Preprint
Data Descriptor

This version is not peer-reviewed.

Dataset and AI Workflow for Deep Learning Image Classification of Ulcerative Colitis and Colorectal Cancer

A peer-reviewed article of this preprint also exists.

Submitted:

21 February 2025

Posted:

25 February 2025

Read the latest preprint version here

Abstract

Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract characterized by the deregulation of immuno-oncology markers. IBD includes ulcerative colitis and Chron disease. Chronic active inflammation is a risk factor for the development of colorectal cancer (CRC). Deep learning is a form of machine learning that is applicable to computer vision, and it includes algorithms and workflows used for image processing, analysis, visualization, and algorithm development. This publication of Data Descriptor type, describes a dataset of histological images of ulcerative colitis, colorectal cancer (adenocarcinoma), and colon control. The samples were stained with hematoxylin and eosin (H&E), and immunohistochemically analyzed for LAIR1 and TOX2 markers. The methods used for collecting and producing the data, analysis using convolutional neural networks (CNNs), where the dataset can be found, and information about its use are also described.

Keywords: 
;  ;  ;  ;  ;  ;  ;  ;  ;  ;  ;  

1. Summary

1.1. Background of Ulcerative Colitis

Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract with systemic repercussions that is characterized by relapsing and remitting episodes of inflammation [1,2]. IBD includes two types: ulcerative colitis, which affects the colon, and Chron disease, which can affect any part of the gastrointestinal tract from the mouth until the perianal area [1,2,3,4,5,6,7,8,9].
The causes and pathogenesis of inflammatory bowel disease are unclear. However, it appears to be a combination of factors from the environment, including the microbiome [10,11], genetic susceptibility [12,13], and immune system (both systemic and gut) [13,14,15,16]. The prevalence of inflammatory bowel disease has increased globally in recent decades, particularly in industrialized countries [17,18,19,20,21]. Clinical risk factors include smoking [22], low physical activity, low fiber intake, high fats, and low vitamin D [23], sleep deprivation, previous acute gastroenteritis, antibiotic use, and early life exposures [24].
In normal conditions and in healthy status, the intestinal barrier is kept thanks to the mucus layer and epithelial cells that create bonds using the tight junctions. This barrier is supported by the presence of IgA and several antimicrobial factors. The immune response is initiated by dendritic cells that acquire, process, and present the antigens to B and T cells (lymphocytes) [25]. The cause of ulcerative colitis is still not completely understood. Ulcerative colitis is associated with a damage of the barrier of the mucosa, with an alteration of the microflora and the generation of an uncontrolled immune reaction. Several subsets of CD4-positive T cell are believed to participate in the pathogenesis of ulcerative colitis including Th1, Th2, Th9, Th17, Th22, TFh cells, and Tregs [26,27].Th9 are associated with continuous apoptosis of the enterocytes and inhibit mucosal healing [25,28]. IL-13 and NK/T cells are also involved in the epithelial injury [25]. Innate immune cells contribute to the cytokine production and continuous inflammation [14,15,29]. The injury of the mucosa is associated with dysbiosis [30], which is defined by an alteration of the composition of the mucosa, with changes of the diversity, increased potentially pathogenic bacteria, and reduced beneficial bacteria [31]. Figure 1 and Figure 2 show an example of ulcerative colitis and immunohistochemistry of the cells of the microenvironment involved in the pathogenesis.
The disease activity of ulcerative colitis is being recorded to properly treat patients and perform clinical trials. Usually, the severity is classified as mild, moderate, and severe. The available classifications include the Mayo [32], Montreal [33], Truelove, and Witts classifications [34]. The grade of mucosal inflammation can be assessed using endoscopy to record the degree of mucosal ulceration and the extent of disease. This can be graded using the Mayo score [32] and Baron score. The degree of histological severity can be evaluated using the Geboes score [35].
Microscopic (histologic) evaluation of ulcerative colitis shows active chronic colitis in patients with untreated disease. Disease activity is defined by neutrophil infiltration of the lamina propria, cryptitis, crypt abscess, and ulceration. The inflammation is limited to the mucosa and submucosa. Compared with Crohn disease, this condition is associated with a lack of transmural inflammation, granuloma, and fissuring ulcers. Dysplasia may be present in long-term disease [36,37,38].
Disease activity can be evaluated using various parameters. Inactive disease lacks neutrophils. If the activity affects less than 50% of the mucosa, it is called mild. When it is > 50% and there are crypt abscesses, it is called moderate. Finally, severe activity is characterized by surface ulcerations and erosion [39].
The first-line therapy for the induction and maintenance of remission of mild to moderate UC is 5-aminosalicylic acid [40]. The initial therapy for ulcerative colitis is topical mesalamine, which is available as a suppository or enema, and it is recommended in cases of ulcerative proctitis or proctosigmoiditis. If topical mesalamine is not tolerated, an alternative therapy is topical glucocorticoids (i.e. hydrocortisone). A combination of 5-ASA and rectal mesalamine may be used for left-sided or extensive colitis. The 5-ASA formulations include mesalamine, sulfasalazine, and diazo-bonded 5-ASA. Patients who do not respond may subsequently be treated with glucocorticoids such as budesonide, which may escalate to prednisone or biological agents (anti-TNF, anti-integrin, anti-IL12/23, S1P modulators, JAK inhibitors) [40,41,42,43]. Patients who do not respond to oral glucocorticoids may require intravenous glucocorticoids. In some cases, surgery is performed [40,41,42,43]. Fecal microbiota transplantation is also available [44].

1.2. Background of Colorectal Cancer

Colorectal cancer (CRC) is a common disease characterized by environmental and genetic factors. It is the third most frequently diagnosed cancer worldwide [45]. There are several factors associated with early onset of CRC, including hereditary syndromes (familial adenomatous polyposis and Lynch syndrome), metabolic dysregulation, history of CRC in first-degree relative; alcohol consumption, lack of regular use of nonsteroidal anti-inflammatory drugs (NSAIDs), and vitamin D intake. Other known risk factors of CRC include inflammatory bowel disease (ulcerative colitis and Crohn disease), abdominopelvic radiation, obesity, diabetes mellitus, insulin resistance, red and processed meat, and tobacco [46,47,48,49]. The diagnosis is usually made by colonoscopy, and treatment includes surgical resection, adjuvant chemotherapy, and postoperative radiation therapy [46]. Adenocarcinoma is the most common histological subtype of CRC. Adenocarcinoma is a glandular neoplasm. Most CRC cases are moderately differentiated with simple, complex, or slightly irregular tubules, and loss of nuclear polarity. The glands are often cribriform, with necrosis, inflammatory cells, and marked desmoplasia [50,51,52].

1.3. Background of Computer Vision for Deep Learning Image Classification

Image processing involves algorithms and workflows used for image processing, analysis, visualization, and algorithm development. Computer vision workflows [53] with deep learning include several types of functions, such as image classification, object detection, and instance segmentation, automated visual inspection, semantic segmentation, and video classification [54,55,56].
A pretrained neural network that has already learned how to extract the characteristics of natural images can be used as a starting point to handle new images. Using a pretrained image classification network [57], the learning time is shorter, and training is usually easier. The transfer learning process takes layers from an already trained neural network and calibrates the parameters on a new dataset [54,55,56,58,59,60,61,62,63,64,65]. The analysis includes a series of steps, including preprocessing the data, import pre-trained networks from platforms such as TensorFlow 2 [66], TensorFlow-Keras [67], Pythorch [68], and ONNX [69], building network, selecting training options, improving the network performance by tuning hyperparameters, visualizing and verifying network behavior during and after training, and exporting the network to other platforms if necessary[54,55,56,58,59,60,61,62,63,64,65].
The most important features of a neural network are accuracy, speed, and size. A good neural network is characterized by being fast and having good performance (i.e. accurate). Neural networks that are accurate in ImageNet can be used to classify other images using transfer learning or feature extraction. Among the various neural networks, the best examples are GoogLeNet, ResNet-18, NobileNet, ResNet-50, ResNet-101, and Inception-v3 [56]. Figure 4 shows the general design of a CNN, and Figure 5 the original RestNet-18 architecture [70].

