Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
1.
J Xray Sci Technol ; 32(1): 31-51, 2024.
Article in English | MEDLINE | ID: mdl-37980593

ABSTRACT

BACKGROUND: Esophageal cancer (EC) is aggressive cancer with a high fatality rate and a rapid rise of the incidence globally. However, early diagnosis of EC remains a challenging task for clinicians. OBJECTIVE: To help address and overcome this challenge, this study aims to develop and test a new computer-aided diagnosis (CAD) network that combines several machine learning models and optimization methods to detect EC and classify cancer stages. METHODS: The study develops a new deep learning network for the classification of the various stages of EC and the premalignant stage, Barrett's Esophagus from endoscopic images. The proposed model uses a multi-convolution neural network (CNN) model combined with Xception, Mobilenetv2, GoogLeNet, and Darknet53 for feature extraction. The extracted features are blended and are then applied on to wrapper based Artificial Bee Colony (ABC) optimization technique to grade the most accurate and relevant attributes. A multi-class support vector machine (SVM) classifies the selected feature set into the various stages. A study dataset involving 523 Barrett's Esophagus images, 217 ESCC images and 288 EAC images is used to train the proposed network and test its classification performance. RESULTS: The proposed network combining Xception, mobilenetv2, GoogLeNet, and Darknet53 outperforms all the existing methods with an overall classification accuracy of 97.76% using a 3-fold cross-validation method. CONCLUSION: This study demonstrates that a new deep learning network that combines a multi-CNN model with ABC and a multi-SVM is more efficient than those with individual pre-trained networks for the EC analysis and stage classification.


Subject(s)
Barrett Esophagus , Esophageal Neoplasms , Humans , Barrett Esophagus/diagnostic imaging , Support Vector Machine , Early Detection of Cancer , Neural Networks, Computer , Esophageal Neoplasms/diagnostic imaging
2.
Saudi J Gastroenterol ; 29(5): 269-277, 2023.
Article in English | MEDLINE | ID: mdl-37787347

ABSTRACT

Since the emergence of artificial intelligence (AI) in medicine, endoscopy applications in gastroenterology have been at the forefront of innovations. The ever-increasing number of studies necessitates the need to organize and classify applications in a useful way. Separating AI capabilities by computer aided detection (CADe), diagnosis (CADx), and quality assessment (CADq) allows for a systematic evaluation of each application. CADe studies have shown promise in accurate detection of esophageal, gastric and colonic neoplasia as well as identifying sources of bleeding and Crohn's disease in the small bowel. While more advanced CADx applications employ optical biopsies to give further information to characterize neoplasia and grade inflammatory disease, diverse CADq applications ensure quality and increase the efficiency of procedures. Future applications show promise in advanced therapeutic modalities and integrated systems that provide multimodal capabilities. AI is set to revolutionize clinical decision making and performance of endoscopy.


Subject(s)
Colonic Neoplasms , Crohn Disease , Endoscopy, Gastrointestinal , Humans , Artificial Intelligence , Colonic Neoplasms/diagnostic imaging , Crohn Disease/diagnostic imaging , Endoscopy , Gastroenterology , Barrett Esophagus/diagnostic imaging
3.
Epigenomics ; 15(15): 759-767, 2023 08.
Article in English | MEDLINE | ID: mdl-37661863

