Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 The techniques developed for automatic analysis and evaluation of HIs help the pathologists in objective diagnosis for disease and decreased human error. A reference guide to recent literature methods for analyzing HIs manifests itself in the categorizati + 2990 word(s) 2990 2020-11-24 10:31:41 |
2 format correction -188 word(s) 2802 2020-11-25 09:29:25 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Elazab, N.; Soliman, H.; El-Sappagh, S.; Islam, S.M.R.; Elmogy, M. Objective Diagnosis for Histopathological Images. Encyclopedia. Available online: https://encyclopedia.pub/entry/3199 (accessed on 28 March 2024).
Elazab N, Soliman H, El-Sappagh S, Islam SMR, Elmogy M. Objective Diagnosis for Histopathological Images. Encyclopedia. Available at: https://encyclopedia.pub/entry/3199. Accessed March 28, 2024.
Elazab, Naira, Hassan Soliman, Shaker El-Sappagh, S. M. Riazul Islam, Mohammed Elmogy. "Objective Diagnosis for Histopathological Images" Encyclopedia, https://encyclopedia.pub/entry/3199 (accessed March 28, 2024).
Elazab, N., Soliman, H., El-Sappagh, S., Islam, S.M.R., & Elmogy, M. (2020, November 24). Objective Diagnosis for Histopathological Images. In Encyclopedia. https://encyclopedia.pub/entry/3199
Elazab, Naira, et al. "Objective Diagnosis for Histopathological Images." Encyclopedia. Web. 24 November, 2020.
Objective Diagnosis for Histopathological Images
Edit

Histopathology refers to the examination by a pathologist of biopsy samples. Histopathology images are captured by a microscope to locate, examine, and classify many diseases, such as different cancer types. They provide a detailed view of different types of diseases and their tissue status. These images are an essential resource with which to define biological compositions or analyze cell and tissue structures. This imaging modality is very important for diagnostic applications.
The analysis of histopathology images is a prolific and relevant research area supporting disease diagnosis. In this paper, the challenges of histopathology image analysis are evaluated. An extensive review of conventional and deep learning techniques that have been applied in histological image analyses is presented. This entry summarizes many current datasets and highlights important challenges and constraints with recent deep learning techniques, alongside possible future research avenues. Despite the progress made in this research area so far, it is still a significant area of open research because of the variety of imaging techniques and disease-specific characteristics. 

medical image analysis histopathology image analysis conventional machine learning methods deep learning methods computer-assisted diagnosis

1. Introduction 

Medical Images are a fundamental section of each patient's digital health file. Such images are produced by individual radiologists who are restricted by speed, professional weaknesses, or a lack of practice. It requires decades and reasonable financial resources to train a radiologist. Additionally, some medical care methods outsource radiology confirmations to less economically developed nations, such as India, via teleradiology. A late or incorrect analysis can cause injury to the patient. Thus, it would be beneficial for medical imaging (MI) analyses to be performed by automatic, precise, and effective machine learning (ML) algorithms. MI analysis is a significant research area for ML, in part because the information is somewhat organized and labeled; i.e., this is probable if the patient was examined in a region with good ML systems [1]. That is significant for two reasons. First, with regards to real patient metrics, MI analysis is a litmus check regarding whether ML techniques would, in actuality, improve individual outcomes and survival. Second, it provides a testbed for human–ML interactions—i.e., how responsive is an individual likely to be to the health changing possibilities being put forward or aided by a nonhuman actor [2]. In recent years, ML has shown significant advances. For a wide variety of applications, including image recognition, medical diagnosis, defect identification and construction health assessments, the potential of this field has also expanded. These new developments in ML are due to many factors, like the creation of self-learning mathematical models that enable computer techniques to execute particular (human-like) tasks based solely on learned patterns, in addition to the increase in the computer power that supports these models' analytical capabilities [3].

There are many imaging types, and their use is becoming more widespread. Types of MI include ultrasound, X-ray, magnetic resonance imaging (MRI), retinal scans, histopathology images (HI), computed tomography (CT), positron emission tomography (PET), and dermoscopy images. Some examples of MIs are shown in Figure 1. Many of these types analyze numerous organs, such as CT and MRI, whereas others are organ-specific, such as retinal and dermoscopy images [4]. The quantity of produced information from each analysis stage differs depending on nature of the MI and the tested organs. HIs are useful for biological studies and to make medical decisions. In addition, they are generally utilized to provide "ground truths" (GTs) for other modalities of MI, such as MRI. A histology slide is a digital record a few megabytes in size, while a magnetic resonance image can be several hundred megabytes. This has a technical effect on how the data is preprocessed and on the architecture design of the algorithm in terms of processor and storage limitations [5].