1.4. Dataset and Research Project Description

Ulcerative colitis is a chronic inflammatory bowel disease associated with a high risk of colorectal cancer. This study used convolutional neural networks and computer vision to classify histological images of ulcerative colitis, colorectal cancer (adenocarcinoma), and colon control.
The series included 35 patients with of ulcerative colitis, 18 with colorectal cancer, and 21 with colon control. Hematoxylin and eosin (H&E) glass slides were converted into high-resolution digital data by high-speed scanning. The whole-tissue slides were split into image patches of 224×224 pixels at 200× magnification and 150 dpi, and the ResNet-18 network was retrained to classify the 3 types of diagnosis. This transfer learning experiment also used other pretrained CNNs for performance comparison, and the gradient-weighted class activation mapping (Grad-CAM) heatmap technique was used to understand the classification decisions.
Additionally, immunohistochemical analysis of two new immuno-oncology markers, LAIR1 and TOX2 were analyzed in ulcerative colitis to differentiate between mesalazine-responsive and steroid-requiring patients. LAIR1 an inhibitory receptor that plays a constitutive negative regulatory role in the cytolytic function of natural killer (NK) cells, B-cells and T-cells [71,72,73]. TOX2 is a new marker similar to PD-1 in the co-inhibitory pathway [71,74].
Statistical analyses showed that steroid-requiring ulcerative colitis was characterized by higher endoscopic Baron and histologic Geboes scores and LAIR1 expression, but lower TOX2 expression in isolated lymphoid follicles. The CNN managed to classify the 3 diagnoses with >99% accuracy and the Grad-CAM confirmed which parts of the images were relevant for the classification. In conclusion, the study showed that CNNs are essential tools for deep learning image recognition.
These results were published as preprints [75].
This paper is the companion manuscript of the recently published article “Ulcerative Colitis, LAIR1 and TOX2 Expression, and Colorectal Cancer Deep Learning Image Classification Using Convolutional Neural Networks” published in Cancers 2024, 16, 4230. https://doi.org/10.3390/cancers16244230.

2. Data Description

The dataset contains image patches of ulcerative colitis, colorectal cancer, and colon control. The image patches were split from whole-tissue images and had a size of 224 × 224. The original magnification of the images was 200× and a dpi of 150. The image patches are anonymized. After splitting the images, the patches were filtered. The criteria were as follows: (1) image patches of only 243×243 size; (2) image patches of more than 5-31 KB that contain tissue at least 20-30% of viable tissue; (3) image patches with diagnostic areas; (4) image patches without artifacts, including broken tissue, folded areas, incorrectly stained tissue, and smashed/crushed tissue. The steps 3 and 4 were manually performed by the doctor specialist in pathology (MD PhD).
The investigations were carried out following the rules of the Declaration of Helsinki of 1975 (website: https://www.wma.net/policies-post/wma-declaration-of-helsinki/; last accessed on December 12, 2024), revised in 2008. Approval from an ethics committee was obtained before undertaking the research (protocol code IRB14R-080, IRB20-156, and 13R-119).

3. Methods

3.1. Hematoxylin and Eosin (H&E) Staining

H&E staining is widely used in histological and cytological applications, both in fixed paraffin-embedded and frozen tissue sections. Hematoxylin produces an intense blue staining of the nuclei. Eosin stains the cytoplasm, collagen, muscle, and erythrocytes in light pink/rose. Staining was performed using a Leica ST5010-CV5030 integrated stainer workstation (Leica Biosystems K.K., Shinjuku, Tokyo, 169-0075, Japan).

3.2. Score Evaluation

The endoscopic scores for chronic inflammatory bowel disease were evaluated using the Baron Score, which classifies mucosal changes into 3 grades. Ulcerative colitis histological assessment was performed using the Geboes score. Appendix Table A1 and Table A2 present the Baron and Geboes scores, respectively.

3.2. Immunohistochemistry

The immunohistochemistry of LAIR1 and TOX2 was performed using a Bond-Max fully automated immunohistochemistry and in situ hybridization staining system following the manufacturer’s instructions (Leica Biosystems K.K.). Visualization of the primary antibody bound to the tissue sections was performed using BOND Polymer Refine Detection (DS9800, Leica Biosystems K.K.). The BOND staining mode was single, i.e. the application of a single marker and chromogen to a single slide. The protocol sequence was the following: preparation (removal of wax), heat-induced epitope retrieval (HIER), probe application, probe removal, and staining. The staining protocol included the following steps: marker, post primary, polymer, mixed DAB, BOND-PRIME hematoxylin. Coverslipping was achieved using a Leica CV5030 fully automated glass coverslipper (Leica Biosystems K.K.).
The primary antibody targeted the leukocyte-associated immunoglobulin-like receptor (LAIR1/CD305) created by the Monoclonal Antibodies Core Unit, located at the Spanish National Cancer Research Center (CNIO: Centro Nacional de Investigaciones Oncologicas; C/ Melchor Fernandez Almagro, 3, E-28029 Madrid, Spain). LAIR1 is a rat monoclonal antibody, clone JAVI82A, antigen used: RBL-1-LAIR1-MYCDDK transfected cells and last booster with LAIR1 recombinant (Gln22-His163, with a C-terminal 6-His tag); isotype IgG2a; reactivity, human; localization, membrane.
The primary antibody TOX2 targeted the TOX High Mobility Group Box Family Member 2 and was also developed by CNIO. Properties: clone name TOM924D, rat monoclonal, IgG2b K, antigen HIS-SUMO-hTOX2-Strep-tag2 full-length protein, human reactivity, nuclear localization.
Rabbit Anti-Rat IgG Antibody (H+L), Mouse Adsorbed, Unconjugated (#AI-4001-.5, Vector Laboratories, Inc., Newark, CA 94560, United States) was used as a linker between primary antibodies and BOND Polymer Refine Detection.
The immunohistochemistry of both antibodies was first tested in reactive lymphoid tissue (tonsils) and small and large intestine tissue controls (Appendix Figure 1). Four examples of biopsies of colon of ulcerative colitis stained with H&E, TOX2 and LAIR1 and high infiltration of positive cells are show in Appendix Figures A1, B1, and C1.

3.3. Whole-Slide Imaging

Whole-slide imaging was performed using a Hamamatsu NanoZoomer S360 scanner (Hamamatsu Photonics K.K., Hamamatsu City, 431-3196, Japan) that rapidly scanned glass slides to convert them to digital data, NZAcquire 1.2.0 software (Hamamatsu Photonics K.K.), and a Dell Precision 5820 Tower equipped with an Intel(R) Xeon(R) W-2135 CPU @ 3.70 GHz 3.70 GHz, 32.0 GB RAM, 64 bits workstation system. The operating system was Windows 10 Pro for Workstations (version 1803, build 17134.1).
The acquisition method was performed following the manufacturer’s instructions. The scan was batch scan-type, at 400× magnification, and single z stack. The number of focus points was defined automatically using NZAcquire software (Figure 6).

3.4. Digital Image Quantification

Conventional immunohistochemical analysis was performed using digital image quantification with Fiji software, as previously described [76,77,78,79]. In summary, quantification was performed in the blue stack, min and max thresholds were set, pixels were measured, and percentages were calculated.

3.5. Image Classification Using CNNs

A convolutional neural network (CNN) was designed based on transfer learning from ResNet-18; and trained to classify the 3 types of image patches; ulcerative colitis (n= 9,281), colon control (n = 12,246), and colorectal cancer (n = 63,725). The CNN was designed using MATLAB (R2023b, update 9, 23.2.0.2668659). The CNN was also trained to differentiate between mesalazine-responsive and steroid-requiring ulcerative colitis based on H&E, LAIR1, and TOX2 staining (Figure 7).
The data were partitioned into a training set (70% of the image patches) to train the network, a validation set (10%) to test the performance of the network during training, and a test set (20%) as a holdout (new data) to test the performance on new data.
The arrange of the images was randomized to guarantee that the CNN learned the classes at a more even rate. Transfer learning, which is the procedure of re-using and adjusting a pre-trained network, was performed on ResNet-18. For this purpose, the fully connected and classification layers of the ResNet-18 were deleted and replaced with new layers. These new layers had an output size of 2. The training did not used augmentation technique. To avoid overfitting, the initial learning rate was set to 0.001. The maximum numbers of epochs was five [75].
Data normalization was applied to the input images. The detailed description of the data normalization is shown in Appendix Table A3 [80,81].
The trained network was characterized by 71 layers. The first layer was the “ImageInputLayer”, and the last one the “ClassificationOutputLayer” (Figure 7). Parameters are shown in Table 1.
The code that was used is shown in Appendix Table A4
In the analysis setup, the image patches of the 3 diagnoses were pooled in 3 different folders. Later, the content of each folder was split into training, validation, and test sets. As a result, no image patches were repeated in different folders. Nonetheless, some researchers think that this strategy could create an information leak. Therefore, an additional and independent test set of 10 cases of colorectal cancer (adenocarcinoma) were classified to confirm the performance of the trained CNN. In this analysis, each patient was analyzed independently [75].
Of note, other types of CNNs were tested in this study. LAIR1 and TOX2 biomarkers were included into the CNN training in an independent analysis from the H&E staining. A differentiation between steroid-requiring (SR) and mesalazine-responsive ulcerative colitis using LAIR1 and TOX2 immunohistochemistry.
The performance parameters were the following: accuracy, precision, recall, F1-Score, specificity, and false positive rate.
The performance of ResNet-18 was compared with the one of other CNNs under same experimental conditions, including DenseNet-201, ResNet-50, Inception-v3, ResNet-101, ShuffleNet, MobileNet-v2, NasNet-Large, GoogLeNet-Places365, VGG-19, EfficientNet-b0, AlexNet, Xception, VGG-16, GoogLeNet, and NasNet-Mobile.

