponding authors: Cătălin-Andrei Duței and Teodora-Ecaterina Mănuc.
Author contributions: Duței CA and Mănuc M designed the review; Duței CA wrote the manuscript; Mănuc TE, Chifulescu AE and Grama FA selected the data and analytic tools; all authors have read and approved the final manuscript.
Conflict-of-interest statement: The authors declare that they have no 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 on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Cătălin-Andrei Duței, MD, Doctor, Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, Blv. Eroii Sanitari, nr 8, Bucharest 050474, Romania. catalin-andrei.dutei@drd.umfcd.ro
Received: June 27, 2025
Revised: August 13, 2025
Accepted: October 10, 2025
Published online: November 14, 2025
Processing time: 139 Days and 17.1 Hours

Abstract

Colorectal cancer is a major cause of cancer-related mortality worldwide, underscoring the importance of early and effective colorectal cancer screening to improve survival rates. Traditional colorectal cancer screening methods include non-invasive tests, such as the fecal immunochemical test (FIT), as well as diagnostic procedures like colonoscopy. Colonoscopy remains the gold standard for detecting and treating precancerous polyps and early-stage cancer, regardless of whether it is used as the first screening test or the second test following a positive FIT. However, its effectiveness can be affected by factors such as operator skill, patient variability, and limited lesion visibility, resulting in a significant rate of missed lesion rates and highlighting the need for more efficient and accurate screening techniques. This review is aimed to assess the current challenges of traditional screening methods with the impact of artificial intelligence (AI) in the diagnostic flow. The literature on AI-powered tools for colorectal cancer screening, including novel applications, emerging programs, and recent guidelines, has been reviewed to highlight both the advantages and limitations of implementing this technology in healthcare. Recent advances in AI have introduced soft AI colonoscopy, with the purpose of improving lesion recognition (computer-aided detection) and/or improving optical diagnosis (computer-aided diagnosis). AI-powered colonoscopy systems employ deep learning algorithms to analyze real-time endoscopic images, enhancing detection rates for adenomas, serrated lesions and cancer by reducing human error. AI-assisted colonoscopy enhances adenoma detection, enabling earlier intervention and improved patient outcomes. The benefits are particularly pronounced for less-experienced practitioners, as the detection rates for AI-assisted colonoscopy are similar to experts. AI integration also helps in the teaching process, in developing standardized procedures, and improving screening procedure accuracy and efficiency across different healthcare providers. However, there are challenges and limitations, such as the cost of AI implementation, data privacy concerns, and the need for extensive clinical validation. As AI technology continues to evolve, its transformation of the colorectal cancer screening system could revolutionize the field, making early detection more accessible and reducing mortality, on the condition that the above issues are addressed before widespread use.

Key Words: Colorectal cancer screening; Colorectal cancer; Artificial intelligence; Artificial intelligence powered colonoscopy; Adenoma detection rate

Core Tip: Colorectal cancer screening has evolved considerably in recent years, with recommended procedures increasingly incorporated into clinical guidelines to enhance early detection. Advances in artificial intelligence have enabled its integration across multiple stages of screening, improving diagnostic accuracy, supporting clinician performance, enhancing imaging, and optimizing patient preparation. However, limitations remain, and artificial intelligence applications must continue to be refined and adjusted for real-world use, particularly with regard to social, privacy and financial considerations.



INTRODUCTION

Cancer represents a major global health burden, affecting social, clinical, psychological and economic domains, and demands the mobilization of every available resource to address it effectively. According to the Agency for Research on Cancer, there were 20 million new cases of cancer per year in 2022, with over 9.7 million cancer-related deaths per year. Colorectal cancer (CRC) is a major cause of cancer-related death worldwide, requiring early and effective CRC screening to improve survival rates. Worldwide, CRC ranks third in incidence, accounting for 1926425 new cases (9.6%) across both sexes, 1069446 in males and 856979 in females, according to the most recent Global Cancer Statistics report (2022). In terms of mortality, CRC ranks as the second leading cause of cancer-related death worldwide, accounting for approximately 904019 deaths annually (9.3%)[1]. In the United States, the burden is similarly high, with CRC representing nearly 10% of all cancer cases and responsible for 8%-9% of cancer- related deaths[1-3]. According to the 2022 Global Cancer Observatory statistics, in the United States, breast cancer ranks first in incidence (12%), followed by lung cancer (11%) and CRC (10%). However, in terms of mortality, CRC ranks second (9%), after lung cancer (18%)[1,4-6]. The Indian Council of Medical Research reports annual incidence rates (AIRs) for colon and rectal cancer in male patients of 4.4 and 4.1 per 100000, respectively. In comparison, the AIR for colon cancer in female patients is approximately 3.9 per 100000[7]. These data have prompted countries worldwide to implement improved screening strategies. In the European Union, the target is to achieve screening coverage of 90% among the eligible population (ages 50-74), using the fecal immunochemical test (FIT) test as the initial method, followed by colonoscopy for positive cases. In the United States, recommendations differ slightly: Individuals at average risk (those aged 45 and older) are advised to undergo either a stool-based test [FIT, fecal occult blood test (FOBT) or DNA-based test] or a visual examination (colonoscopy, rectosigmoidoscopy or computed tomography colonography). Those at higher risk should begin screening earlier and use more specific diagnostic methods[1].

Background data on traditional and innovative screening methods

Traditional CRC screening methods include non-invasive stool-based tests, such as the guaiac FOBT, FIT, and multi-targeted stool DNA test, as well as more accurate diagnostic procedures like colonoscopy. Whether used as a primary screening tool or as a follow-up after a positive FIT, colonoscopy remains the gold standard for detecting and removing precancerous polyps and identifying early-stage cancers. However, its effectiveness can be affected by factors such as operator skill, patient variability and lesion visibility. The persistence of missed lesions underscores the need for more efficient and accurate screening techniques[8]. Studies have shown that up to 22% of polyps may be missed during CRC screening colonoscopies, and approximately 8% of cancers develop within 3 years following a screening colonoscopy[9,10].

Studies indicate that several biomarkers are already in use for diagnosing CRC; however, their specificity for CRC cells remains limited. These include carbohydrate antigen (CA) 19-9, a tetrasaccharide expressed on the surface of pancreatic and colonic adenocarcinoma cells, and CA125, which is detected in both colonic and ovarian cancers and has also been associated with endometriosis. DNA-based molecular biomarkers function by detecting methylated DNA in stool samples. Improved diagnostic accuracy can be achieved by combining the FIT with a multi-target stool DNA test to detect and quantify hypermethylated DNA. Stool DNA testing demonstrates higher sensitivity than FIT alone (92.3% vs 73.8%); however, it is also associated with a higher rate of false positives. Another serum-based blood test developed and studied in the United States is Epi proColon, which uses a real-time polymerase chain reaction (PCR) technique to detect methylated SEPT9, a biomarker associated with CRC. However, its sensitivity and specificity are relatively low: 68.2% and 79.1% respectively, regardless of cancer stage. Due to its higher rate of false positives, Epi proColon performs less effectively than FIT and colonoscopy. Other methylation markers that have been investigated include SFRO2, VIM, FBN2, TCERG1 and serum vimentin methylation. Their diagnostic performance for early-stage CRC detection generally falls within a moderate range of specificity and sensitivity.

In addition to biomarkers, circulating tumor cells (CTC) have been studied, analyzed and demonstrated promise in cancer detection. However, this technique is limited by the low concentration of CTCs in the blood (1 CTC per 109 red blood cells) and by variability in sampling methods. The efficiency of CTC capture and enrichment largely depends on their physical and biological properties, including density, size, deformability, electrophoresis and affinity-based capture. Common selection approaches involve identifying cells that are CD45-negative and EPCAM-positive[11-23].

Another promising biomarker under investigation is tumor-associated circulating transcripts. These RNA-based liquid biopsies leverage artificial intelligence (AI) models as generalized linear models, random forest (RF), gradient boosting machine (GBM), deep neural networks (DNN), and automated machine learning. Among these, the DNN model has demonstrated superior performance, achieving optimal sensitivity (85.7%) and specificity (90.9%) for the detection of early-stage CRC. Of the ten markers studied, EPCAM, NPTN, KRT19, MKI67 and VIM were found to be highly expressed in CRC cell lines, whereas FOXA2 and MCAM showed lower expression levels. In contrast, ERBB2, TERT and SNAI2 were undetectable[24-26]. SEPTIN9 is another biomarker found to be useful in more advanced pathological stages of CRC, as levels of methylated SEPTIN9 in peripheral blood increase with disease progression. Its reported sensitivity for detecting CRC ranges from 75% to 79.3%, whereas its sensitivity for identifying adenomas remains suboptimal at approximately 27%[27,28].

In recent years, the gut microbiota has been intensely studied as a key to early CRC and adenoma detection. The gut microbiota can influence several processes, including metabolic regulation, inflammation control, and epigenetic reprogramming. There is increasing evidence that the gut microbiota can be leveraged to identify individuals at high risk for CRC and early detection. After performing 16S rRNA sequencing on fecal samples from individuals with positive FIT, researchers found a significant increase in the abundance of Proteobacteria in those with colonic lesions compared with healthy controls. Another study identified elevated levels of Fusobacterium nucleatum in the oral cavity, tumor tissue, and feces of individuals with colonic lesions. The presence of this bacterium demonstrated a sensitivity of 82% and specificity of 62% for distinguishing colorectal adenomatous polyposis from healthy individuals, and a sensitivity of 66% with specificity of 90% for differentiating adenomas from CRC[29-35].

The detection of free fatty acids (FFAs) in serum represents another promising approach for early CRC diagnosis, as CRC patients exhibit significantly lower FFA levels compared to healthy individuals. Combined analysis of FFAs such as C16:1, C18:3, C18:2, C18:1, C20:4, and C22:6 has achieved a sensitivity of up to 84.6% and specificity of 89.8% for early CRC detection. Furthermore, the combination of C16:1, C18:3 and C18:2 demonstrated a sensitivity of 70% and a specificity of 81% in distinguishing benign intestinal diseases from CRC[36].

THE PROMISING ROLE OF AI IN CRC

Since their invention, computers have evolved into diverse branches and become integral to nearly all scientific, engineering and healthcare fields. They enable the development of programs designed to replicate aspects of human intelligence and behavior. In healthcare, AI has emerged as a powerful tool, particularly in the prevention, diagnosis and management of diseases, including CRC. AI operates through two pathways: Virtual and physical. The virtual pathway encompasses machine learning and deep learning, with the latter representing a specialized subset of the former. Machine learning involves processing input data to identify patterns using statistical and analytical techniques, without the need for explicit programming[37]. In unsupervised machine learning, the program autonomously identifies patterns within data using statistical methods, without predefined labels or outcomes. In contrast, supervised learning relies on known outputs to guide the training process toward specific goals. Deep learning employs artificial neural networks to model complex relationships. It includes DNNs for feed-forward processing, recurrent neural networks for sequential data analysis, and convolutional neural networks (CNNs) for interpreting grid-like structures. These architectures are particularly well-suited for tasks such as biomedical signal processing, feature learning, and medical image recognition[38-44].

Recent advancements in AI have led to the development of soft AI colonoscopy, designed to enhance both lesion recognition through computer-aided detection (CADe), and optical diagnosis through computer-aided diagnosis (CADx). These AI-powered colonoscopy systems employ deep learning algorithms to analyze real-time endoscopic images, thereby improving detection rates for adenomas, serrated lesions and CRC. Studies have shown that AI-assisted colonoscopy improves adenoma detection enabling earlier intervention and leading to better patient outcomes. The benefits are particularly notable for less-experienced clinicians, as the detection rates approach those of experts. Moreover, AI integration can support training and education, promote standardized procedures, and enhance the consistency and reliability of screenings across healthcare settings, thereby improving accuracy and efficiency. As AI technology continues to advance, its application in CRC screening has the potential to revolutionize early detection, reduce CRC-related mortality, and reshape the future of cancer prevention and care[41].

Despite these benefits, challenges such as the cost of AI implementation, data privacy concerns and the need for extensive clinical validation must be addressed before widespread use. Studies indicate that the application of AI in healthcare attracted approximately 6.6 billion USD in investments from public and commercial sectors in 2021. In the United States alone, the use of AI is projected to generate annual savings of up to 150 billion USD by 2026, reflecting its growing impact on healthcare efficiency and cost reduction[11]. In this review, we present the latest advances in the field, along with with future applications and expected limitations in CRC screening.

HOW CAN AI BE INTEGRATED IN CRC SCREENING?
Enhancing the diagnostic accuracy of FIT?

The preferred method of CRC screening is FOBT. Most studies indicate that the traditional guaiac-based FOBT is inferior to the FIT, making FIT the preferred method for CRC screening. More recently, the introduction of the ColonView FIT test has further advanced screening accuracy and convenience[45-54]. To improve adenoma detection methods, various studies have assessed different AI-based enhancements, such as logistic regression (LR), support vector machine (SVM), neural network (NN), RF and GBM[55]. All tested AI models demonstrated superiority in colorectal neoplasia screening, primarily evaluated through the adenoma detection rate (ADR), as adenomas are the precursor of CRC. ADR remains the key performance metric and a critical endpoint for reducing CRC incidence[56]. Among the various models assessed, GBM/RF/NN algorithms were identified as the most effective[55]. The diagnostic accuracy of various AI models surpassed that of conventional low-risk/high-risk FOBT and FIT tests. Specifically, GBM, RF, NN, SVM, and LR models achieved sensitivities of 85%/82%/68%/60%/55% specificities of 74%/72%/76%/69%/77%, and overall efficiencies of 79%/76%/73%/65%/67%, respectively[55].

Helping improve bowel preparation?

In CRC screening, the preferred initial method is a fecal-based test, with patients who test positive subsequently undergoing a full colonoscopy. In the United States, however, screening often begins directly with colonoscopy. In both approaches, a major problem is inadequate bowel preparation, which occurs in approximately 25% of cases and frequently necessitates a repeat procedure. Additional limitations include longer procedure times and a reduced ADR, reported to be as low as 48% under suboptimal conditions[57-59]. The potential role of AI in improving bowel preparation for colonoscopy remains under investigation. Recent studies have evaluated ChatGPT-generated pre-colonoscopy preparation prompts, which can be tailored to individual patients and healthcare providers[60-62]. As anticipated, AI has also been applied to develop bowel preparation scoring systems[61]. Moreover, analyses of prompt disagreement have identified several common issues, including: Overly long or short preparation times, dietary misunderstandings, improper timing or medication use, unclear pre-procedure clearance instructions, confusing sequence of steps, mismatched preparation types, excessive medical jargon, lack of readability at a sixth-grade reading level, and unclear guidance on fasting requirements[62].