ABSTRACT

Aim: DNA methylation is involved in esophageal adenocarcinoma (EAC) and Barrett's esophagus (BE). Microarchitectures of on-neoplastic BE associated with DNA methylation status were examined using magnifying narrow-band imaging (NBI) endoscopy. Patients and methods: Using biopsies from non-neoplastic BE without cancer (n = 66; N group), with EAC (n = 27; ADJ group) and EAC tissue (n = 22; T group), methylation of N33, DPYS, SLC16A12, miR124a3 and miR34bc genes were quantified. Magnifying NBI features of non-neoplastic BE were classified according to their morphologies. Results: The ADJ and T groups presented higher DNA methylation compared with the N group. Magnifying NBI endoscopic features of non-neoplastic BE also correlated with DNA methylation as an independent factor. Conclusion: Microarchitectures of BE visualized by magnifying NBI endoscopy correlated with DNA methylation.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Humans , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/genetics , Barrett Esophagus/pathology , DNA Methylation , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/genetics , Adenocarcinoma/pathology
5.
London; NICE; Feb. 8, 2023. 29 p.
Non-conventional in English | BIGG - GRADE guidelines | ID: biblio-1415735

ABSTRACT

This guideline covers monitoring, treatment and follow-up for people aged 18 and over with Barrett's oesophagus and stage 1 oesophageal adenocarcinoma. It includes advice on endoscopic and non-endoscopic techniques. It aims to improve outcomes by ensuring the most effective investigations and treatments are used.


Subject(s)
Humans , Adolescent , Adult , Barrett Esophagus/diagnostic imaging , Esophageal Neoplasms/prevention & control , Esophagoscopes/standards
6.
J Nucl Med ; 64(5): 803-808, 2023 05.
Article in English | MEDLINE | ID: mdl-36604181

ABSTRACT

Esophageal adenocarcinoma causes 6% of cancer-related deaths worldwide. Near-infrared fluorescence molecular endoscopy (NIR-FME) uses a tracer that targets overexpressed proteins. In this study, we aimed to investigate the feasibility of an epidermal growth factor receptor (EGFR)-targeted tracer, cetuximab-800CW, to improve detection of early-stage esophageal adenocarcinoma. Methods: We validated EGFR expression in 73 esophageal tissue sections. Subsequently, we topically administered cetuximab-800CW and performed high-definition white-light endoscopy (HD-WLE), narrow-band imaging, and NIR-FME in 15 patients with Barrett esophagus (BE). Intrinsic fluorescence values were quantified using multidiameter single-fiber reflectance and single-fiber fluorescence spectroscopy. Back-table imaging, histopathologic examination, and EGFR immunohistochemistry on biopsy samples collected during NIR-FME procedures were performed and compared with in vivo imaging results. Results: Immunohistochemical preanalysis showed high EGFR expression in 67% of dysplastic tissue sections. NIR-FME visualized all 12 HD-WLE-visible lesions and 5 HD-WLE-invisible dysplastic lesions, with increased fluorescence signal in visible dysplastic BE lesions compared with nondysplastic BE as shown by multidiameter single-fiber reflectance/single-fiber fluorescence, reflecting a target-to-background ratio of 1.5. Invisible dysplastic lesions also showed increased fluorescence, with a target-to-background ratio of 1.67. Immunohistochemistry analysis showed EGFR overexpression in 16 of 17 (94%) dysplastic BE lesions, which all showed fluorescence signal. Conclusion: This study has shown that NIR-FME using cetuximab-800CW can improve detection of dysplastic lesions missed by HD-WLE and narrow-band imaging.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Humans , Cetuximab , Fluorescence , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/pathology , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/pathology , Endoscopy , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , ErbB Receptors/metabolism
7.
Mol Imaging Biol ; 25(1): 18-35, 2023 02.
Article in English | MEDLINE | ID: mdl-35764908