Figure 1. Examples of some medical image types: (a) MRI scan of the left side of a brain; (b) an axial CT brain scan; (c) an axial CT lung scan; (d) chest x-ray; (e) a histology slide with high-grade glioma.

Pathology analyses are traditionally executed by an individual pathologist observing a dyed specimen on a glass slide with a microscope. Lately, efforts have been made to record the whole slide with a reader and save it as an electronic picture, called a whole slide image (WSI) [6].

Digitizing pathology is just one recent development that produces high levels of visible information designed for automated diagnoses. It enables us to see and understand pathologic cell and muscle samples in good quality images with assistance from personal computer tools. It also brings about the possibility of applying image analysis techniques. Such techniques would assist pathologists and support their explanations, such as hosting and grading. Various classification and segmentation methods for HI have already been discussed in this review. We present and compare conventional techniques and deep learning (DL) methods to choose the most appropriate method for histopathology issues [7].

Natural microscopic architecture data and their features at nuclei, tissue, and different organ levels could be key to illness expansion and infection treatment analysis. Additionally, to examine and diagnose the histological image of biologic microscopic, pathologists have identified the morphological features of tissue that show the current presence of infection, such as cancer [8].

Some characteristics of disease, such as tumor-infiltrating lymphocytes, might be deduced from HI alone. Additionally, HI analysis, which is called the "gold standard" in many disease diagnoses, is nearly included in all kinds of cancer detection and treatment procedures. HI needs specific analysis with respect to organs and a specific task for the visualization of various tissue components under a microscope. With one or more stains, the sections are dyed. These are staining attempts to uncover cellular elements. The contrast is shown by using counterstains [9].

Efficient ML algorithms are presented and used in HI analysis to help pathologists to acquire a quick, stable, and quantified examination result for a more accurate diagnosis. Many different traditional and deep learning methods support the pathologists in accessing more tissues to determine the internal relationship between the visual images and the specific illness. Additionally, since the ML techniques are generally semi- or fully automated, they are effective, encouraging technical feasibility for histopathology examination within the recent big data age [10].

On the other hand, most of the HI analysis stages are based on mathematical basics. Mathematical operations and functions are applied to all analysis stages, starting from the preprocessing to diagnosis stages to provide an intensive analysis for HIs. Figure 2 illustrates the main phases of a common histopathological images pipeline based on conventional ML techniques. First, HIs are supplied to the system as a 2D array for grayscale images or a 3D array for colored images. Then, the preprocessing stage applies some linear algebra operations on the image array to enhance the image quality. This stage helps to distinguish significant structures from others in the processed images. Third, the segmentation stage is applied to differentiate the cells from other background objects by applying some state-of-the-art mathematical algorithms, such as thresholding, level set, watershed transform, and intensity and texture homogeneity transforms. Fourth, the feature extraction stage extracts the most significant features in the segmented images instead of processing each pixel, which reduces the system's computation complexity. Besides, most handcrafted features are based on applying some mathematical techniques to detect the changes in the intensity, color, or texture of the pixels. Common derivative techniques are utilized to detect these changes by applying first or second derivatives to pixel values. Finally, the diagnosis stage is applied to classify or cluster the processed images, depending on the extracted features. The classification and clustering techniques are based on applying some mathematical operations that distinguish the processed images based on the extracted features.

Figure 2. An overview of the HI analysis pipeline.

2. Histopathological Image Overview

HI has natural and abnormal biological structures, as well as morphological and architectural features defined by pathologists, based on their knowledge. Even given the tissue area, some structures are small, and related patterns typically have high visual appearance variability. In biological systems and anatomy, most visual variability is inherent [11].

Next to obtaining electronic HI via the biopsy test, the guide analysis of images contributes to variability in diagnosis and treatment. To get over this issue, CAD techniques are applied to provide an objective examination of disease. The fundamental steps necessary for applying the CAD examination system appear in Figure 2. This includes electronic image handling methods, such as segmentation, feature extraction, and classification [12].