3.6. Computational Requirements

All analyses were performed using a desktop computer equipped with an AMD Ryzen 9 7950X CPU, 128 Gb of RAM (Crucial Desktop DDR5-4800 UDIMM 1.1V CL40, CT2K32G48C40U5 x2), and an Nvidia GeForce RTX 4090 graphics card (ASUS ROG Strix GeForce RTX® 4090 OC Edition 24GB GDDR6X).

Supplementary Materials

The following supporting information can be downloaded at the website of this paper posted on Preprints.org. Image_patches_examples_datav2-3439728.

Author Contributions

Conceptualization, J.C.; methodology, J.C., G.R. and R.H.; software, J.C.; formal analysis, J.C.; primary antibodies, J.C. and G.R.; writing—original draft preparation, J.C.; writing—review, J.C. and R.H.; funding acquisition, J.C. and R.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Education, Culture, Sports, Science, and Technology of Japan; and KAKEN grants 23K06454, 18K15100, and 15K19061. R.H. is funded by the University of Sharjah (grant no: 24010902153) and ASPIRE, the technology program management pillar of Abu Dhabi’s Advanced Technology Research Council (ATRC), via the ASPIRE Precision Medicine Research Institute Abu Dhabi (VRI-20–10).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board (Ethics Committee) of TOKAI UNIVERSITY, SCHOOL OF MEDICINE (protocol code IRB14R-080, IRB20-156, and 13R-119).

Informed Consent Statement

Informed consent was obtained from all participants.

Data Availability Statement

All data, including methodology, are available upon request to Joaquim Carreras (joaquim.carreras@tokai.ac.jp). Data is located at Zenodo CERN and OpenAIRE Open Science repository: Carreras, J. (2024). Image dataset (Version 1) [Data set]. Zenodo. https://doi.org/10.5281/zenodo.14429385. The samples are anonymized and all metadata are erased. The data is under license Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) (Website: https://creativecommons.org/licenses/by-nc-nd/4.0/deed.en; last accessed on December 30, 2024).