ABSTRACT

Flexible high-definition white-light endoscopy is the current gold standard in screening for cancer and its precursor lesions in the gastrointestinal tract. However, miss rates are high, especially in populations at high risk for developing gastrointestinal cancer (e.g., inflammatory bowel disease, Lynch syndrome, or Barrett's esophagus) where lesions tend to be flat and subtle. Fluorescence molecular endoscopy (FME) enables intraluminal visualization of (pre)malignant lesions based on specific biomolecular features rather than morphology by using fluorescently labeled molecular probes that bind to specific molecular targets. This strategy has the potential to serve as a valuable tool for the clinician to improve endoscopic lesion detection and real-time clinical decision-making. This narrative review presents an overview of recent advances in FME, focusing on probe development, techniques, and clinical evidence. Future perspectives will also be addressed, such as the use of FME in patient stratification for targeted therapies and potential alliances with artificial intelligence. KEY MESSAGES: • Fluorescence molecular endoscopy is a relatively new technology that enables safe and real-time endoscopic lesion visualization based on specific molecular features rather than on morphology, thereby adding a layer of information to endoscopy, like in PET-CT imaging. • Recently the transition from preclinical to clinical studies has been made, with promising results regarding enhancing detection of flat and subtle lesions in the colon and esophagus. However, clinical evidence needs to be strengthened by larger patient studies with stratified study designs. • In the future fluorescence molecular endoscopy could serve as a valuable tool in clinical workflows to improve detection in high-risk populations like patients with Barrett's esophagus, Lynch syndrome, and inflammatory bowel syndrome, where flat and subtle lesions tend to be malignant up to five times more often. • Fluorescence molecular endoscopy has the potential to assess therapy responsiveness in vivo for targeted therapies, thereby playing a role in personalizing medicine. • To further reduce high miss rates due to human and technical factors, joint application of artificial intelligence and fluorescence molecular endoscopy are likely to generate added value.


Subject(s)
Barrett Esophagus , Colorectal Neoplasms, Hereditary Nonpolyposis , Humans , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/pathology , Artificial Intelligence , Positron Emission Tomography Computed Tomography , Endoscopy, Gastrointestinal/methods , Endoscopy/methods , Molecular Imaging/methods
8.
World J Gastroenterol ; 29(48): 6198-6207, 2023 Dec 28.
Article in English | MEDLINE | ID: mdl-38186865

ABSTRACT

BACKGROUND: Barrett's esophagus (BE), which has increased in prevalence worldwide, is a precursor for esophageal adenocarcinoma. Although there is a gap in the detection rates between endoscopic BE and histological BE in current research, we trained our artificial intelligence (AI) system with images of endoscopic BE and tested the system with images of histological BE. AIM: To assess whether an AI system can aid in the detection of BE in our setting. METHODS: Endoscopic narrow-band imaging (NBI) was collected from Chung Shan Medical University Hospital and Changhua Christian Hospital, resulting in 724 cases, with 86 patients having pathological results. Three senior endoscopists, who were instructing physicians of the Digestive Endoscopy Society of Taiwan, independently annotated the images in the development set to determine whether each image was classified as an endoscopic BE. The test set consisted of 160 endoscopic images of 86 cases with histological results. RESULTS: Six pre-trained models were compared, and EfficientNetV2B2 (accuracy [ACC]: 0.8) was selected as the backbone architecture for further evaluation due to better ACC results. In the final test, the AI system correctly identified 66 of 70 cases of BE and 85 of 90 cases without BE, resulting in an ACC of 94.37%. CONCLUSION: Our AI system, which was trained by NBI of endoscopic BE, can adequately predict endoscopic images of histological BE. The ACC, sensitivity, and specificity are 94.37%, 94.29%, and 94.44%, respectively.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Humans , Barrett Esophagus/diagnostic imaging , Artificial Intelligence , Esophageal Neoplasms/diagnostic imaging , Adenocarcinoma/diagnostic imaging , Hospitals, University
9.
World J Gastroenterol ; 28(32): 4516-4526, 2022 Aug 28.
Article in English | MEDLINE | ID: mdl-36157931