HI analysis contains the computations executed at various zoom scales (×2, ×4.5, ×10, ×20, and ×40) for multivariate mathematical examination, analysis, and classification. It could be achieved at a lower zoom for tissue stage examination. Demir et al. [13] presented tissue stage and cell stage examination techniques for cancer diagnosis. They examined HI by applying preprocessing, feature extraction, and classification strategies. The new improvement in electronic pathology requirements for the growth of quantitative and automatic digital image examination methods aids pathologists in understanding the number of digitized HIs [14].

3. Conventional Machine Learning Methods

CAD systems played an essential role and have become an important research topic in HI and diagnostics. Various image processing techniques were applied to examine the disease's diagnosis and prognosis for these HIs. Various image processing and computer vision (CV) techniques have been implemented for gland and nuclei segmentation, cell kind recognition, or classification to extract quantitative measurements of disease characteristics from HIs and automatically assess whether or not a disease exists inside examined samples. It could help to determine the degree of seriousness of the disease, whether present in the sample. Conventional ML methods often contain a few steps to manage HI, as shown in Figure 3.

Mathematics 08 01863 g003 550

Figure 3. The conventional machine learning methods for HI.

4. Deep Learning Methods

Recently, DL techniques have often been studied in the effective form of ML methods. Within the last few years, DL techniques outperformed traditional ML methods in varied fields, such as CV, natural language processing (NLP), biomedical fields, and automated analysis for HI [7]. DL methods in the CV are derived from the structure levels for nonlinear transformations on natural input pixels. This structure formed significantly abstract representations, which could be realized in a hierarchical style [15]. A typical instance of a commonly applied structure is the CNN [16].

Multiple criteria can be considered when using the DL techniques to deal with histopathology, since accomplishing the method is partly due to the task-species setting. Among the principal features of HI is that appropriate styles be determined by the magnification stage. The key factors are the size of the patch given to the network, the localization of parts in the image where appropriate histopathology originals can be found, and the homogeneity of staining for WSI [17]. The network structure represents an important position, while many studies keep predefined system structures, as illustrated in Figure 4.

Figure 4. The typical deep learning steps for HI analysis.

The majority of the DL techniques for localizing, classifying, and segmenting HI are somewhat recent. Deep neural techniques are stated in the new literature of HI analysis, such as [6][13][18]. For example, Irshad et al. [19] were the first mentioned in a review. The critical patterns from an exhaustive analysis of different nuclei identification, segmentation, and classification approaches utilized in HI, specifically in H&E staining protocols, were described and discussed in this review. Ciresan et al. [20] presented one of the first significant efforts to utilize the deep method in mitosis recognition for HI analysis. Arevalo et al. [21] presented a hybrid illustration method to the basal cell of carcinoma areas and utilized a topographic unsupervised technique and a case of characteristic illustrations. They increased the classifier's efficiency by 6% regarding traditional structure-based discrete cosine transform (DCT). Nayak et al. [22] presented an alternative method for the unsupervised Boltzmann technique for understanding image signatures. They classified images of the cancer genome atlas (TCGA) for apparent cell-kidney cancer and glioblastoma variform. The last stage was created utilizing the classifier of multi-class support vector machines (SVM) techniques.

Approaches that rely on Generative Adversarial Networks (GANs) are likely to minimize the need for large volumes of manual notations. Not only have recent innovations enhanced initiatives but so have new technologies. Now, unattended techniques may carry out various tasks for which supervised methods are indispensable. The latest state-of-the-art advances in histopathological images of GANs were summarized in [23]. The overview of the discussed studies is summarized in Table 1.

Table 1. Overview of supervised and unsupervised learning models based on DL techniques.

Study

Organ

Staining

Potential Usage

Method

Supervised Learning

Litjens et al. [24]

different tissue

H&E

Prostate and breast carcinoma detection

Convolutional Neural Network based on pixel classifier

Nagpal et al. [25]

Prostate

H&E

Anticipating Gleason indicator

CNN based on sectional Gleason model classifier + k-nearst neighbors (KNN) based on Gleason grade anticipation

Zhao et al. [26]

Breast

H&E

Metastasis Detection + classification

Characteristic pyramid collecting based on the fully convolutional network (FCN) system with the synergistic training technique

Xing et al. [27]

different tissue

H&E, Immunohistochemistry (IHC)

Segmentation of nuclei

CNN + selection based on sparse form Pattern

Gu et al. [28]

Breast

H&E

Tumor detection

U-Net based on multiple resolution model with multiple encoders and a singular decoder  system

