Abstract
The explosive growth of artificial intelligence (AI) technologies, especially deep learning methods, has been translated at revolutionary speed to efforts in AI-assisted healthcare. New applications of AI to renal pathology have recently become available, driven by the successful AI deployments in digital pathology. However, synergetic developments of renal pathology and AI require close interdisciplinary collaborations between computer scientists and renal pathologists. Computer scientists should understand that not every AI innovation is translatable to renal pathology, while renal pathologists should capture high-level principles of the relevant AI technologies. Herein, we provide an integrated review on current and possible future applications in AI-assisted renal pathology, by including perspectives from computer scientists and renal pathologists. First, the standard stages, from data collection to analysis, in full-stack AI-assisted renal pathology studies are reviewed. Second, representative renal pathology-optimized AI techniques are introduced. Last, we review current clinical AI applications, as well as promising future applications with the recent advances in AI.
Keywords: artificial intelligence, renal pathology, machine learning, deep learning
1. Introduction
Artificial intelligence (AI) represents any technique that enables computers to mimic human intelligence1, 2. The explosive development of its sub-discipline of Machine Learning3, especially Deep Learning4, has led to paradigm shifts in our daily routines ranging from computer vision5, natural language processing6, social media7, autonomous driving8 and healthcare9. AI, for years, has generated enthusiasm in healthcare for its potential to support clinical diagnosis, patient management, and therapeutic planning9, 10. The increasing availability of enormous data (e.g., imaging, omics data, electronic medical record, clinical outcomes, etc.) and unprecedented computing power (e.g., big data management, high-performance computing, high-end networking, etc.) are driving large-scale and data-driven solutions to a variety of imaging-related fields, including, but not limited to, radiology11, surgical intervention12, oncology13 and digital pathology14, 15.
Renal pathology is a subspecialty of general pathology that characterizes medical and transplant kidney diseases based on diagnostic assessment from light microscopy (LM), immunofluorescence (IF) microscopy, and electron microscopy (EM). Renal pathologists examine the renal biopsy to render a definitive clinicopathologic diagnosis, closely working with nephrologists. New applications of AI to renal pathology have recently become available, driven by the successful AI deployments in digital pathology16 and the more widespread use of digital diagnostic imaging, in particular, the recent advances in whole slide imaging (WSI) technology17.
Machine learning is typically regarded as a subset of AI, which focuses on empowering computer systems with the ability to “learn from data”3, rather than mimicking other aspects of human intelligence, such as body motion or human emotion. Since most AI techniques in renal pathology are developed to learn essential knowledge from data, AI and machine learning are often used as interchangeable terms in this context.
Deep learning is the latest breakthrough of AI/machine learning, which offers an unparalleled ability to efficiently manage patients, accelerate diagnosis, and guide treatments18. The performance of a deep learning model is typically constantly improved when more training examples are provided, while traditional AI can easily hit a learning plateau without further improvements19. Interestingly, the core computation algorithms of deep learning, such as artificial neural networks (ANN)20, 21 and convolutional neural networks (CNN), were proposed in the 1990s or earlier22. The exceptional learning capability of deep learning is largely driven by recent advances in computing software23 and hardware24, supporting deeper layers25, more parameters26 and large-scale training data27. With unprecedented learning capability, AI can learn extensive medical knowledge from large-scale renal pathology data, to support clinical decisions and reduce diagnostic and therapeutic errors in clinical practice.
Renal pathologists play a critical role in the work-up of patients to render a correct biopsy diagnosis, with prognostic assessment, activity and chronicity grading and give accurate information to nephrologists to guide their choice of effective therapy. However, these tasks require both task-specific machine learning techniques for AI and deep clinical understanding. Therefore, the synergetic developments of renal pathology and AI require close interdisciplinary collaborations between computer scientists, nephrologists and renal pathologists16. To design an optimal AI algorithm for renal pathology, computer scientists should comprehend the current practice scenarios and barriers in renal pathology, while renal pathologists should understand high-level principles of the different AI technologies. Herein, we review the recent synergetic advances in AI-assisted renal pathology, with integrated perspectives from both computer scientists and renal pathologists. First, we review the standard stages in full-stack AI-assisted renal pathology studies, from data collection to analysis. Second, we introduce representative renal pathology-optimized AI techniques beyond using off-the-shelf algorithms. Last, we review the current clinical AI applications and promising future applications with recent advances in AI. The goal of this review is to provide an integrated vision of AI-assisted renal pathology by combining engineering and clinical perspectives, across research design, algorithm developments and clinical applications.
2. Standard Workflow of AI-assisted Renal Pathology
The standard workflow of performing an AI-assisted renal pathology study typically consists of four stages: (1) problem definition, (2) data collection and annotation, (3) AI development and training and (4) data fusion and analysis. An example of the workflow for a biopsy using AI integrated with clinical information and genotype is illustrated in Fig. 1. Briefly, AI can be used to count the percentage of glomeruli with global glomerulosclerosis (GS), with the fine-grained classification of these globally sclerotic glomeruli as obsolescent, solidified or disappearing. Obsolescent GS is associated with normal aging, while solidified/disappearing GS is characteristic of hypertension-associated disease28. The upper panel in Fig. 1 shows a representative pipeline of AI-assisted GS characterization, using a WSI image acquired from anAfrica American man with a biopsy diagnosis of hypertension-associated nephropathy. The image features can be further aggregated with genetic and clinical features by another AI fusion algorithm, shown in this example as a computer-aided diagnosis system (Fig. 1). For this patient, the morphologic diagnosis is arterionephrosclerosis, with 4/19 glomeruli GS in a solidified pattern and one disappearing GS, the end stage of a solidified glomerulus as it becomes contiguous with the fibrotic interstitium, with additional characteristic vascular lesions. Here we have integrated these quantitative AI-derived assessments of morphologic lesions, with the patient’s ethnicity (Africa American), and genotype, with two ApoL1 high risk variants.
Figure 1.

A representative workflow of AI-assisted renal pathology. In the upper panel, AI is used to count the percentage of glomeruli with global glomerulosclerosis (GS), with the fine-grained classification of obsolescent, solidified or disappearing types. Then, the image features are further aggregated with genetic and clinical features by another AI fusion algorithm. The lower left panel shows the representative tasks of AI-assisted quantification, while the lower right panel shows the representation modalities for AI-based multi-modal data fusion.
2.1. Problem Definition
AI projects in renal pathology are commonly driven by specific problems from scientific research or clinical practice. There are two main motivations for employing AI in renal pathology studies. The first motivation is to help pathologists accomplish specific tasks with less manual efforts, higher efficiency and even better performance29–32. A straightforward example is to compute the percentage of sclerotic glomeruli in a biopsy. The second motivation is to offer new capabilities to investigate problems that are unscalable or impractical for pathologists. For example, AI can “visualize” all voxels inside a 3D volume simultaneously, using advanced 3D microscopy imaging33–35, while the human vision is limited by understanding serial 2D sections or projections.
2.2. Data Collection and Annotation
Once the problem is defined, the next step is to collect corresponding renal tissue samples, including standard tissue sections, digitizing metadata, digital imaging of stained tissue sections, and quality control36, 37. The learning strategies can be classified as three types: supervised learning, semi-supervised learning, and unsupervised learning38. The key differences are whether human annotations are required for training AI to understand the digitized images. The “supervision” can be simply interpreted as annotations in AI. Briefly, the supervised learning methods utilize well-annotated data to train AI systems, while the unsupervised learning methods encourage the AI systems to learn clinically relevant information from unlabeled data. The semi-supervised learning strategy utilizes both annotated and unlabeled data. Supervised learning is still the most prevalent design of AI-assisted solutions for renal pathology, due to its superior performance of distinguishing subtle morphological differences that are difficult to recognize by unsupervised self-learning. Moreover, even for the unsupervised learning algorithms, annotated data are still needed for validation purposes.
