Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
J Transl Med ; 22(1): 443, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730319

ABSTRACT

BACKGROUND: The immune microenvironment impacts tumor growth, invasion, metastasis, and patient survival and may provide opportunities for therapeutic intervention in pancreatic ductal adenocarcinoma (PDAC). Although never studied as a potential modulator of the immune response in most cancers, Keratin 17 (K17), a biomarker of the most aggressive (basal) molecular subtype of PDAC, is intimately involved in the histogenesis of the immune response in psoriasis, basal cell carcinoma, and cervical squamous cell carcinoma. Thus, we hypothesized that K17 expression could also impact the immune cell response in PDAC, and that uncovering this relationship could provide insight to guide the development of immunotherapeutic opportunities to extend patient survival. METHODS: Multiplex immunohistochemistry (mIHC) and automated image analysis based on novel computational imaging technology were used to decipher the abundance and spatial distribution of T cells, macrophages, and tumor cells, relative to K17 expression in 235 PDACs. RESULTS: K17 expression had profound effects on the exclusion of intratumoral CD8+ T cells and was also associated with decreased numbers of peritumoral CD8+ T cells, CD16+ macrophages, and CD163+ macrophages (p < 0.0001). The differences in the intratumor and peritumoral CD8+ T cell abundance were not impacted by neoadjuvant therapy, tumor stage, grade, lymph node status, histologic subtype, nor KRAS, p53, SMAD4, or CDKN2A mutations. CONCLUSIONS: Thus, K17 expression correlates with major differences in the immune microenvironment that are independent of any tested clinicopathologic or tumor intrinsic variables, suggesting that targeting K17-mediated immune effects on the immune system could restore the innate immunologic response to PDAC and might provide novel opportunities to restore immunotherapeutic approaches for this most deadly form of cancer.


Subject(s)
Keratin-17 , Pancreatic Neoplasms , Humans , Keratin-17/metabolism , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Tumor Microenvironment/immunology , Female , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Male , CD8-Positive T-Lymphocytes/immunology , Macrophages/metabolism , Macrophages/immunology , Middle Aged , Aged , Receptors, Cell Surface , Antigens, Differentiation, Myelomonocytic , Antigens, CD
2.
IEEE Winter Conf Appl Comput Vis ; 2024: 5170-5179, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38808304

ABSTRACT

To achieve high-quality results, diffusion models must be trained on large datasets. This can be notably prohibitive for models in specialized domains, such as computational pathology. Conditioning on labeled data is known to help in data-efficient model training. Therefore, histopathology reports, which are rich in valuable clinical information, are an ideal choice as guidance for a histopathology generative model. In this paper, we introduce PathLDM, the first text-conditioned Latent Diffusion Model tailored for generating high-quality histopathology images. Leveraging the rich contextual information provided by pathology text reports, our approach fuses image and textual data to enhance the generation process. By utilizing GPT's capabilities to distill and summarize complex text reports, we establish an effective conditioning mechanism. Through strategic conditioning and necessary architectural enhancements, we achieved a SoTA FID score of 7.64 for text-to-image generation on the TCGA-BRCA dataset, significantly outperforming the closest text-conditioned competitor with FID 30.1.

3.
Res Sq ; 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38464123

ABSTRACT

Background: The immune microenvironment impacts tumor growth, invasion, metastasis, and patient survival and may provide opportunities for therapeutic intervention in pancreatic ductal adenocarcinoma (PDAC). Although never studied as a potential modulator of the immune response in most cancers, Keratin 17 (K17), a biomarker of the most aggressive (basal) molecular subtype of PDAC, is intimately involved in the histogenesis of the immune response in psoriasis, basal cell carcinoma, and cervical squamous cell carcinoma. Thus, we hypothesized that K17 expression could also impact the immune cell response in PDAC, and that uncovering this relationship could provide insight to guide the development of immunotherapeutic opportunities to extend patient survival. Methods: Multiplex immunohistochemistry (mIHC) and automated image analysis based on novel computational imaging technology were used to decipher the abundance and spatial distribution of T cells, macrophages, and tumor cells, relative to K17 expression in 235 PDACs. Results: K17 expression had profound effects on the exclusion of intratumoral CD8 + T cells and was also associated with decreased numbers of peritumoral CD8 + T cells, CD16 + macrophages, and CD163 + macrophages (p < 0.0001). The differences in the intratumor and peritumoral CD8 + T cell abundance were not impacted by neoadjuvant therapy, tumor stage, grade, lymph node status, histologic subtype, nor KRAS, p53, SMAD4, or CDKN2A mutations. Conclusions: Thus, K17 expression correlates with major differences in the immune microenvironment that are independent of any tested clinicopathologic or tumor intrinsic variables, suggesting that targeting K17-mediated immune effects on the immune system could restore the innate immunologic response to PDAC and might provide novel opportunities to restore immunotherapeutic approaches for this most deadly form of cancer.