Tellez et al. [29]

Breast

H&E

Detection of Mitosis

Train of Convolutional Network applying H&E registered to PHH3 slides as a reference

Wei et al. [30]

Lung

H&E

Histological subtypes of lung gland classifier

ResNet-18 on the basis of  patch classification

Song et al. [31]

Cervix

Papanicolaou (Pap), H&E

Cells Segmentation

Multiple level CNN system

Agarwalla et al. [32]

Breast

H&E

Segmentation of tumor

CNN and 2D- Long short-term memory (LSTM) to representing training and context collecting

Ding et al. [33]

Colon

H&E

Glands segmentation

Multiple level FCN network with a high-resolution section to avoid the lost in highest pooling layers

Bejnordi et al. [34]

Breast

H&E

Invasive Carcinoma detection

Multiple level CNN which first determines tumor-associated stromal modifications and more categorize into normal/benign versus invasive carcinoma

Seth et al. [35]

Breast

H&E

Ductal carcinoma in-situ (DCIS) segmentation

Compared UNets learned in many resolutions

Unsupervised Learning

Xu et al. [36]

Breast

H&E

Segmentation of nuclei

Stacked sparse autoencoders

Bulten and Litjens [37]

Prostate

H&E

Tumor classification

Convolutional adversarial Autoencoders

Hou et al. [38]

Breast

H&E

Segmentation and detection of nuclei

Sparse autoencoder

Sari and Gunduz-Demir [39]

Colon

H&E

Feature extraction and classification

Restricted Boltzmann + clustering

Gadermayr et al. [40]

Kidney

Stain agnostic

Object of interest segmentation in WSIs

CycleGAN + UNet segmentation

Gadermayr et al. [41]

Kidney

Periodic acid–Schiff (PAS), H&E

Glomeruli segmentation

CycleGAN

5. Datasets

The size of the datasets given to researchers for training and testing their methods has dramatically increased in the latest challenges. There is a set of public databases in the electronic pathology subject that include manual annotations for HI, as listed in Table 2 and Table 3 [39]. They might help the examination objectively. Slide issue (stain) and image issue (image resolution, zoom level) are similar. However, all these databases are targeted to specific diseases. These databases do not handle several tasks. Additionally, there are many high scale HI datasets, which include WSIs of high resolutions.

TCGA [42] includes around 10,000 images from different types of cancer. Genotype-Tissue Expression (GTE) [40] includes around 20,000 WSIs from different tissues. The Stanford Tissue Microarray Database (TMAD) is available for researchers to access images of microarrays for tissue. It provides images of archiving 349 distinguished probes on 1488 microarray slides of tissue [41]. The CAMELYON dataset is a collection of WSI tissues for the sentinel lymph node. It contains CAMELYON16 and CAMELYON17 challenges that include 399 WSI and 1000 WSI, respectively. The data are currently accessed via registration on the CAMELYON17 website [43]. The Breast Cancer Histopathological Image (BreakHis) contains 9109 macroscopic images for the tissue of the breast tumor obtained from 82 patients in various magnifying factors (40X, 100X, 200X). Up to now, it includes samples of 2480 benign and 5429 malignant WSIs [44].

Table 2. Some common downloadable WSI databases.

Table 3. Some publicly available hand-annotated histopathological images.

Datasets

No of Images

Staining

Organs

Potential Usage

KIMIA960 [53][54]

960

H&E/IHC

Different tissue

Classification

Bio-segmentation [55][56]

58

H&E

Breast

Classification

Bioimaging challenge 2015 [57]

269

H&E

Breast

Classification

GlaS [58]

165

H&E

Colorectal

Gland segmentation

BreakHis [59]

7909

H&E

Breast

Classification

Jakob Nikolas et al. [54][60]

100

IHC

Colorectal

Detection of blood vessel

MITOS-ATYPIA-14 [61]

4240

H&E

Breast

Detection of mitosis, classification

Kumar et al. [53][62]

30

H&E

Different cancer

Segmentation of Nuclear

MITOS [63]

100

H&E

Breast

Detection of mitosis

Janowczyk et al. [64][65]

374

H&E

Lymphoma

classification

Janowczyk et al. [64][65]

85

H&E

Colorectal

Segmentation of gland

Ma et al. [66]

81

IHC

Breast

TIL analysis

Linder et al. [67][68]