When various specialists, including senior physicians, fellows, junior doctors and representatives of the American Gastroenterological Association, evaluated the ChatGPT-generated pre-colonoscopy bowel preparation prompts, they found the material to be easy to understand, scientifically accurate and effective in addressing common patient questions about colonoscopy. Notably, post-procedure outcomes demonstrated significant improvement, with surveillance management compliance reaching nearly 90% and accuracy approximately 85%. Furthermore, several studies have shown that providing ChatGPT with contextualized medical guidelines enables it to generate highly accurate recommendations for colonoscopy screening intervals[63-67]. For instance, two commonly used bowel preparation instructions given to patients were found to exceed the recommended sixth-grade reading level, scoring 8.5 and 7.8 on the Flesch-Kincaid reading scale and 10.5 and 8.6 on the Gunning Fog Index, respectively. In comparison, the AI-generated preparation prompts were rated as scientifically accurate, more patient-friendly, and better aligned with clinical standards. However, they were noted to be slightly too complex, indicating room for further refinement[62].

Increasing ADR?

The ADR is a critical quality indicator in colonoscopy, as higher ADRs are directly associated with reduced overall risk and burden of CRC. Improving ADR relies heavily on maintaining high-quality colonoscopy performance. However, despite ongoing advances, approximately 27% of polyps are still missed during colonoscopy, largely due to non-endoscopic factors, such as inadequate bowel preparation and varying levels of endoscopist expertise[68]. Several studies have explored strategies to address this issue, with recent research focusing on the integration of AI into colonoscopy[69-78].

One such method is ALPHAON, an AI-based CADe algorithm with exceptional performance in polyp detection. It has demonstrated an accuracy of 0.97 [95% confidence interval (CI): 0.96-0.99], sensitivity of 0.91 (95%CI: 0.85-0.97), specificity of 0.99 (95%CI: 0.97-0.99), and an area under the ROC curve of 0.967. Additionally, it enhances outcomes across experience levels. Among trainees, it achieved an accuracy of 0.95 (95%CI: 0.93-0.96) and sensitivity of 0.80 (95%CI: 0.74-0.86), while among experts, performance was slightly higher, with an accuracy of 0.96 (95%CI: 0.94-0.97) and sensitivity of 0.84 (95%CI: 0.79-0.90). Although ALPHAON demonstrates impressive diagnostic performance, it still has notable limitations. Currently, it cannot classify polyps or provide pathological information, though future developments may allow its transition into a CADx system. Another limitation involves false-positive detections, such as bubbles, mucosal folds, and tags, which can increase withdrawal time, lead to unnecessary biopsies, and contribute to operator distraction and fatigue[79-81]. Multiple randomized controlled trials have demonstrated that incorporating CADe significantly improves both ADR and polyp detection rates compared to conventional colonoscopy alone[82-85]. However, while numerous trials have evaluated different AI algorithms at varying levels, the findings remain variable and inconclusive, underscoring the need for further validation and standardization(Table 1)[86-108].

Table 1 Trials using computer-aided detection system for colorectal lesions[86-108].
Ref.
Year
Application used
Patients (n)
Findings
Wang et al[86]2019Endo Screener1058AI system significantly increased ADR (29.1% vs 20.3%; P < 0.001)
Su et al[87]2019AQCS623AI significantly increased ADR (28.9% vs 16.5%; P < 0.001)
Liu et al[88]2020Henan Tongyu1026AI significantly increased ADR (39% vs 24%; P < 0.001)
Gong et al[89]2020ENDOANGEL704AI significantly increased ADR (16% vs 8%; P = 0.001)
Repici et al[90]2020GI genius685AI significantly increased ADR (54.8% vs 40.4%)
Wang et al[91]2020Endo Screener369AMR was significantly lower with AI (13.9% vs 40.0%; P < 0.001)
Wang et al[92]2020Endo Screener962AI system significantly increased ADR rather than the sham system (34% vs 28%; P = 0.030)
Kamba et al[93]2021Endo Screener358AMR was significantly lower with AI (13.8% vs 40.6%; P < 0.001)
Xu et al[94]2021Endo Screener2325AI system did not significantly increase PDR (38.8% vs 36.2%; P = 0.183)
Luo et al[95]2021Endo Screener150AI system significantly increased PDR (38.7% vs 34.0%; P < 0.001)
Shaukat et al[96]2022Endo Screener1359AI system significantly increased adenomas per colonoscopy (1.05 vs 0.83; P = 0.002)
Wallace et al[97]2022GI genius230AMR was significantly lower with AI (15.5% vs 32.4%; P < 0.001)
Glissen Brown et al[98]2022Endo Screener223AMR was significantly lower with AI than with high-definition white light colonoscopy (20.12% vs 31.25%; P = 0.025)
Lui et al[99]2023Endo Screener216AI significantly increased ADR in proximal colon (44.7% vs 34.6%)
Ahmad et al[100]2023GI genius614AI system did not significantly increase ADR (71.4% vs 65.0%; P = 0.09)
Mangas-Sanjuan et al[101]2023GI genius3213AI system did not significantly increase advanced colorectal neoplasia detection rate (34.8% vs 34.6%; P = 0.91)
Karsenti et al[102]2023GI genius2015AI system slightly increased ADR (37.5% vs 33.7%; P = 0.051)
Wei et al[103]2023Endo Vigilant769AI system did not significantly increase ADR (35.9% vs 37.2%; P = 0.774)
Nakashima et al[104]2023CAD EYE415AI system significantly increased ADR (59.4% vs 47.6%; P = 0.018)
Gimeno-García et al[105]2024ENDO-AID370AI system significantly increased ADR (55.1% vs 43.8%; P = 0.029)
Yao et al[106]2024ENDOANGEL685AMR was significantly lower with AI (18.82% vs 43.69%; P < 0.001)
Schöler et al[107]2024CAD EYE286AI system did not significantly increase ADR (43% vs 41%; P = 0.696)
Yamaguchi et al[108]2024CAD EYE231AMR was significantly lower with AI (25.6% vs 38.6%; P = 0.033)

Some studies are advancing FocuU2Net, an innovative bi-level nested U-structure integrated with a dual-attention mechanism[109]. The model integrates focus gate modules for spatial and channel-wise attention and residual U-blocks with multi-scale receptive fields for capturing diverse contextual information. Comprehensive evaluations on five benchmark datasets: Kvasir-SEG, CVC-ClinicDB, CVC-ColonDB, and EndoScene demonstrate Dice score improvements of 3.14% to 43.59% over state-of-the-art models, with an 85% success rate in cross-dataset validations, significantly surpassing prior competing models with sub-5% success rates (Table 2)[110]. Compared to more recent methods such as SR-AttNet, PolypSegNet, CSAP-UNet, I2-UNet, segment anything model 2 (SAM2) and segment anything in medical images (MedSAM), FocusU2Net maintains competitive performance, highlighting its relevance and effectiveness in the evolving research landscape[110]. When discussing deep learning-based approaches for polyp segmentation approaches, several architectures are noteworthy[110]: Traditional encoder-decoder architectures (U-Net, U-Net++, ResU-Net++, ResUNet, MultiResUNet) offer key advantages including efficient feature propagation through skip connection, volumetric skip connection with deep supervision, nested skip connections with residual learning, enhanced gradient flow, and multiscale feature fusion. However, these architectures can face certain challenges, particularly in capturing broader contextual information, preserving fine structural details, and accurately segmenting complex or irregularly shaped polyps. Additionally, multi-scale feature handling remains a technical limitation.

Table 2 Performance comparison of studied models[110].
Model (year)CVC-ClinicDB
Kvasir-SEG
CVC-ColonDB
EndoScene
Dice
IoU
Rec
Prec
Dice
IoU
Rec
Prec
Dice
IoU
Rec
Prec
Dice
IoU
Rec
Prec
UNet (2015)29.8319.5775.0722.7639.7627.5384.7831.2815.389.8785.32114.5329.2119.5458.7325.88
U2Net (2020)91.0684.6191.5892.32286.5277.4585.8989.882.2873.4379.5388.43290.9685.53291.2392.172
I2UNet (2024)92.32287.60294.14178.0187.7584.45186.7088.5975.0565.9469.8949.5087.4177.8792.38277.53
SR-AttNet (2023)85.2076.7988.9284.1488.0280.8388.37290.89283.35276.42284.0983.4191.03285.0490.9992.02
FoccusU2Net (proposed)93.6189.3193.1293.3189.8184.3288.98291.14286.4177.6184.2293.2193.6188.1193.7194.81

Modified encoder-decoder architectures (DilatedRes-fully convolutional network, PolypSegNet, SR-AttNet, U2 Net, I2 Net, PSPNet, DC-UNet, MSRFNet, BA-Net) benefit from features such as dilated residual convolution (a deep-learning tool to look at a wider area of an image without losing details) to improve fully convolutional network, skip connections via multi-scale context fusion, stretch-relax attention with feature-to-mask extraction, nested U-structure to capture rich contextual information, dual-path U-Net for low-level details, high-level semantics, pyramid scene parsing network for pixel-level prediction, efficient residual CNN for improved gradients, multi-scale feature fusion by multiple dual shot face detectors blocks, boundary-aware modules to explicitly find edge-details, but fail to excel because of poor generalization, risk of overfitting (learning the training data too well, but performing poorly on new, unseen data), feature redundancy, semantic gap in encoding–decoding.

Transfer learning-based approaches (DoubleUNet, HarDNet-MSEG, Inception U-Net) represent another important category of deep learning models for polyp segmentation. These methods leverage pretrained feature extractors to enhance performance and efficiency, offering several advantages: Atrous spatial pyramid pooling for improved multi-scale context capture in semantic segmentation networks, stacked U-Net architectures, high-performance backbones with cascaded decoders for refined feature extraction, and multi-modality fusion with deep encoding, enabling integration of information from multiple data sources. However, these models also present notable limitations, including limited interpretability, high computational cost, overfitting and adaptability issues.

Transformer-based approaches (SwinE-Net, TransUnet, SwinPA-Net) have some advantages, such as EfficientNet with Swin transformer [vision transformer (ViT) architecture] for global context, transformer-CNN hybrid encoder with U-Net Decoder, Swin transformer with local pyramid attention module. Limitations include lack of inductive biases (guiding principles for a model to predict new data more reliably), high memory demand, and limited spatial understanding.

Attention-based approaches (FocusU-Net, FANet, ResGANet, PraNet, CSAP-UNet), have benefits from dual attention gate (a mechanism for image segmentation) with short-range skip connections, feedback module with hard attention to refine features, lightweight ResNet with modular group attention blocks, parallel partial decoder with reverse attention (a mechanism for image segmentation used in difficult regions), parallel CNN and transformer with self-attention. Challenges include limited fine-grained feature capture, imbalanced local-global context, class imbalance issues and training instability (when the learning process fails or becomes erratic) and overfitting.

The advantages of foundation model-based approaches (SAM, SAM2, MedSAM) include: Zero-shot transfer to various tasks, evaluation of SAM for polyp segmentation and capability of universal medical image segmentation. Challenges include poor model interpretability, boundary refinement issues, poor generalization and high cost.

Another such program is DeepLabV3+, with an encoder-decoder structure and ResNet architecture. The Kvasir-SEG polyp dataset was used to train the learning-machine, and the model proved high performance metrics such as mean Dice similarity coefficient: 0.9873, mean intersection over union (IoU): 0.9751, making it suitable for polyp segmentation. DeepLabv3+’s performance had higher indicators in polyp segmentation compared to studies conducted with U-Net and similar structures in the assessment of colonoscopic images[111].

In addition, a distinct model known as DenseNet-169-based online Tigerclaw fuzzy region segmentation, originally developed for dermoscopy imaging, has demonstrated remarkable performance, achieving 98.9% accuracy and high classification robustness for benign lesions, and 97.4% accuracy for malignant lesions, provided that the input data are properly preprocessed and segmented. This model shows strong potential for real-time precision diagnosis of CRC and other malignancies[112].

Cecal intubation rate (CIR) is also a key quality indicator, as it is associated with a better standardized colonoscopy, better ADR and advanced adenoma detection (ADDR), which is absolutely necessary in screening colonoscopy. Until present, cecal intubation has been a self-acknowledged feature, but AI can help assist colonoscopy and achieve better CIR and ADR by combining the two techniques[113]. All of the above considered led to lower rates of post-colonoscopy CRC[114,115]. In one study, an AI-based cecum recognition system, known as the AI-common reporting standard, was employed for post-hoc verification during CRC screening colonoscopies. While it did not further improve CIR, it significantly increased both the ADR and ADDR by 5%, with the most notable improvements observed in the proximal colon, the region where post-colonoscopy CRC most commonly arises[113].

Designing applications, programs and networks for CRC screening enhancement and analysis involving AI

Selvaraj et al[6] explored a more practical real-time CRC screening AI colonoscopy application with a three-step approach: (1) Automated polyp segmentation using CRPU-Net architecture; (2) Analysis of machine learning for binary classification of segmented regions using scale-invariant feature transform and oriented fast and rotated brief descriptors, followed by feature fusion and dimensionality reduction through principal component analysis to create an optimal feature representation for classification; and (3) Comparison of deep learning exploration for automated classification using state of the art and ViT model.

Additionally, they considered the development of a hybrid model colorectal polyp (CRP)-ViT by potentially combining the ViT model with deep learning architectures. As a result, CRPU-Net achieved outstanding segmentation performance with 96.56% accuracy, 95.40% IoU and 97.39% Dice coefficient. Even more, the CRP-ViT model demonstrated promising results in binary classification with 96.59% accuracy, 96.38% sensitivity, 96.80% specificity, 96.82% precision and 96.36% negative predictive value, with the endpoint being the implementation of CRPU-Net and CRP-ViT, AI systems designed for real-time polyp segmentation and potential precancerous polyp classification[6].

Another way to achieve the quality indicators in CRC screening colonoscopy is using new state-of-the-art 4k image colonoscopes with AI aid computer aided detection endo-aid CADe. Orzeszko et al[106] examined two groups with 1000 participants each, with one group using CADe. The study found that only polyp detection rate was higher in the CADe group. There was no significant difference in any other segment or parameter[116,117].

Venkatayogi et al[118] have suggested the use of transfer learning and a vision-based surface tactile sensor (VS-TS) to classify polyps detected during colonoscopy. VS-TS includes a deformable silicone membrane, an optics module, and an array of light-emitting diodes to enhance polyp illumination during imaging. The sensor detects deformation of the silicone when touching polyp phantoms, and each phantom presses against the sensor. In this way, polyps can be identified and characterized according to the detailed textural patterns. Such textural features provided by extraction by the VS-TS can be used and integrated in a SVM algorithm for polyp classification, which requires data from the ImageNet dataset and a preceding training of ResNet-18. Classification occurs on the basis of two versions of ResNet-18: The first uses random weights (ResNet1) and the second, the preceding training on ImageNet (ResNet2). ResNet2 demonstrates an accuracy rate of 91.93%, higher than that of ResNet1, with an accuracy of 54.95%[117,118].