4.
Cancer Inform ; 23: 11769351231223806, 2024.
Article in English | MEDLINE | ID: mdl-38322427

ABSTRACT

Large-scale, multi-site collaboration is becoming indispensable for a wide range of research and clinical activities in oncology. To facilitate the next generation of advances in cancer biology, precision oncology and the population sciences it will be necessary to develop and implement data management and analytic tools that empower investigators to reliably and objectively detect, characterize and chronicle the phenotypic and genomic changes that occur during the transformation from the benign to cancerous state and throughout the course of disease progression. To facilitate these efforts it is incumbent upon the informatics community to establish the workflows and architectures that automate the aggregation and organization of a growing range and number of clinical data types and modalities ranging from new molecular and laboratory tests to sophisticated diagnostic imaging studies. In an attempt to meet those challenges, leading health care centers across the country are making steep investments to establish enterprise-wide, data warehouses. A significant limitation of many data warehouses, however, is that they are designed to support only alphanumeric information. In contrast to those traditional designs, the system that we have developed supports automated collection and mining of multimodal data including genomics, digital pathology and radiology images. In this paper, our team describes the design, development and implementation of a multi-modal, Clinical & Research Data Warehouse (CRDW) that is tightly integrated with a suite of computational and machine-learning tools to provide actionable insight into the underlying characteristics of the tumor environment that would not be revealed using standard methods and tools. The System features a flexible Extract, Transform and Load (ETL) interface that enables it to adapt to aggregate data originating from different clinical and research sources depending on the specific EHR and other data sources utilized at a given deployment site.

5.
Nat Methods ; 21(2): 182-194, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38347140

ABSTRACT

Validation metrics are key for tracking scientific progress and bridging the current chasm between artificial intelligence research and its translation into practice. However, increasing evidence shows that, particularly in image analysis, metrics are often chosen inadequately. Although taking into account the individual strengths, weaknesses and limitations of validation metrics is a critical prerequisite to making educated choices, the relevant knowledge is currently scattered and poorly accessible to individual researchers. Based on a multistage Delphi process conducted by a multidisciplinary expert consortium as well as extensive community feedback, the present work provides a reliable and comprehensive common point of access to information on pitfalls related to validation metrics in image analysis. Although focused on biomedical image analysis, the addressed pitfalls generalize across application domains and are categorized according to a newly created, domain-agnostic taxonomy. The work serves to enhance global comprehension of a key topic in image analysis validation.


Subject(s)
Artificial Intelligence
6.
Nat Methods ; 21(2): 195-212, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38347141

ABSTRACT

Increasing evidence shows that flaws in machine learning (ML) algorithm validation are an underestimated global problem. In biomedical image analysis, chosen performance metrics often do not reflect the domain interest, and thus fail to adequately measure scientific progress and hinder translation of ML techniques into practice. To overcome this, we created Metrics Reloaded, a comprehensive framework guiding researchers in the problem-aware selection of metrics. Developed by a large international consortium in a multistage Delphi process, it is based on the novel concept of a problem fingerprint-a structured representation of the given problem that captures all aspects that are relevant for metric selection, from the domain interest to the properties of the target structure(s), dataset and algorithm output. On the basis of the problem fingerprint, users are guided through the process of choosing and applying appropriate validation metrics while being made aware of potential pitfalls. Metrics Reloaded targets image analysis problems that can be interpreted as classification tasks at image, object or pixel level, namely image-level classification, object detection, semantic segmentation and instance segmentation tasks. To improve the user experience, we implemented the framework in the Metrics Reloaded online tool. Following the convergence of ML methodology across application domains, Metrics Reloaded fosters the convergence of validation methodology. Its applicability is demonstrated for various biomedical use cases.