ABSTRACT

Barrett's esophagus (BE) is a condition that results from replacement of the damaged normal squamous esophageal mucosa to intestinal columnar mucosa and is the most significant predisposing factor for development of esophageal adenocarcinoma. Current guidelines recommend endoscopic evaluation for screening and surveillance based on various risk factors which has limitations such as invasiveness, availability of a trained specialist, patient logistics and cost. Trans-nasal endoscopy is a less invasive modality but still has similar limitations such as limited availability of trained specialist and costs. Non-endoscopic modalities, in comparison, require minimal intervention, can be done in an office visit and has the potential to be a more ideal choice for mass public screening and surveillance, particularly in patents at low risk for BE. These include newer generations of esophageal capsule endoscopy which provides direct visualization of BE, and tethered capsule endomicroscopy which can obtain high-resolution images of the esophagus. Various cell collection devices coupled with biomarkers have been used for BE screening. Cytosponge, in combination with TFF3, as well as EsophaCap and EsoCheck have shown promising results in various studies when used with various biomarkers. Other modalities including circulatory microRNAs and volatile organic compounds that have demonstrated favorable outcomes. Use of these cell collection methods for BE surveillance is a potential area of future research.


Subject(s)
Barrett Esophagus , Esophageal Neoplasms , MicroRNAs , Volatile Organic Compounds , Barrett Esophagus/diagnostic imaging , Biomarkers , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/etiology , Esophagoscopy/adverse effects , Humans
10.
Sci Rep ; 12(1): 16314, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36175457

ABSTRACT

Volumetric laser endomicroscopy (VLE) is an advanced endoscopic imaging tool that can improve dysplasia detection in Barrett's esophagus (BE). However, VLE scans generate 1200 cross-sectional images that can make interpretation difficult. The impact of a new VLE artificial intelligence algorithm called Intelligent Real-time Image Segmentation (IRIS) is not well-characterized. This is a randomized prospective cross-over study of BE patients undergoing endoscopy who were randomized to IRIS-enhanced or unenhanced VLE first followed by the other (IRIS-VLE vs. VLE-IRIS, respectively) at expert BE centers. The primary outcome was image interpretation time, which served as a surrogate measure for ease of interpretation. The secondary outcome was diagnostic yield of dysplasia for each imaging modality. 133 patients were enrolled. 67 patients were randomized to VLE-IRIS and 66 to IRIS-VLE. Total interpretation time did not differ significantly between groups (7.8 min VLE-IRIS vs. 7 min IRIS-VLE, P = 0.1), however unenhanced VLE interpretation time was significantly shorter in the IRIS-VLE group (2.4 min vs. 3.8 min, P < 0.01). When IRIS was used first, 100% of dysplastic areas were identified, compared with 76.9% when VLE was the first interpretation modality (P = 0.06). IRIS-enhanced VLE reduced the time of subsequent unenhanced VLE interpretation, suggesting heightened efficiency and improved dysplasia detection. It was also able to identify all endoscopically non-visible dysplastic areas.


Subject(s)
Barrett Esophagus , Artificial Intelligence , Barrett Esophagus/diagnostic imaging , Cross-Over Studies , Humans , Hyperplasia , Lasers , Prospective Studies
11.
J Gastroenterol Hepatol ; 37(11): 2113-2119, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35997124

ABSTRACT

BACKGROUND AND AIM: Gastric and esophageal cancers are associated with high morbidity in India. In the absence of formal screening programs in India, it is essential that all elective esophago-gastro-duodenoscopies (EGDs), irrespective of indication, be also considered an opportunity to screen for premalignant lesions. With this premise, we tried to assess the adherence to best practices in the detection of premalignant upper gastro-intestinal lesions (PMUGIL) among endoscopists in India. We also evaluated the adequacy of training, availability of appropriate facilities, and differences between teaching and non-teaching centers. METHODS: We disbursed a survey among endoscopists working in India, through the membership database of the Society of Gastrointestinal Endoscopists of India, by email and instant messaging. The responses were collected and subsequently analyzed. RESULTS: We obtained a total of 422 eligible responses. The adherence to best practices assessed was lower than the set threshold in all except one parameter in both teaching centers and non-teaching centers. Only 58.5% of endoscopists had received training in the detection of PMUGIL. Appropriate image enhanced endoscopy (IEE) facilities were available to only 58.05% of surveyed endoscopists. CONCLUSIONS: Strategies to improve detection of PMUGIL should be directed at improving adherence to best practices, ensuring adequate training of endoscopists in the evaluation of PMUGIL and improving infrastructure.