1377

IHC

Colorectal

Segmentation of epithelium and stroma

References

  1. Litjens, G.; Kooi, T.; Bejnordi, B.E.; Setio, A.A.A.; Ciompi, F.; Ghafoorian, M.; van der Laak, J.A.; van Ginneken, B.; Sanchez, I.C. A survey on deep learning in medical image analysis. Med. Image Anal. 2017, 42, 60–88.
  2. Ker, J.; Wang, L.; Rao, J.; Lim, T. Deep Learning Applications in Medical Image Analysis. IEEE Access 2017, 6, 9375–9389.
  3. Perez, H.; Tah, J. Improving the Accuracy of Convolutional Neural Networks by Identifying and Removing Outlier Images in Datasets Using t-SNE. Mathematics 2020, 8, 662.
  4. Suzuki, K. Overview of deep learning in medical imaging. Radiol. Phys. Technol. 2017, 10, 257–273.
  5. Pantanowitz, L. Digital images and the future of digital pathology. J. Pathol. Inform. 2010, 1.
  6. Gurcan, M.N.; Boucheron, L.E.; Can, A.; Madabhushi, A.; Rajpoot, N.M.; Yener, B. Histopathological image analysis: A review. IEEE Rev. Biomed. Eng. 2009, 2, 147–171.
  7. Greenspan, H.; Ginneken, B.; van Summers, R.M. Guest Editorial Deep Learning in Medical Imaging: Overview and Future Promise of an Exciting New Technique. IEEE Trans Med. Imaging 2016, 35, 1153–1159.
  8. Rubin, R.; Strayer, D.S.; Rubin, E. Rubin’s Pathology: Clinicopathologic Foundations of Medicine; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2008.
  9. Hewitson, T.; Darby, I.; Walker, J. Histology Protocols. In Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2010.
  10. Li, C.; Chen, H.; Li, X.; Xu, N.; Hu, Z.; Xue, D.; Qi, S.; Ma, H.; Zhang, L.; Sun, H. A review for cervical histopathology image analysis using machine vision approaches. Artif. Intell. Rev. 2020, 53, 4821–4862.
  11. He, L.; Long, L.R.; Antani, S.; Thoma, G.R. Histology image analysis for carcinoma detection and grading. Comput. Methods Programs Biomed. 2012, 107, 538–556.
  12. Ghaznavi, F.; Evans, A.; Madabhushi, A.; Feldman, M. Digital Imaging in Pathology: Whole-Slide Imaging and Beyond. Annu. Rev. Pathol. Mech. Dis. 2013, 8, 331–359.
  13. Demir, C.; Yener, B. Automated Cancer Diagnosis Based on Histopathological Images: A Systematic Survey; Technical Report for Rensselaer Polytechnic Institute: New York, NY, USA, 2005.
  14. Belsare, A. Histopathological Image Analysis Using Image Processing Techniques: An Overview. Signal Image Process. Int. J. 2012, 3, 23–36.
  15. Krizhevsky, A.; Sutskever, I.; Hinton, G.E. ImageNet classification with deep convolutional neural networks. In Proceedings of the Advances in Neural Information Processing Systems (NIPS), Lake Tahoe, NV, USA, 3–6 December 2012; pp. 1097–1105.
  16. Le Cun, Y.; Bottou, L.; Bengio, Y.; Haffner, P. Gradient-based learning applied to document recognition. Proc. IEEE 1998, 86, 2278–2324.
  17. Lecun, Y.; Bengio, Y.; Hinton, G. Deep learning. Nature 2015, 521, 436–444.
  18. Arevalo, J.; Cruz-Roa, A. Histopathology image representation for automatic analysis: A state-of-the-art review. Rev. Med. 2014, 22, 79–91.
  19. Irshad, H.; Veillard, A.; Roux, L.; Racoceanu, D. Methods for Nuclei Detection, Segmentation, and Classification in Digital Histopathology: A Review—Current Status and Future Potential. IEEE Rev. Biomed. Eng. 2013, 7, 97–114.
  20. Ciresan, D.C.; Giusti, A.; Gambardella, L.M.; Schmidhuber, J. Mitosis Detection in Breast Cancer Histology Images with Deep Neural Networks. In Proceedings of the International Conference on Medical Image Computing and Computer-Assisted Intervention, Nagoya, Japan, 22–26 September 2013; Springer: Berlin/Heidelberg, Germany, 2013; Volume 2, pp. 411–418.
  21. Arevalo, J.; Cruz-Roa, A.; González, F.A. Hybrid image representation learning model with invariant features for basal cell carcinoma detection. In Proceedings of the IX International Seminar on Medical Information Processing and Analysis, Mexico City, Mexico, 11–14 November2013; SPIE: Bellingham, WA, USA, 2013; Volume 8922, p. 89220M.
  22. Nayak, N.; Chang, H.; Borowsky, A.; Spellman, P.T.; Parvin, B. Classification of tumor histopathology via sparse feature learning. In Proceedings of the 2013 IEEE 10th International Symposium on Biomedical Imaging, San Francisco, CA, USA, 7–11 April 2013; IEEE: Piscataway, NJ, USA, 2013; pp. 410–413.
  23. Tschuchnig, M.E.; Oostingh, G.J.; Gadermayr, M. Generative Adversarial Networks in Digital Pathology: A Survey on Trends and Future Potential. arXiv 2020, arXiv:abs/2004.14936.
  24. Xing, F.; Xie, Y.; Yang, L. An Automatic Learning-Based Framework for Robust Nucleus Segmentation. IEEE Trans. Med Imaging 2015, 35, 550–566.
  25. Gu, F.; Burlutskiy, N.; Andersson, M.; Wilén, L.K. Multi-resolution Networks for Semantic Segmentation in Whole Slide Images. In Lecture Notes in Computer Science; Springer Science and Business Media LLC: Berlin/Heidelberg, Germany, 2018; pp. 11–18.
  26. Tellez, D.; Balkenhol, M.; Otte-Holler, I.; van de Loo, R.; Vogels, R.; Bult, P.; Wauters, C.; Vreuls, W.; Mol, S.; Karssemeijer, N.; et al. Whole-Slide Mitosis Detection in H&E Breast Histology Using PHH3 as a Reference to Train Distilled Stain-Invariant Convolutional Networks. IEEE Trans. Med Imaging 2018, 37, 2126–2136.
  27. Wei, J.W.; Tafe, L.J.; Linnik, Y.A.; Vaickus, L.J.; Tomita, N.; Hassanpour, S. Pathologist-level classification of histologic patterns on resected lung adenocarcinoma slides with deep neural networks. Sci. Rep. 2019, 9, 1–8.
  28. Song, Y.; Tan, E.-L.; Jiang, X.; Cheng, J.-Z.; Ni, D.; Chen, S.; Lei, B.Y.; Wang, T. Accurate Cervical Cell Segmentation from Overlapping Clumps in Pap Smear Images. IEEE Transact. Med. Imaging 2017, 36, 288–300.
  29. Agarwalla, A.; Shaban, M.; Rajpoot, N.M. Representation-Aggregation Networks for Segmentation of Multi-Gigapixel Histology Images. arXiv 2017, arXiv:1707-08814.
  30. Ding, H.; Pan, Z.; Cen, Q.; Li, Y.; Chen, S. Multi-scale fully convolutional network for gland segmentation using three-class classification. Neurocomputing 2020, 380, 150–161.
  31. Bejnordi, B.E.; Zuidhof, G.C.A.; Balkenhol, M.; Hermsen, M.; Bult, P.; Ginneken, B.; van Karssemeijer, N.; Litjens, G.J.S.; Laak, J. Context-aware stacked convolutional neural networks for classification of breast carcinomas in whole-slide histopathology images. J. Med. Imaging 2017, 4, 044504.
  32. Seth, N.; Akbar, S.; Nofech-Mozes, S.; Salama, S.; Martel, A.L. Automated Segmentation of DCIS in Whole Slide Images. In Case-Based Reasoning Research and Development; Springer Science and Business Media LLC: Berlin/Heidelberg, Germany, 2019; pp. 67–74.
  33. Xu, J.; Xiang, L.; Liu, Q.; Gilmore, H.; Wu, J.; Tang, J.; Madabhushi, A. Stacked Sparse Autoencoder (SSAE) for Nuclei Detection on Breast Cancer Histopathology Images. IEEE Trans. Med Imaging 2015, 35, 119–130.
  34. Bulten, W.; Litjens, G. Unsupervised Prostate Cancer Detection on H&E using Convolutional Adversarial Autoencoders. arXiv 2018, arXiv:1804.07098.