Subject(s)
Algorithms , Image Processing, Computer-Assisted , Machine Learning , Semantics
7.
NPJ Precis Oncol ; 8(1): 9, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38200147

ABSTRACT

Digital pathology has seen a proliferation of deep learning models in recent years, but many models are not readily reusable. To address this challenge, we developed WSInfer: an open-source software ecosystem designed to streamline the sharing and reuse of deep learning models for digital pathology. The increased access to trained models can augment research on the diagnostic, prognostic, and predictive capabilities of digital pathology.

8.
ArXiv ; 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-36945687

ABSTRACT

Validation metrics are key for the reliable tracking of scientific progress and for bridging the current chasm between artificial intelligence (AI) research and its translation into practice. However, increasing evidence shows that particularly in image analysis, metrics are often chosen inadequately in relation to the underlying research problem. This could be attributed to a lack of accessibility of metric-related knowledge: While taking into account the individual strengths, weaknesses, and limitations of validation metrics is a critical prerequisite to making educated choices, the relevant knowledge is currently scattered and poorly accessible to individual researchers. Based on a multi-stage Delphi process conducted by a multidisciplinary expert consortium as well as extensive community feedback, the present work provides the first reliable and comprehensive common point of access to information on pitfalls related to validation metrics in image analysis. Focusing on biomedical image analysis but with the potential of transfer to other fields, the addressed pitfalls generalize across application domains and are categorized according to a newly created, domain-agnostic taxonomy. To facilitate comprehension, illustrations and specific examples accompany each pitfall. As a structured body of information accessible to researchers of all levels of expertise, this work enhances global comprehension of a key topic in image analysis validation.

9.
Comput Methods Programs Biomed ; 239: 107631, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37271050

ABSTRACT

BACKGROUND AND OBJECTIVE: Histopathology is the gold standard for diagnosis of many cancers. Recent advances in computer vision, specifically deep learning, have facilitated the analysis of histopathology images for many tasks, including the detection of immune cells and microsatellite instability. However, it remains difficult to identify optimal models and training configurations for different histopathology classification tasks due to the abundance of available architectures and the lack of systematic evaluations. Our objective in this work is to present a software tool that addresses this need and enables robust, systematic evaluation of neural network models for patch classification in histology in a light-weight, easy-to-use package for both algorithm developers and biomedical researchers. METHODS: Here we present ChampKit (Comprehensive Histopathology Assessment of Model Predictions toolKit): an extensible, fully reproducible evaluation toolkit that is a one-stop-shop to train and evaluate deep neural networks for patch classification. ChampKit curates a broad range of public datasets. It enables training and evaluation of models supported by timm directly from the command line, without the need for users to write any code. External models are enabled through a straightforward API and minimal coding. As a result, Champkit facilitates the evaluation of existing and new models and deep learning architectures on pathology datasets, making it more accessible to the broader scientific community. To demonstrate the utility of ChampKit, we establish baseline performance for a subset of possible models that could be employed with ChampKit, focusing on several popular deep learning models, namely ResNet18, ResNet50, and R26-ViT, a hybrid vision transformer. In addition, we compare each model trained either from random weight initialization or with transfer learning from ImageNet pretrained models. For ResNet18, we also consider transfer learning from a self-supervised pretrained model. RESULTS: The main result of this paper is the ChampKit software. Using ChampKit, we were able to systemically evaluate multiple neural networks across six datasets. We observed mixed results when evaluating the benefits of pretraining versus random intialization, with no clear benefit except in the low data regime, where transfer learning was found to be beneficial. Surprisingly, we found that transfer learning from self-supervised weights rarely improved performance, which is counter to other areas of computer vision. CONCLUSIONS: Choosing the right model for a given digital pathology dataset is nontrivial. ChampKit provides a valuable tool to fill this gap by enabling the evaluation of hundreds of existing (or user-defined) deep learning models across a variety of pathology tasks. Source code and data for the tool are freely accessible at https://github.com/SBU-BMI/champkit.