Subject(s)
Endoscopy, Gastrointestinal , Esophageal Neoplasms , Stomach Neoplasms , Humans , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/prevention & control , Stomach Neoplasms/diagnostic imaging , Stomach Neoplasms/epidemiology , Stomach Neoplasms/prevention & control , Barrett Esophagus/diagnostic imaging
14.
United European Gastroenterol J ; 10(6): 528-537, 2022 07.
Article in English | MEDLINE | ID: mdl-35521666

ABSTRACT

BACKGROUND AND AIMS: Seattle protocol biopsies for Barrett's Esophagus (BE) surveillance are labour intensive with low compliance. Dysplasia detection rates vary, leading to missed lesions. This can potentially be offset with computer aided detection. We have developed convolutional neural networks (CNNs) to identify areas of dysplasia and where to target biopsy. METHODS: 119 Videos were collected in high-definition white light and optical chromoendoscopy with i-scan (Pentax Hoya, Japan) imaging in patients with dysplastic and non-dysplastic BE (NDBE). We trained an indirectly supervised CNN to classify images as dysplastic/non-dysplastic using whole video annotations to minimise selection bias and maximise accuracy. The CNN was trained using 148,936 video frames (31 dysplastic patients, 31 NDBE, two normal esophagus), validated on 25,161 images from 11 patient videos and tested on 264 iscan-1 images from 28 dysplastic and 16 NDBE patients which included expert delineations. To localise targeted biopsies/delineations, a second directly supervised CNN was generated based on expert delineations of 94 dysplastic images from 30 patients. This was tested on 86 i-scan one images from 28 dysplastic patients. FINDINGS: The indirectly supervised CNN achieved a per image sensitivity in the test set of 91%, specificity 79%, area under receiver operator curve of 93% to detect dysplasia. Per-lesion sensitivity was 100%. Mean assessment speed was 48 frames per second (fps). 97% of targeted biopsy predictions matched expert and histological assessment at 56 fps. The artificial intelligence system performed better than six endoscopists. INTERPRETATION: Our CNNs classify and localise dysplastic Barrett's Esophagus potentially supporting endoscopists during surveillance.


Subject(s)
Barrett Esophagus , Esophageal Neoplasms , Artificial Intelligence , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/pathology , Biopsy/methods , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/pathology , Humans , Neural Networks, Computer
15.
World J Gastroenterol ; 28(11): 1113-1122, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35431503

ABSTRACT

Barrett's esophagus (BE) is a well-established risk factor for esophageal adenocarcinoma. It is recommended that patients have regular endoscopic surveillance, with the ultimate goal of detecting early-stage neoplastic lesions before they can progress to invasive carcinoma. Detection of both dysplasia or early adenocarcinoma permits curative endoscopic treatments, and with this aim, thorough endoscopic assessment is crucial and improves outcomes. The burden of missed neoplasia in BE is still far from being negligible, likely due to inappropriate endoscopic surveillance. Over the last two decades, advanced imaging techniques, moving from traditional dye-spray chromoendoscopy to more practical virtual chromoendoscopy technologies, have been introduced with the aim to enhance neoplasia detection in BE. As witnessed in other fields, artificial intelligence (AI) has revolutionized the field of diagnostic endoscopy and is set to cover a pivotal role in BE as well. The aim of this commentary is to comprehensively summarize present evidence, recent research advances, and future perspectives regarding advanced imaging technology and AI in BE; the combination of computer-aided diagnosis to a widespread adoption of advanced imaging technologies is eagerly awaited. It will also provide a useful step-by-step approach for performing high-quality endoscopy in BE, in order to increase the diagnostic yield of endoscopy in clinical practice.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Artificial Intelligence , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/epidemiology , Endoscopy/methods , Esophageal Neoplasms/diagnosis , Esophagoscopy/methods , Humans
16.
Endoscopy ; 54(12): 1198-1204, 2022 12.
Article in English | MEDLINE | ID: mdl-35299273