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Endoscopic Baron score.
Table A1. Endoscopic Baron score.
0 Normal: matte mucosa, ramifying vascular pattern clearly visible, no spontaneous bleeding, no bleeding to light touch.
1 Abnormal, but non-hemorrhagic: appearance between 0 and 2.
2 Moderately hemorrhagic: bleeding to light touch, but no spontaneous bleeding ahead of the instrument on initial inspection
3 Severely hemorrhagic: spontaneous bleeding ahead of instrument at initial inspection and bleeding to light touch
Table A2. Histologic Geboes score.
Table A2. Histologic Geboes score.
Grade 0 Structural (architectural changes)
Subgrades
0 No abnormality
0.1 Mild abnormality
0.2 Mild or moderate diffuse or multifocal abnormalities
0.3 Severe diffuse or multifocal abnormalities
Grade 1 Chronic inflammatory infiltrate
Subgrades
1 No increase
1.1 Mild but unequivocal increase
1.2 Moderate increase
1.3 Marked increase
Grade 2 Lamina propria neutrophils and eosinophils
2A Eosinophils
2A.0 No increase
2A.1 Mild but unequivocal increase
2A.2 Moderate increase
2A.3 Marked increase
2B Neutrophils
2B.0 No increase
2B.1 Mild but unequivocal increase
2B.2 Moderate increase
2B.3 Marked increase
Grade 3 Neutrophils in epithelium
Subgrades
3.0 None
3.1 < 5% Crypts involves
3.2 < 50% Crypts involves
3.3 > 50% Crypts involves
Grade 4 Crypt destruction
Subgrades
4.0 None
4.1 Probable — local excess of neutrophils in part of crypt
4.2 Probable — marked attenuation
4.3 Unequivocal crypt destruction
Grade 5 Erosion or ulceration
Subgrades
5.0 No erosion, ulceration, or granulation tissue
5.1 Recovering epithelium + adjacent inflammation
5.2 Probable erosion focally stripped
5.3 Unequivocal erosion
5.4 Ulcer or granulation tissue
Table A3. Data normalization.
Table A3. Data normalization.
Data normalization was applied to the input images: imageInputLayer (an image input layer
inputs 2-D images to a neural network and applies data normalization), and
batchNormalizationLayer (a batch normalization layer normalizes a mini-batch of data across all
observations for each channel independently. To speed up training of the convolutional
neural network and reduce the sensitivity to network initialization, batch
normalization layers are used between convolutional layers and nonlinearities, such as
ReLU layers. Layer = batchNormalizationLayer (Name, Value) creates a batch
normalization layer and sets the optional TrainedMean, TrainedVariance, Epsilon, Parameters and
Initialization, Learning Rate and Regularization, and Name properties using one or
more name-value pairs. After normalization, the layer scales the input with a learnable
scale factor γ and shifts it by a learnable offset β) [80,81]
Table A4. Code.
Table A4. Code.
Ⓐ To load training setup data:
trainingSetup = load("…”)
Ⓑ Import data:
imdsTrain = trainingSetup.imdsTrain;
imdsValidation = trainingSetup.imdsValidation;
Ⓒ Resize the images to match the network input layer:
augimdsTrain = augmentedImageDatastore([224 224 3],imdsTrain);
augimdsValidation = augmentedImageDatastore([224 224 3],imdsValidation);
Ⓓ Set training options:
opts = trainingOptions("sgdm",...
  "ExecutionEnvironment","auto",...
  "InitialLearnRate",0.001,...
  "MaxEpochs",5,...
  "Shuffle","every-epoch",...
  "Plots","training-progress",...
  "ValidationData",augimdsValidation);
Ⓔ Create layer graph:
lgraph = layerGraph();
Ⓕ Add layer branches:
tempLayers = [
  imageInputLayer([224 224 3],"Name","data","Normalization","zscore","Mean",trainingSetup.data.Mean,"StandardDeviation",trainingSetup.data.StandardDeviation)
  convolution2dLayer([7 7],64,"Name","conv1","BiasLearnRateFactor",0,"Padding",[3 3 3 3],"Stride",[2 2],"Bias",trainingSetup.conv1.Bias,"Weights",trainingSetup.conv1.Weights)
  batchNormalizationLayer("Name","bn_conv1","Offset",trainingSetup.bn_conv1.Offset,"Scale",trainingSetup.bn_conv1.Scale,"TrainedMean",trainingSetup.bn_conv1.TrainedMean,"TrainedVariance",trainingSetup.bn_conv1.TrainedVariance)
  reluLayer("Name","conv1_relu")
  maxPooling2dLayer([3 3],"Name","pool1","Padding",[1 1 1 1],"Stride",[2 2])];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
  convolution2dLayer([3 3],64,"Name","res2a_branch2a","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res2a_branch2a.Bias,"Weights",trainingSetup.res2a_branch2a.Weights)
  batchNormalizationLayer("Name","bn2a_branch2a","Offset",trainingSetup.bn2a_branch2a.Offset,"Scale",trainingSetup.bn2a_branch2a.Scale,"TrainedMean",trainingSetup.bn2a_branch2a.TrainedMean,"TrainedVariance",trainingSetup.bn2a_branch2a.TrainedVariance)
  reluLayer("Name","res2a_branch2a_relu")
  convolution2dLayer([3 3],64,"Name","res2a_branch2b","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res2a_branch2b.Bias,"Weights",trainingSetup.res2a_branch2b.Weights)
  batchNormalizationLayer("Name","bn2a_branch2b","Offset",trainingSetup.bn2a_branch2b.Offset,"Scale",trainingSetup.bn2a_branch2b.Scale,"TrainedMean",trainingSetup.bn2a_branch2b.TrainedMean,"TrainedVariance",trainingSetup.bn2a_branch2b.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
additionLayer(2,"Name","res2a")
reluLayer("Name","res2a_relu")];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([3 3],64,"Name","res2b_branch2a","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res2b_branch2a.Bias,"Weights",trainingSetup.res2b_branch2a.Weights)
batchNormalizationLayer("Name","bn2b_branch2a","Offset",trainingSetup.bn2b_branch2a.Offset,"Scale",trainingSetup.bn2b_branch2a.Scale,"TrainedMean",trainingSetup.bn2b_branch2a.TrainedMean,"TrainedVariance",trainingSetup.bn2b_branch2a.TrainedVariance)
reluLayer("Name","res2b_branch2a_relu")
convolution2dLayer([3 3],64,"Name","res2b_branch2b","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res2b_branch2b.Bias,"Weights",trainingSetup.res2b_branch2b.Weights)
batchNormalizationLayer("Name","bn2b_branch2b","Offset",trainingSetup.bn2b_branch2b.Offset,"Scale",trainingSetup.bn2b_branch2b.Scale,"TrainedMean",trainingSetup.bn2b_branch2b.TrainedMean,"TrainedVariance",trainingSetup.bn2b_branch2b.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
additionLayer(2,"Name","res2b")
reluLayer("Name","res2b_relu")];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([3 3],128,"Name","res3a_branch2a","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Stride",[2 2],"Bias",trainingSetup.res3a_branch2a.Bias,"Weights",trainingSetup.res3a_branch2a.Weights)
batchNormalizationLayer("Name","bn3a_branch2a","Offset",trainingSetup.bn3a_branch2a.Offset,"Scale",trainingSetup.bn3a_branch2a.Scale,"TrainedMean",trainingSetup.bn3a_branch2a.TrainedMean,"TrainedVariance",trainingSetup.bn3a_branch2a.TrainedVariance)
reluLayer("Name","res3a_branch2a_relu")
convolution2dLayer([3 3],128,"Name","res3a_branch2b","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res3a_branch2b.Bias,"Weights",trainingSetup.res3a_branch2b.Weights)
batchNormalizationLayer("Name","bn3a_branch2b","Offset",trainingSetup.bn3a_branch2b.Offset,"Scale",trainingSetup.bn3a_branch2b.Scale,"TrainedMean",trainingSetup.bn3a_branch2b.TrainedMean,"TrainedVariance",trainingSetup.bn3a_branch2b.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([1 1],128,"Name","res3a_branch1","BiasLearnRateFactor",0,"Stride",[2 2],"Bias",trainingSetup.res3a_branch1.Bias,"Weights",trainingSetup.res3a_branch1.Weights)
batchNormalizationLayer("Name","bn3a_branch1","Offset",trainingSetup.bn3a_branch1.Offset,"Scale",trainingSetup.bn3a_branch1.Scale,"TrainedMean",trainingSetup.bn3a_branch1.TrainedMean,"TrainedVariance",trainingSetup.bn3a_branch1.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
additionLayer(2,"Name","res3a")
reluLayer("Name","res3a_relu")];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([3 3],128,"Name","res3b_branch2a","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res3b_branch2a.Bias,"Weights",trainingSetup.res3b_branch2a.Weights)
batchNormalizationLayer("Name","bn3b_branch2a","Offset",trainingSetup.bn3b_branch2a.Offset,"Scale",trainingSetup.bn3b_branch2a.Scale,"TrainedMean",trainingSetup.bn3b_branch2a.TrainedMean,"TrainedVariance",trainingSetup.bn3b_branch2a.TrainedVariance)
reluLayer("Name","res3b_branch2a_relu")
convolution2dLayer([3 3],128,"Name","res3b_branch2b","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res3b_branch2b.Bias,"Weights",trainingSetup.res3b_branch2b.Weights)
batchNormalizationLayer("Name","bn3b_branch2b","Offset",trainingSetup.bn3b_branch2b.Offset,"Scale",trainingSetup.bn3b_branch2b.Scale,"TrainedMean",trainingSetup.bn3b_branch2b.TrainedMean,"TrainedVariance",trainingSetup.bn3b_branch2b.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
additionLayer(2,"Name","res3b")
reluLayer("Name","res3b_relu")];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([3 3],256,"Name","res4a_branch2a","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Stride",[2 2],"Bias",trainingSetup.res4a_branch2a.Bias,"Weights",trainingSetup.res4a_branch2a.Weights)
batchNormalizationLayer("Name","bn4a_branch2a","Offset",trainingSetup.bn4a_branch2a.Offset,"Scale",trainingSetup.bn4a_branch2a.Scale,"TrainedMean",trainingSetup.bn4a_branch2a.TrainedMean,"TrainedVariance",trainingSetup.bn4a_branch2a.TrainedVariance)