Subject(s)
Neoplasms , Neural Networks, Computer , Humans , Algorithms , Software , Histological Techniques
10.
Bioinformatics ; 39(4)2023 04 03.
Article in English | MEDLINE | ID: mdl-36943380

ABSTRACT

MOTIVATION: Deep learning attained excellent results in digital pathology recently. A challenge with its use is that high quality, representative training datasets are required to build robust models. Data annotation in the domain is labor intensive and demands substantial time commitment from expert pathologists. Active learning (AL) is a strategy to minimize annotation. The goal is to select samples from the pool of unlabeled data for annotation that improves model accuracy. However, AL is a very compute demanding approach. The benefits for model learning may vary according to the strategy used, and it may be hard for a domain specialist to fine tune the solution without an integrated interface. RESULTS: We developed a framework that includes a friendly user interface along with run-time optimizations to reduce annotation and execution time in AL in digital pathology. Our solution implements several AL strategies along with our diversity-aware data acquisition (DADA) acquisition function, which enforces data diversity to improve the prediction performance of a model. In this work, we employed a model simplification strategy [Network Auto-Reduction (NAR)] that significantly improves AL execution time when coupled with DADA. NAR produces less compute demanding models, which replace the target models during the AL process to reduce processing demands. An evaluation with a tumor-infiltrating lymphocytes classification application shows that: (i) DADA attains superior performance compared to state-of-the-art AL strategies for different convolutional neural networks (CNNs), (ii) NAR improves the AL execution time by up to 4.3×, and (iii) target models trained with patches/data selected by the NAR reduced versions achieve similar or superior classification quality to using target CNNs for data selection. AVAILABILITY AND IMPLEMENTATION: Source code: https://github.com/alsmeirelles/DADA.


Subject(s)
Deep Learning , Neural Networks, Computer , Software , Image Processing, Computer-Assisted , Data Curation
11.
Article in English | MEDLINE | ID: mdl-38741683

ABSTRACT

In digital pathology, the spatial context of cells is important for cell classification, cancer diagnosis and prognosis. To model such complex cell context, however, is challenging. Cells form different mixtures, lineages, clusters and holes. To model such structural patterns in a learnable fashion, we introduce several mathematical tools from spatial statistics and topological data analysis. We incorporate such structural descriptors into a deep generative model as both conditional inputs and a differentiable loss. This way, we are able to generate high quality multi-class cell layouts for the first time. We show that the topology-rich cell layouts can be used for data augmentation and improve the performance of downstream tasks such as cell classification.

12.
Proc Mach Learn Res ; 227: 74-94, 2023 Jul.
Article in English | MEDLINE | ID: mdl-38817539

ABSTRACT

Multiplex Immunohistochemistry (mIHC) is a cost-effective and accessible method for in situ labeling of multiple protein biomarkers in a tissue sample. By assigning a different stain to each biomarker, it allows the visualization of different types of cells within the tumor vicinity for downstream analysis. However, to detect different types of stains in a given mIHC image is a challenging problem, especially when the number of stains is high. Previous deep-learning-based methods mostly assume full supervision; yet the annotation can be costly. In this paper, we propose a novel unsupervised stain decomposition method to detect different stains simultaneously. Our method does not require any supervision, except for color samples of different stains. A main technical challenge is that the problem is underdetermined and can have multiple solutions. To conquer this issue, we propose a novel inversion regulation technique, which eliminates most undesirable solutions. On a 7-plexed IHC images dataset, the proposed method achieves high quality stain decomposition results without human annotation.

13.
Bioinformatics ; 38(14): 3629-3637, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35674341

ABSTRACT

MOTIVATION: Whole slide tissue images contain detailed data on the sub-cellular structure of cancer. Quantitative analyses of this data can lead to novel biomarkers for better cancer diagnosis and prognosis and can improve our understanding of cancer mechanisms. Such analyses are challenging to execute because of the sizes and complexity of whole slide image data and relatively limited volume of training data for machine learning methods. RESULTS: We propose and experimentally evaluate a multi-resolution deep learning method for breast cancer survival analysis. The proposed method integrates image data at multiple resolutions and tumor, lymphocyte and nuclear segmentation results from deep learning models. Our results show that this approach can significantly improve the deep learning model performance compared to using only the original image data. The proposed approach achieves a c-index value of 0.706 compared to a c-index value of 0.551 from an approach that uses only color image data at the highest image resolution. Furthermore, when clinical features (sex, age and cancer stage) are combined with image data, the proposed approach achieves a c-index of 0.773. AVAILABILITY AND IMPLEMENTATION: https://github.com/SBU-BMI/deep_survival_analysis.


Subject(s)
Breast Neoplasms , Deep Learning , Humans , Female , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Machine Learning , Survival Analysis , Image Processing, Computer-Assisted/methods
14.
Front Med (Lausanne) ; 9: 894430, 2022.
Article in English | MEDLINE | ID: mdl-35712087

ABSTRACT

Background: Deep learning methods have demonstrated remarkable performance in pathology image analysis, but they are computationally very demanding. The aim of our study is to reduce their computational cost to enable their use with large tissue image datasets. Methods: We propose a method called Network Auto-Reduction (NAR) that simplifies a Convolutional Neural Network (CNN) by reducing the network to minimize the computational cost of doing a prediction. NAR performs a compound scaling in which the width, depth, and resolution dimensions of the network are reduced together to maintain a balance among them in the resulting simplified network. We compare our method with a state-of-the-art solution called ResRep. The evaluation is carried out with popular CNN architectures and a real-world application that identifies distributions of tumor-infiltrating lymphocytes in tissue images. Results: The experimental results show that both ResRep and NAR are able to generate simplified, more efficient versions of ResNet50 V2. The simplified versions by ResRep and NAR require 1.32× and 3.26× fewer floating-point operations (FLOPs), respectively, than the original network without a loss in classification power as measured by the Area under the Curve (AUC) metric. When applied to a deeper and more computationally expensive network, Inception V4, NAR is able to generate a version that requires 4× lower than the original version with the same AUC performance. Conclusions: NAR is able to achieve substantial reductions in the execution cost of two popular CNN architectures, while resulting in small or no loss in model accuracy. Such cost savings can significantly improve the use of deep learning methods in digital pathology. They can enable studies with larger tissue image datasets and facilitate the use of less expensive and more accessible graphics processing units (GPUs), thus reducing the computing costs of a study.

15.
Cancers (Basel) ; 14(9)2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35565277

ABSTRACT

Tumor-infiltrating lymphocytes (TILs) have been established as a robust prognostic biomarker in breast cancer, with emerging utility in predicting treatment response in the adjuvant and neoadjuvant settings. In this study, the role of TILs in predicting overall survival and progression-free interval was evaluated in two independent cohorts of breast cancer from the Cancer Genome Atlas (TCGA BRCA) and the Carolina Breast Cancer Study (UNC CBCS). We utilized machine learning and computer vision algorithms to characterize TIL infiltrates in digital whole-slide images (WSIs) of breast cancer stained with hematoxylin and eosin (H&E). Multiple parameters were used to characterize the global abundance and spatial features of TIL infiltrates. Univariate and multivariate analyses show that large aggregates of peritumoral and intratumoral TILs (forests) were associated with longer survival, whereas the absence of intratumoral TILs (deserts) is associated with increased risk of recurrence. Patients with two or more high-risk spatial features were associated with significantly shorter progression-free interval (PFI). This study demonstrates the practical utility of Pathomics in evaluating the clinical significance of the abundance and spatial patterns of distribution of TIL infiltrates as important biomarkers in breast cancer.

16.
Comput Methods Programs Biomed ; 220: 106828, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35500506

ABSTRACT

BACKGROUND AND OBJECTIVE: Deep learning methods have demonstrated remarkable performance in pathology image analysis, but they require a large amount of annotated training data from expert pathologists. The aim of this study is to minimize the data annotation need in these analyses. METHODS: Active learning (AL) is an iterative approach to training deep learning models. It was used in our context with a Tumor Infiltrating Lymphocytes (TIL) classification task to minimize annotation. State-of-the-art AL methods were evaluated with the TIL application and we have proposed and evaluated a more efficient and effective AL acquisition method. The proposed method uses data grouping based on imaging features and model prediction uncertainty to select meaningful training samples (image patches). RESULTS: An experimental evaluation with a collection of cancer tissue images shows that: (i) Our approach reduces the number of patches required to attain a given AUC as compared to other approaches, and (ii) our optimization (subpooling) leads to AL execution time improvement of about 2.12×. CONCLUSIONS: This strategy enabled TIL based deep learning analyses using smaller annotation demand. We expect this approach may be used to build other analyses in digital pathology with fewer training samples.


Subject(s)
Lymphocytes, Tumor-Infiltrating , Neoplasms , Humans , Image Processing, Computer-Assisted , Lymphocytes, Tumor-Infiltrating/pathology , Neoplasms/diagnostic imaging , Neoplasms/pathology , Problem-Based Learning
17.
J Pathol Inform ; 13: 5, 2022.
Article in English | MEDLINE | ID: mdl-35136672

ABSTRACT

BACKGROUND: Population-based state cancer registries are an authoritative source for cancer statistics in the United States. They routinely collect a variety of data, including patient demographics, primary tumor site, stage at diagnosis, first course of treatment, and survival, on every cancer case that is reported across all U.S. states and territories. The goal of our project is to enrich NCI's Surveillance, Epidemiology, and End Results (SEER) registry data with high-quality population-based biospecimen data in the form of digital pathology, machine-learning-based classifications, and quantitative histopathology imaging feature sets (referred to here as Pathomics features). MATERIALS AND METHODS: As part of the project, the underlying informatics infrastructure was designed, tested, and implemented through close collaboration with several participating SEER registries to ensure consistency with registry processes, computational scalability, and ability to support creation of population cohorts that span multiple sites. Utilizing computational imaging algorithms and methods to both generate indices and search for matches makes it possible to reduce inter- and intra-observer inconsistencies and to improve the objectivity with which large image repositories are interrogated. RESULTS: Our team has created and continues to expand a well-curated repository of high-quality digitized pathology images corresponding to subjects whose data are routinely collected by the collaborating registries. Our team has systematically deployed and tested key, visual analytic methods to facilitate automated creation of population cohorts for epidemiological studies and tools to support visualization of feature clusters and evaluation of whole-slide images. As part of these efforts, we are developing and optimizing advanced search and matching algorithms to facilitate automated, content-based retrieval of digitized specimens based on their underlying image features and staining characteristics. CONCLUSION: To meet the challenges of this project, we established the analytic pipelines, methods, and workflows to support the expansion and management of a growing repository of high-quality digitized pathology and information-rich, population cohorts containing objective imaging and clinical attributes to facilitate studies that seek to discriminate among different subtypes of disease, stratify patient populations, and perform comparisons of tumor characteristics within and across patient cohorts. We have also successfully developed a suite of tools based on a deep-learning method to perform quantitative characterizations of tumor regions, assess infiltrating lymphocyte distributions, and generate objective nuclear feature measurements. As part of these efforts, our team has implemented reliable methods that enable investigators to systematically search through large repositories to automatically retrieve digitized pathology specimens and correlated clinical data based on their computational signatures.

18.
IEEE Open J Eng Med Biol ; 3: 218-226, 2022.
Article in English | MEDLINE | ID: mdl-36860498

ABSTRACT

Histopathologic evaluation of Hematoxylin & Eosin (H&E) stained slides is essential for disease diagnosis, revealing tissue morphology, structure, and cellular composition. Variations in staining protocols and equipment result in images with color nonconformity. Although pathologists compensate for color variations, these disparities introduce inaccuracies in computational whole slide image (WSI) analysis, accentuating data domain shift and degrading generalization. Current state-of-the-art normalization methods employ a single WSI as reference, but selecting a single WSI representative of a complete WSI-cohort is infeasible, inadvertently introducing normalization bias. We seek the optimal number of slides to construct a more representative reference based on composite/aggregate of multiple H&E density histograms and stain-vectors, obtained from a randomly selected WSI population (WSI-Cohort-Subset). We utilized 1,864 IvyGAP WSIs as a WSI-cohort, and built 200 WSI-Cohort-Subsets varying in size (from 1 to 200 WSI-pairs) using randomly selected WSIs. The WSI-pairs' mean Wasserstein Distances and WSI-Cohort-Subsets' standard deviations were calculated. The Pareto Principle defined the optimal WSI-Cohort-Subset size. The WSI-cohort underwent structure-preserving color normalization using the optimal WSI-Cohort-Subset histogram and stain-vector aggregates. Numerous normalization permutations support WSI-Cohort-Subset aggregates as representative of a WSI-cohort through WSI-cohort CIELAB color space swift convergence, as a result of the law of large numbers and shown as a power law distribution. We show normalization at the optimal (Pareto Principle) WSI-Cohort-Subset size and corresponding CIELAB convergence: a) Quantitatively, using 500 WSI-cohorts; b) Quantitatively, using 8,100 WSI-regions; c) Qualitatively, using 30 cellular tumor normalization permutations. Aggregate-based stain normalization may contribute in increasing computational pathology robustness, reproducibility, and integrity.

19.
Comput Methods Programs Biomed ; 208: 106291, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34333205

ABSTRACT

BACKGROUND AND OBJECTIVE: Computerized pathology image analysis is an important tool in research and clinical settings, which enables quantitative tissue characterization and can assist a pathologist's evaluation. The aim of our study is to systematically quantify and minimize uncertainty in output of computer based pathology image analysis. METHODS: Uncertainty quantification (UQ) and sensitivity analysis (SA) methods, such as Variance-Based Decomposition (VBD) and Morris One-At-a-Time (MOAT), are employed to track and quantify uncertainty in a real-world application with large Whole Slide Imaging datasets - 943 Breast Invasive Carcinoma (BRCA) and 381 Lung Squamous Cell Carcinoma (LUSC) patients. Because these studies are compute intensive, high-performance computing systems and efficient UQ/SA methods were combined to provide efficient execution. UQ/SA has been able to highlight parameters of the application that impact the results, as well as nuclear features that carry most of the uncertainty. Using this information, we built a method for selecting stable features that minimize application output uncertainty. RESULTS: The results show that input parameter variations significantly impact all stages (segmentation, feature computation, and survival analysis) of the use case application. We then identified and classified features according to their robustness to parameter variation, and using the proposed features selection strategy, for instance, patient grouping stability in survival analysis has been improved from in 17% and 34% for BRCA and LUSC, respectively. CONCLUSIONS: This strategy created more robust analyses, demonstrating that SA and UQ are important methods that may increase confidence digital pathology.


Subject(s)
Image Processing, Computer-Assisted , Humans , Uncertainty
20.
Sci Rep ; 11(1): 2809, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33531581

ABSTRACT

Accurate prognostic biomarkers in early-stage melanoma are urgently needed to stratify patients for clinical trials of adjuvant therapy. We applied a previously developed open source deep learning algorithm to detect tumor-infiltrating lymphocytes (TILs) in hematoxylin and eosin (H&E) images of early-stage melanomas. We tested whether automated digital (TIL) analysis (ADTA) improved accuracy of prediction of disease specific survival (DSS) based on current pathology standards. ADTA was applied to a training cohort (n = 80) and a cutoff value was defined based on a Receiver Operating Curve. ADTA was then applied to a validation cohort (n = 145) and the previously determined cutoff value was used to stratify high and low risk patients, as demonstrated by Kaplan-Meier analysis (p ≤ 0.001). Multivariable Cox proportional hazards analysis was performed using ADTA, depth, and ulceration as co-variables and showed that ADTA contributed to DSS prediction (HR: 4.18, CI 1.51-11.58, p = 0.006). ADTA provides an effective and attainable assessment of TILs and should be further evaluated in larger studies for inclusion in staging algorithms.


Subject(s)
Image Processing, Computer-Assisted , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/mortality , Skin Neoplasms/mortality , Skin/pathology , Adult , Aged , Aged, 80 and over , Biopsy , Chemotherapy, Adjuvant , Clinical Decision-Making/methods , Deep Learning , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Melanoma/diagnosis , Melanoma/pathology , Melanoma/therapy , Middle Aged , Neoplasm Staging , Patient Selection , Prognosis , ROC Curve , Retrospective Studies , Risk Assessment/methods , Skin/cytology , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...