Annotations in renal pathology are typically achieved from manual efforts from pathologists39, 40. The annotations train the AI algorithms to understand different levels of image concepts41. For example, the annotations can be offered at image-level (for image classification), region-level (for object detection) or pixel-level (for image segmentation). However, inadequate annotations are one major bottleneck in training deep learning algorithms in renal pathology42, as precise annotations require extensive clinical knowledges from trained practitioners, rather than using crowdsourcing annotations from lay person annotators. For renal pathologists, it is not only tedious but unscalable to annotate large-scale data for training deep learning algorithms, with the additional challenges of inter-rater variabilities and protocol consistency. Fortunately, more feasible AI-assisted annotation algorithms have been proposed to reduce the manual workload for human annotators, such as human-in-the-loop annotation or AI-based interactive annotation37, 38. Meanwhile, a collaborative data curation pipeline is appealing to distribute the workload between clinical experts and lay annotators.
2.3. AI Development and Training
In healthcare, AI refers to computer techniques that mimic human physicians’ decisions and actions. Most AI algorithms in renal pathology have thus been invented to understand biopsies by simulating the behaviors of renal pathologists, including object detection (e.g., identify glomeruli, tubules, arteries), classification (e.g., phenotyping, diagnosis, therapeutic planning), segmentation (e.g., trace contours of objects), synthesis (e.g., staining), and rendering diagnosis (e.g., fuse information from multiple modalities or resources)29–31, 43–53. Historically, such learning algorithms were built upon traditional machine learning approaches, by capturing human-designed imaging features among digital microscope images (e.g., color, edge, shape). However, such methods were usually limited by the lack of generalizability and completeness of the hand-crafted features. Deep learning-based AI algorithms are now increasingly used to learn discriminative features from large-scale training images16, 32, 54. In the following sections, we introduce four standard techniques in AI-assisted renal pathology, which utilize data-driven features in different ways.
2.3.1. Classification
Image classification is a fundamental task in computer vision to provide a specific label for each image4. Specific to renal pathology, the label can be a phenotype, a diagnosis, or a therapeutic plan. Recently, deep learning-based AI has been widely used for different classification tasks in renal pathology, which typically consist of two parts: feature extraction and diagnosis43, 44, 55–58. In contrast to previous AI algorithms, deep learning algorithms can extract multi-scale features (from local to global), which benefit from the deeper (more layers) network design. In a canonical CNN design59, the initial layers focus on extracting local image features (e.g., color, shape, and morphology of a nucleus), while the deeper layers extract global image features (e.g., staining, shape, and morphology of a glomerulus). Then, a final diagnosis is typically made by aggregating the multi-scale features using a multi-layered perceptron (MLP) network60. The process is similar to rendering a biopsy interpretation by a renal pathologist, where the pathologist might first screen the local and global image features, and then aggregate all evidences to achieve a final diagnosis.
2.3.2. Detection
Detection is regarded as a higher-level visual perception compared with classification, since it offers a region-level classification of each detected object (e.g., a glomerulus within a rectangular bounding box), instead of image-level characterization in classification45–48, 61, 62. In AI implementation, a feature extraction step is first performed to obtain local and global features, which is similar to the feature extractor used in classification. Next, key landmarks of each object (e.g., corners of bounding boxes) are achieved by aggregating extracted features from multi-scale data. Last, a classification sub-network is employed to provide a label for each object, within each bounding box. Using this design, many deep learning-based detection algorithms (e.g., Faster-RCNN63) have outperformed traditional model-based approaches in terms of accuracy and efficiency64, 65. Some AI approaches have even provided real-time capability of object detections, which have led to a new line of products, called the smart microscope66. The smart microscope might change the current paradigm of inspecting biopsies, as it offers real-time support to pathologists analyzing samples, detecting objects, rendering diagnosis and generating reports66. Using the smart microscope, AI quantifications are conducted when pathologists perform routine pathology workflow, such as visual inspection with a microscope, without extra effort.
2.3.3. Segmentation
Image segmentation is the most detailed quantification of an image since each pixel is assigned with a label, as opposed to image-level or region-level characterization31, 43, 49–53, 67–74. Technically, the segmentation task can be regarded as an extreme case of classification or detection, where the classification is performed on each pixel. Pixel-level quantification provides us the precise spatial and quantitative measurements of objects at different scales. However, extensive pixel-level classification is time consuming. To speed up such processes, fully convolutional network (FCN)-based75 approaches were proposed to perform classification on all pixels simultaneously. Recently, to achieve instance object level segmentation, FCN networks have been incorporated with detection networks to form holistic instance segmentation approaches (e.g., Mask-RCNN76). Although segmentation is not typically involved in routine pathology workflow, this approach provides a promising direction to replace the current semi-quantitative phenotyping methods with fully-quantitative solutions.
2.3.4. Synthesis
Image synthesis is one class of AI algorithms to synthesize the image from one appearance to another, which is typically referred to as generative adversarial networks (GAN)77–79. Instead of providing labels for image, region, or pixel, the aim of image synthesis is to generate a new “real look” image from another existing image, or even from a random noise or a prior shape. In renal pathology, image synthesis has been used for virtual staining, which transfers the appearance of a digitized tissue from one stain to another78, 80–84. As the synthetic images are generated digitally rather than physically, there is almost no extra imaging cost to generate large-scale new data as sample size augmentation to train a deep neural network. For instance, GAN technologies were utilized to develop a generalizable glomerular segmentation model83, without the need of large databases with different staining patterns85. Synthetic images have been used in next-generation renal histomorphometry32, to perform label-free object classification79, 86, detection87 and segmentation88–90 from renal pathological images. The summarization of AI methods in renal pathology is provided in Table 1.
Table 1.
AI methods in renal pathology
| Methodology | Authors | Journal | Year | Tools | Applied task | Number of WSIs or cases |
|---|---|---|---|---|---|---|
| Classification | Ginley et al. [43] | J. Am. Soc. Nephrol. | 2019 | RNN, CNN | Glomerulus classification, Multiclass segmentation of renal morphology | 54 WSIs, human 25 WSIs, mouse |
| Uchino et al. [44] | International Journal of Medical Informatics | 2020 | CNN | Glomerulus classification | 283 cases, human | |
| Ginley et al. [55] | Proc SPIE Int Soc Opt Eng | 2020 | RNN, CNN | Glomerulus segmentation, Recurrent biopsy classification, Glomerular component analysis | 82 WSIs (65 cases), human | |
| Zee et al. [56] | Arch Pathol Lab Med | 2018 | Scoring System | Glomerulus morphology assessment | 236 WSIs (glomeruli), human | |
| Chagas et al. [57] | Artif Intell Med. | 2020 | CNN, SVM | Glomerulus hypercellularity | 811 WSIs (glomeruli), human | |
| Ledbetter et al. [58] | arXiv. | 2017 | CNN | Prediction of kidney function | 80 cases, human | |
| Detection | Temerinac-Ott et al. [45] | Proc Int Symp Image Signal Process Anal | 2017 | CNN, HOG | Glomerulus detection | 6 cases, human |
| Bukowy et al. [46] | J Am Soc Nephrol | 2018 | CNN | Glomerulus detection | 87 WSIs, human | |
| Simon et al. [47] | Sci Rep | 2018 | SVM, CNN | Glomerulus detection | 15 WSIs, mouse 25 WSIs, human |
|
| Marée et al. [48] | Proc IEEE Int Symp Biomed Imaging | 2016 | Commercial software | Glomerulus detection | 200 WSIs, human | |
| Kato et al. [61] | Bmc Bioinformatics | 2015 | SVM, HOG | Glomerulus detection | 20 WSIs, mouse | |
| Gallego et al. [62] | J Imaging | 2018 | CNN | Glomerulus detection, Glomerulus classification | 108 WSIs, human | |
| Yang et al. [91] | Med Image Comput Comput Assist Interv | 2020 | CNN | Glomerulus detection | 42 WSIs, human | |
| Segmentation | Hermsen et al. [31] | J Am Soc Nephrol | 2019 | CNN | Multiclass segmentation of nephrectomy and transplant biopsies | 50 WSIs, human |
| Ginley et al. [43] | J Am Soc Nephrol | 2019 | RNN, CNN | Glomerulus segmentation, Multiclass segmentation of renal morphology | 54 WSIs, human 25 WSIs, mouse |
|
| Bueno et al. [49] | Comput Methods Programs Biomed | 2020 | CNN | Glomerulus segmentation, Glomerulus classification | 47 WSIs, human | |
| Kannan et al. [50] | Kidney Int Rep | 2019 | CNN | Glomerulus segmentation | 275 WSIs (171 cases), human | |
| Gadermayr et al. [51] | arXiv preprint | 2017 | CNN | Glomerulus segmentation | 24 WSIs, mouse | |
| Gadermayr et al. [52] | Comput Biol Med | 2017 | SVM | Glomerulus detection, Glomerulus segmentation | 8 WSIs from mouse kidneys | |
| Ginley et al. [53] | arXiv preprint | 2020 | CNN | Multiclass segmentation of renal morphology | 65 cases, human | |
| Santo et al. [67] | Proc SPIE Int Soc Opt Eng | 2020 | Feature engineering | LN biopsies segmentation | 21 WSIs, human | |
| Gupta et al. [68] | Proc Machine Learning Res | 2019 | CNN | Glomerulus segmentation | 22 WSIs, mouse | |
| Ginley et al. [69] | J Med Imaging | 2017 | Gabor Filterbank, Statistical Testing | Glomerulus segmentation | 1000 images, mouse | |
| Sarder et al. [70] | Proc SPIE Int Soc Opt Eng | 2020 | Gabor Filterbank, Statistical Testing | Glomerulus segmentation | 15 WSIs, mouse | |
| Tey et al. [71] | Comput Methods Programs Biomed | 2018 | Alternating Decision Trees, SVM | Glomerulus segmentation | 286 WSIs (70 cases), human | |
| Altini et al. [72] | Electronics | 2020 | CNN | Glomerulus segmentation, Glomerulus classification | 26 WSIs (19 cases), human | |
| Bouteldja et al. [73] | J Am Soc Nephrol | 2020 | CNN | Kidney tissue segmentation | 168 WSIs, mouse | |
| Jha et al. [92] | J Med Imaging | 2020 | CNN | Glomerulus segmentation | 1454 images, human | |
| Jayapandian et al. [74] | Kidney Int | 2020 | CNN | Kidney tissue segmentation | 459 WSIs (125 cases), human | |
| Synthesis | Murali et al. [78] | Proc SPIE Int Soc Opt Eng | 2020 | GAN | Renal histopathology images synthesis | 20K images, human |
| Lutnick et al. [79] | Proc SPIE Int Soc Opt Eng. | 2020 | VAE-GAN | Glomerulus images synthetic, Glomerulus classification | 59930 images, human 27508 images, mouse |
|
| Zhang et al. [81] | Light: Sci & Appl | 2020 | GAN | Digital stain | 12 WSIs, human | |
| Gadermayr et al. [82] | Med Image Comput Comput Assist Interv | 2018 | GAN, CNN | Digital stain, Glomerulus segmentation | 59 WSIs, mouse | |
| Gadermayr et al. [83] | IEEE Trans Med Imaging | 2019 | GAN, CNN | Digital stain, Glomerulus segmentation | 41 WSIs, mouse | |
| de Bel et al. [84] | Med Imaging Deep Learning | 2020 | GAN | Digital stain | 64 WSIs, human | |
| Wu et al. [86] | Proc Conf AAAI Artif Intell | 2019 | GAN, CNN | Digital stain, Glomerulus classification | 209 cases, human | |
| Gadermayr et al. [88] | Med Imaging Deep Learning | 2019 | GAN | Glomerulus synthetic segmentation | 23 WSIs, mouse | |
| Gupta et al. [89] | Med Image Comput Comput Assist Interv | 2019 | GAN, CNN | Digital stain, Glomerulus segmentation | 59 WSIs, mouse | |
| Mei et al. [90] | IEEE ICASSP | 2019 | GAN, CNN | Digital stain, Glomerulus segmentation | 819 images, human |
CNN = convolutional neural network, GAN = generative adversarial network, VAE = variational autoencoder, SVM = support vector machine, RNN = recurrent neural network, HOG = histogram of oriented gradients.
WSI = whole slide imaging, LN = lupus nephritis.
2.4. AI-based Data Fusion and Analysis
The same lesion by light microscopy may be due to varied underlying diseases, e.g. nodular glomerulosclerosis in diabetic nephropathy vs light chain deposition disease or other entities. Renal pathologists thus integrate various data to make diagnoses, including light microscopy, immunofluorescence, electron microscopy and clinical setting, and even specific genetic testing93, 94. AI algorithms have led to successful clinical applications by aggregating heterogeneous healthcare data, advanced by fusing data with different dimensions and representations in a fully quantitative and data-driven manner95. AI-assisted multimodal data fusion and analysis algorithms can also play important roles in renal pathology. One AI-based multi-modal data fusion strategy is “late fusion”96, which is similar to human decision making. Briefly, different AI networks are typically employed to extract the useful knowledge (features) from different modalities95. Then, such features are concatenated and aggregated (e.g., using MLP60) into final outcomes, analogous to decision-making from comprehensive clinical assessments integrated with various tests by physicians. AI-based approaches can also perform “early fusion”96, such as conducting pixel-wise alignment and integration between different imaging modalities97, or mapping single-cell RNA-seq with pathological images98.
3. Renal Pathology Optimized Deep Learning
Currently, a majority of AI-assisted renal pathology studies utilize off-the-shelf deep learning algorithms that have been developed in the computer vision community, such as Faster-RCNN63, Mask-RCNN76, U-Net99 etc. The rationale is that both renal pathology and computer vision fields deal with similar targets -- images. Therefore, it is a natural and reasonable choice to apply prevalent algorithms in computer vision to renal pathology. However, computer vision-oriented AI algorithms are not necessarily optimized for biomedical objects. Therefore, deep learning optimized for renal pathology is appealing to further boost the performance of AI in renal pathology. In this section, we review cases of such optimized deep learning algorithms, including optimized data representation, optimized algorithm and optimized dimensionality (Fig. 2).
Figure 2.

Illustration of AI algorithms optimized for renal pathology. This figure compares off-the-shelf AI algorithms in computer vision and AI algorithms optimized for renal pathology. The optimized AI algorithms have been developed to address unique challenges in renal pathology, including optimized data representation, optimized algorithm and optimized dimensionality.
3.1. Optimized Representation
Representation means the format in which data are stored, processed, and transmitted to efficiently convey quantitative information. In AI-assisted renal pathology, the “computer vision” oriented representation is not necessarily optimized for biomedical objects. For instance, the widely used rectangular encoder of representing glomerular objects yielded many false positive due to the extensive variations in e.g. number of diverse pathogenetic mechanisms100. Therefore, Kato et al. proposed a new renal pathology optimized descriptor to perform comprehensive glomerular detection61. Similarly, Simon et al. introduced a circular neighborhood image features extractor, which achieved superior detection performance of spherical shape glomeruli47. In the deep learning era, the computer vision oriented bounding boxes representation is employed in most glomerular detection studies. However, the bounding box might not be the most effective representation for the ball-shaped glomeruli, which yields less reproducibility with image rotation, even for the same tissue. Recently, we proposed a new circle representation91 as an optimized data representation for glomerular detection, which notably improved the detection performance.
3.2. Optimized Algorithm
An obvious obstacle that limits the performance of off-the-shelf deep learning algorithms in renal pathology applications is the fundamental difference in image resolution. In computer vision, even a high-resolution image (e.g., “4K” resolution, 3840×2160 pixels) is relatively small compared with “Gigapixel” WSI (the resolution can be higher than 32,768×32,768 pixels). Therefore, the scale of AI algorithms in computer vision might not be optimized for renal pathology. To bridge the gap of resolution, use of tiling and down-sampling are typically inevitable when deploying standard computer vision AI approaches to renal pathology. For example, a single glomerulus in a 40× WSI can have a resolution of more than 1000×1000 pixels, yet its feature maps are compressed to 28×28 pixels in the standard Mask-RCNN segmentation approach76. Therefore, AI algorithms optimized for renal pathology are required to address the unique challenges in high resolution diagnostic imaging. In this regard, Gadermayr et al. proposed a cascade segmentation method to first segment the glomeruli on the low resolution image using a shallower CNN, and then segment the high resolution glomerular regions with a deeper CNN51. Recently, we proposed a “detect-then-segment” instance segmentation approach92 to balance the computation efficiency without significantly compromising image resolution for analysis of renal pathology WSI.
3.3. Optimized Dimensionality
Dimensionality is a critical property when designing a proper strategy of understanding any types of imaging data. In computer vision, the input images for AI are typically 2D natural images, acquired from digital cameras. In radiology, the input images are typically 3D volumes, acquired from computed tomography (CT) or magnetic resonance imaging (MRI). By contrast, in renal pathology, a set of serial sections from the same tissue can be acquired as a stack of images from WSI. However, the stacks of 2D images are not aligned perfectly as they are for radiological images, due to large tissue deformation, missing areas of tissues, and artifacts from WSI. Therefore, such images are typically processed as independent 2D images when AI algorithms are deployed. However, if the 3D context of glomeruli can be quantified holistically from all 2D WSI serial sections, the robustness and reproducibility of glomerular phenotyping would be improved significantly. The advantages of 3D quantification have been elucidated using advanced microscopy imaging techniques, such as confocal microcopy101, electron microscopy35, 102, 103 and automated tape collecting microtome104. To further enable the 3D quantification of glomeruli for serial WSI images, we proposed an AI method105 to perform 3D identification and association of glomeruli. Using this method, the time needed to quantify atubular glomeruli, (i.e. those without patent connection through the proximal tubule outlet) in a 3D stack of whole mouse kidney sections form one kidney was reduced from 30 hours (done maually by humans) to 30 mins.
4. Clinical AI Applications in Renal Pathology
AI-assisted pathologists demonstrated higher accuracy than either AI alone or the pathologist alone in detecting cancer micrometastases106. New molecular technologies and the big data revolution are paving the way to a new era in renal pathology. In this section, the applications of AI in renal pathology are reviewed, from integrated pathology to precision medicine (Fig. 3).
Figure 3.

AI applications in renal pathology and nephrology. An example of applications of AI in renal pathology is presented, from integrated pathology to precision medicine.
4.1. From Morphologic to Integrated Pathology
Current histopathological categories used in renal pathology are irrespective of different molecular mechanisms. A diagnosis is made based on the following flow: recognition of injury patterns, grouping/clustering of patterns and integration with clinical data. AI could assist in pattern recognition and clustering. By using CNN technology, 10 different types of renal structures could be segmented, including e.g. glomeruli, Bowman’s capsule, proximal tubules, distal tubules, arteries and interstitium31. Recurrent neural network (RNN) technology was successfully used to classify diabetic nephropathy43. Adversarial learning could play a potentially important role in robust AI, especially considering disease variability and the challenges of rare disease. For instance, if the AI system has self-awareness of uncertainty using adversarial learning as a posterior probability, the computer would request help from human experts to reduce inaccurate diagnoses and reduce the workload of pathologists by letting AI figure out the “easy” cases. In addition, AI could distill more useful information from gigapixel images than the human eye, which means more sub-visual morphology could be discovered by AI. An algorithm based on the auto-encoder network, which is an unsupervised deep learning technique that learns how to efficiently extract (encode) intrinsic features from high-dimensional images, will be more stable and effective for such efforts107. Therefore, morphology profiles, named morphomics, can be expanded by AI. The identification of previously underrecognized patterns, and their potential correlations with outcomes, treatment responses or even specific injury pathways, will change paradigms for renal pathology categorization. Moreover, the deep Cox model108 and multimodal learning could be used for prediction of prognosis.
AI also could contribute to a move from morphologic classification to mechanism-based disease definition. One major concern of morphology-based diagnosis is that one disease can present different patterns and one injury pattern can be induced by different diseases. IgA nephropathy shows a broad range of glomerular changes by light microscopy, from normal or just mesangial cell proliferation to sclerosis or even crescents, while cellular crescents could be related to e.g. anti-GBM antibody-mediated disease or ANCA vasculitis. Morphology-based disease definitions/classifications have as a goal to show some clinical relevance. The Columbia classification of FSGS has distinctive correlation to clinical features and outcomes, although the etiologies may be quite varied, with potential added insights providing better personalized treatment approaches109. On the other hand, the old classification of membranoproliferative glomerulonephritis (MPGN) based on morphology in EM did not allow recognition of specific etiologies underlying the lesions. The current MPGN classification adding immunofluorescence findings allows recognition and distinction of MPGN lesions driven by complement dysregulation vs. immune complexes or monoclonal immunoglobulin deposition, which significantly fits in implication for therapy. Diseases have molecular signatures, which can potentially be detected by genomic, epigenomic, proteomic and/or metabolomic profiles. Morphologic changes, assessed and aided by sub-visual signals detectable by AI, could be linked to these molecular features, to achieve more accurate, comprehensive, and clinically relevant pathology diagnosis/classification.
This integrated pathology approach will require matching and clustering multi-dimensional data (morphomics, molecular omics, and clinical data) (Fig. 3). Linear or non-linear data transformations are widely used to cluster. If data have a complex structure, these techniques would be unsatisfying for clustering.
4.2. From Descriptive to Quantitative Pathology
Although most pathology reports are descriptive, some morphologic scores have been incorporated into clinical practice, such as activity/chronicity indices of lupus nephritis (LN) and MEST-C score of IgA nephropathy. These scores are useful in predicting renal outcomes110, 111. Other parameters, such as fractional interstitial area, average glomerular tuft volume, and cortical density of glomeruli, which only could be measured by morphometry on scanned slides and cannot be precisely achieved with the human eye, correlate with various aspects of renal function112. Quantifying lesions may be time-consuming and have variable reproducibility. For instance, the chronicity index for LN showed a moderate interobserver agreement (intraclass correlation coefficient 0.494) when assessed by five specialized renal pathologists113. The benefits of transforming from descriptive pathology to quantified lesions are obvious. Image quantification is more objective than description and can overcome the observer’s potential bias. Compared to the current semi-quantitation system often used, e.g. 0–4+, the use of continuous variable quantitation is more sensitive and may be more useful for prediction of outcome. With the common use of WSI, enhancement of computer hardware and server capacity, AI-assisted quantitative analysis of pathological images can be realized. A recent study showed that AI can quantify interstitial fibrosis in renal biopsy specimens better than pathologists114. Thus, AI-assisted pathological quantitation reduces human effort, and can ensure consistency, reproducibility and standardization.
4.3. Towards Precision Medicine
Precision medicine aims to classify patients into mechanistically defined subgroups with different disease susceptibility, prognosis and therapeutic responsiveness. Review of failed clinical trials sometimes shows positive results in subgroups of patients although results were negative in the overall group115. In oncology, disease is now not categorized by tissue of origin, e.g. cancer of the lung, but rather by mutation that can be targeted by a specific drug, e.g. ALK mutation positive. The feasible strategies to tailor an individualized treatment are as follows: clarify the molecular pathology spectrum of a certain disease; develop appropriate drugs for single or multiple spectra; evaluate the spectrum in the individual patient by integrating morphomics, molecular omics, and clinical data; and finally choose treatment drugs to achieve “the right treatment for the right patient”. The combination and analysis of these large-scaled, disparate dimensional datasets are challenging. AI and use of tailored different algorithms could be used for this strategy (Fig. 3).
Deep reinforcement learning is a promising method in precision medicine, since this approach has unique advantages to explore the problem set by itself in an unsupervised manner, to for instance determine optimal chemotherapy drug dosage and optimal scheduling of radiation therapy for cancer treatment116, 117. Deep reinforcement has been used to select optimal image patches for automated immunohistochemical scoring118. One machine learning algorithm LOBICO, a logic optimization for binary input to continuous input, has been successfully applied to merge omics data and identify biomarkers predicting drug response in cancer cells119.
Currently, most AI-based computer-aided diagnosis systems function as black boxes, with limited capability of explanation and reasoning in terms of why the computer system made specific decisions. Recently, a new direction of algorithms, named explainable AI, has emerged to provide diagnostic evidence that can be verified by humans, accelerating deployment of AI in practice120. For instance, the AI attentions from a classification task can be visualized using CAM121, GradCAM122 or Lime123 tools, which highlights the regions that the AI algorithm is “looking at” when making decision. Ultimately, with stronger interpretation capability, explainable AI techniques would contribute to discover sub-visual biomarkers, which will play a vital role in the development of therapies, personalized diagnosis and treatment of diseases.
Recent AI-based clustering technologies also provide powerful means to group histopathological images with similar morphological and pathological features across large-scale patient cohorts. One example would be large-scale histopathological image retrieval systems. Using deep texture representation algorithms, such web-based systems could help clinicians to retrieves similar histopathological images from a large database, even with the patients’ diagnoses, treatments, and prognoses as references. The challenges and proposed solutions of AI-based techniques are summarized in Table 2.
Table 2.
Some of the limitations of AI methods and proposed solutions
| Methodology | Limitation | Explanation | Proposed Solution |
|---|---|---|---|
| Classification | Class imbalance | The anatomy of interest or pathological tissues only occupy a very small portion of WSI images. | Weight rebalance techniques can be used to assign higher penalties when images in minor classes are misclassified. |
| Detection | Balance recall and precision | To balance false positive and false negative is a dilemma since only positive samples are typically annotated in a standard object detection task. | To actively mine hard negative samples is an effective solution to enlarge decision margins in detection tasks, to reduce ambiguities. |
| Segmentation | Annotated data | Pixel-wise annotations for training a segmentation AI are resource intensive for digital renal pathology. | Semi-supervised learning algorithms can utilize a smaller cohort of annotated data and a larger cohort of unannotated data, to enhance segmentation performance. |
| Synthesis | Hallucination | Hallucination is a major limitation of applying image synthesis. It is challenging to validate whether the synthetic images are biologically plausible. | Instead of directly using synthetic images in diagnosis, synthetic images can be used as an intermediate modality for downstream tasks with transfer learning. |
| Shared limitations across methodologies | Generalizability | The AI model, trained from specific cohorts or sites, might not generalize well on other cohorts or sites. | Federated learning introduces a new computing solution to train a generalizable deep learning model using heterogenous multi-site datasets in an efficient manner without moving patients’ data outside each site. |
| Dimensional issue | Images can be Gigapixel size, leading to fundamental computing challenges when training and deploying models at scale. | Patch-wise training can reduce the input size for an AI model. Parallel computing algorithms with more processor cores can accelerate the processing speed on large images. | |
| Explainability | Most AI algorithms currently function as “black boxes”, with limited capability of explaining why certain decisions are made. | Attention-based mechanisms and recent explainable AI techniques are promising solutions to provide stronger interpretability of AI models. | |
| Data fusion | Effective algorithms are needed to aggregate different data including light, immunofluorescence and electron microscopy, with clinical setting, and even specific genetic testing. | Multi-task learning and multi-modality fusion algorithms can normalize and aggregate different feature representations to support holistic clinical decisions with multi-modality data. |
5. Summary
AI technologies are playing increasingly important roles in renal pathology. These approaches have a strong potential to shift the current clinical paradigms in terms of accelerating workflow, supporting clinical decision-making and determining personalized treatment plans. However, there are still essential challenges that hinder the broader applications of AI in renal pathology, such as limited annotated data, complicated multi-modal data, and lack of clinical validation. Therefore, more seamless collaboration between pathologists and engineers will leverage further advances in developing next generation AI algorithms in renal pathology, with potential for major impacts in diagnosis and understanding of kidney diseases.
Acknowledgments:
This work was supported in part by NIH NIDDK DK56942 (ABF).
Footnotes
Disclosure statement
The authors have declared that no conflict of interest exists.
References
- 1.Poole D, Mackworth A, Goebel R. Computational Intelligence. Oxford University Press; 1998. [Google Scholar]
- 2.Russell S, Norvig P. Artificial intelligence: a modern approach. Prentice Hall; 2002. [Google Scholar]
- 3.Bishop CM. Pattern recognition and machine learning. Springer; 2006. [Google Scholar]
- 4.Goodfellow I, Bengio Y, Courville A, et al. Deep learning. MIT Press Cambridge; 2016. [Google Scholar]
- 5.Voulodimos A, Doulamis N, Doulamis A, et al. Deep learning for computer vision: A brief review. Comput Intell Neurosci. 2018; 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Young T, Hazarika D, Poria S, et al. Recent trends in deep learning based natural language processing. IEEE Comput Intell Mag. 2018; 13: 55–75. [Google Scholar]
- 7.Zafarani R, Abbasi MA, Liu H. Social media mining: an introduction. Cambridge University Press; 2014. [Google Scholar]
- 8.Grigorescu S, Trasnea B, Cocias T, et al. A survey of deep learning techniques for autonomous driving. J Field Robotics 2020; 37: 362–386. [Google Scholar]
- 9.Esteva A, Robicquet A, Ramsundar B, et al. A guide to deep learning in healthcare. Nat Med. 2019; 25: 24–29. [DOI] [PubMed] [Google Scholar]
- 10.Jiang F, Jiang Y, Zhi H, et al. Artificial intelligence in healthcare: past, present and future. Stroke Vasc Neurol. 2017; 2: 230–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hosny A, Parmar C, Quackenbush J, et al. Artificial intelligence in radiology. Nat Rev Cancer 2018; 18: 500–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hashimoto DA, Rosman G, Rus D, et al. Artificial intelligence in surgery: promises and perils. Ann Surg. 2018; 268: 70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Londhe VY, Bhasin B. Artificial intelligence and its potential in oncology. Drug Discov Today. 2019; 24: 228–232. [DOI] [PubMed] [Google Scholar]
- 14.Niazi MKK, Parwani AV, Gurcan MN. Digital pathology and artificial intelligence. Lancet Oncol. 2019; 20: e253–e261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tizhoosh HR, Pantanowitz L. Artificial intelligence and digital pathology: challenges and opportunities. J Pathol Inform. 2018; 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Becker JU, Mayerich D, Padmanabhan M, et al. Artificial intelligence and machine learning in nephropathology. Kidney Int. 2020; 98: 65–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ghaznavi F, Evans A, Madabhushi A, et al. Digital imaging in pathology: whole-slide imaging and beyond. Annu Rev Pathol. 2013; 8: 331–359. [DOI] [PubMed] [Google Scholar]
- 18.Miotto R, Wang F, Wang S, et al. Deep learning for healthcare: review, opportunities and challenges. Brief Bioinform. 2018; 19: 1236–1246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Dargan S, Kumar M, Ayyagari MR, et al. A survey of deep learning and its applications: A new paradigm to machine learning. Arch Comput Method E. 2019: 1–22. [Google Scholar]
- 20.Werbos PJ. Backpropagation through time: what it does and how to do it. P IEEE. 1990; 78: 1550–1560. [Google Scholar]
- 21.Zurada JM. Introduction to artificial neural systems, West Publishing Co; 1992. [Google Scholar]
- 22.LeCun Y, Boser B, Denker JS, et al. Backpropagation applied to handwritten zip code recognition. Neural Comput.1989; 1: 541–551. [Google Scholar]
- 23.Shi S, Wang Q, Xu P, et al. Benchmarking state-of-the-art deep learning software tools. IEEE CCBD. 2016; 99–104. [Google Scholar]
- 24.LeCun Y Deep Learning Hardware: Past, Present, and Future. Proc IEEE Int Solid-State Circuits Conf. 2019; 12–19. [Google Scholar]
- 25.Krizhevsky A, Sutskever I, Hinton GE. Imagenet classification with deep convolutional neural networks. Adv Neural Inf Process Syst. 2012; 1097–1105. [Google Scholar]
- 26.Brown TB, Mann B, Ryder N, et al. Language models are few-shot learners. arXiv preprint arXiv:200514165 2020. [Google Scholar]
- 27.Deng J, Dong W, Socher R, et al. Imagenet: A large-scale hierarchical image database. Proc IEEE Comput Soc Conf Comput Vis Pattern Recognit. 2009; 248–255. [Google Scholar]
- 28.Marcantoni C, Ma L-J, Federspiel C, et al. Hypertensive nephrosclerosis in African Americans versus caucasians. Kidney Int. 2002; 62: 172–180. [DOI] [PubMed] [Google Scholar]
- 29.Marsh JN, Matlock MK, Kudose S, et al. Deep learning global glomerulosclerosis in transplant kidney frozen sections. IEEE Trans Med Imaging. 2018; 37: 2718–2728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Barros GO, Navarro B, Duarte A, et al. PathoSpotter-K: A computational tool for the automatic identification of glomerular lesions in histological images of kidneys. Sci Rep. 2017; 7: 46769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hermsen M, de Bel T, Den Boer M, et al. Deep Learning–Based Histopathologic Assessment of Kidney Tissue. J Am Soc Nephrol. 2019; 30: 1968–1979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Santo BA, Rosenberg AZ, Sarder P. Artificial intelligence driven next-generation renal histomorphometry. Curr Opin Nephrol Hypertens. 2020; 29: 265–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Østergaard MV, Sembach FE, Skytte JL, et al. Automated image analyses of glomerular hypertrophy in a mouse model of diabetic nephropathy. Kidney360. 2020: 10.34067/KID.0001272019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chen H-H, Lee T-T, Chen A, et al. 3D digital pathology for a chemical-functional analysis of glomeruli in health and pathology. Anal Chem. 2018; 90: 3811–3818. [DOI] [PubMed] [Google Scholar]
- 35.Torkamani N, Jerums G, Crammer P, et al. Three dimensional glomerular reconstruction: A novel approach to evaluate renal microanatomy in diabetic kidney disease. Sci Rep. 2019; 9: 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Niel O, Bastard P. Artificial intelligence in nephrology: core concepts, clinical applications, and perspectives. Am J Kidney Dis. 2019; 74: 803–810. [DOI] [PubMed] [Google Scholar]
- 37.Barisoni L, Gimpel C, Kain R, et al. Digital pathology imaging as a novel platform for standardization and globalization of quantitative nephropathology. Clin Kidney J. 2017; 10: 176–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yadav V, Bethard S. A survey on recent advances in named entity recognition from deep learning models. arXiv preprint arXiv:191011470 2019. [Google Scholar]
- 39.Lutnick B, Ginley B, Govind D, et al. An integrated iterative annotation technique for easing neural network training in medical image analysis. Nat Mach Intell. 2019; 1: 112–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zhu Z, Lu Y, Deng R, et al. EasierPath: An Open-source Tool for Human-in-the-loop Deep Learning of Renal Pathology. arXiv preprint arXiv:200713952 2020. [Google Scholar]
- 41.Rangan GK, Tesch GH. Quantification of renal pathology by image analysis. Nephrology 2007; 12: 553–558. [DOI] [PubMed] [Google Scholar]
- 42.Tschuchnig ME, Oostingh GJ, Gadermayr M. Generative Adversarial Networks in Digital Pathology: A Survey on Trends and Future Potential. arXiv preprint arXiv:200414936 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ginley B, Lutnick B, Jen K-Y, et al. Computational segmentation and classification of diabetic glomerulosclerosis. J Am Soc Nephrol. 2019; 30: 1953–1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Uchino E, Suzuki K, Sato N, et al. Classification of glomerular pathological findings using deep learning and nephrologist-AI collective intelligence approach. medRxiv. 2020: 2019.2012. 2030.19016162. [DOI] [PubMed] [Google Scholar]
- 45.Temerinac-Ott M, Forestier G, Schmitz J, et al. Detection of glomeruli in renal pathology by mutual comparison of multiple staining modalities. Proc Int Symp Image Signal Process Anal. 2009; 19–24. [Google Scholar]
- 46.Bukowy JD, Dayton A, Cloutier D, et al. Region-based convolutional neural nets for localization of glomeruli in trichrome-stained whole kidney sections. J Am Soc Nephrol. 2018; 29: 2081–2088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Simon O, Yacoub R, Jain S, et al. Multi-radial LBP features as a tool for rapid glomerular detection and assessment in whole slide histopathology images. Sci Rep. 2018; 8: 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Marée R, Dallongeville S, Olivo-Marin J-C, et al. An approach for detection of glomeruli in multisite digital pathology. Proc IEEE Int Symp Biomed Imaging. 2016; 1033–1036. [Google Scholar]
- 49.Bueno G, Fernandez-Carrobles MM, Gonzalez-Lopez L, et al. Glomerulosclerosis identification in whole slide images using semantic segmentation. Comput Methods Programs Biomed. 2020; 184: 105273. [DOI] [PubMed] [Google Scholar]
- 50.Kannan S, Morgan LA, Liang B, et al. Segmentation of glomeruli within trichrome images using deep learning. Kidney Int Rep. 2019; 4: 955–962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gadermayr M, Dombrowski A-K, Klinkhammer BM, et al. CNN cascades for segmenting whole slide images of the kidney. arXiv preprint arXiv:170800251 2017. [Google Scholar]
- 52.Gadermayr M, Eschweiler D, Jeevanesan A, et al. Segmenting renal whole slide images virtually without training data. Comput Biol Med. 2017; 90: 88–97. [DOI] [PubMed] [Google Scholar]
- 53.Ginley B, Jen K-Y, Rosenberg A, et al. Neural network segmentation of interstitial fibrosis, tubular atrophy, and glomerulosclerosis in renal biopsies. arXiv preprint arXiv:200212868 2020. [Google Scholar]
- 54.Barisoni L, Lafata KJ, Hewitt SM, et al. Digital pathology and computational image analysis in nephropathology. Nat Rev Nephrol. 2020: 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ginley B, Jen K-Y, Rosenberg A, et al. Fully automated classification of glomerular lesions in lupus nephritis. Proc SPIE Int Soc Opt Eng. 2020; 11320:113200Y. [PMC free article] [PubMed] [Google Scholar]
- 56.Zee J, Hodgin JB, Mariani LH, et al. Reproducibility and feasibility of strategies for morphologic assessment of renal biopsies using the Nephrotic Syndrome Study Network digital pathology scoring system. Arch Pathol Lab Med. 2018; 142: 613–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chagas P, Souza L, Araújo I, et al. Classification of glomerular hypercellularity using convolutional features and support vector machine. Artif Intell Med. 2020; 103: 101808. [DOI] [PubMed] [Google Scholar]
- 58.Ledbetter D, Ho L, Lemley KV. Prediction of kidney function from biopsy images using convolutional neural networks. arXiv preprint arXiv:1702.01816 2017. [Google Scholar]
- 59.He K, Zhang X, Ren S, et al. Deep residual learning for image recognition. Proc IEEE Comput Soc Conf Comput Vis Pattern Recognit. 2016; 770–778. [Google Scholar]
- 60.Hastie T, Tibshirani R, Friedman J. The elements of statistical learning: data mining, inference, and prediction. Springer Science & Business Media, 2009. [Google Scholar]
- 61.Kato T, Relator R, Ngouv H, et al. Segmental HOG: new descriptor for glomerulus detection in kidney microscopy image. Bmc Bioinformatics. 2015; 16: 316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gallego J, Pedraza A, Lopez S, et al. Glomerulus classification and detection based on convolutional neural networks. J. Imaging 2018; 4: 20. [Google Scholar]
- 63.Ren S, He K, Girshick R, et al. Faster r-cnn: Towards real-time object detection with region proposal networks. Adv Neural Inf Process Syst. 2015; 91–99. [DOI] [PubMed] [Google Scholar]
- 64.Zhao ZQ, Zheng P, Xu ST, et al. Object detection with deep learning: A review. IEEE Trans Neural Netw Learn Syst. 2019; 30: 3212–3232. [DOI] [PubMed] [Google Scholar]
- 65.Wu X, Sahoo D, Hoi SC. Recent advances in deep learning for object detection. Neurocomputing. 2020. [Google Scholar]
- 66.Zhang Y, Chen H, Wei Y, et al. From whole slide imaging to microscopy: Deep microscopy adaptation network for histopathology cancer image classification. Med Image Comput Comput Assist Interv. 2019; 360–368. [Google Scholar]
- 67.Santo BA, Segal BH, Tomaszewski JE, et al. Neutrophil Extracellular Traps (NETs): an unexplored territory in renal pathobiology, a pilot computational study. Proc SPIE Int Soc Opt Eng. 2020; 11320: 1132012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Gupta L, Klinkhammer BM, Boor P, et al. Iterative learning to make the most of unlabeled and quickly obtained labeled data in histology. Proc Machine Learning Res. 2019; 102: 215–224. [Google Scholar]
- 69.Ginley B, Tomaszewski JE, Yacoub R, et al. Unsupervised labeling of glomerular boundaries using Gabor filters and statistical testing in renal histology. J Med Imaging 2017; 4: 021102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Sarder P, Ginley B, Tomaszewski JE. Automated renal histopathology: Digital extraction and quantification of renal pathology. Proc SPIE Int Soc Opt Eng. 2020; 9791: 97910F. [Google Scholar]
- 71.Tey WK, Kuang YC, Ooi MP-L, et al. Automated quantification of renal interstitial fibrosis for computer-aided diagnosis: A comprehensive tissue structure segmentation method. Comput Methods Programs Biomed. 2018; 155: 109–120. [DOI] [PubMed] [Google Scholar]
- 72.Altini N, Cascarano GD, Brunetti A, et al. A Deep Learning Instance Segmentation Approach for Global Glomerulosclerosis Assessment in Donor Kidney Biopsies. Electronics. 2020; 9: 1768. [Google Scholar]
- 73.Bouteldja N, Klinkhammer BM, Bülow RD, et al. Deep Learning–Based Segmentation and Quantification in Experimental Kidney Histopathology. J Am Soc Nephrol. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Jayapandian CP., Chen Y, Janowczyk AR, et al. Development and evaluation of deep learning–based segmentation of histologic structures in the kidney cortex with multiple histologic stains. Kidney Int. 2020; August 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Long J, Shelhamer E, Darrell T. Fully convolutional networks for semantic segmentation. Proc IEEE Comput Soc Conf Comput Vis Pattern Recognit. 2015; 3431–3440. [DOI] [PubMed] [Google Scholar]
- 76.He K, Gkioxari G, Dollár P, et al. Mask r-cnn. Proc IEEE Int Conf Comput Vis. 2017; 2961–2969. [Google Scholar]
- 77.Goodfellow I, Pouget-Abadie J, Mirza M, et al. Generative adversarial nets. Adv Neural Inf Process Syst. 2014; 2672–2680 [Google Scholar]
- 78.Murali LK, Lutnick B, Ginley B, et al. Generative modeling for renal microanatomy. Proc SPIE Int Soc Opt Eng. 2020; 11320:113200F. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lutnick B, Ginley B, Jen K-Y, et al. Generative modeling for label-free glomerular modeling and classification. Proc SPIE Int Soc Opt Eng. 2020; 11320: 1132007. [PMC free article] [PubMed] [Google Scholar]
- 80.Benjdira B, Bazi Y, Koubaa A, et al. Unsupervised domain adaptation using generative adversarial networks for semantic segmentation of aerial images. Remote Sens. 2019; 11: 1369. [Google Scholar]
- 81.Zhang Y, de Haan K, Rivenson Y, et al. Digital synthesis of histological stains using micro-structured and multiplexed virtual staining of label-free tissue. Light: Sci & Appl. 2020; 9: 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Gadermayr M, Appel V, Klinkhammer BM, et al. Which way round? a study on the performance of stain-translation for segmenting arbitrarily dyed histological images. Med Image Comput Comput Assist Interv. 2018; 165–173. [Google Scholar]
- 83.Gadermayr M, Gupta L, Appel V, et al. Generative adversarial networks for facilitating stain-independent supervised and unsupervised segmentation: a study on kidney histology. IEEE Trans Med Imaging. 2019; 38: 2293–2302. [DOI] [PubMed] [Google Scholar]
- 84.de Bel T, Hermsen M, Kers J, et al. Stain-Transforming Cycle-Consistent Generative Adversarial Networks for Improved Segmentation of Renal Histopathology. Med Imaging Deep Learning. 2020; 151–163. [Google Scholar]