reluLayer("Name","res4a_branch2a_relu")
convolution2dLayer([3 3],256,"Name","res4a_branch2b","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res4a_branch2b.Bias,"Weights",trainingSetup.res4a_branch2b.Weights)
batchNormalizationLayer("Name","bn4a_branch2b","Offset",trainingSetup.bn4a_branch2b.Offset,"Scale",trainingSetup.bn4a_branch2b.Scale,"TrainedMean",trainingSetup.bn4a_branch2b.TrainedMean,"TrainedVariance",trainingSetup.bn4a_branch2b.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([1 1],256,"Name","res4a_branch1","BiasLearnRateFactor",0,"Stride",[2 2],"Bias",trainingSetup.res4a_branch1.Bias,"Weights",trainingSetup.res4a_branch1.Weights)
batchNormalizationLayer("Name","bn4a_branch1","Offset",trainingSetup.bn4a_branch1.Offset,"Scale",trainingSetup.bn4a_branch1.Scale,"TrainedMean",trainingSetup.bn4a_branch1.TrainedMean,"TrainedVariance",trainingSetup.bn4a_branch1.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
additionLayer(2,"Name","res4a")
reluLayer("Name","res4a_relu")];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([3 3],256,"Name","res4b_branch2a","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res4b_branch2a.Bias,"Weights",trainingSetup.res4b_branch2a.Weights)
batchNormalizationLayer("Name","bn4b_branch2a","Offset",trainingSetup.bn4b_branch2a.Offset,"Scale",trainingSetup.bn4b_branch2a.Scale,"TrainedMean",trainingSetup.bn4b_branch2a.TrainedMean,"TrainedVariance",trainingSetup.bn4b_branch2a.TrainedVariance)
reluLayer("Name","res4b_branch2a_relu")
convolution2dLayer([3 3],256,"Name","res4b_branch2b","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res4b_branch2b.Bias,"Weights",trainingSetup.res4b_branch2b.Weights)
batchNormalizationLayer("Name","bn4b_branch2b","Offset",trainingSetup.bn4b_branch2b.Offset,"Scale",trainingSetup.bn4b_branch2b.Scale,"TrainedMean",trainingSetup.bn4b_branch2b.TrainedMean,"TrainedVariance",trainingSetup.bn4b_branch2b.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
additionLayer(2,"Name","res4b")
reluLayer("Name","res4b_relu")];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([3 3],512,"Name","res5a_branch2a","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Stride",[2 2],"Bias",trainingSetup.res5a_branch2a.Bias,"Weights",trainingSetup.res5a_branch2a.Weights)
batchNormalizationLayer("Name","bn5a_branch2a","Offset",trainingSetup.bn5a_branch2a.Offset,"Scale",trainingSetup.bn5a_branch2a.Scale,"TrainedMean",trainingSetup.bn5a_branch2a.TrainedMean,"TrainedVariance",trainingSetup.bn5a_branch2a.TrainedVariance)
reluLayer("Name","res5a_branch2a_relu")
convolution2dLayer([3 3],512,"Name","res5a_branch2b","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res5a_branch2b.Bias,"Weights",trainingSetup.res5a_branch2b.Weights)
batchNormalizationLayer("Name","bn5a_branch2b","Offset",trainingSetup.bn5a_branch2b.Offset,"Scale",trainingSetup.bn5a_branch2b.Scale,"TrainedMean",trainingSetup.bn5a_branch2b.TrainedMean,"TrainedVariance",trainingSetup.bn5a_branch2b.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([1 1],512,"Name","res5a_branch1","BiasLearnRateFactor",0,"Stride",[2 2],"Bias",trainingSetup.res5a_branch1.Bias,"Weights",trainingSetup.res5a_branch1.Weights)
batchNormalizationLayer("Name","bn5a_branch1","Offset",trainingSetup.bn5a_branch1.Offset,"Scale",trainingSetup.bn5a_branch1.Scale,"TrainedMean",trainingSetup.bn5a_branch1.TrainedMean,"TrainedVariance",trainingSetup.bn5a_branch1.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
additionLayer(2,"Name","res5a")
reluLayer("Name","res5a_relu")];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
convolution2dLayer([3 3],512,"Name","res5b_branch2a","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res5b_branch2a.Bias,"Weights",trainingSetup.res5b_branch2a.Weights)
batchNormalizationLayer("Name","bn5b_branch2a","Offset",trainingSetup.bn5b_branch2a.Offset,"Scale",trainingSetup.bn5b_branch2a.Scale,"TrainedMean",trainingSetup.bn5b_branch2a.TrainedMean,"TrainedVariance",trainingSetup.bn5b_branch2a.TrainedVariance)
reluLayer("Name","res5b_branch2a_relu")
convolution2dLayer([3 3],512,"Name","res5b_branch2b","BiasLearnRateFactor",0,"Padding",[1 1 1 1],"Bias",trainingSetup.res5b_branch2b.Bias,"Weights",trainingSetup.res5b_branch2b.Weights)
batchNormalizationLayer("Name","bn5b_branch2b","Offset",trainingSetup.bn5b_branch2b.Offset,"Scale",trainingSetup.bn5b_branch2b.Scale,"TrainedMean",trainingSetup.bn5b_branch2b.TrainedMean,"TrainedVariance",trainingSetup.bn5b_branch2b.TrainedVariance)];
lgraph = addLayers(lgraph,tempLayers);
tempLayers = [
additionLayer(2,"Name","res5b")
reluLayer("Name","res5b_relu")
globalAveragePooling2dLayer("Name","pool5")
fullyConnectedLayer(3,"Name","fc")
softmaxLayer("Name","prob")
classificationLayer("Name","classoutput")];
lgraph = addLayers(lgraph,tempLayers);
Ⓖ Clean up helper variable:
clear tempLayers;
Ⓗ Connect layer branches:
lgraph = connectLayers(lgraph,"pool1","res2a_branch2a");
lgraph = connectLayers(lgraph,"pool1","res2a/in2");
lgraph = connectLayers(lgraph,"bn2a_branch2b","res2a/in1");
lgraph = connectLayers(lgraph,"res2a_relu","res2b_branch2a");
lgraph = connectLayers(lgraph,"res2a_relu","res2b/in2");
lgraph = connectLayers(lgraph,"bn2b_branch2b","res2b/in1");
lgraph = connectLayers(lgraph,"res2b_relu","res3a_branch2a");
lgraph = connectLayers(lgraph,"res2b_relu","res3a_branch1");
lgraph = connectLayers(lgraph,"bn3a_branch2b","res3a/in1");
lgraph = connectLayers(lgraph,"bn3a_branch1","res3a/in2");
lgraph = connectLayers(lgraph,"res3a_relu","res3b_branch2a");
lgraph = connectLayers(lgraph,"res3a_relu","res3b/in2");
lgraph = connectLayers(lgraph,"bn3b_branch2b","res3b/in1");
lgraph = connectLayers(lgraph,"res3b_relu","res4a_branch2a");
lgraph = connectLayers(lgraph,"res3b_relu","res4a_branch1");
lgraph = connectLayers(lgraph,"bn4a_branch2b","res4a/in1");
lgraph = connectLayers(lgraph,"bn4a_branch1","res4a/in2");
lgraph = connectLayers(lgraph,"res4a_relu","res4b_branch2a");
lgraph = connectLayers(lgraph,"res4a_relu","res4b/in2");
lgraph = connectLayers(lgraph,"bn4b_branch2b","res4b/in1");
lgraph = connectLayers(lgraph,"res4b_relu","res5a_branch2a");
lgraph = connectLayers(lgraph,"res4b_relu","res5a_branch1");
lgraph = connectLayers(lgraph,"bn5a_branch2b","res5a/in1");
lgraph = connectLayers(lgraph,"bn5a_branch1","res5a/in2");
lgraph = connectLayers(lgraph,"res5a_relu","res5b_branch2a");
lgraph = connectLayers(lgraph,"res5a_relu","res5b/in2");
lgraph = connectLayers(lgraph,"bn5b_branch2b","res5b/in1");
Ⓘ Train network:
[net, traininfo] = trainNetwork(augimdsTrain,lgraph,opts);
Figure A1. This figure shows the H&E staining of 4 cases of endoscopic biopsies of ulcerative colitis. The area of interest for AI analysis is delineated in yellow.
Figure A1. This figure shows the H&E staining of 4 cases of endoscopic biopsies of ulcerative colitis. The area of interest for AI analysis is delineated in yellow.
Preprints 150152 g0a1
Figure B1. Immunohistochemistry of TOX2 and LAIR1 in reactive tonsil and colonic mucosa control. TOX2 (left column) is compatible with the staining pattern of PD-1 and in the germinal centers identified follicular T helper cells. TOX2-positive cells (right column) were also identified in the lamina propria of the mucosa. LAIR1 is an inhibitory receptor found on peripheral mononuclear cells, including natural killer cells, T cells, and B cells. LAIR1 had a pattern compatible with macrophages/dendritic cells in the germinal centers and the interfollicular areas. Additionally, the mantle zones that include naïve B lymphocytes, were also positive for LAIR1. In the lamina propria of colonic mucosa, LAIR1 marked cells with macrophage/dendritic cells morphology.
Figure B1. Immunohistochemistry of TOX2 and LAIR1 in reactive tonsil and colonic mucosa control. TOX2 (left column) is compatible with the staining pattern of PD-1 and in the germinal centers identified follicular T helper cells. TOX2-positive cells (right column) were also identified in the lamina propria of the mucosa. LAIR1 is an inhibitory receptor found on peripheral mononuclear cells, including natural killer cells, T cells, and B cells. LAIR1 had a pattern compatible with macrophages/dendritic cells in the germinal centers and the interfollicular areas. Additionally, the mantle zones that include naïve B lymphocytes, were also positive for LAIR1. In the lamina propria of colonic mucosa, LAIR1 marked cells with macrophage/dendritic cells morphology.
Preprints 150152 g0b1
Figure C1. Immunohistochemistry of TOX2 and LAIR1 in ulcerative colitis. This figure shows the staining of TOX2 and LAIR1 in the mucosa of 4 biopsies. The cases had a high infiltration of TOX2-positive and LAIR1-positive cells in the lamina propria.
Figure C1. Immunohistochemistry of TOX2 and LAIR1 in ulcerative colitis. This figure shows the staining of TOX2 and LAIR1 in the mucosa of 4 biopsies. The cases had a high infiltration of TOX2-positive and LAIR1-positive cells in the lamina propria.
Preprints 150152 g0c1