  35. Hou, L.; Agarwal, A.; Samaras, D.; Kurc, T.M.; Gupta, R.R.; Saltz, J.H. Robust Histopathology Image Analysis: To Label or to Synthesize? In Proceedings of the 2019 IEEE/CVF Conference on Computer Vision and Pattern Recognition (CVPR), Long Beach, CA, USA, 16–20 June 2019; Institute of Electrical and Electronics Engineers (IEEE): Piscataway, NJ, USA, 2019; pp. 8525–8534.
  36. Sari, C.T.; Gunduz-Demir, C. Unsupervised Feature Extraction via Deep Learning for Histopathological Classification of Colon Tissue Images. IEEE Transact. Med. Imaging 2019, 38, 1139–1149.
  37. Gadermayr, M.; Gupta, L.; Appel, V.; Boor, P.; Klinkhammer, B.M.; Merhof, D. Generative Adversarial Networks for Facilitating Stain-Independent Supervised and Unsupervised Segmentation: A Study on Kidney Histology. IEEE Trans. Med Imaging 2019, 38, 2293–2302.
  38. Gadermayr, M.; Gupta, L.; Klinkhammer, B.M.; Boor, P.; Merhof, D. Unsupervisedly Training GANs for Segmenting Digital Pathology with Automatically Generated Annotations. arXiv 2018, arXiv:1805.10059.
  39. Komura, D.; Ishikawa, S. Machine Learning Methods for Histopathological Image Analysis. Comput. Struct. Biotechnol. J. 2018, 16, 34–42.
  40. Search Home—Biospecimen Research Database. Available online: https://brd.nci.nih.gov/brd/image-search/searchhome (accessed on 1 October 2020).
  41. TMAD Main Menu. Available online: https://tma.im/cgi-bin/home.pl (accessed on 1 October 2020).
  42. Weinstein, J.N.; Collisson, E.A.; Mills, G.B.; Shaw, K.R.M.; Ozenberger, B.A.; Ellrott, K.; Shmulevich, I.; Sander, C.; Stuart, J.M. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 2013, 45, 1113–1120.
  43. Home—CAMELYON17—Grand Challenge. Available online: https://camelyon17.grand-challenge.org/ (accessed on 1 October 2020).
  44. Breast Cancer Histopathological Database (BreakHis)—Laboratório Visão Robótica e Imagem. Available online: https://web.inf.ufpr.br/vri/databases/breast-cancer-histopathological-database-breakhis/ (accessed on 1 October 2020).
  45. Bentaieb, A.; Li-Chang, H.; Huntsman, D.; Hamarneh, G. A structured latent model for ovarian carcinoma subtyping from histopathology slides. Med Image Anal. 2017, 39, 194–205.
  46. Search GDC. Available online: https://portal.gdc.cancer.gov/legacy-archive/search/f (accessed on 1 October 2020).
  47. Ovarian Carcinomas Histopathology Dataset. Available online: http://ensc-mica-www02.ensc.sfu.ca/download/ (accessed on 1 October 2020).
  48. Babaie, M.; Kalra, S.; Sriram, A.; Mitcheltree, C.; Zhu, S.; Khatami, A.; Rahnamayan, S.; Tizhoosh, H.R. Classification and Retrieval of Digital Pathology Scans: A New Dataset. In Proceedings of the 2017 IEEE Conference on Computer Vision and Pattern Recognition Workshops (CVPRW), Honolulu, HI, USA, 21–16 July 2017; Institute of Electrical and Electronics Engineers (IEEE): Piscataway, NJ, USA, 2017; pp. 760–768.
  49. Marinelli, R.J.; Montgomery, K.; Liu, C.L.; Shah, N.H.; Prapong, W.; Nitzberg, M.; Zachariah, Z.K.; Sherlock, G.; Natkunam, Y.; West, R.B.; et al. The Stanford Tissue Microarray Database. Nucleic Acids Res. 2007, 36, D871–D877.
  50. Kumar, N.; Verma, R.; Sharma, S.; Bhargava, S.; Vahadane, A.; Sethi, A. A Dataset and a Technique for Generalized Nuclear Segmentation for Computational Pathology. IEEE Trans. Med Imaging 2017, 36, 1550–1560.
  51. Pathology Images: KIMIA Path960—Kimia Lab. Available online: https://kimialab.uwaterloo.ca/kimia/index.php/pathology-images-kimia-path960/ (accessed on 1 October 2020).