Another network, called area-boundary constraint network, was developed, which focuses on the polyp itself with its characteristics and boundaries, using one encoder for capitalization and two decoders, one for the polyp and the other for the boundary, to raise segmentation effectiveness. It uses selective kernel modules to obtain more accurate and dynamic images of polyps for better segmentation. For training, attentive but diverse network uses 3 databases: EndoScene, Kvasir-SEG and ETIS-Larib[119-121].

A newer way to perform polyp segmentation was proposed by Elkarazle et al[122], which uses an upgraded version of the multi-scale attention network (MA-NET) aided by a modified Mix-ViT transformer, enhancing its capability to obtain ultrafine-grained visual classification, and therefore more accurate segmentation of difficult polyp types. By adapting the Mix-ViT transformer for this specific application, one can replace the typical convolution-based encoder in MA-NET and leads to better feature extraction at multiple scales. Its advantage is the ability to distinguish between polypoid and non-polypoid regions. The network can enhance features by including a layer that applies contrast-limited adaptive histogram equalization in the CIELAB color for input images and provides a proper solution for segmenting polyps that are small and flat. The necessary training of the model includes Kvasir-SEG and CVC-ClinicDB datasets, with added cross-validation on CVC-ColonDB and ETIS-LaribDB for robustness and applicability[122-124].

Does colon capsule endoscopy have a role in CRC screening?

One neglected technique of colon visualization is colon capsule endoscopy (CCE). The results were initially disappointing, and it was expensive because of the need to use one capsule for each patient, and each lesion needed a follow-up colonoscopy. During the COVID-19 pandemic, the technique was revisited with better equipment, cheaper and more accurate images, and some promising results were achieved. Researchers are considering coupling it with AI to achieve better polyp matching, which opens up a novel field in endoscopy with potential to address challenges regarding the polyp matching within the same video. Other studies found different figures with more promising uses in CCE. However, all studies found CCE in CRC screening requires some adjustments, including better equipment, lower cost and more input from AI programs[125-127]. Despite the proven potential of AI, refining such techniques is crucial to make CCE more reliable than conventional colonoscopy for lower gastrointestinal diagnostics (Table 3)[128-138].

Table 3 The comparison between ileo-colonoscopy and colon capsule endoscopy[128-138].

CCE
Colonoscopy
Extent of gastrointestinal tract examinedGastric antrum, small bowel and colon on CCETerminal ileum and colon only for colonoscopy
Patient safetyCCE has non-invasive with minimal capsule retention risk, reliant on patient selection (0.73%-2%)Colonoscopy is invasive with perforation risk: 88 per 100000 people (0.88%)
Bowel preparation requirementAdditional low residue diet or high-volume laxative e.g., polyethylene glycol in addition to standard bowel preparation for CCEStandard bowel preparation including volume bowel preparation in standard colonoscopy
Ability in taking biopsies and therapyUnable to take biopsies or perform therapeutics with capsuleAble to take biopsies or perform therapeutics with the colonoscope
LocalizationNo scope guided for localization of pathology other than visual landmarks such as ileocecal valve, appendiceal orifice and anal cushion in video-capsuleScope guided is available for more accurate localization of the pathologies within the colon at colonoscopy
Procedure timeCCE has an average reading time: 45-60 minutesColonoscopy has an average 30 minutes procedural slots
Improving conduct in the post-screening steps: Polypectomy, surgery, lymph node removal

Considering that screening colonoscopy includes the resection of polyps greater than 1 cm, of which more than 13% are T1 cancers, the question of when to address them via surgery remains. The challenge offers AI an opportunity to create an artificial NN to predict the risk of recurrence using colonoscopy and pathology images. Su et al[139] found that the AI model prevented more than 34.9% of unnecessary surgical resections compared with United States guidelines in all enrolled patients.

Following endoscopic resection of T1 CRC, some procedures require further surgery for lymph node metastasis. An AI model can be employed here as well to reduce unnecessary surgery after endoscopic resection. This possibility has been explored using four classifiers: Regularized LR classifier, RF classifier, CatBoost classifier (CBC), and the voting classifier. Among these, CBC showed the best response based on the Japanese Society for Cancer of the Colon and Rectum guidelines. Furthermore, it demonstrated that by training some AI models, unnecessary surgery can be avoided[140].

An interesting application explored by Liu et al[141] is Fovea-UNet, a deep learning model inspired by the human eye fovea. Its aim is to detect and segment lymph node metastases of CRC using CT images. The study used a dataset containing 81 whole slide images (WSI) of metastases from 624 metastatic regions that were extracted and labeled, as well as divided into a training set 57 WSIs (451 metastatic regions) and a test set 24 WSIs (173 metastatic regions). The study directs the model’s focus on the important areas, using a special module adjusted according to feature relevance. In the feature extraction process, they involve a lightweight backbone that is adjusted to a regularization strategy (GhostNet backbone) to cut on demands, while maintaining extraction efficiency. The proposed model has shown its superiority in the segmentation process, promising 79.38% IoU and 88.51% Dice similarity coefficient, better than other models[141].

Histopathology standardization in CRC screening

When going further and asking AI to look into pathology and predict precancerous and cancerous lesions, unfortunately all of the potential AI engines failed to be more accurate than specialists[142]. However, AI can perform better in diagnosis using a deep learning model that classifies WSIs into various categories, differentiating the stage of CCR and the normal tissue sample. This model includes a CNN architecture, such as ResNet, which has proven superior in dealing with multiple images, and creates three frameworks: The first uses image level with low resolution to detect potential cancer; the second uses cellular level images that employ the CNN to differentiate cancer from noncancer; and the third combines the first two for better diagnosis, reaching 94.6% accuracy on the Cancer Genome Atlas (images from Cancer Genome Atlas) dataset and 92.0% accuracy on an external dataset with real life images from hospitals[143].

To better train AI models, some researchers have provided histological images of CRC samples from a dataset available by Kather with 5000 tissue tiles, to help distinguish between tumor and stroma and integrate the data through a transfer-learning-based binary classifier. Such framework employs four different CNN architectures: Visual geometry group (VGG) 19, EfficientNetB1, InceptionResNetV2 and DenseNet121, with background information from ImageNet dataset. Tuning the classifier on specific features of the CRC dataset, one can also retain the learned features. The results of these architectures are of higher accuracy: 96.4%, 96.87%, and 97.65%, for VGG19, EfficientNetB1 and InceptionResNetV2 respectively, which go beyond the presented reference values of the study. In conclusion, applying transfer-learning using pre-trained CNNs works as a booster for classifying tumor and stroma regions in terms of histological CRC images[144,145].

One can go further towards feature extraction and use optimal deep feature fusion approach on biomedical images method, median filtering to eliminate noise, and a combination of three deep learning models: MobileNet, SqueezeNet and SE-ResNet. A selection of hyperparameters can be performed using the osprey optimization algorithm; finally, a deep belief network model is engaged to better classify CRC. The technique was tested on the Warwick-QU dataset and achieved 99.39%accuracy[146].

Imaging

In the process of CRC screening, imaging of the colon (computer-tomography and magnetic resonance imaging) is also useful, especially for patients who are ineligible for colonoscopy (if there are fixations that cannot be passed or if anesthesia is not recommended), or in the future, when more noninvasive techniques for screening are expected to be developed. Even in this field, the results are relatively subjective and tend to be operator-dependent. Under such circumstances, the AI intervention may optimize diagnosis, proven by some studies that use RK-Net with unsupervised learning (UL) techniques and deep learning. The network takes cluster images, performs extraction using Mo-bileNetV2 and learns to identify cancerous tissue. The benefits of using UL are reduced training time by selecting only the most relevant clusters, reducing costs and time for training, while achieving 95% accuracy[147].

REAL-LIFE AI-ASSISTED GUIDELINES ACCORDING TO RECOMMENDATIONS OF WORLDWIDE MAJOR ASSOCIATIONS

AI inevitably remains an intriguing field due to its variability, training capacity and impact on various medical societies across the world. Table 4 summarizes the guidelines provided by several important associations in terms of CRC screening.

Table 4 Real-life recommendations of world-wide major associations[148-153].
Organization
Starting age
Screening methods
Interval
AI integration/position
American Cancer Society45Colonoscopy, stool-based (FIT/FIT-DNA), CT-colonographyColonoscopy: 10 years; FIT: Annually; FIT-DNA: Every 3-yearsSupports emerging AI technologies in clinical research and potential guideline updates
American Gastroenterological Association45Colonoscopy preferred; other options acceptable for shared decision-makingBased on methodDraft guidelines recommend AI-assisted colonoscopy (CADe) as a quality enhancing tool
European Society of Gastrointestinal Endoscopy50 (varies by country)FIT-based programs, colonoscopy in select settingFIT: Every two years; colonoscopy: Every ten yearsEncourages integration of CADe systems in clinical practice to improve adenoma detection rates
National Comprehensive Cancer Network45Colonoscopy, stool-based tests, CT colonographyColonoscopy: Ten years; FIT/FIT-DNA: Per testSupports AI use for colonoscopy enhancement particularly in high-risk population
World Health Organization50 (resource dependent)FIT-based screening for population level programsFIT: Every two yearsRecognizes potential role of AI in expanding access and accuracy in low-resource setting; encourages cost-effective AI research
Japanese Society of Gastroenterology40 (or even earlier for workplace health checks)FITFIT: AnnuallyJapan’s first major AI-colonoscopy system, EndoBRAIN, received regulatory approval and reimbursement starting in 2024
Food and Drug Administration45 (earlier initiation for those with higher risk factors)Colonoscopy, FIT, stool DNA-FIT, CT colonography, flexible sigmoidoscopyColonoscopy: Every ten years; FIT: Annually; Stool DNA-FIT: Every 1-3 years; CT colonography: Every five years; Flexible sigmoidoscopy: Every 5-10 yearsFDA has cleared CADe systems for real-time detection assistance during colonoscopy, but explicitly prohibits there use for lesion diagnosis or automated clinical decision making
American Gastroenterological Association guideline

Recommendation (with grade: No recommendation-very low certainty of evidence): In adults undergoing colonoscopy, the American Gastroenterological Association (AGA) makes no recommendation on the use of CADe-assisted colonoscopy[148]. Currently, there are draft guidelines that suggest CADe as a quality-enhancing tool, especially in polyp finding in adults[148].

Remarks[148]: CADe is already acknowledged by the AGA to improve ADR, but overall, there is a very low certainty of evidence on long-term outcomes for decision making in CRC incidence and mortality[148]. An update is planned for this recommendation because CADe continues to improve as an AI application[148]. There are evidence gaps on data from studies referring to the impact on advanced adenomas, difficult-to-detect adenomas, access to colonoscopy, patient values and preferences, cost-effectiveness or impact on long term outcomes[148].

The combination between CADe and computer-aided diagnostics may add to post-polypectomy surveillance in the context of this technology, and further studies may be applied on diagnosing and resecting polyps with CADe alone[148].

One of the most important issues is the concern for security in patient privacy, data security and ownership of AI platforms used in healthcare. According to some authors, arising healthcare errors should be a responsibility shared between the doctors who used the device and the designers[148].

There are several CADe systems on the healthcare market with periodic updates, but these should be more transparent and accompanied by training tools and facilities. Even for educational purposes, there are authors who believe it would negatively impact skill development[148].

The American Society for Gastrointestinal Endoscopy AI task force statements

Current advances in AI allow for the development of AI-based algorithms that can be applied to endoscopy to augment endoscopist performance in lesion detection and characterization. This is equivalent to subjectivity reduction in reporting quality and helping to build smart endoscopy algorithms, which can subsequently optimize workflow in endoscopy, including improved documentation[149].

Using AI and machine learning aids in predictive models, diagnosis, and prognosis, but high-quality data with multidimensionality are needed for risk prediction and forecasting of specific clinical conditions and their outcomes, whenever using machine learning methods[149].

Large data pool and cloud-based tools are essential for the clinical research in gastroenterology to go a step further. To understand maximal extent of the diseases and make treatment decisions, multimodal data are required for training AI[149].

Understanding how to evaluate AI algorithms in the gastroenterology literature and clinical trials is important for gastroenterologists, trainees, and researchers, therefore implying educational efforts to be made by gastroenterology societies[149].

AI algorithms have to be transparent, easy to interpret and explain, so they can be adopted in the clinical practice of gastrointestinal endoscopy. Regarding the payment of AI in endoscopy, a thorough evaluation of AI systems needs to be performed to purchase the most reliable and cost-effective endoscopy guide. Studies to guide reimbursement may also be needed[149].

Evidence standards for AI in gastroenterology are not yet well-defined and need further development. Several aspects still remain to be dealt with and require a balanced view of AI technologies and an active collaboration between the medical team and staff from technology, industry, computer science fields, last but not least researchers. This is critical for a suitable advancement of AI in gastroenterology[149].

European Society of Gastrointestinal Endoscopy position statement

The general belief of this panel is that patients would opt-in for CADe assistance for screening or surveillance colonoscopy, during their procedure, on condition that they are adequately informed. They should be made aware of both potential benefits and limitations, in the case of such measure for reducing CRC incidence and mortality[150]. However, this recommendation is weak because it bears a significant uncertainty of evidence, low absolute benefits for CRC incidence and mortality, and it increases the patient burden associated with CADe use, especially by over-diagnosing polyps and the need for more surveillance by colonoscopy[150].

Japanese Society of Gastroenterology statement

Medical innovation is often most exciting in its early stages, particularly during the design and promotion of preliminary products. However, before new medical devices can be implemented in clinical practice, they must undergo extensive bureaucratic processes that are both time-consuming and expensive. However, these challenges should not discourage the development of brilliant ideas in medicine. To maintain high standards and strict regulations, the regulatory authorities should be involved from the start to find solutions. For example, in Japan, the Pharmaceuticals and Medical Devices Agency, Medical Device Agency and the Ministry of Health, Labor and Welfare provide opportunities for consulting in matters of regulatory approval and reimbursement applications. Several public bodies, such as Japan Agency for Medical Research and Development, offer support, including financial support, in the whole process of implementing new medical devices and applications, with an even greater opening towards challenging projects[151].

Finally, even after achieving regulatory approval and reimbursement, the journey is not over: Any new medical or AI device should be continuously monitored for benefits and harms that potentially arise during use, and this also requires a tremendous effort, all for the optimization of healthcare[151].

In 2025, the data are promising, but in a real medical setting, there is one question to be answered regarding the AI assistance, before implementation: “Who is ultimately responsible for a clinical result?” Even with its continuous improvement and refinement, AI cannot avoid false-positive or false-negative diagnosis, thus human doctors must take responsibility for the final clinical decision. The Japan Gastroenterological Endoscopy Society provides guidelines for the clinical use of AI software (https://www.jges.net/wp-content/uploads/2023/05/AIsoft.pdf). The guidelines clearly state that physicians are responsible for diagnosis, and that AI should only be used adjunctively to enhance performance in various clinical settings and contribute to the better health of people around the world. Japan’s first major AI-colonoscopy system, EndoBRAIN, received regulatory approval and reimbursement starting in 2024[152].

In conclusion, Japan, as a leader in the field of gastrointestinal endoscopy, takes on the responsibility to demonstrate the clinical and economic benefits of AI in gastro-intestinal endoscopic diagnosis, to initiate social implementation and to build further evidence[152].

Food and Drug Administration

In January 2021, the Food and Drug Administration (FDA) launched its AI/machine learning based software as a medical device action plan, which sets the foundation for regulating AI in healthcare. Its main points are: A tailored regulatory framework for such software, adherence to good machine learning practices, patient-centered transparency, algorithmic bias control, robust performance and real-world performance monitoring.

The AI tools for CRC detection cleared by the FDA are: Gastrointestinal genius (which assists real time colonoscopy interpretation with a raise in ADR from 42% to 55.1%); SKOUT system (a CADe device for trained gastroenterologists to flag potential polyps in real time colonoscopy); Olympus CADDIE (the first cloud-based AI platform for CRC colonoscopy assistance allowing remote updating)[153].

STATE OF THE ART

In 2025, ChatGPT (version 4.5) was evaluated for geographic and linguistic accuracy to provide recommendations for CRC screening and monitoring. Due to detected variations across countries and languages, the AI model requires further validation according to each context. Large language models need to be equipped with high-quality clinical data, sensitive to the region they originate in, so that they can be used in real-world settings in a reliable manner. The characteristics they still lack are standardization and transparency. All these could be enhanced by reducing present limitations in data sourcing, time and version differences and title-recommendation discrepancies. Healthcare professionals should ponder all input when using AI-generated advice in clinical practice[154].

It has been demonstrated that deep learning, radiomics and multimodal data integration may achieve similar results as expert endoscopists in matters of endoscopic image examinations. However, the challenges that arise are due to limited model generalization (fragmented datasets), algorithmic limitations in rare conditions, insufficient training data and ethical issues. To overcome these, multicenter databases should be acquired, AI tools in prospective trials should be validated, and programs to raise clinical trust, unified ethical/regulatory frameworks should be developed[155].

AI models can also assist polyp classification in computed tomography colonography, aid in and select the referral of patients for endoscopy (as a second reader). In terms of economic impact, such aid would result in cost-effectiveness. According to guidelines, the recommendations are the following: The resection of CRPs ≥ 10 mm and CRPs of 6-9 mm depending on age or other conditions should be followed by endoscopy, while polyps less than ≤ 5 mm should undergo screening computed tomography colonography. However, these recommendations have proven to not be cost-effective, only gaining 464407 United States dollar per life-year, compared to 59015 United States dollar for polyps with a size of 6-9 mm and 151 United States dollar cost savings for each patient, for polyps larger than 10 mm[156-160]. AI provides a benefit as a second reader for polyp classification, turning endoscopic referral after computed tomography colonography into a more effective tool when analyzing and diagnosing adenomatous and non-adenomatous CRPs[161]. Furthermore, CADe using computed tomography colonography data could improve sensitivity by analyzing images obtained in two positions. At the same time, both sensibility and sensitivity can be increased by a deep learning algorithm that integrates training to detect small and large polyps, low-dose pretreatment, non-uniform intestinal dilatation, or low-dose imaging[162,163].

An intriguing recent study describes how four AI models integrated with ColonView test results and clinical features of the patients have outperformed the diagnostic accuracy of the conventional LR method in FIT-based screening. These models are: SVM, NN, RF and GBM and confirm the augmentation of the diagnostic accuracy rate when combined with non-AI/clinically obtained data[164].

Increasing ADR and polypectomy rate in AI-assisted colonoscopy is a goal for all gastroenterologists worldwide. In Asia, the use of AI-aided colonoscopy (real life) has brought about a significant difference for endoscopists, raising ADR from 24.3% to 30.4% and the polypectomy rate from 28.4% to 33.6%[165].

Finally, the tendency for such studies is also remarked regionally in small countries in certain regions; an example is Romania: South-West Oltenia studied and suggested a refined framework for CRC risk stratification, using NN-based composite scoring for stratification of FIT[166].

CLINICAL AND ECONOMIC IMPACT OF IMPLEMENTING AI

Beyond all scientific fields of CRC screening in which AI has been involved, rather complex and sometimes hard to implement, the real-life clinical impact should encourage further research and innovative ideas. In matters of clinical aspect, AI has increased the ADR, to reduce missed lesion rates, by extending the spectrum of colors, areas that are difficult to visualize, flat polyps that are hard to identify. Moreover, it improves in vivo polyp characterization by increasing the prediction rate for the stage of polyp/cancer, solidifying the link to histopathology, better identifying hyperplastic polyps to resect and not collect (to reduce the number of unnecessary polypectomies), predicting bleeding in performing a polypectomy, and in the end stratifying the risk for every individual patient.

Another benefit is that it standardizes colonoscopy quality by reducing inter-individual variability, raising examination quality, aligning the description language (for both colonoscopy and histopathology), and raising colonoscopy quality for beginners.

Finally, it integrates and promotes CRC screening by identifying the population who needs screening, and highlighting the risk population and raising awareness. It may also reduce the rate of duplication or omission and correct the weak points of screening.

It also has a significant economic impact by reducing the cost of double colonoscopy when bowel preparation is weak, reducing unnecessary histopathological assessment of polypectomies in patients who do not require it, and raising the effectiveness of the procedures, shortening their duration, and possibly reducing their number or the interval between re-screening. The list continues, with a reduction of the number of unnecessary surgeries, prioritization of cases that need emergency colonoscopy or additional attention from an expert, rather than a beginner. In the end, reducing the costs of advanced CRC by detecting early lesions, and either standardizing the treatment in each patient case or adjusting to its stage should not be neglected.

LIMITATIONS

This review has primarily focused on the benefits of AI assistance during various steps of CRC screening, diagnosis and medical conduct. However, there are also limitations that should be mentioned. First, there is no perfect technology, especially in the field of medicine. Deep learning mechanisms perform well given that they are provided sufficient data, such as thousands of images with polyps, histopathology, computed tomography and magnetic resonance imaging image slides. Considering that even in real life cases, polyp classification and recurrence pattern prediction are still challenges, even for experts, slight differences are likely to arise in deep learning. While expert opinions can be accompanied by arguments, AI may fail to provide reasonable explanation.

Another aspect to be considered is population variability due to age, gender, and race. In this case, AI algorithms can have biased results; for example, NNs can perform well for a certain population (for which the program was trained) but make wrong decisions for another (for which the data input is low). Besides population, there is significant variability between endoscopes that can alter and bias the algorithms.

Also, results can be influenced by situations when the human eye proves its superiority to the AI due to its fast adaptability. This is the case when AI cannot discriminate and analyze correctly due to endoscopic factors such as peristaltic movements, gas bubbles, light variations, contrast, leading to lower performance.

Last but not least, there are still economic and ethical perspective problems to be addressed: The responsibility of AI errors (for example, missed cancerous lesions), patient data confidentiality, workforce reduction in the case of AI overtaking, and the financial burden. Patients have become increasingly preoccupied by the confidentiality of their personal information, every medical procedure requires a signed consent; involving computer programs raises new worries among patients the fear that their personal data may virtually leak worldwide.

If AI is to be officially involved in diagnostic procedures, the financial expenses may initially increase, as most programs to be implemented come with high costs. Therefore, the questions are: What healthcare department decides which programs and devices should be implemented in the patient workflow; how they can be made available worldwide, who should pay for them, and what happens in the scenario in which human task force is replaced by AI?

CONCLUSION

AI has the potential to significantly transform CRC screening at multiple stages of the patient diagnostic process. Its integration begins with improving diagnostic accuracy of noninvasive tests, such as FIT, by applying algorithms that optimize threshold settings and patient stratification. In addition, AI-assisted tools can support bowel preparation, guiding patients through personalized instructions to ensure high-quality colonoscopy outcomes. During endoscopic evaluation, AI has the greatest impact by increasing ADR through real-time image analysis, better characterizing detected polyps and reducing the rate of missing precancerous lesions. It can also reduce the number of unnecessary interventions endoscopic and surgical procedures. Beyond detection, AI contributes to therapeutic decision-making in the post-screening stages, helping in surgical planning and lymph node removal, enhancing both safety and outcomes by reducing unnecessary interventions. Other technologies that are less user-friendly, such as colonic capsule endoscopy, offer a promising alternative screening approach when combined with AI, especially in populations with limited access to traditional colonoscopy. In imaging, AI improves the accuracy of interpretation, particularly in computed tomography colonography and capsule endoscopy, thereby expanding screening capabilities beyond conventional methods. In addition, AI facilitates standardization of histopathological analysis, reducing interobserver variability and improving the consistency of CRC diagnostics. The development of AI-based applications and networks can streamline the entire CRC screening process, optimizing workflows, ensuring compliance with follow-up and facilitating data sharing for population-wide screening programs. From a broader perspective, the implementation of AI in CRC screening promises positive clinical and economic impact, with the potential for earlier detection, reduced interval cancers, and cost savings due to more efficient use of resources. There are, however, limitations that remain, including the generalization of algorithms, data privacy concerns, regulatory hurdles and the need for extensive clinical validation. In conclusion, AI holds the promise of substantially enhancing the effectiveness, efficiency and equity of CRC screening, but its full potential will only be achieved through careful integration, rigorous evaluations and ethical consideration.

Footnotes

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

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Romania

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade B

Novelty: Grade B, Grade B, Grade C

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

Scientific Significance: Grade B, Grade B, Grade D

P-Reviewer: Al-Bayati K, MD, Researcher, Canada; Rizwan M, PhD, Pakistan S-Editor: Fan M L-Editor: Filipodia P-Editor: Yu HG

References
1.  Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5690]  [Cited by in RCA: 9836]  [Article Influence: 9836.0]  [Reference Citation Analysis (3)]
2.  Issa IA, Noureddine M. Colorectal cancer screening: An updated review of the available options. World J Gastroenterol. 2017;23:5086-5096.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 434]  [Cited by in RCA: 398]  [Article Influence: 49.8]  [Reference Citation Analysis (11)]
3.  Tariq H, Kamal MU, Sapkota B, ElShikh F, Pirzada UA, Pullela N, Azam S, Zhang A, Baiomi A, Abbas H, Makker J, Balar B, Ihimoyan A, Daniel M, Dev A. Evaluation of the combined effect of factors influencing bowel preparation and adenoma detection rates in patients undergoing colonoscopy. BMJ Open Gastroenterol. 2019;6:e000254.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
4.  Siegel RL, Wagle NS, Cercek A, Smith RA, Jemal A. Colorectal cancer statistics, 2023. CA Cancer J Clin. 2023;73:233-254.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1681]  [Reference Citation Analysis (3)]
5.  Fu J, Gao Y, Zhou P, Huang Y, Jiao J, Lin S, Wang Y, Guo Y. D2polyp-Net: A cross-modal space-guided network for real-time colorectal polyp detection and diagnosis. Biomed Signal Proces. 2024;91:105934.  [PubMed]  [DOI]  [Full Text]
6.  Selvaraj J, Umapathy S, Amarnath Rajesh N. Artificial intelligence based real time colorectal cancer screening study: Polyp segmentation and classification using multi-house database. Biomed Signal Proces. 2025;99:106928.  [PubMed]  [DOI]  [Full Text]
7.  Vaid AK, Mohapatra PN, Desai C. Indian consensus statement on the management of metastatic colorectal cancer. Int J Adv Med. 2021;8:1775.  [PubMed]  [DOI]  [Full Text]
8.  Indian Council of Medical Research  Consensus Document for Management of Colorectal Cancer. [cited September 25, 2025]. Available from: https://www.icmr.gov.in/icmrobject/custom_data/pdf/resource-guidelines/Colorectal%20Cancer_0.pdf.  [PubMed]  [DOI]
9.  van Rijn JC, Reitsma JB, Stoker J, Bossuyt PM, van Deventer SJ, Dekker E. Polyp miss rate determined by tandem colonoscopy: a systematic review. Am J Gastroenterol. 2006;101:343-350.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 878]  [Cited by in RCA: 923]  [Article Influence: 48.6]  [Reference Citation Analysis (0)]
10.  Morris EJ, Rutter MD, Finan PJ, Thomas JD, Valori R. Post-colonoscopy colorectal cancer (PCCRC) rates vary considerably depending on the method used to calculate them: a retrospective observational population-based study of PCCRC in the English National Health Service. Gut. 2015;64:1248-1256.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 81]  [Cited by in RCA: 111]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
11.  Forbes Insights  AI And Healthcare: A Giant Opportunity. [cited September 30, 2025]. Available from: https://www.forbes.com/sites/insights-intelai/2019/02/11/ai-and-healthcare-a-giant-opportunity/?sh=12813fe94c68.  [PubMed]  [DOI]
12.  Morson B. President's address. The polyp-cancer sequence in the large bowel. Proc R Soc Med. 1974;67:451-457.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 142]  [Cited by in RCA: 125]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
13.  Scarà S, Bottoni P, Scatena R. CA 19-9: Biochemical and Clinical Aspects. Adv Exp Med Biol. 2015;867:247-260.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 144]  [Cited by in RCA: 230]  [Article Influence: 25.6]  [Reference Citation Analysis (1)]
14.  Fiala L, Bob P, Raboch J. Oncological markers CA-125, CA 19-9 and endometriosis. Medicine (Baltimore). 2018;97:e13759.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
15.  Bottoni P, Scatena R. The Role of CA 125 as Tumor Marker: Biochemical and Clinical Aspects. Adv Exp Med Biol. 2015;867:229-244.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 83]  [Cited by in RCA: 123]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
16.  Imperiale TF, Ransohoff DF, Itzkowitz SH, Levin TR, Lavin P, Lidgard GP, Ahlquist DA, Berger BM. Multitarget stool DNA testing for colorectal-cancer screening. N Engl J Med. 2014;370:1287-1297.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1015]  [Cited by in RCA: 1265]  [Article Influence: 115.0]  [Reference Citation Analysis (1)]
17.  Vakil N, Ciezki K, Huq N, Singh M. Multitarget stool DNA testing for the prevention of colon cancer: outcomes in a large integrated healthcare system. Gastrointest Endosc. 2020;92:334-341.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
18.  Shirley M. Epi proColon(®) for Colorectal Cancer Screening: A Profile of Its Use in the USA. Mol Diagn Ther. 2020;24:497-503.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
19.  Potter NT, Hurban P, White MN, Whitlock KD, Lofton-Day CE, Tetzner R, Koenig T, Quigley NB, Weiss G. Validation of a real-time PCR-based qualitative assay for the detection of methylated SEPT9 DNA in human plasma. Clin Chem. 2014;60:1183-1191.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 149]  [Cited by in RCA: 219]  [Article Influence: 19.9]  [Reference Citation Analysis (0)]
20.  Yi JM, Dhir M, Guzzetta AA, Iacobuzio-Donahue CA, Heo K, Yang KM, Suzuki H, Toyota M, Kim HM, Ahuja N. DNA methylation biomarker candidates for early detection of colon cancer. Tumour Biol. 2012;33:363-372.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 53]  [Cited by in RCA: 59]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
21.  Ferreira MM, Ramani VC, Jeffrey SS. Circulating tumor cell technologies. Mol Oncol. 2016;10:374-394.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 357]  [Cited by in RCA: 384]  [Article Influence: 42.7]  [Reference Citation Analysis (0)]
22.  Agarwal A, Balic M, El-Ashry D, Cote RJ. Circulating Tumor Cells: Strategies for Capture, Analyses, and Propagation. Cancer J. 2018;24:70-77.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 49]  [Cited by in RCA: 53]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
23.  Kumar A, Aravind N, Gillani T, Kumar D. Artificial intelligence breakthrough in diagnosis, treatment, and prevention of colorectal cancer – A comprehensive review. Biomed Signal Proces. 2025;101:107205.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
24.  Han J, Park S, Kim LA, Chung SH, Kim TI, Lee JM, Kim JK, Park JJ, Lee H. Machine Learning-Enabled Non-Invasive Screening of Tumor-Associated Circulating Transcripts for Early Detection of Colorectal Cancer. Int J Mol Sci. 2025;26:1477.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
25.  Alzubaidi L, Zhang J, Humaidi AJ, Al-Dujaili A, Duan Y, Al-Shamma O, Santamaría J, Fadhel MA, Al-Amidie M, Farhan L. Review of deep learning: concepts, CNN architectures, challenges, applications, future directions. J Big Data. 2021;8:53.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3068]  [Cited by in RCA: 1200]  [Article Influence: 300.0]  [Reference Citation Analysis (0)]
26.  Sjöstedt E, Zhong W, Fagerberg L, Karlsson M, Mitsios N, Adori C, Oksvold P, Edfors F, Limiszewska A, Hikmet F, Huang J, Du Y, Lin L, Dong Z, Yang L, Liu X, Jiang H, Xu X, Wang J, Yang H, Bolund L, Mardinoglu A, Zhang C, von Feilitzen K, Lindskog C, Pontén F, Luo Y, Hökfelt T, Uhlén M, Mulder J. An atlas of the protein-coding genes in the human, pig, and mouse brain. Science. 2020;367:eaay5947.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 192]  [Cited by in RCA: 704]  [Article Influence: 140.8]  [Reference Citation Analysis (0)]
27.  Tóth K, Sipos F, Kalmár A, Patai AV, Wichmann B, Stoehr R, Golcher H, Schellerer V, Tulassay Z, Molnár B. Detection of methylated SEPT9 in plasma is a reliable screening method for both left- and right-sided colon cancers. PLoS One. 2012;7:e46000.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 142]  [Cited by in RCA: 154]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
28.  Jin P, Kang Q, Wang X, Yang L, Yu Y, Li N, He YQ, Han X, Hang J, Zhang J, Song L, Han Y, Sheng JQ. Performance of a second-generation methylated SEPT9 test in detecting colorectal neoplasm. J Gastroenterol Hepatol. 2015;30:830-833.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 112]  [Cited by in RCA: 116]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
29.  Villéger R, Lopès A, Veziant J, Gagnière J, Barnich N, Billard E, Boucher D, Bonnet M. Microbial markers in colorectal cancer detection and/or prognosis. World J Gastroenterol. 2018;24:2327-2347.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 98]  [Cited by in RCA: 89]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
30.  Zhang J, Hasty J, Zarrinpar A. Live bacterial therapeutics for detection and treatment of colorectal cancer. Nat Rev Gastroenterol Hepatol. 2024;21:295-296.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
31.  Nakatsu G, Li X, Zhou H, Sheng J, Wong SH, Wu WK, Ng SC, Tsoi H, Dong Y, Zhang N, He Y, Kang Q, Cao L, Wang K, Zhang J, Liang Q, Yu J, Sung JJ. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat Commun. 2015;6:8727.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 558]  [Cited by in RCA: 523]  [Article Influence: 52.3]  [Reference Citation Analysis (0)]
32.  Yazici C, Wolf PG, Kim H, Cross TL, Vermillion K, Carroll T, Augustus GJ, Mutlu E, Tussing-Humphreys L, Braunschweig C, Xicola RM, Jung B, Llor X, Ellis NA, Gaskins HR. Race-dependent association of sulfidogenic bacteria with colorectal cancer. Gut. 2017;66:1983-1994.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 111]  [Cited by in RCA: 160]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
33.  Tilg H, Adolph TE, Gerner RR, Moschen AR. The Intestinal Microbiota in Colorectal Cancer. Cancer Cell. 2018;33:954-964.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 398]  [Cited by in RCA: 561]  [Article Influence: 80.1]  [Reference Citation Analysis (0)]
34.  McCoy AN, Araújo-Pérez F, Azcárate-Peril A, Yeh JJ, Sandler RS, Keku TO. Fusobacterium is associated with colorectal adenomas. PLoS One. 2013;8:e53653.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 406]  [Cited by in RCA: 402]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
35.  Zhang X, Zhang Y, Gui X, Zhang Y, Zhang Z, Chen W, Zhang X, Wang Y, Zhang M, Shang Z, Xin Y, Zhang Y. Salivary Fusobacterium nucleatum serves as a potential biomarker for colorectal cancer. iScience. 2022;25:104203.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 23]  [Reference Citation Analysis (0)]
36.  Zhang Y, He C, Qiu L, Wang Y, Qin X, Liu Y, Li Z. Serum Unsaturated Free Fatty Acids: A Potential Biomarker Panel for Early-Stage Detection of Colorectal Cancer. J Cancer. 2016;7:477-483.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 40]  [Cited by in RCA: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
37.  Yeo YH, Samaan JS, Ng WH, Ting PS, Trivedi H, Vipani A, Ayoub W, Yang JD, Liran O, Spiegel B, Kuo A. Assessing the performance of ChatGPT in answering questions regarding cirrhosis and hepatocellular carcinoma. Clin Mol Hepatol. 2023;29:721-732.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 177]  [Cited by in RCA: 352]  [Article Influence: 176.0]  [Reference Citation Analysis (0)]
38.  Hamet P, Tremblay J. Artificial intelligence in medicine. Metabolism. 2017;69S:S36-S40.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 728]  [Cited by in RCA: 878]  [Article Influence: 109.8]  [Reference Citation Analysis (0)]
39.  Sivapalaratnam S. Artificial intelligence and machine learning in haematology. Br J Haematol. 2019;185:207-208.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11]  [Cited by in RCA: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
40.  El Hajjar A, Rey JF. Artificial intelligence in gastrointestinal endoscopy: general overview. Chin Med J (Engl). 2020;133:326-334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 33]  [Cited by in RCA: 72]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
41.  Mitsala A, Tsalikidis C, Pitiakoudis M, Simopoulos C, Tsaroucha AK. Artificial Intelligence in Colorectal Cancer Screening, Diagnosis and Treatment. A New Era. Curr Oncol. 2021;28:1581-1607.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 25]  [Cited by in RCA: 142]  [Article Influence: 35.5]  [Reference Citation Analysis (0)]
42.  Deo RC. Machine Learning in Medicine. Circulation. 2015;132:1920-1930.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1155]  [Cited by in RCA: 2031]  [Article Influence: 225.7]  [Reference Citation Analysis (6)]
43.  Ruffle JK, Farmer AD, Aziz Q. Artificial Intelligence-Assisted Gastroenterology- Promises and Pitfalls. Am J Gastroenterol. 2019;114:422-428.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 71]  [Cited by in RCA: 98]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
44.  Goyal H, Mann R, Gandhi Z, Perisetti A, Ali A, Aman Ali K, Sharma N, Saligram S, Tharian B, Inamdar S. Scope of Artificial Intelligence in Screening and Diagnosis of Colorectal Cancer. J Clin Med. 2020;9:3313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 41]  [Cited by in RCA: 45]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
45.  Pohl H, Anderson JC, Aguilera-Fish A, Calderwood AH, Mackenzie TA, Robertson DJ. Recurrence of Colorectal Neoplastic Polyps After Incomplete Resection. Ann Intern Med. 2021;174:1377-1384.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 31]  [Cited by in RCA: 38]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
46.  Halloran SP. Intelligent Use of the Fecal Immunochemical Test in Population-Based Screening. Ann Intern Med. 2018;169:496-497.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
47.  US Preventive Services Task Force, Davidson KW, Barry MJ, Mangione CM, Cabana M, Caughey AB, Davis EM, Donahue KE, Doubeni CA, Krist AH, Kubik M, Li L, Ogedegbe G, Owens DK, Pbert L, Silverstein M, Stevermer J, Tseng CW, Wong JB. Screening for Colorectal Cancer: US Preventive Services Task Force Recommendation Statement. JAMA. 2021;325:1965-1977.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 402]  [Cited by in RCA: 1223]  [Article Influence: 305.8]  [Reference Citation Analysis (0)]
48.  Bresalier RS, Senore C, Young GP, Allison J, Benamouzig R, Benton S, Bossuyt PMM, Caro L, Carvalho B, Chiu HM, Coupé VMH, de Klaver W, de Klerk CM, Dekker E, Dolwani S, Fraser CG, Grady W, Guittet L, Gupta S, Halloran SP, Haug U, Hoff G, Itzkowitz S, Kortlever T, Koulaouzidis A, Ladabaum U, Lauby-Secretan B, Leja M, Levin B, Levin TR, Macrae F, Meijer GA, Melson J, O'Morain C, Parry S, Rabeneck L, Ransohoff DF, Sáenz R, Saito H, Sanduleanu-Dascalescu S, Schoen RE, Selby K, Singh H, Steele RJC, Sung JJY, Symonds EL, Winawer SJ; Members of the World Endoscopy Colorectal Cancer Screening New Test Evaluation Expert Working Group. An efficient strategy for evaluating new non-invasive screening tests for colorectal cancer: the guiding principles. Gut. 2023;72:1904-1918.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 36]  [Cited by in RCA: 30]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
49.  Robertson DJ, Dominitz JA, Beed A, Boardman KD, Del Curto BJ, Guarino PD, Imperiale TF, LaCasse A, Larson MF, Gupta S, Lieberman D, Planeta B, Shaukat A, Sultan S, Menees SB, Saini SD, Schoenfeld P, Goebel S, von Rosenvinge EC, Baffy G, Halasz I, Pedrosa MC, Kahng LS, Cassim R, Greer KB, Kinnard MF, Bhatt DB, Dunbar KB, Harford WV Jr, Mengshol JA, Olson JE, Patel SG, Antaki F, Fisher DA, Sullivan BA, Lenza C, Prajapati DN, Wong H, Beyth R, Lieb JG 2nd, Manlolo J, Ona FV, Cole RA, Khalaf N, Kahi CJ, Kohli DR, Rai T, Sharma P, Anastasiou J, Hagedorn C, Fernando RS, Jackson CS, Jamal MM, Lee RH, Merchant F, May FP, Pisegna JR, Omer E, Parajuli D, Said A, Nguyen TD, Tombazzi CR, Feldman PA, Jacob L, Koppelman RN, Lehenbauer KP, Desai DS, Madhoun MF, Tierney WM, Ho MQ, Hockman HJ, Lopez C, Carter Paulson E, Tobi M, Pinillos HL, Young M, Ho NC, Mascarenhas R, Promrat K, Mutha PR, Pandak WM Jr, Shah T, Schubert M, Pancotto FS, Gawron AJ, Underwood AE, Ho SB, Magno-Pagatzaurtundua P, Toro DH, Beymer CH, Kaz AM, Elwing J, Gill JA, Goldsmith SF, Yao MD, Protiva P, Pohl H, Kyriakides T; CONFIRM Study Group. Baseline Features and Reasons for Nonparticipation in the Colonoscopy Versus Fecal Immunochemical Test in Reducing Mortality From Colorectal Cancer (CONFIRM) Study, a Colorectal Cancer Screening Trial. JAMA Netw Open. 2023;6:e2321730.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 18]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
50.  Lauby-Secretan B, Vilahur N, Bianchini F, Guha N, Straif K; International Agency for Research on Cancer Handbook Working Group. The IARC Perspective on Colorectal Cancer Screening. N Engl J Med. 2018;378:1734-1740.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 224]  [Cited by in RCA: 242]  [Article Influence: 34.6]  [Reference Citation Analysis (0)]
51.  European Colorectal Cancer Screening Guidelines Working Group; von Karsa L, Patnick J, Segnan N, Atkin W, Halloran S, Lansdorp-Vogelaar I, Malila N, Minozzi S, Moss S, Quirke P, Steele RJ, Vieth M, Aabakken L, Altenhofen L, Ancelle-Park R, Antoljak N, Anttila A, Armaroli P, Arrossi S, Austoker J, Banzi R, Bellisario C, Blom J, Brenner H, Bretthauer M, Camargo Cancela M, Costamagna G, Cuzick J, Dai M, Daniel J, Dekker E, Delicata N, Ducarroz S, Erfkamp H, Espinàs JA, Faivre J, Faulds Wood L, Flugelman A, Frkovic-Grazio S, Geller B, Giordano L, Grazzini G, Green J, Hamashima C, Herrmann C, Hewitson P, Hoff G, Holten I, Jover R, Kaminski MF, Kuipers EJ, Kurtinaitis J, Lambert R, Launoy G, Lee W, Leicester R, Leja M, Lieberman D, Lignini T, Lucas E, Lynge E, Mádai S, Marinho J, Maučec Zakotnik J, Minoli G, Monk C, Morais A, Muwonge R, Nadel M, Neamtiu L, Peris Tuser M, Pignone M, Pox C, Primic-Zakelj M, Psaila J, Rabeneck L, Ransohoff D, Rasmussen M, Regula J, Ren J, Rennert G, Rey J, Riddell RH, Risio M, Rodrigues V, Saito H, Sauvaget C, Scharpantgen A, Schmiegel W, Senore C, Siddiqi M, Sighoko D, Smith R, Smith S, Suchanek S, Suonio E, Tong W, Törnberg S, Van Cutsem E, Vignatelli L, Villain P, Voti L, Watanabe H, Watson J, Winawer S, Young G, Zaksas V, Zappa M, Valori R. European guidelines for quality assurance in colorectal cancer screening and diagnosis: overview and introduction to the full supplement publication. Endoscopy. 2013;45:51-59.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 80]  [Cited by in RCA: 206]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
52.  Meklin J, Syrjänen K, Eskelinen M. Colorectal Cancer Screening With Traditional and New-generation Fecal Immunochemical Tests: A Critical Review of Fecal Occult Blood Tests. Anticancer Res. 2020;40:575-581.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
53.  Meklin J, SyrjÄnen K, Eskelinen M. Fecal Occult Blood Tests in Colorectal Cancer Screening: Systematic Review and Meta-analysis of Traditional and New-generation Fecal Immunochemical Tests. Anticancer Res. 2020;40:3591-3604.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 40]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
54.  Syrjänen K, Eskelinen M, Meklin J, Hendolin P, Eskelinen M, Suovaniemi O. Colorectal Cancer Screening by Fecal Immunochemical Tests (FIT): Considerations on Sampling and Markers (Hb and Hb/Hp Complex) of Fecal Occult Blood (FOB). Anticancer Res. 2024;44:1513-1523.  [PubMed]  [DOI]  [Full Text]
55.  Eskelinen M, Selander T, Guimarães DP, Kaarniranta K, Syrjänen K, Eskelinen M. Artificial Intelligence Models Could Enhance the Diagnostic Accuracy (DA) of Fecal Immunochemical Test (FIT) in the Detection of Colorectal Adenoma in a Screening Setting. Anticancer Res. 2025;45:267-275.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
56.  Clark JC, Collan Y, Eide TJ, Estève J, Ewen S, Gibbs NM, Jensen OM, Koskela E, MacLennan R, Simpson JG. Prevalence of polyps in an autopsy series from areas with varying incidence of large-bowel cancer. Int J Cancer. 1985;36:179-186.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 166]  [Cited by in RCA: 157]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
57.  Kastenberg D, Bertiger G, Brogadir S. Bowel preparation quality scales for colonoscopy. World J Gastroenterol. 2018;24:2833-2843.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 104]  [Cited by in RCA: 171]  [Article Influence: 24.4]  [Reference Citation Analysis (10)]
58.  Chen G, Zhao Y, Xie F, Shi W, Yang Y, Yang A, Wu D. Educating Outpatients for Bowel Preparation Before Colonoscopy Using Conventional Methods vs Virtual Reality Videos Plus Conventional Methods: A Randomized Clinical Trial. JAMA Netw Open. 2021;4:e2135576.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 8]  [Cited by in RCA: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
59.  Donovan K, Manem N, Miller D, Yodice M, Kabbach G, Feustel P, Tadros M. The Impact of Patient Education Level on Split-Dose Colonoscopy Bowel Preparation for CRC Prevention. J Cancer Educ. 2022;37:1083-1088.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 9]  [Cited by in RCA: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
60.  Kröner PT, Engels MM, Glicksberg BS, Johnson KW, Mzaik O, van Hooft JE, Wallace MB, El-Serag HB, Krittanawong C. Artificial intelligence in gastroenterology: A state-of-the-art review. World J Gastroenterol. 2021;27:6794-6824.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 28]  [Cited by in RCA: 94]  [Article Influence: 23.5]  [Reference Citation Analysis (7)]
61.  Kerbage A, Kassab J, El Dahdah J, Burke CA, Achkar JP, Rouphael C. Accuracy of ChatGPT in Common Gastrointestinal Diseases: Impact for Patients and Providers. Clin Gastroenterol Hepatol. 2024;22:1323-1325.e3.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 32]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
62.  Wilkoff MH, Piniella NR, Advani R. Can Artificial Intelligence Create an Accurate Colonoscopy Bowel Preparation Prompt? Gastro Hep Adv. 2025;4:100566.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
63.  Lim DYZ, Tan YB, Koh JTE, Tung JYM, Sng GGR, Tan DMY, Tan CK. ChatGPT on guidelines: Providing contextual knowledge to GPT allows it to provide advice on appropriate colonoscopy intervals. J Gastroenterol Hepatol. 2024;39:81-106.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 32]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
64.  Gorelik Y, Ghersin I, Maza I, Klein A. Harnessing language models for streamlined postcolonoscopy patient management: a novel approach. Gastrointest Endosc. 2023;98:639-641.e4.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 28]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
65.  Gilson A, Safranek CW, Huang T, Socrates V, Chi L, Taylor RA, Chartash D. How Does ChatGPT Perform on the United States Medical Licensing Examination (USMLE)? The Implications of Large Language Models for Medical Education and Knowledge Assessment. JMIR Med Educ. 2023;9:e45312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 518]  [Cited by in RCA: 874]  [Article Influence: 437.0]  [Reference Citation Analysis (0)]
66.  Russell L, Mathura P, Lee A, Dhaliwal R, Kassam N, Kohansal A. Patient-centered approaches to targeting incomplete bowel preparations for inpatient colonoscopies. Ann Gastroenterol. 2021;34:547-551.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
67.  Keswani RN, Crockett SD, Calderwood AH. AGA Clinical Practice Update on Strategies to Improve Quality of Screening and Surveillance Colonoscopy: Expert Review. Gastroenterology. 2021;161:701-711.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 25]  [Cited by in RCA: 104]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
68.  Zhao S, Wang S, Pan P, Xia T, Chang X, Yang X, Guo L, Meng Q, Yang F, Qian W, Xu Z, Wang Y, Wang Z, Gu L, Wang R, Jia F, Yao J, Li Z, Bai Y. Magnitude, Risk Factors, and Factors Associated With Adenoma Miss Rate of Tandem Colonoscopy: A Systematic Review and Meta-analysis. Gastroenterology. 2019;156:1661-1674.e11.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 205]  [Cited by in RCA: 398]  [Article Influence: 66.3]  [Reference Citation Analysis (0)]
69.  Mori Y, Neumann H, Misawa M, Kudo SE, Bretthauer M. Artificial intelligence in colonoscopy - Now on the market. What's next? J Gastroenterol Hepatol. 2021;36:7-11.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 28]  [Cited by in RCA: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
70.  Bi WL, Hosny A, Schabath MB, Giger ML, Birkbak NJ, Mehrtash A, Allison T, Arnaout O, Abbosh C, Dunn IF, Mak RH, Tamimi RM, Tempany CM, Swanton C, Hoffmann U, Schwartz LH, Gillies RJ, Huang RY, Aerts HJWL. Artificial intelligence in cancer imaging: Clinical challenges and applications. CA Cancer J Clin. 2019;69:127-157.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 848]  [Cited by in RCA: 864]  [Article Influence: 144.0]  [Reference Citation Analysis (3)]
71.  Milea D, Najjar RP, Zhubo J, Ting D, Vasseneix C, Xu X, Aghsaei Fard M, Fonseca P, Vanikieti K, Lagrèze WA, La Morgia C, Cheung CY, Hamann S, Chiquet C, Sanda N, Yang H, Mejico LJ, Rougier M-B, Kho R, Thi Ha Chau T, Singhal S, Gohier P, Clermont-Vignal C, Cheng C-Y, Jonas JB, Yu-Wai-Man P, Fraser CL, Chen JJ, Ambika S, Miller NR, Liu Y, Newman NJ, Wong TY, Biousse V; BONSAI Group. Artificial Intelligence to Detect Papilledema from Ocular Fundus Photographs. N Engl J Med. 2020;382:1687-1695.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 155]  [Cited by in RCA: 233]  [Article Influence: 46.6]  [Reference Citation Analysis (0)]
72.  Li X, Zhang S, Zhang Q, Wei X, Pan Y, Zhao J, Xin X, Qin C, Wang X, Li J, Yang F, Zhao Y, Yang M, Wang Q, Zheng Z, Zheng X, Yang X, Whitlow CT, Gurcan MN, Zhang L, Wang X, Pasche BC, Gao M, Zhang W, Chen K. Diagnosis of thyroid cancer using deep convolutional neural network models applied to sonographic images: a retrospective, multicohort, diagnostic study. Lancet Oncol. 2019;20:193-201.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 163]  [Cited by in RCA: 273]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
73.  Esteva A, Kuprel B, Novoa RA, Ko J, Swetter SM, Blau HM, Thrun S. Dermatologist-level classification of skin cancer with deep neural networks. Nature. 2017;542:115-118.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5683]  [Cited by in RCA: 5500]  [Article Influence: 687.5]  [Reference Citation Analysis (0)]
74.  Liang H, Tsui BY, Ni H, Valentim CCS, Baxter SL, Liu G, Cai W, Kermany DS, Sun X, Chen J, He L, Zhu J, Tian P, Shao H, Zheng L, Hou R, Hewett S, Li G, Liang P, Zang X, Zhang Z, Pan L, Cai H, Ling R, Li S, Cui Y, Tang S, Ye H, Huang X, He W, Liang W, Zhang Q, Jiang J, Yu W, Gao J, Ou W, Deng Y, Hou Q, Wang B, Yao C, Liang Y, Zhang S, Duan Y, Zhang R, Gibson S, Zhang CL, Li O, Zhang ED, Karin G, Nguyen N, Wu X, Wen C, Xu J, Xu W, Wang B, Wang W, Li J, Pizzato B, Bao C, Xiang D, He W, He S, Zhou Y, Haw W, Goldbaum M, Tremoulet A, Hsu CN, Carter H, Zhu L, Zhang K, Xia H. Evaluation and accurate diagnoses of pediatric diseases using artificial intelligence. Nat Med. 2019;25:433-438.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 257]  [Cited by in RCA: 322]  [Article Influence: 53.7]  [Reference Citation Analysis (0)]
75.  Kim YJ, Bae JP, Chung JW, Park DK, Kim KG, Kim YJ. New polyp image classification technique using transfer learning of network-in-network structure in endoscopic images. Sci Rep. 2021;11:3605.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
76.  Calderaro J, Kather JN. Artificial intelligence-based pathology for gastrointestinal and hepatobiliary cancers. Gut. 2021;70:1183-1193.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 40]  [Cited by in RCA: 71]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
77.  Le Berre C, Sandborn WJ, Aridhi S, Devignes MD, Fournier L, Smaïl-Tabbone M, Danese S, Peyrin-Biroulet L. Application of Artificial Intelligence to Gastroenterology and Hepatology. Gastroenterology. 2020;158:76-94.e2.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 230]  [Cited by in RCA: 339]  [Article Influence: 67.8]  [Reference Citation Analysis (1)]
78.  Lee J, Lee H, Chung JW. The Role of Artificial Intelligence in Gastric Cancer: Surgical and Therapeutic Perspectives: A Comprehensive Review. J Gastric Cancer. 2023;23:375-387.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
79.  Yao X, Saha A, Saravanan S, Low A, Sussman J. A Study Protocol for a Comprehensive Evaluation of Two Artificial Intelligence-Based Tools in Title and Abstract Screening for the Development of Evidence-Based Cancer Guidelines. Cancer Innov. 2025;4:e70021.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
80.  Sun K, Wang Y, Qu R, Yang Q, Luo R, Jiang Z, Wang H, Fu W. Comprehensive application of artificial intelligence in colorectal cancer: A review. iScience. 2025;28:112980.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
81.  Lee H, Chung JW, Kim KO, Kwon KA, Kim JH, Yun SC, Jung SW, Sheeraz A, Yoon YJ, Kim JH, Kayasseh MA. Validation of Artificial Intelligence Computer-Aided Detection of Colonic Neoplasm in Colonoscopy. Diagnostics (Basel). 2024;14:2762.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
82.  Ashat M, Klair JS, Singh D, Murali AR, Krishnamoorthi R. Impact of real-time use of artificial intelligence in improving adenoma detection during colonoscopy: A systematic review and meta-analysis. Endosc Int Open. 2021;9:E513-E521.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 15]  [Cited by in RCA: 34]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
83.  Barua I, Vinsard DG, Jodal HC, Løberg M, Kalager M, Holme Ø, Misawa M, Bretthauer M, Mori Y. Artificial intelligence for polyp detection during colonoscopy: a systematic review and meta-analysis. Endoscopy. 2021;53:277-284.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 86]  [Cited by in RCA: 166]  [Article Influence: 41.5]  [Reference Citation Analysis (1)]