References

  1. Hodson, R. Inflammatory bowel disease. Nature 2016, 540, S97. [CrossRef]
  2. Sairenji, T.; Collins, K.L.; Evans, D.V. An Update on Inflammatory Bowel Disease. Prim Care 2017, 44, 673-692. [CrossRef]
  3. Bruner, L.P.; White, A.M.; Proksell, S. Inflammatory Bowel Disease. Prim Care 2023, 50, 411-427. [CrossRef]
  4. Din, S.; Wong, K.; Mueller, M.F.; Oniscu, A.; Hewinson, J.; Black, C.J.; Miller, M.L.; Jimenez-Sanchez, A.; Rabbie, R.; Rashid, M.; et al. Mutational Analysis Identifies Therapeutic Biomarkers in Inflammatory Bowel Disease-Associated Colorectal Cancers. Clin Cancer Res 2018, 24, 5133-5142. [CrossRef]
  5. Halliday, G.; Porter, R.J.; Black, C.J.; Arends, M.J.; Din, S. c-MET immunohistochemical expression in sporadic and inflammatory bowel disease associated lesions. World J Gastroenterol 2022, 28, 1338-1346. [CrossRef]
  6. Hemmer, A.; Forest, K.; Rath, J.; Bowman, J. Inflammatory Bowel Disease: A Concise Review. S D Med 2023, 76, 416-423.
  7. Khor, B.; Gardet, A.; Xavier, R.J. Genetics and pathogenesis of inflammatory bowel disease. Nature 2011, 474, 307-317. [CrossRef]
  8. Porter, R.J.; Arends, M.J.; Churchhouse, A.M.D.; Din, S. Inflammatory Bowel Disease-Associated Colorectal Cancer: Translational Risks from Mechanisms to Medicines. J Crohns Colitis 2021, 15, 2131-2141. [CrossRef]
  9. Zhang, Y.Z.; Li, Y.Y. Inflammatory bowel disease: pathogenesis. World J Gastroenterol 2014, 20, 91-99. [CrossRef]
  10. Singh, N.; Bernstein, C.N. Environmental risk factors for inflammatory bowel disease. United European Gastroenterol J 2022, 10, 1047-1053. [CrossRef]
  11. Qiu, P.; Ishimoto, T.; Fu, L.; Zhang, J.; Zhang, Z.; Liu, Y. The Gut Microbiota in Inflammatory Bowel Disease. Front Cell Infect Microbiol 2022, 12, 733992. [CrossRef]
  12. Jarmakiewicz-Czaja, S.; Zielinska, M.; Sokal, A.; Filip, R. Genetic and Epigenetic Etiology of Inflammatory Bowel Disease: An Update. Genes (Basel) 2022, 13. [CrossRef]
  13. Graham, D.B.; Xavier, R.J. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 2020, 578, 527-539. [CrossRef]
  14. Saez, A.; Gomez-Bris, R.; Herrero-Fernandez, B.; Mingorance, C.; Rius, C.; Gonzalez-Granado, J.M. Innate Lymphoid Cells in Intestinal Homeostasis and Inflammatory Bowel Disease. Int J Mol Sci 2021, 22. [CrossRef]
  15. Saez, A.; Herrero-Fernandez, B.; Gomez-Bris, R.; Sanchez-Martinez, H.; Gonzalez-Granado, J.M. Pathophysiology of Inflammatory Bowel Disease: Innate Immune System. Int J Mol Sci 2023, 24. [CrossRef]
  16. Lu, Q.; Yang, M.F.; Liang, Y.J.; Xu, J.; Xu, H.M.; Nie, Y.Q.; Wang, L.S.; Yao, J.; Li, D.F. Immunology of Inflammatory Bowel Disease: Molecular Mechanisms and Therapeutics. J Inflamm Res 2022, 15, 1825-1844. [CrossRef]
  17. Kaplan, G.G.; Windsor, J.W. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2021, 18, 56-66. [CrossRef]
  18. Agrawal, M.; Jess, T. Implications of the changing epidemiology of inflammatory bowel disease in a changing world. United European Gastroenterol J 2022, 10, 1113-1120. [CrossRef]
  19. Narula, N.; Wong, E.C.L.; Dehghan, M.; Mente, A.; Rangarajan, S.; Lanas, F.; Lopez-Jaramillo, P.; Rohatgi, P.; Lakshmi, P.V.M.; Varma, R.P.; et al. Association of ultra-processed food intake with risk of inflammatory bowel disease: prospective cohort study. BMJ 2021, 374, n1554. [CrossRef]
  20. Burisch, J.; Zhao, M.; Odes, S.; De Cruz, P.; Vermeire, S.; Bernstein, C.N.; Kaplan, G.G.; Duricova, D.; Greenberg, D.; Melberg, H.O.; et al. The cost of inflammatory bowel disease in high-income settings: a Lancet Gastroenterology & Hepatology Commission. Lancet Gastroenterol Hepatol 2023, 8, 458-492. [CrossRef]
  21. Buie, M.J.; Quan, J.; Windsor, J.W.; Coward, S.; Hansen, T.M.; King, J.A.; Kotze, P.G.; Gearry, R.B.; Ng, S.C.; Mak, J.W.Y.; et al. Global Hospitalization Trends for Crohn's Disease and Ulcerative Colitis in the 21st Century: A Systematic Review With Temporal Analyses. Clin Gastroenterol Hepatol 2023, 21, 2211-2221. [CrossRef]
  22. Wijnands, A.M.; Elias, S.G.; Dekker, E.; Fidder, H.H.; Hoentjen, F.; Ten Hove, J.R.; Maljaars, P.W.J.; van der Meulen-de Jong, A.E.; Mooiweer, E.; Ouwehand, R.J.; et al. Smoking and colorectal neoplasia in patients with inflammatory bowel disease: Dose-effect relationship. United European Gastroenterol J 2023, 11, 612-620. [CrossRef]
  23. Ham, N.S.; Hwang, S.W.; Oh, E.H.; Kim, J.; Lee, H.S.; Park, S.H.; Yang, D.H.; Ye, B.D.; Byeon, J.S.; Myung, S.J.; et al. Influence of Severe Vitamin D Deficiency on the Clinical Course of Inflammatory Bowel Disease. Dig Dis Sci 2021, 66, 587-596. [CrossRef]
  24. Mark A Peppercorn, Adam S Cheifetz. Definitions, epidemiology, and risk factors for inflammatory bowel disease. In: UpToDate, Sunanda V Kane (Ed), Wolters Kluwer. (Accessed on December 11, 2024.).
  25. Ungaro, R.; Mehandru, S.; Allen, P.B.; Peyrin-Biroulet, L.; Colombel, J.F. Ulcerative colitis. Lancet 2017, 389, 1756-1770. [CrossRef]
  26. Gomez-Bris, R.; Saez, A.; Herrero-Fernandez, B.; Rius, C.; Sanchez-Martinez, H.; Gonzalez-Granado, J.M. CD4 T-Cell Subsets and the Pathophysiology of Inflammatory Bowel Disease. Int J Mol Sci 2023, 24. [CrossRef]
  27. Cui, G.; Yuan, A.; Sorbye, S.W.; Florholmen, J. Th9 and Th17 Cells in Human Ulcerative Colitis-Associated Dysplastic Lesions. Clin Med Insights Oncol 2024, 18, 11795549241301358. [CrossRef]
  28. Gerlach, K.; Lechner, K.; Popp, V.; Offensperger, L.; Zundler, S.; Wiendl, M.; Becker, E.; Atreya, R.; Rath, T.; Neurath, M.F.; et al. The JAK1/3 inhibitor tofacitinib suppresses T cell homing and activation in chronic intestinal inflammation. J Crohns Colitis 2020. [CrossRef]
  29. Mitsialis, V.; Wall, S.; Liu, P.; Ordovas-Montanes, J.; Parmet, T.; Vukovic, M.; Spencer, D.; Field, M.; McCourt, C.; Toothaker, J.; et al. Single-Cell Analyses of Colon and Blood Reveal Distinct Immune Cell Signatures of Ulcerative Colitis and Crohn's Disease. Gastroenterology 2020, 159, 591-608 e510. [CrossRef]
  30. Shmuel-Galia, L.; Humphries, F.; Lei, X.; Ceglia, S.; Wilson, R.; Jiang, Z.; Ketelut-Carneiro, N.; Foley, S.E.; Pechhold, S.; Houghton, J.; et al. Dysbiosis exacerbates colitis by promoting ubiquitination and accumulation of the innate immune adaptor STING in myeloid cells. Immunity 2021, 54, 1137-1153 e1138. [CrossRef]
  31. DeGruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current Understanding of Dysbiosis in Disease in Human and Animal Models. Inflamm Bowel Dis 2016, 22, 1137-1150. [CrossRef]
  32. Schroeder, K.W.; Tremaine, W.J.; Ilstrup, D.M. Coated oral 5-aminosalicylic acid therapy for mildly to moderately active ulcerative colitis. A randomized study. N Engl J Med 1987, 317, 1625-1629. [CrossRef]
  33. Silverberg, M.S.; Satsangi, J.; Ahmad, T.; Arnott, I.D.; Bernstein, C.N.; Brant, S.R.; Caprilli, R.; Colombel, J.F.; Gasche, C.; Geboes, K.; et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Can J Gastroenterol 2005, 19 Suppl A, 5A-36A. [CrossRef]
  34. Truelove, S.C.; Witts, L.J. Cortisone in ulcerative colitis; final report on a therapeutic trial. Br Med J 1955, 2, 1041-1048. [CrossRef]
  35. Geboes, K.; Riddell, R.; Ost, A.; Jensfelt, B.; Persson, T.; Lofberg, R. A reproducible grading scale for histological assessment of inflammation in ulcerative colitis. Gut 2000, 47, 404-409. [CrossRef]
  36. Fabian, O.; Kamaradova, K. Morphology of inflammatory bowel diseases (IBD). Cesk Patol 2022, 58, 27-37.
  37. Feakins, R.M. Ulcerative colitis or Crohn's disease? Pitfalls and problems. Histopathology 2014, 64, 317-335. [CrossRef]