  52. Gelasca, E.D.; Byun, J.; Obara, B.; Manjunath, B.S. Evaluation and benchmark for biological image segmentation. In Proceedings of the 2008 15th IEEE International Conference on Image Processing, San Diego, CA, USA, 12–15 October 2008; Institute of Electrical and Electronics Engineers (IEEE): Pisctaway, NJ, USA, 2008; pp. 1816–1819.
  53. Bio-Segmentation Center for Bio-Image Informatics UC Santa Barbara. Available online: https://bioimage.ucsb.edu/research/bio-segmentation (accessed on 1 October 2020).
  54. Bioimaging Challenge 2015 Breast Histology Dataset—Datasets CKAN. Available online: https://rdm.inesctec.pt/dataset/nis-2017-003 (accessed on 1 October 2020).
  55. BIALab@Warwick: GlaS Challenge Contest. Available online: https://warwick.ac.uk/fac/sci/dcs/research/tia/glascontest/ (accessed on 1 October 2020).
  56. Kather, J.N.; Marx, A.; Reyes-Aldasoro, C.C.; Schad, L.R.; Zoellner, F.G.; Weis, C.-A. Continuous representation of tumor microvessel density and detection of angiogenic hotspots in histological whole-slide images. Oncotarget 2015, 6, 19163–19176.
  57. Dataset—MITOS-ATYPIA-14—Grand Challenge. Available online: https://mitos-atypia-14.grand-challenge.org/dataset/ (accessed on 1 October 2020).
  58. Nucleisegmentation. Available online: https://nucleisegmentationbenchmark.weebly.com/ (accessed on 1 October 2020).
  59. Dataset Tumor Proliferation Assessment Challenge 2016. Available online: http://tupac.tue-image.nl/node/3 (accessed on 1 October 2020).
  60. Janowczyk, A.; Madabhushi, A. Deep learning for digital pathology image analysis: A comprehensive tutorial with selected use cases. J. Pathol. Informat. 2016, 7.
  61. Andrew Janowczyk—Tidbits from Along the Way. Available online: http://www.andrewjanowczyk.com/ (accessed on 1 October 2020).
  62. Ma, Z.; Shiao, S.L.; Yoshida, E.J.; Swartwood, S.; Huang, F.; Doche, M.E.; Chung, A.P.; Knudsen, B.S.; Gertych, A. Data integration from pathology slides for quantitative imaging of multiple cell types within the tumor immune cell infiltrate. Diagn. Pathol. 2017, 12.
  63. Roux, L.; Racoceanu, D.; Loménie, N.; Kulikova, M.; Irshad, H.; Klossa, J.; Capron, F.; Genestie, C.; Le Naour, G.; Gurcan, M.N. Mitosis detection in breast cancer histological images An ICPR 2012 contest. J. Pathol. Inform. 2013, 4, 8.
  64. Linder, N.; Konsti, J.; Turkki, R.; Rahtu, E.; Lundin, M.; Nordling, S.; Haglund, C.; Ahonen, T.; Pietikäinen, M.; Lundin, J. Identification of tumor epithelium and stroma in tissue microarrays using texture analysis. Diagn. Pathol. 2012, 7, 22.
  65. Egfr Colon Stroma Classification. Available online: http://fimm.webmicroscope.net/supplements/epistroma (accessed on 1 October 2020).
  66. Jimenez-del-Toro, O.; Otálora, S.; Andersson, M.; Eurén, K.; Hedlund, M.; Rousson, M.; Müller, H.; Atzori, M. Chapter 10—Analysis of Histopathology Images: From Traditional Machine Learning to Deep Learning; Academic Press: Cambridge, MA, USA, 2017; pp. 281–314.
  67. Zhang, Y.; Zhang, B.; Coenen, F.; Xiao, J.; Lu, W. One-class kernel subspace ensemble for medical image classification. EURASIP J. Adv. Signal Process. 2014, 2014, 17.
  68. Xia, Y.; Cao, X.; Wen, F.; Hua, G.; Sun, J. Learning Discriminative Reconstructions for Unsupervised Outlier Removal. In Proceedings of the 2015 IEEE International Conference on Computer Vision (ICCV), Las Condes, Chile, 11–15 December 2015; Institute of Electrical and Electronics Engineers (IEEE): Piscataway, NJ, USA, 2015; pp. 1511–1519.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 1.2K
Revisions: 2 times (View History)
Update Date: 29 Jan 2021
1000/1000