84.  Gadi SRV, Mori Y, Misawa M, East JE, Hassan C, Repici A, Byrne MF, von Renteln D, Hewett DG, Wang P, Saito Y, Matsubayashi CO, Ahmad OF, Sharma P, Gross SA, Sengupta N, Mansour N, Cherubini A, Dinh NN, Xiao X, Mountney P, González-Bueno Puyal J, Little G, LaRocco S, Conjeti S, Seibt H, Zur D, Shimada H, Berzin TM, Glissen Brown JR. Creating a standardized tool for the evaluation and comparison of artificial intelligence-based computer-aided detection programs in colonoscopy: a modified Delphi approach. Gastrointest Endosc. 2025;102:109-116.e2.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
85.  Rondonotti E, Di Paolo D, Rizzotto ER, Alvisi C, Buscarini E, Spadaccini M, Tamanini G, Paggi S, Amato A, Scardino G, Romeo S, Alicante S, Ancona F, Guido E, Marzo V, Chicco F, Agazzi S, Rosa C, Correale L, Repici A, Hassan C, Radaelli F; AIFIT Study Group. Efficacy of a computer-aided detection system in a fecal immunochemical test-based organized colorectal cancer screening program: a randomized controlled trial (AIFIT study). Endoscopy. 2022;54:1171-1179.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 49]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
86.  Wang P, Berzin TM, Glissen Brown JR, Bharadwaj S, Becq A, Xiao X, Liu P, Li L, Song Y, Zhang D, Li Y, Xu G, Tu M, Liu X. Real-time automatic detection system increases colonoscopic polyp and adenoma detection rates: a prospective randomised controlled study. Gut. 2019;68:1813-1819.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 398]  [Cited by in RCA: 559]  [Article Influence: 93.2]  [Reference Citation Analysis (0)]
87.  Su JR, Li Z, Shao XJ, Ji CR, Ji R, Zhou RC, Li GC, Liu GQ, He YS, Zuo XL, Li YQ. Impact of a real-time automatic quality control system on colorectal polyp and adenoma detection: a prospective randomized controlled study (with videos). Gastrointest Endosc. 2020;91:415-424.e4.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 153]  [Cited by in RCA: 215]  [Article Influence: 43.0]  [Reference Citation Analysis (0)]
88.  Liu WN, Zhang YY, Bian XQ, Wang LJ, Yang Q, Zhang XD, Huang J. Study on detection rate of polyps and adenomas in artificial-intelligence-aided colonoscopy. Saudi J Gastroenterol. 2020;26:13-19.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 82]  [Cited by in RCA: 130]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
89.  Gong D, Wu L, Zhang J, Mu G, Shen L, Liu J, Wang Z, Zhou W, An P, Huang X, Jiang X, Li Y, Wan X, Hu S, Chen Y, Hu X, Xu Y, Zhu X, Li S, Yao L, He X, Chen D, Huang L, Wei X, Wang X, Yu H. Detection of colorectal adenomas with a real-time computer-aided system (ENDOANGEL): a randomised controlled study. Lancet Gastroenterol Hepatol. 2020;5:352-361.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 298]  [Cited by in RCA: 273]  [Article Influence: 54.6]  [Reference Citation Analysis (0)]
90.  Repici A, Badalamenti M, Maselli R, Correale L, Radaelli F, Rondonotti E, Ferrara E, Spadaccini M, Alkandari A, Fugazza A, Anderloni A, Galtieri PA, Pellegatta G, Carrara S, Di Leo M, Craviotto V, Lamonaca L, Lorenzetti R, Andrealli A, Antonelli G, Wallace M, Sharma P, Rosch T, Hassan C. Efficacy of Real-Time Computer-Aided Detection of Colorectal Neoplasia in a Randomized Trial. Gastroenterology. 2020;159:512-520.e7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 237]  [Cited by in RCA: 411]  [Article Influence: 82.2]  [Reference Citation Analysis (0)]
91.  Wang P, Liu P, Glissen Brown JR, Berzin TM, Zhou G, Lei S, Liu X, Li L, Xiao X. Lower Adenoma Miss Rate of Computer-Aided Detection-Assisted Colonoscopy vs Routine White-Light Colonoscopy in a Prospective Tandem Study. Gastroenterology. 2020;159:1252-1261.e5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 81]  [Cited by in RCA: 153]  [Article Influence: 30.6]  [Reference Citation Analysis (0)]
92.  Wang P, Liu X, Berzin TM, Glissen Brown JR, Liu P, Zhou C, Lei L, Li L, Guo Z, Lei S, Xiong F, Wang H, Song Y, Pan Y, Zhou G. Effect of a deep-learning computer-aided detection system on adenoma detection during colonoscopy (CADe-DB trial): a double-blind randomised study. Lancet Gastroenterol Hepatol. 2020;5:343-351.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 164]  [Cited by in RCA: 305]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
93.  Kamba S, Tamai N, Saitoh I, Matsui H, Horiuchi H, Kobayashi M, Sakamoto T, Ego M, Fukuda A, Tonouchi A, Shimahara Y, Nishikawa M, Nishino H, Saito Y, Sumiyama K. Reducing adenoma miss rate of colonoscopy assisted by artificial intelligence: a multicenter randomized controlled trial. J Gastroenterol. 2021;56:746-757.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 21]  [Cited by in RCA: 81]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
94.  Xu L, He X, Zhou J, Zhang J, Mao X, Ye G, Chen Q, Xu F, Sang J, Wang J, Ding Y, Li Y, Yu C. Artificial intelligence-assisted colonoscopy: A prospective, multicenter, randomized controlled trial of polyp detection. Cancer Med. 2021;10:7184-7193.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 33]  [Cited by in RCA: 34]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
95.  Luo Y, Zhang Y, Liu M, Lai Y, Liu P, Wang Z, Xing T, Huang Y, Li Y, Li A, Wang Y, Luo X, Liu S, Han Z. Artificial Intelligence-Assisted Colonoscopy for Detection of Colon Polyps: a Prospective, Randomized Cohort Study. J Gastrointest Surg. 2021;25:2011-2018.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 31]  [Cited by in RCA: 66]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
96.  Shaukat A, Lichtenstein DR, Somers SC, Chung DC, Perdue DG, Gopal M, Colucci DR, Phillips SA, Marka NA, Church TR, Brugge WR; SKOUT™ Registration Study Team. Computer-Aided Detection Improves Adenomas per Colonoscopy for Screening and Surveillance Colonoscopy: A Randomized Trial. Gastroenterology. 2022;163:732-741.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 84]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
97.  Wallace MB, Sharma P, Bhandari P, East J, Antonelli G, Lorenzetti R, Vieth M, Speranza I, Spadaccini M, Desai M, Lukens FJ, Babameto G, Batista D, Singh D, Palmer W, Ramirez F, Palmer R, Lunsford T, Ruff K, Bird-Liebermann E, Ciofoaia V, Arndtz S, Cangemi D, Puddick K, Derfus G, Johal AS, Barawi M, Longo L, Moro L, Repici A, Hassan C. Impact of Artificial Intelligence on Miss Rate of Colorectal Neoplasia. Gastroenterology. 2022;163:295-304.e5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 195]  [Cited by in RCA: 149]  [Article Influence: 49.7]  [Reference Citation Analysis (1)]
98.  Glissen Brown JR, Mansour NM, Wang P, Chuchuca MA, Minchenberg SB, Chandnani M, Liu L, Gross SA, Sengupta N, Berzin TM. Deep Learning Computer-aided Polyp Detection Reduces Adenoma Miss Rate: A United States Multi-center Randomized Tandem Colonoscopy Study (CADeT-CS Trial). Clin Gastroenterol Hepatol. 2022;20:1499-1507.e4.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 27]  [Cited by in RCA: 129]  [Article Influence: 43.0]  [Reference Citation Analysis (0)]
99.  Lui TKL, Hang DV, Tsao SKK, Hui CKY, Mak LLY, Ko MKL, Cheung KS, Thian MY, Liang R, Tsui VWM, Yeung CK, Dao LV, Leung WK. Computer-assisted detection versus conventional colonoscopy for proximal colonic lesions: a multicenter, randomized, tandem-colonoscopy study. Gastrointest Endosc. 2023;97:325-334.e1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
100.  Ahmad A, Wilson A, Haycock A, Humphries A, Monahan K, Suzuki N, Thomas-Gibson S, Vance M, Bassett P, Thiruvilangam K, Dhillon A, Saunders BP. Evaluation of a real-time computer-aided polyp detection system during screening colonoscopy: AI-DETECT study. Endoscopy. 2023;55:313-319.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 44]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
101.  Mangas-Sanjuan C, de-Castro L, Cubiella J, Díez-Redondo P, Suárez A, Pellisé M, Fernández N, Zarraquiños S, Núñez-Rodríguez H, Álvarez-García V, Ortiz O, Sala-Miquel N, Zapater P, Jover R; CADILLAC study investigators. Role of Artificial Intelligence in Colonoscopy Detection of Advanced Neoplasias : A Randomized Trial. Ann Intern Med. 2023;176:1145-1152.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 44]  [Article Influence: 22.0]  [Reference Citation Analysis (3)]
102.  Karsenti D, Tharsis G, Perrot B, Cattan P, Percie du Sert A, Venezia F, Zrihen E, Gillet A, Lab JP, Tordjman G, Cavicchi M. Effect of real-time computer-aided detection of colorectal adenoma in routine colonoscopy (COLO-GENIUS): a single-centre randomised controlled trial. Lancet Gastroenterol Hepatol. 2023;8:726-734.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 31]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
103.  Wei MT, Shankar U, Parvin R, Abbas SH, Chaudhary S, Friedlander Y, Friedland S. Evaluation of Computer-Aided Detection During Colonoscopy in the Community (AI-SEE): A Multicenter Randomized Clinical Trial. Am J Gastroenterol. 2023;118:1841-1847.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 47]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
104.  Nakashima H, Kitazawa N, Fukuyama C, Kawachi H, Kawahira H, Momma K, Sakaki N. Clinical Evaluation of Computer-Aided Colorectal Neoplasia Detection Using a Novel Endoscopic Artificial Intelligence: A Single-Center Randomized Controlled Trial. Digestion. 2023;104:193-201.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 25]  [Reference Citation Analysis (0)]
105.  Gimeno-García AZ, Hernández Negrin D, Hernández A, Nicolás-Pérez D, Rodríguez E, Montesdeoca C, Alarcon O, Romero R, Baute Dorta JL, Cedrés Y, Castillo RD, Jiménez A, Felipe V, Morales D, Ortega J, Reygosa C, Quintero E, Hernández-Guerra M. Usefulness of a novel computer-aided detection system for colorectal neoplasia: a randomized controlled trial. Gastrointest Endosc. 2023;97:528-536.e1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 40]  [Reference Citation Analysis (0)]
106.  Yao L, Li X, Wu Z, Wang J, Luo C, Chen B, Luo R, Zhang L, Zhang C, Tan X, Lu Z, Zhu C, Huang Y, Tan T, Liu Z, Li Y, Li S, Yu H. Effect of artificial intelligence on novice-performed colonoscopy: a multicenter randomized controlled tandem study. Gastrointest Endosc. 2024;99:91-99.e9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 20]  [Cited by in RCA: 27]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
107.  Schöler J, Alavanja M, de Lange T, Yamamoto S, Hedenström P, Varkey J. Impact of AI-aided colonoscopy in clinical practice: a prospective randomised controlled trial. BMJ Open Gastroenterol. 2024;11:e001247.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 6]  [Cited by in RCA: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
108.  Yamaguchi D, Shimoda R, Miyahara K, Yukimoto T, Sakata Y, Takamori A, Mizuta Y, Fujimura Y, Inoue S, Tomonaga M, Ogino Y, Eguchi K, Ikeda K, Tanaka Y, Takedomi H, Hidaka H, Akutagawa T, Tsuruoka N, Noda T, Tsunada S, Esaki M. Impact of an artificial intelligence-aided endoscopic diagnosis system on improving endoscopy quality for trainees in colonoscopy: Prospective, randomized, multicenter study. Dig Endosc. 2024;36:40-48.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 33]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
109.  Dumachi AI, Buiu C. Applications of Machine Learning in Cancer Imaging: A Review of Diagnostic Methods for Six Major Cancer Types. Electronics. 2024;13:4697.  [PubMed]  [DOI]  [Full Text]
110.  Ovi TB, Bashree N, Nyeem H, Wahed MA. FocusU(2)Net: Pioneering dual attention with gated U-Net for colonoscopic polyp segmentation. Comput Biol Med. 2025;186:109617.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
111.  Oğuz FE, Alkan A. AI-Enhanced Interface for Colonic Polyp Segmentation Using DeepLabv3+ with Comparative Backbone Analysis. Biomed Phys Eng Express.  2024.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
112.  Ashwini A, Purushothaman K, Rosi A, Vaishnavi T. Artificial Intelligence based real-time automatic detection and classification of skin lesion in dermoscopic samples using DenseNet-169 architecture. J Intell Fuzzy Syst. 2023;45:6943-6958.  [PubMed]  [DOI]  [Full Text]
113.  Hsu WF, Chang WY, Kuo CY, Chang LC, Lin HH, Wu MS, Chiu HM. Effect of a novel artificial intelligence-based cecum recognition system on adenoma detection metrics in a screening colonoscopy setting. Gastrointest Endosc. 2025;101:452-455.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
114.  Baxter NN, Sutradhar R, Forbes SS, Paszat LF, Saskin R, Rabeneck L. Analysis of administrative data finds endoscopist quality measures associated with postcolonoscopy colorectal cancer. Gastroenterology. 2011;140:65-72.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 365]  [Cited by in RCA: 405]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
115.  Lee TJ, Rutter MD, Blanks RG, Moss SM, Goddard AF, Chilton A, Nickerson C, McNally RJ, Patnick J, Rees CJ. Colonoscopy quality measures: experience from the NHS Bowel Cancer Screening Programme. Gut. 2012;61:1050-1057.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 209]  [Cited by in RCA: 255]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
116.  Orzeszko Z, Gach T, Bogacki P, Markowska B, Solecki R, Szura M. Effect of artificial intelligence implementation to the latest generation 4K colonoscopy. Pol Przegl Chir. 2024;96:24-30.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
117.  Vázquez D, Bernal J, Sánchez FJ, Fernández-Esparrach G, López AM, Romero A, Drozdzal M, Courville A. A Benchmark for Endoluminal Scene Segmentation of Colonoscopy Images. J Healthc Eng. 2017;2017:4037190.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 302]  [Cited by in RCA: 188]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
118.  Venkatayogi N, Kara OC; Bonyun J, Ikoma N, Alambeigi F.   Classification of Colorectal Cancer Polyps via Transfer Learning and Vision-Based Tactile Sensing. Available from: arXiv:2211.04573.  [PubMed]  [DOI]  [Full Text]
119.  Fang Y, Zhu D, Yao J, Yuan Y, Tong K. ABC-Net: Area-Boundary Constraint Network With Dynamical Feature Selection for Colorectal Polyp Segmentation. IEEE Sens J. 2021;21:11799-11809.  [PubMed]  [DOI]  [Full Text]
120.  Jha D, Smedsrud PH, Riegler MA, Halvorsen P, Lange TD, Johansen D, Johansen HD.   Kvasir-SEG: A Segmented Polyp Dataset. Available from: arXiv:1911.07069.  [PubMed]  [DOI]  [Full Text]
121.  Silva J, Histace A, Romain O, Dray X, Granado B. Toward embedded detection of polyps in WCE images for early diagnosis of colorectal cancer. Int J Comput Assist Radiol Surg. 2014;9:283-293.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 234]  [Cited by in RCA: 289]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
122.  Elkarazle K, Raman V, Then P, Chua C. Improved Colorectal Polyp Segmentation Using Enhanced MA-NET and Modified Mix-ViT Transformer. IEEE Access. 2023;11:69295-69309.  [PubMed]  [DOI]  [Full Text]
123.  Bernal J, Sánchez FJ, Fernández-Esparrach G, Gil D, Rodríguez C, Vilariño F. WM-DOVA maps for accurate polyp highlighting in colonoscopy: Validation vs. saliency maps from physicians. Comput Med Imaging Graph. 2015;43:99-111.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 336]  [Cited by in RCA: 474]  [Article Influence: 47.4]  [Reference Citation Analysis (0)]
124.  Bernal J, Sánchez J, Vilariño F. Towards automatic polyp detection with a polyp appearance model. Pattern Recogn. 2012;45:3166-3182.  [PubMed]  [DOI]  [Full Text]
125.  Eliakim R, Yassin K, Niv Y, Metzger Y, Lachter J, Gal E, Sapoznikov B, Konikoff F, Leichtmann G, Fireman Z, Kopelman Y, Adler SN. Prospective multicenter performance evaluation of the second-generation colon capsule compared with colonoscopy. Endoscopy. 2009;41:1026-1031.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 204]  [Cited by in RCA: 212]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
126.  Igawa A, Oka S, Tanaka S, Otani I, Kunihara S, Chayama K. Evaluation for the Clinical Efficacy of Colon Capsule Endoscopy in the Detection of Laterally Spreading Tumors. Digestion. 2017;95:43-48.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
127.  Eliakim R, Fireman Z, Gralnek IM, Yassin K, Waterman M, Kopelman Y, Lachter J, Koslowsky B, Adler SN. Evaluation of the PillCam Colon capsule in the detection of colonic pathology: results of the first multicenter, prospective, comparative study. Endoscopy. 2006;38:963-970.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 239]  [Cited by in RCA: 223]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
128.  Koulaouzidis A, Dabos K, Philipper M, Toth E, Keuchel M. How should we do colon capsule endoscopy reading: a practical guide. Ther Adv Gastrointest Endosc. 2021;14:26317745211001983.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 23]  [Cited by in RCA: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
129.  Spada C, Hassan C, Galmiche JP, Neuhaus H, Dumonceau JM, Adler S, Epstein O, Gay G, Pennazio M, Rex DK, Benamouzig R, de Franchis R, Delvaux M, Devière J, Eliakim R, Fraser C, Hagenmuller F, Herrerias JM, Keuchel M, Macrae F, Munoz-Navas M, Ponchon T, Quintero E, Riccioni ME, Rondonotti E, Marmo R, Sung JJ, Tajiri H, Toth E, Triantafyllou K, Van Gossum A, Costamagna G; European Society of Gastrointestinal Endoscopy. Colon capsule endoscopy: European Society of Gastrointestinal Endoscopy (ESGE) Guideline. Endoscopy. 2012;44:527-536.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 203]  [Cited by in RCA: 180]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
130.  Thorndal C, Selnes O, Lei II, Schostek S, Koulaouzidis A. Retention of endoscopic capsules in diverticula: Literature review of a capsule endoscopy rarity. Endosc Int Open. 2024;12:E788-E796.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
131.  Kindt IS, Martiny FHJ, Gram EG, Bie AKL, Jauernik CP, Rahbek OJ, Nielsen SB, Siersma V, Bang CW, Brodersen JB. The risk of bleeding and perforation from sigmoidoscopy or colonoscopy in colorectal cancer screening: A systematic review and meta-analyses. PLoS One. 2023;18:e0292797.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
132.  Lei II, Thorndal C, Manzoor MS, Parsons N, Noble C, Huhulea C, Koulaouzidis A, Arasaradnam RP. The Diagnostic Accuracy of Colon Capsule Endoscopy in Inflammatory Bowel Disease-A Systematic Review and Meta-Analysis. Diagnostics (Basel). 2024;14:2056.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
133.  van Riswijk MLM, van Keulen KE, Siersema PD. Efficacy of ultra-low volume (≤1 L) bowel preparation fluids: Systematic review and meta-analysis. Dig Endosc. 2022;34:13-32.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
134.  Tan X, Yang W, Wichmann D, Huang C, Mothes B, Grund KE, Chen Z, Chen Z. Magnetic endoscopic imaging as a rational investment for specific colonoscopies: a systematic review and meta-analysis. Expert Rev Gastroenterol Hepatol. 2021;15:447-458.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
135.  Lei II, Nia GJ, White E, Wenzek H, Segui S, Watson AJM, Koulaouzidis A, Arasaradnam RP. Clinicians' Guide to Artificial Intelligence in Colon Capsule Endoscopy-Technology Made Simple. Diagnostics (Basel). 2023;13:1038.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
136.  Barclay RL, Vicari JJ, Doughty AS, Johanson JF, Greenlaw RL. Colonoscopic withdrawal times and adenoma detection during screening colonoscopy. N Engl J Med. 2006;355:2533-2541.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 911]  [Cited by in RCA: 957]  [Article Influence: 50.4]  [Reference Citation Analysis (0)]
137.  Schelde-Olesen B, Bjørsum-Meyer T, Koulaouzidis A, Buijs MM, Herp J, Kaalby L, Baatrup G, Deding U. Interobserver agreement on landmark and flexure identification in colon capsule endoscopy. Tech Coloproctol. 2023;27:1219-1225.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
138.  Moug SJ, Fountas S, Johnstone MS, Bryce AS, Renwick A, Chisholm LJ, McCarthy K, Hung A, Diament RH, McGregor JR, Khine M, Saldanha JD, Khan K, Mackay G, Leitch EF, McKee RF, Anderson JH, Griffiths B, Horgan A, Lockwood S, Bisset C, Molloy R, Vella M. Analysis of lesion localisation at colonoscopy: outcomes from a multi-centre U.K. study. Surg Endosc. 2017;31:2959-2967.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
139.  Su J, Liu Z, Li H, Kang L, Huang K, Wu J, Huang H, Ling F, Yao X, Huang C. Artificial intelligence-based model to predict recurrence after local excision in T1 rectal cancer. Eur J Surg Oncol. 2025;51:109717.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
140.  Baek JE, Yi H, Hong SW, Song S, Lee JY, Hwang SW, Park SH, Yang DH, Ye BD, Myung SJ, Yang SK, Kim N, Byeon JS. Artificial Intelligence Models May Aid in Predicting Lymph Node Metastasis in Patients with T1 Colorectal Cancer. Gut Liver. 2025;19:69-76.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
141.  Liu Y, Wang J, Wu C, Liu L, Zhang Z, Yu H. Fovea-UNet: detection and segmentation of lymph node metastases in colorectal cancer with deep learning. Biomed Eng Online. 2023;22:74.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
142.  Faa G, Fraschini M, Didaci L, Saba L, Scartozzi M, Orvieto E, Rugge M. "Artificial histology" in colonic Neoplasia: A critical approach. Dig Liver Dis. 2025;57:663-668.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
143.  Zhou C, Jin Y, Chen Y, Huang S, Huang R, Wang Y, Zhao Y, Chen Y, Guo L, Liao J. Histopathology classification and localization of colorectal cancer using global labels by weakly supervised deep learning. Comput Med Imaging Graph. 2021;88:101861.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 11]  [Cited by in RCA: 41]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
144.  Tamang LD, Kim MT, Kim SJ, Kim BW.   Tumor-Stroma Classification in Colorectal Cancer Patients with Transfer Learning based Binary Classifier. Proceedings of the 2021 International Conference on Information and Communication Technology Convergence (ICTC); 2021 Oct 20-22; Jeju Island, Korea. IEEE, 2021: 1645-1648.  [PubMed]  [DOI]
145.  Kather JN, Weis CA, Bianconi F, Melchers SM, Schad LR, Gaiser T, Marx A, Zöllner FG. Multi-class texture analysis in colorectal cancer histology. Sci Rep. 2016;6:27988.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 227]  [Cited by in RCA: 191]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
146.  Alotaibi SR, Alohali MA, Maashi M, Alqahtani H, Alotaibi M, Mahmud A. Advances in colorectal cancer diagnosis using optimal deep feature fusion approach on biomedical images. Sci Rep. 2025;15:4200.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
147.  Guo J, Cao W, Nie B, Qin Q. Unsupervised Learning Composite Network to Reduce Training Cost of Deep Learning Model for Colorectal Cancer Diagnosis. IEEE J Transl Eng Health Med. 2023;11:54-59.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
148.  Sultan S, Shung DL, Kolb JM, Foroutan F, Hassan C, Kahi CJ, Liang PS, Levin TR, Siddique SM, Lebwohl B. AGA Living Clinical Practice Guideline on Computer-Aided Detection-Assisted Colonoscopy. Gastroenterology. 2025;168:691-700.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
149.  ASGE AI Task Force, Parasa S, Berzin T, Leggett C, Gross S, Repici A, Ahmad OF, Chiang A, Coelho-Prabhu N, Cohen J, Dekker E, Keswani RN, Kahn CE, Hassan C, Petrick N, Mountney P, Ng J, Riegler M, Mori Y, Saito Y, Thakkar S, Waxman I, Wallace MB, Sharma P. Consensus statements on the current landscape of artificial intelligence applications in endoscopy, addressing roadblocks, and advancing artificial intelligence in gastroenterology. Gastrointest Endosc. 2025;101:2-9.e1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 19]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
150.  Bretthauer M, Ahmed J, Antonelli G, Beaumont H, Beg S, Benson A, Bisschops R, De Cristofaro E, Gibbons E, Häfner M, Karsenti D, Laquière A, Loly JP, O'Reilly SM, Pellisé M, Grubelic Ravic K, Triantafyllou K, Tziatzios G, Valente R, Walter BM, Wiesand M, Lorenzo-Zúñiga V, Gralnek IM. Use of computer-assisted detection (CADe) colonoscopy in colorectal cancer screening and surveillance: European Society of Gastrointestinal Endoscopy (ESGE) Position Statement. Endoscopy. 2025;57:667-673.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 5]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
151.  Misawa M, Kudo SE, Mori Y. Implementation of Artificial Intelligence in Colonoscopy Practice in Japan. JMA J. 2025;8:60-63.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
152.  Tsuji Y, Fujishiro M. AI Era Is Coming: The Implementation of AI Medical Devices to Endoscopy. JMA J. 2025;8:64-65.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
153.  Aso MC, Sostres C, Lanas A. Artificial Intelligence in GI endoscopy: what to expect. Front Med (Lausanne). 2025;12:1588873.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
154.  Zeng A, Steinke J, Bocse HF, De Pastena M. Dr. LLM Will See You Now: The Ability of ChatGPT to Provide Geographically Tailored Colorectal Cancer Screening and Surveillance Recommendations. J Clin Med. 2025;14:5101.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
155.  Chen ZL, Wang C, Wang F. Revolutionizing gastroenterology and hepatology with artificial intelligence: From precision diagnosis to equitable healthcare through interdisciplinary practice. World J Gastroenterol. 2025;31:108021.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 3]  [Reference Citation Analysis (5)]
156.  Spada C, Hassan C, Bellini D, Burling D, Cappello G, Carretero C, Dekker E, Eliakim R, de Haan M, Kaminski MF, Koulaouzidis A, Laghi A, Lefere P, Mang T, Milluzzo SM, Morrin M, McNamara D, Neri E, Pecere S, Pioche M, Plumb A, Rondonotti E, Spaander MC, Taylor S, Fernandez-Urien I, van Hooft JE, Stoker J, Regge D. Imaging alternatives to colonoscopy: CT colonography and colon capsule. European Society of Gastrointestinal Endoscopy (ESGE) and European Society of Gastrointestinal and Abdominal Radiology (ESGAR) Guideline - Update 2020. Eur Radiol. 2021;31:2967-2982.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 49]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
157.  Yee J, Dachman A, Kim DH, Kobi M, Laghi A, McFarland E, Moreno C, Park SH, Pickhardt PJ, Plumb A, Pooler BD, Zalis M, Chang KJ. Erratum for: CT Colonography Reporting and Data System (C-RADS): Version 2023 Update. Radiology. 2024;310:e249004.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
158.  Rex DK, Boland CR, Dominitz JA, Giardiello FM, Johnson DA, Kaltenbach T, Levin TR, Lieberman D, Robertson DJ. Colorectal Cancer Screening: Recommendations for Physicians and Patients from the U.S. Multi-Society Task Force on Colorectal Cancer. Am J Gastroenterol. 2017;112:1016-1030.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 368]  [Cited by in RCA: 491]  [Article Influence: 61.4]  [Reference Citation Analysis (0)]
159.  Wolf AMD, Fontham ETH, Church TR, Flowers CR, Guerra CE, LaMonte SJ, Etzioni R, McKenna MT, Oeffinger KC, Shih YT, Walter LC, Andrews KS, Brawley OW, Brooks D, Fedewa SA, Manassaram-Baptiste D, Siegel RL, Wender RC, Smith RA. Colorectal cancer screening for average-risk adults: 2018 guideline update from the American Cancer Society. CA Cancer J Clin. 2018;68:250-281.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 945]  [Cited by in RCA: 1338]  [Article Influence: 191.1]  [Reference Citation Analysis (0)]
160.  Pickhardt PJ, Hassan C, Laghi A, Zullo A, Kim DH, Iafrate F, Morini S. Small and diminutive polyps detected at screening CT colonography: a decision analysis for referral to colonoscopy. AJR Am J Roentgenol. 2008;190:136-144.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 76]  [Cited by in RCA: 66]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
161.  Grosu S, Fabritius MP, Winkelmann M, Puhr-Westerheide D, Ingenerf M, Maurus S, Graser A, Schulz C, Knösel T, Cyran CC, Ricke J, Kazmierczak PM, Ingrisch M, Wesp P. Effect of artificial intelligence-aided differentiation of adenomatous and non-adenomatous colorectal polyps at CT colonography on radiologists' therapy management. Eur Radiol. 2025;35:4091-4099.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
162.  Endo S, Nagata K, Utano K, Nozu S, Yasuda T, Takabayashi K, Hirayama M, Togashi K, Ohira H. Development and validation of computer-aided detection for colorectal neoplasms using deep learning incorporated with computed tomography colonography. BMC Gastroenterol. 2025;25:149.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
163.  Tsukanov VV, Vasyutin AV, Kasparov EV, Tonkikh JL. Is the use of artificial intelligence the main stage for detecting polyps during colonoscopy? World J Gastroenterol. 2025;31:106500.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
164.  Eskelinen M, Selander T, Guimarães DP, Kaarniranta K, Syrjänen K, Eskelinen M. Four Different Artificial Intelligence Models Versus Logistic Regression to Enhance the Diagnostic Accuracy of Fecal Immunochemical Test in the Detection of Colorectal Carcinoma in a Screening Setting. Anticancer Res. 2025;45:2477-2491.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
165.  Tun HM, Rahman HA, Naing L, Malik OA. Artificial intelligence utilization in cancer screening program across ASEAN: a scoping review. BMC Cancer. 2025;25:703.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
166.  Bocioagă AG, Oancea CN, Rădulescu D, Ungureanu BS, Iovănescu VF, Florescu DN, Doica IP, Sacerdoțianu VM, Streba L, Ciurea T, Gheonea DI. Neural Network-Based Composite Risk Scoring for Stratification of Fecal Immunochemical Test-Positive Patients in Colorectal Cancer Screening: Findings from South-West Oltenia. Cancers (Basel). 2025;17:1868.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]