  38. El-Zimaity, H.; Shaffer, S.R.; Riddell, R.H.; Pai, R.K.; Bernstein, C.N. Beyond Neutrophils for Predicting Relapse and Remission in Ulcerative Colitis. J Crohns Colitis 2023, 17, 767-776. [CrossRef]
  39. Gupta, R.B.; Harpaz, N.; Itzkowitz, S.; Hossain, S.; Matula, S.; Kornbluth, A.; Bodian, C.; Ullman, T. Histologic inflammation is a risk factor for progression to colorectal neoplasia in ulcerative colitis: a cohort study. Gastroenterology 2007, 133, 1099-1105; quiz 1340-1091. [CrossRef]
  40. Gros, B.; Kaplan, G.G. Ulcerative Colitis in Adults: A Review. JAMA 2023, 330, 951-965. [CrossRef]
  41. Russell D Cohen, Adam C Stein. Management of moderate to severe ulcerative colitis in adults. In: UpToDate, Sunanda V Kane (Ed), Wolters Kluwer. (Accessed on December, 2024.).
  42. Jana Al Hashash, Miguel Regueiro.Medical management of low-risk adult patients with mild to moderate ulcerative colitis. In: UpToDate, Kristen M Robson (Ed), Wolters Kluwer. (Accessed on December 11, 2024.).
  43. Le Berre, C.; Honap, S.; Peyrin-Biroulet, L. Ulcerative colitis. Lancet 2023, 402, 571-584. [CrossRef]
  44. Marshall, D.A.; MacDonald, K.V.; Kao, D.; Bernstein, C.N.; Kaplan, G.G.; Jijon, H.; Hazlewood, G.; Panaccione, R.; Nasser, Y.; Raman, M.; et al. Patient preferences for active ulcerative colitis treatments and fecal microbiota transplantation. Ther Adv Chronic Dis 2024, 15, 20406223241239168. [CrossRef]
  45. Global Cancer Observatory. International Agency for Research on Cancer. World Health Organization. Available at: https://gco.iarc.fr/ (Accessed on December 13, 2023). Available online: (accessed on.
  46. Finlay A Macrae. Epidemiology and risk factors for colorectal cancer. In: UpToDate, Richard M Goldberg, David Seres (Eds), Wolters Kluwer. (Accessed on December 12, 2024.).
  47. Baidoun, F.; Elshiwy, K.; Elkeraie, Y.; Merjaneh, Z.; Khoudari, G.; Sarmini, M.T.; Gad, M.; Al-Husseini, M.; Saad, A. Colorectal Cancer Epidemiology: Recent Trends and Impact on Outcomes. Curr Drug Targets 2021, 22, 998-1009. [CrossRef]
  48. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467-1480. [CrossRef]
  49. Patel, S.G.; Karlitz, J.J.; Yen, T.; Lieu, C.H.; Boland, C.R. The rising tide of early-onset colorectal cancer: a comprehensive review of epidemiology, clinical features, biology, risk factors, prevention, and early detection. Lancet Gastroenterol Hepatol 2022, 7, 262-274. [CrossRef]
  50. Sullivan, B.A.; Noujaim, M.; Roper, J. Cause, Epidemiology, and Histology of Polyps and Pathways to Colorectal Cancer. Gastrointest Endosc Clin N Am 2022, 32, 177-194. [CrossRef]
  51. Yokoyama, S.; Watanabe, T.; Fujita, Y.; Matsumura, S.; Ueda, K.; Nagano, S.; Kinoshita, I.; Murakami, D.; Tabata, H.; Tsuji, T.; et al. Histology of metastatic colorectal cancer in a lymph node. PLoS One 2023, 18, e0284536. [CrossRef]
  52. Nagtegaal, I.D.; Hugen, N. The Increasing Relevance of Tumour Histology in Determining Oncological Outcomes in Colorectal Cancer. Curr Colorectal Cancer Rep 2015, 11, 259-266. [CrossRef]
  53. Gulsoy, T.; Baykal Kablan, E. FocalNeXt: A ConvNeXt augmented FocalNet architecture for lung cancer classification from CT-scan images. Expert Systems with Applications 2025, 261. [CrossRef]
  54. Taatjes, D.J.; Bouffard, N.A.; Barrow, T.; Devitt, K.A.; Gardner, J.A.; Braet, F. Quantitative pixel intensity- and color-based image analysis on minimally compressed files: implications for whole-slide imaging. Histochem Cell Biol 2019, 152, 13-23. [CrossRef]
  55. Hofener, H.; Homeyer, A.; Weiss, N.; Molin, J.; Lundstrom, C.F.; Hahn, H.K. Deep learning nuclei detection: A simple approach can deliver state-of-the-art results. Comput Med Imaging Graph 2018, 70, 43-52. [CrossRef]
  56. MathWorks. MATLAB for Artificial Intelligence. Design AI models and AI-driven systems. Website: https://www.mathworks.com/ (Accessed on December 12, 2024). Available online: (accessed on.
  57. Ewaeed, N.A.; Abed, H.N.; Abed, S.N. Detecting and Classifying Household Insects in Iraq by using Transfer Learning Models. Journal of Advanced Research in Applied Sciences and Engineering Technology 2025, 50, 21-33. [CrossRef]
  58. Xu, H.; Usuyama, N.; Bagga, J.; Zhang, S.; Rao, R.; Naumann, T.; Wong, C.; Gero, Z.; Gonzalez, J.; Gu, Y.; et al. A whole-slide foundation model for digital pathology from real-world data. Nature 2024, 630, 181-188. [CrossRef]
  59. Das, N.; Das, S. Attention-UNet architectures with pretrained backbones for multi-class cardiac MR image segmentation. Curr Probl Cardiol 2024, 49, 102129. [CrossRef]
  60. Jiang, X.; Hu, Z.; Wang, S.; Zhang, Y. Deep Learning for Medical Image-Based Cancer Diagnosis. Cancers (Basel) 2023, 15. [CrossRef]
  61. Weller, J.H.; Scheese, D.; Tragesser, C.; Yi, P.H.; Alaish, S.M.; Hackam, D.J. Artificial Intelligence vs. Doctors: Diagnosing Necrotizing Enterocolitis on Abdominal Radiographs. J Pediatr Surg 2024, 59, 161592. [CrossRef]
  62. Khan, H.A.; Jue, W.; Mushtaq, M.; Mushtaq, M.U. Brain tumor classification in MRI image using convolutional neural network. Math Biosci Eng 2020, 17, 6203-6216. [CrossRef]
  63. Karimi, D.; Dou, H.; Gholipour, A. Medical Image Segmentation Using Transformer Networks. IEEE Access 2022, 10, 29322-29332. [CrossRef]
  64. Wang, X.; Yang, S.; Zhang, J.; Wang, M.; Zhang, J.; Yang, W.; Huang, J.; Han, X. Transformer-based unsupervised contrastive learning for histopathological image classification. Med Image Anal 2022, 81, 102559. [CrossRef]
  65. Lama, N.; Kasmi, R.; Hagerty, J.R.; Stanley, R.J.; Young, R.; Miinch, J.; Nepal, J.; Nambisan, A.; Stoecker, W.V. ChimeraNet: U-Net for Hair Detection in Dermoscopic Skin Lesion Images. J Digit Imaging 2023, 36, 526-535. [CrossRef]
  66. TensorFlow. Website: https://www.tensorflow.org/ (Accessed on December 12, 2024). Available online: (accessed on.
  67. TensorFlow Keras Basic image classification. Website: https://www.tensorflow.org/tutorials/keras/classification (Accessed on December 12, 2024). Available online: (accessed on.
  68. PyTorch, get started. Website: https://pytorch.org/ (Accessed on December 12, 2024). Available online: (accessed on.
  69. ONNX, open neural network exchange. Website: https://onnx.ai/ (Accessed on December 12, 2024). Available online: (accessed on.
  70. Kaiming He, Xiangyu Zhang, Shaoqing Ren, Jian Sun. Deep Residual Learning for Image Recognition. arXiv:1512.03385v1.
  71. UniProt, C. UniProt: the Universal Protein Knowledgebase in 2023. Nucleic Acids Res 2023, 51, D523-D531. [CrossRef]
  72. Peng, D.H.; Rodriguez, B.L.; Diao, L.; Chen, L.; Wang, J.; Byers, L.A.; Wei, Y.; Chapman, H.A.; Yamauchi, M.; Behrens, C.; et al. Collagen promotes anti-PD-1/PD-L1 resistance in cancer through LAIR1-dependent CD8(+) T cell exhaustion. Nat Commun 2020, 11, 4520. [CrossRef]
  73. Van Laethem, F.; Donaty, L.; Tchernonog, E.; Lacheretz-Szablewski, V.; Russello, J.; Buthiau, D.; Almeras, M.; Moreaux, J.; Bret, C. LAIR1, an ITIM-Containing Receptor Involved in Immune Disorders and in Hematological Neoplasms. Int J Mol Sci 2022, 23. [CrossRef]
  74. Xu, W.; Zhao, X.; Wang, X.; Feng, H.; Gou, M.; Jin, W.; Wang, X.; Liu, X.; Dong, C. The Transcription Factor Tox2 Drives T Follicular Helper Cell Development via Regulating Chromatin Accessibility. Immunity 2019, 51, 826-839 e825. [CrossRef]
  75. Carreras, J.; Roncador, G.; Hamoudi, R. Ulcerative Colitis, LAIR1 and TOX2 expression and Colorectal Cancer Deep Learning Image Classification Using Convolutional Neural Networks. Preprints 2024, 2024110211. [CrossRef]