ABSTRACT

BACKGROUND: Esophageal adenocarcinoma (EAC) is a molecularly heterogeneous disease with poor prognosis that is rising rapidly in incidence. We aimed to demonstrate specific binding by a peptide heterodimer to Barrett's neoplasia in human subjects. METHODS: Peptide monomers specific for EGFR and ErbB2 were arranged in a heterodimer configuration and labeled with IRDye800. This near-infrared (NIR) contrast agent was topically administered to patients with Barrett's esophagus (BE) undergoing either endoscopic therapy or surveillance. Fluorescence images were collected using a flexible fiber accessory passed through the instrument channel of an upper gastrointestinal endoscope. Fluorescence images were collected from 31 BE patients. A deep learning model was used to segment the target (T) and background (B) regions. RESULTS: The mean target-to-background (T/B) ratio was significantly greater for high grade dysplasia (HGD) and EAC versus BE, low grade dysplasia (LGD), and squamous epithelium. At a T/B ratio of 1.5, sensitivity and specificity of 94.1 % and 92.6 %, respectively, were achieved for the detection of Barrett's neoplasia with an area under the curve of 0.95. No adverse events attributed to the heterodimer were found. EGFR and ErbB2 expression were validated in the resected specimens. CONCLUSIONS: This "first-in-human" clinical study demonstrates the feasibility of detection of early Barrett's neoplasia using a NIR-labeled peptide heterodimer.


Subject(s)
Barrett Esophagus , Esophageal Neoplasms , Precancerous Conditions , Humans , Precancerous Conditions/pathology , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/epidemiology , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/etiology , Hyperplasia , Peptides
17.
Lancet Oncol ; 23(2): 270-278, 2022 02.
Article in English | MEDLINE | ID: mdl-35030332

ABSTRACT

BACKGROUND: Endoscopic surveillance is recommended for patients with Barrett's oesophagus because, although the progression risk is low, endoscopic intervention is highly effective for high-grade dysplasia and cancer. However, repeated endoscopy has associated harms and access has been limited during the COVID-19 pandemic. We aimed to evaluate the role of a non-endoscopic device (Cytosponge) coupled with laboratory biomarkers and clinical factors to prioritise endoscopy for Barrett's oesophagus. METHODS: We first conducted a retrospective, multicentre, cross-sectional study in patients older than 18 years who were having endoscopic surveillance for Barrett's oesophagus (with intestinal metaplasia confirmed by TFF3 and a minimum Barrett's segment length of 1 cm [circumferential or tongues by the Prague C and M criteria]). All patients had received the Cytosponge and confirmatory endoscopy during the BEST2 (ISRCTN12730505) and BEST3 (ISRCTN68382401) clinical trials, from July 7, 2011, to April 1, 2019 (UK Clinical Research Network Study Portfolio 9461). Participants were divided into training (n=557) and validation (n=334) cohorts to identify optimal risk groups. The biomarkers evaluated were overexpression of p53, cellular atypia, and 17 clinical demographic variables. Endoscopic biopsy diagnosis of high-grade dysplasia or cancer was the primary endpoint. Clinical feasibility of a decision tree for Cytosponge triage was evaluated in a real-world prospective cohort from Aug 27, 2020 (DELTA; ISRCTN91655550; n=223), in response to COVID-19 and the need to provide an alternative to endoscopic surveillance. FINDINGS: The prevalence of high-grade dysplasia or cancer determined by the current gold standard of endoscopic biopsy was 17% (92 of 557 patients) in the training cohort and 10% (35 of 344) in the validation cohort. From the new biomarker analysis, three risk groups were identified: high risk, defined as atypia or p53 overexpression or both on Cytosponge; moderate risk, defined by the presence of a clinical risk factor (age, sex, and segment length); and low risk, defined as Cytosponge-negative and no clinical risk factors. The risk of high-grade dysplasia or intramucosal cancer in the high-risk group was 52% (68 of 132 patients) in the training cohort and 41% (31 of 75) in the validation cohort, compared with 2% (five of 210) and 1% (two of 185) in the low-risk group, respectively. In the real-world setting, Cytosponge results prospectively identified 39 (17%) of 223 patients as high risk (atypia or p53 overexpression, or both) requiring endoscopy, among whom the positive predictive value was 31% (12 of 39 patients) for high-grade dysplasia or intramucosal cancer and 44% (17 of 39) for any grade of dysplasia. INTERPRETATION: Cytosponge atypia, p53 overexpression, and clinical risk factors (age, sex, and segment length) could be used to prioritise patients for endoscopy. Further investigation could validate their use in clinical practice and lead to a substantial reduction in endoscopy procedures compared with current surveillance pathways. FUNDING: Medical Research Council, Cancer Research UK, Innovate UK.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , COVID-19 , Esophageal Neoplasms/pathology , Patient Selection , Watchful Waiting/methods , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/metabolism , Aged , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/metabolism , Barrett Esophagus/therapy , Biomarkers/metabolism , COVID-19/prevention & control , Clinical Decision-Making , Clinical Trials as Topic , Cross-Sectional Studies , Decision Trees , Disease Progression , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/metabolism , Esophagoscopy , Feasibility Studies , Female , Humans , Male , Middle Aged , Pilot Projects , Prospective Studies , Retrospective Studies , Risk Assessment , Risk Factors , SARS-CoV-2 , Trefoil Factor-3/metabolism , Tumor Suppressor Protein p53/metabolism
18.
Dig Dis Sci ; 67(10): 4805-4812, 2022 10.
Article in English | MEDLINE | ID: mdl-35084606

ABSTRACT

BACKGROUND AND AIMS: Endoscopic surveillance of Barrett's esophagus (BE) by white light examination is insufficient to diagnose dysplastic change. In this work, we describe an optical imaging method to obtain high-resolution cross-sectional imaging using a paddle-shaped probe affixed to the endoscope tip. METHODS: We integrated Optical Coherence Tomography (OCT), an optical imaging method that produces cross-sectional images, into a paddle probe attached to video endoscope. We acquired images of esophageal epithelium from patients undergoing routine upper GI endoscopy. Images were classified by a reviewer blinded to patient identity and condition, and these results were compared with clinical diagnosis. RESULTS: We successfully captured epithelial OCT images from 30 patients and identified features consistent with both squamous epithelium and Barrett's esophagus. Our blinded image reviewer classified BE versus non-BE with 91.5% accuracy (65/71 image regions), including sensitivity of 84.6% for BE (11/13) and a specificity of 93.1% (54/58). However, in 16 patients, intubation of the probe into the esophagus could not be achieved. CONCLUSIONS: A paddle probe is a feasible imaging format for acquiring cross-sectional OCT images from the esophagus and can provide a structural assessment of BE and non-BE tissue. Probe form factor is the current limiting obstacle, but could be addressed by further miniaturization.


Subject(s)
Barrett Esophagus , Esophageal Neoplasms , Barrett Esophagus/diagnostic imaging , Endoscopes , Endoscopy, Digestive System , Esophagoscopy/methods , Humans , Tomography, Optical Coherence/methods
19.
Dig Dis ; 40(1): 97-105, 2022.
Article in English | MEDLINE | ID: mdl-33794523

ABSTRACT

BACKGROUND: Barrett's esophagus (BE) requires surveillance to identify potential neoplasia at an early stage. The standard surveillance regimen includes random 4-quadrant biopsies by Seattle protocol. Main limitations of random biopsies are high risk of sampling error, difficulties in histology interpretation, common inadequate classification of pathohistological changes, increased risk of bleeding, and time necessary to acquire the final diagnosis. Probe-based confocal laser endomicroscopy (pCLE) has emerged as a potential tool with an aim to overcome these obvious limitations. SUMMARY: pCLE represents a real-time microscopic imaging method that offers evaluation of epithelial and subepithelial structures with 1,000-fold magnification. In theory, pCLE has potential to eliminate the need for biopsy in BE patients. The main advantages would be real-time diagnosis and decision-making, greater diagnostic accuracy, and evaluation of larger area compared to random biopsies. Clinical pCLE studies in the esophagus show high diagnostic accuracy, and its high negative predictive value offers high reliability and confidence to exclude dysplastic and neoplastic lesions. However, it still cannot replace histopathology due to lower positive predictive value and sensitivity. Key Messages: Despite promising results, its role in routine use in patients with BE remains questionable primarily due to lack of well-organized double-blind randomized trials.


Subject(s)
Barrett Esophagus , Esophageal Neoplasms , Barrett Esophagus/diagnostic imaging , Biopsy , Esophageal Neoplasms/diagnostic imaging , Esophagoscopy , Humans , Lasers , Microscopy, Confocal , Randomized Controlled Trials as Topic , Reproducibility of Results
20.
Clin Gastroenterol Hepatol ; 20(4): 756-765.e3, 2022 04.
Article in English | MEDLINE | ID: mdl-33549871

ABSTRACT

BACKGROUND & AIMS: Tethered capsule endomicroscopy (TCE) involves swallowing a small tethered pill that implements optical coherence tomography (OCT) imaging, procuring high resolution images of the whole esophagus. Here, we demonstrate and evaluate the feasibility and safety of TCE and a portable OCT imaging system in patients with Barrett's esophagus (BE) in a multi-center (5-site) clinical study. METHODS: Untreated patients with BE as per endoscopic biopsy diagnosis were eligible to participate in the study. TCE procedures were performed in unsedated patients by either doctors or nurses. After the capsule was swallowed, the device continuously obtained 10-µm-resolution cross-sectional images as it traversed the esophagus. Following imaging, the device was withdrawn through mouth, and disinfected for subsequent reuse. BE lengths were compared to endoscopy findings when available. OCT-TCE images were compared to volumetric laser endomicroscopy (VLE) images from a patient who had undergone VLE on the same day as TCE. RESULTS: 147 patients with BE were enrolled across all sites. 116 swallowed the capsule (79%), 95/114 (83.3%) men and 21/33 (63.6%) women (P = .01). High-quality OCT images were obtained in 104/111 swallowers (93.7%) who completed the procedure. The average imaging duration was 5.55 ± 1.92 minutes. The mean length of esophagus imaged per patient was 21.69 ± 5.90 cm. A blinded comparison of maximum extent of BE measured by OCT-TCE and EGD showed a strong correlation (r = 0.77-0.79). OCT-TCE images were of similar quality to those obtained by OCT-VLE. CONCLUSIONS: The capabilities of TCE to be used across multiple sites, be administered to unsedated patients by either physicians or nurses who are not expert in OCT-TCE, and to rapidly and safely evaluate the microscopic structure of the esophagus make it an emerging tool for screening and surveillance of BE patients. Clinical trial registry website and trial number: NCT02994693 and NCT03459339.


Subject(s)
Barrett Esophagus , Esophageal Neoplasms , Barrett Esophagus/diagnostic imaging , Barrett Esophagus/pathology , Biopsy , Esophageal Neoplasms/pathology , Esophagoscopy/methods , Feasibility Studies , Female , Humans , Male , Tomography, Optical Coherence/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...