- 85.BenTaieb A, Hamarneh G Adversarial stain transfer for histopathology image analysis. IEEE Trans Med Imaging. 2017; 37: 792–802. [DOI] [PubMed] [Google Scholar]
- 86.Wu B, Zhang X, Zhao S, et al. G2C: a generator-to-classifier framework integrating multi-stained visual cues for pathological glomerulus classification. Proc Conf AAAI Artif Intell. 2019; 33: 1214–1221 [Google Scholar]
- 87.Kanayama T, Kurose Y, Tanaka K, et al. Gastric cancer detection from endoscopic images using synthesis by GAN. Med Image Comput Comput Assist Interv. 2019; 530–538. [Google Scholar]
- 88.Gadermayr M, Gupta L, Klinkhammer BM, et al. Unsupervisedly training GANs for segmenting digital pathology with automatically generated annotations. Med Imaging Deep Learning. 2019; 102: 175–184. [Google Scholar]
- 89.Gupta L, Klinkhammer BM, Boor P, et al. GAN-Based Image Enrichment in Digital Pathology Boosts Segmentation Accuracy. Med Image Comput Comput Assist Interv. 2019; 631–639. [Google Scholar]
- 90.Mei K, Zhu C, Jiang L, et al. Cross-stained segmentation from renal biopsy images using multi-level adversarial learning. IEEE ICASSP. 2019; 1424–1428. [Google Scholar]
- 91.Yang H, Deng R, Lu Y, et al. CircleNet: Anchor-free glomerulus detection with circle representation. Med Image Comput Comput Assist Interv. 2020; 35–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Jha A, Yang H, Deng R, et al. Instance segmentation for whole slide imaging: End-to-end or detect-then-segment. J Med Imaging. 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Chang A, Gibson IW, Cohen AH, et al. A position paper on standardizing the nonneoplastic kidney biopsy report. Human pathology. 2012; 43: 1192–1196. [DOI] [PubMed] [Google Scholar]
- 94.Sethi S, Haas M, Markowitz GS, et al. Mayo clinic/renal pathology society consensus report on pathologic classification, diagnosis, and reporting of GN. J Am Soc Nephrol. 2016; 27: 1278–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Cai Q, Wang H, Li Z, et al. A Survey on Multimodal Data-Driven Smart Healthcare Systems: Approaches and Applications. IEEE Access. 2019; 7: 133583–133599. [Google Scholar]
- 96.Domingues I, Müller H, Ortiz A, et al. Guest Editorial: Information Fusion for Medical Data: Early, Late, and Deep Fusion Methods for Multimodal Data. IEEE J Biomed Health Inform. 2020; 24: 14–16. [Google Scholar]
- 97.Lippolis G, Edsjö A, Helczynski L, et al. Automatic registration of multi-modal microscopy images for integrative analysis of prostate tissue sections. BMC cancer. 2013; 13: 408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Moncada R, Wagner F, Chiodin M, et al. Building a tumor atlas: integrating single-cell RNA-Seq data with spatial transcriptomics in pancreatic ductal adenocarcinoma. bioRxiv 2018: 254375. [Google Scholar]
- 99.Ronneberger O, Fischer P, Brox T. U-net: Convolutional networks for biomedical image segmentation. Med Image Comput Comput Assist Interv 2015; 234–241. [Google Scholar]
- 100.D’Agati VD, Mengel M. The rise of renal pathology in nephrology: structure illuminates function. Am J Kidney Dis. 2013; 61: 1016–1025. [DOI] [PubMed] [Google Scholar]
- 101.Zucker RM, Rogers J, Ellis-Hutchings R. Confocal Microscopy of Thick Tissue Sections: 3D Visualization of Rat Kidney Glomeruli. Microscopy and Microanalysis. 2008; 14: 730–731. [Google Scholar]
- 102.Neal CR, Crook H, Bell E, et al. Three-dimensional reconstruction of glomeruli by electron microscopy reveals a distinct restrictive urinary subpodocyte space. J Am Soc Nephrol. 2005; 16: 1223–1235. [DOI] [PubMed] [Google Scholar]
- 103.Arkill KP, Qvortrup K, Starborg T, et al. Resolution of the three dimensional structure of components of the glomerular filtration barrier. BMC nephrology. 2014; 15: 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Terasaki M, Brunson JC, Sardi J. Analysis of the three dimensional structure of the kidney glomerulus capillary network. bioRxiv 2019: 677864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Deng R, Yang H, Jha A, et al. Map3D: Registration based multi-object tracking on 3D serial whole slide images. arXiv preprint arXiv:200606038 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Steiner DF, MacDonald R, Liu Y, et al. Impact of deep learning assistance on the histopathologic review of lymph nodes for metastatic breast cancer. Am J Surg Pathol. 2018; 42: 1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kingma DP, Welling M. Auto-encoding variational bayes. arXiv preprint arXiv:13126114 2013. [Google Scholar]
- 108.Katzman JL, Shaham U, Cloninger A, et al. DeepSurv: personalized treatment recommender system using a Cox proportional hazards deep neural network. BMC Med Res Methodol. 2018; 18: 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Thomas D, Franceschini N, Hogan S, et al. Clinical and pathologic characteristics of focal segmental glomerulosclerosis pathologic variants. Kidney Int. 2006; 69: 920–926. [DOI] [PubMed] [Google Scholar]
- 110.Austin HA, Boumpas DT, Vaughan EM, et al. Predicting renal outcomes in severe lupus nephritis: contributions of clinical and histologic data. Kidney Int. 1994; 45: 544–550. [DOI] [PubMed] [Google Scholar]
- 111.Alamartine E, Sauron C, Laurent B, et al. The use of the Oxford classification of IgA nephropathy to predict renal survival. Clin J Am Soc Nephrol. 2011; 6: 2384–2388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Lemley KV, Bagnasco SM, Nast CC, et al. Morphometry predicts early GFR change in primary proteinuric glomerulopathies: a longitudinal cohort study using generalized estimating equations. PloS One. 2016; 11: e0157148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Grootscholten C, Bajema IM, Florquin S, et al. Interobserver agreement of scoring of histopathological characteristics and classification of lupus nephritis. Nephrol Dial Transplant. 2008; 23: 223–230. [DOI] [PubMed] [Google Scholar]
- 114.Kolachalama VB, Singh P, Lin CQ, et al. Association of pathological fibrosis with renal survival using deep neural networks. Kidney Int Rep. 2018; 3: 464–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Heerspink HJ, Ninomiya T, Persson F, et al. Is a reduction in albuminuria associated with renal and cardiovascular protection? A post hoc analysis of the ALTITUDE trial. Diabetes Obes Metab. 2016; 18: 169–177. [DOI] [PubMed] [Google Scholar]
- 116.Thongprayoon C, Kaewput W, Kovvuru K, et al. : Promises of big data and artificial intelligence in nephrology and transplantation. J Clin Med. 2020; 9: 1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Yu C, Liu J, Nemati S. Reinforcement learning in healthcare: A survey. arXiv preprint arXiv:190808796 2019. [Google Scholar]
- 118.Qaiser T, Rajpoot NM. Learning where to see: A novel attention model for automated immunohistochemical scoring. IEEE Trans Med Imaging. 2019; 38: 2620–2631. [DOI] [PubMed] [Google Scholar]
- 119.Iorio F, Knijnenburg TA, Vis DJ, et al. A landscape of pharmacogenomic interactions in cancer. Cell. 2016; 166: 740–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Holzinger A, Malle B, Kieseberg P, et al. Towards the augmented pathologist: Challenges of explainable-AI in digital pathology. arXiv preprint arXiv:171206657 2017. [Google Scholar]
- 121.Zhang Z, Xie Y, Xing F, et al. MDNet: A Semantically and Visually Interpretable Medical Image Diagnosis Network. IEEE Comput Soc Conf Comput Vis Pattern Recognit. 2017; 6428–6436. [Google Scholar]
- 122.Selvaraju RR, Cogswell M, Das A, et al. Grad-cam: Visual explanations from deep networks via gradient-based localization. IEEE Comput Soc Conf Comput Vis Pattern Recognit. 2017; 618–626. [Google Scholar]
- 123.Ribeiro MT, Singh S, Guestrin C. “ Why should I trust you?” Explaining the predictions of any classifier. Proceedings of the 22nd ACM SIGKDD. 2016; 1135–1144 [Google Scholar]