  76. Carreras, J.; Kikuti, Y.Y.; Bea, S.; Miyaoka, M.; Hiraiwa, S.; Ikoma, H.; Nagao, R.; Tomita, S.; Martin-Garcia, D.; Salaverria, I.; et al. Clinicopathological characteristics and genomic profile of primary sinonasal tract diffuse large B cell lymphoma (DLBCL) reveals gain at 1q31 and RGS1 encoding protein; high RGS1 immunohistochemical expression associates with poor overall survival in DLBCL not otherwise specified (NOS). Histopathology 2017, 70, 595-621. [CrossRef]
  77. Carreras, J.; Kikuti, Y.Y.; Hiraiwa, S.; Miyaoka, M.; Tomita, S.; Ikoma, H.; Ito, A.; Kondo, Y.; Itoh, J.; Roncador, G.; et al. High PTX3 expression is associated with a poor prognosis in diffuse large B-cell lymphoma. Cancer Sci 2022, 113, 334-348. [CrossRef]
  78. Carreras, J.; Yukie Kikuti, Y.; Miyaoka, M.; Hiraiwa, S.; Tomita, S.; Ikoma, H.; Kondo, Y.; Shiraiwa, S.; Ando, K.; Sato, S.; et al. Genomic Profile and Pathologic Features of Diffuse Large B-Cell Lymphoma Subtype of Methotrexate-associated Lymphoproliferative Disorder in Rheumatoid Arthritis Patients. Am J Surg Pathol 2018, 42, 936-950. [CrossRef]
  79. Carreras, J.; Kikuti, Y.Y.; Roncador, G.; Miyaoka, M.; Hiraiwa, S.; Tomita, S.; Ikoma, H.; Kondo, Y.; Ito, A.; Shiraiwa, S.; et al. High Expression of Caspase-8 Associated with Improved Survival in Diffuse Large B-Cell Lymphoma: Machine Learning and Artificial Neural Networks Analyses. BioMedInformatics 2021, 1, 18-46. [CrossRef]
  80. Carreras, J. Celiac Disease Deep Learning Image Classification Using Convolutional Neural Networks. J. Imaging 2024, 10, 200. [CrossRef]
  81. MathWorks. Batch normalization layer. Available online: https://www.mathworks.com/help/deeplearning/ref/nnet.cnn.layer.batchnormalizationlayer.html (accessed on 28 July 2024).
Figure 1. Pathogenesis of ulcerative colitis and cells of the immune microenvironment. Ulcerative colitis is a chronic inflammatory condition that affects the colon. The incidence of ulcerative colitis has been increasing in the last decades. The pathogenesis is multifactorial including genetic predisposition and an alteration of the immune tolerance and homeostasis of the mucosa. Several immune cells are involved. This figure shows the immunohistochemical staining of CD3-positive T lymphocytes, CD8-positive cytotoxic T lymphocytes, FOXP3-positive regulatory T lymphocytes (Tregs), PD-1-positive follicular T helper cells (TFH), and CD68-positive macrophages. H&E, hematoxylin and eosin staining.
Figure 1. Pathogenesis of ulcerative colitis and cells of the immune microenvironment. Ulcerative colitis is a chronic inflammatory condition that affects the colon. The incidence of ulcerative colitis has been increasing in the last decades. The pathogenesis is multifactorial including genetic predisposition and an alteration of the immune tolerance and homeostasis of the mucosa. Several immune cells are involved. This figure shows the immunohistochemical staining of CD3-positive T lymphocytes, CD8-positive cytotoxic T lymphocytes, FOXP3-positive regulatory T lymphocytes (Tregs), PD-1-positive follicular T helper cells (TFH), and CD68-positive macrophages. H&E, hematoxylin and eosin staining.
Preprints 150152 g001
Figure 2. Detailed visualization of some of the immune microenvironment components of ulcerative colitis. This figure shows in higher magnification the details of the tissue distribution of some of the immune microenvironment cells in ulcerative colitis. The lamina propria is characterized by a variable infiltration of CD3+T lymphocytes and CD68+macrophages. Within the CD3+T lymphocytes, cytotoxic CD8+T lymphocytes and FOXP3+regulatory T lymphocytes (Tregs) can easily be found. .
Figure 2. Detailed visualization of some of the immune microenvironment components of ulcerative colitis. This figure shows in higher magnification the details of the tissue distribution of some of the immune microenvironment cells in ulcerative colitis. The lamina propria is characterized by a variable infiltration of CD3+T lymphocytes and CD68+macrophages. Within the CD3+T lymphocytes, cytotoxic CD8+T lymphocytes and FOXP3+regulatory T lymphocytes (Tregs) can easily be found. .
Preprints 150152 g002
Figure 3. Histological features of colorectal cancer. The most frequent histological subtype of colorectal cancer is adenocarcinoma.
Figure 3. Histological features of colorectal cancer. The most frequent histological subtype of colorectal cancer is adenocarcinoma.
Preprints 150152 g003
Figure 4. Structure of a convolutional neural network (CNN). The algorithm of a CNN is characterized by taking an input image and assigning weights and biases to the different components. Then, this is deep learning algorithm performs the image classification. The CNN is comprised of 3 main layers: the convolutional, the pooling, and fully connected layers.
Figure 4. Structure of a convolutional neural network (CNN). The algorithm of a CNN is characterized by taking an input image and assigning weights and biases to the different components. Then, this is deep learning algorithm performs the image classification. The CNN is comprised of 3 main layers: the convolutional, the pooling, and fully connected layers.
Preprints 150152 g004
Figure 5. Original ResNet-18 architecture.
Figure 5. Original ResNet-18 architecture.
Preprints 150152 g005
Figure 6. Whole-slide imaging. The glass slides were converted to digital data using a Hamamatsu NanoZoomer S360 scanner that scanned the slides at 400× magnification. .
Figure 6. Whole-slide imaging. The glass slides were converted to digital data using a Hamamatsu NanoZoomer S360 scanner that scanned the slides at 400× magnification. .
Preprints 150152 g006
Figure 7. Image patches of ulcerative colitis, colorectal cancer (adenocarcinoma), and colon control.
Figure 7. Image patches of ulcerative colitis, colorectal cancer (adenocarcinoma), and colon control.
Preprints 150152 g007
Figure 7. Trained network layers. The CNN was designed using a transfer learning strategy and ResNet-18.
Figure 7. Trained network layers. The CNN was designed using a transfer learning strategy and ResNet-18.
Preprints 150152 g008
Table 1. Design and training parameters.
Table 1. Design and training parameters.
ResNet-18-Based CNN Training (70%) Validation (10%) Training Options
Input type: image patches
Output type: classification
Number of layers: 71
Number of connections: 78
Observations: 59,677
Classes: 3
Ulcerative colitis: 6497
Colorectal cancer: 44,608
Colon control: 8572
Observations: 8525
Classes: 3
Ulcerative colitis: 928
Colorectal cancer: 6372
Colon control: 1225
Solver: sgdm
Initial learning rate: 0.001
MiniBatch size: 128
MaxEpochs: 5
Validation frequency: 50
Iterations: 2330
Iterations per epoch: 466
Additional detailed training options
Import images
Augmentation options: none
Available parameters
Random reflection axis: x, y
Random rotation (degrees): min, max
Random rescaling: min, max
Random horizontal translation (pixels): min, max
Random vertical translation (pixels): min, max
Resize during training to match network input size: yes, no
Solver
Momentum: 0.9
Learn rate
LearnRateSchedule: none
LearnRateDropFactor: 0.1
LearnRateDropPeriod: 10
Normalization and Regularization
L2Regularization: 0.0001
ResetInputNormalization: yes
BatchNormalizationStatistics: population
Mini-Batch:
Shuffle: every-epoch
Validation and Output:
ValidationPatience: Inf
OutputNetwork: last-iteration
Gradient Clipping:
GradientThresholdMethod: I2norm
GradientThreshold: Inf
Hardware:
ExecutionThreshold: auto.
Checkpoint
CheckpointPath: n/a
CheckpointFrequency: 1
CheckpointFrequencyUnit: epoch
Based on transfer learning of ResNet-18. Convolutional neural network, CNN.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated