Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 46
1.
Histopathology ; 84(6): 915-923, 2024 May.
Article En | MEDLINE | ID: mdl-38433289

A growing body of research supports stromal tumour-infiltrating lymphocyte (TIL) density in breast cancer to be a robust prognostic and predicive biomarker. The gold standard for stromal TIL density quantitation in breast cancer is pathologist visual assessment using haematoxylin and eosin-stained slides. Artificial intelligence/machine-learning algorithms are in development to automate the stromal TIL scoring process, and must be validated against a reference standard such as pathologist visual assessment. Visual TIL assessment may suffer from significant interobserver variability. To improve interobserver agreement, regulatory science experts at the US Food and Drug Administration partnered with academic pathologists internationally to create a freely available online continuing medical education (CME) course to train pathologists in assessing breast cancer stromal TILs using an interactive format with expert commentary. Here we describe and provide a user guide to this CME course, whose content was designed to improve pathologist accuracy in scoring breast cancer TILs. We also suggest subsequent steps to translate knowledge into clinical practice with proficiency testing.


Breast Neoplasms , Humans , Female , Pathologists , Lymphocytes, Tumor-Infiltrating , Artificial Intelligence , Prognosis
2.
IEEE Trans Med Imaging ; PP2024 Feb 21.
Article En | MEDLINE | ID: mdl-38381642

Methods for unsupervised domain adaptation (UDA) help to improve the performance of deep neural networks on unseen domains without any labeled data. Especially in medical disciplines such as histopathology, this is crucial since large datasets with detailed annotations are scarce. While the majority of existing UDA methods focus on the adaptation from a labeled source to a single unlabeled target domain, many real-world applications with a long life cycle involve more than one target domain. Thus, the ability to sequentially adapt to multiple target domains becomes essential. In settings where the data from previously seen domains cannot be stored, e.g., due to data protection regulations, the above becomes a challenging continual learning problem. To this end, we propose to use generative feature-driven image replay in conjunction with a dual-purpose discriminator that not only enables the generation of images with realistic features for replay, but also promotes feature alignment during domain adaptation. We evaluate our approach extensively on a sequence of three histopathological datasets for tissue-type classification, achieving state-of-the-art results. We present detailed ablation experiments studying our proposed method components and demonstrate a possible use-case of our continual UDA method for an unsupervised patch-based segmentation task given high-resolution tissue images. Our code is available at: https://github.com/histocartography/multi-scale-feature-alignment.

3.
J Pathol ; 262(3): 271-288, 2024 03.
Article En | MEDLINE | ID: mdl-38230434

Recent advances in the field of immuno-oncology have brought transformative changes in the management of cancer patients. The immune profile of tumours has been found to have key value in predicting disease prognosis and treatment response in various cancers. Multiplex immunohistochemistry and immunofluorescence have emerged as potent tools for the simultaneous detection of multiple protein biomarkers in a single tissue section, thereby expanding opportunities for molecular and immune profiling while preserving tissue samples. By establishing the phenotype of individual tumour cells when distributed within a mixed cell population, the identification of clinically relevant biomarkers with high-throughput multiplex immunophenotyping of tumour samples has great potential to guide appropriate treatment choices. Moreover, the emergence of novel multi-marker imaging approaches can now provide unprecedented insights into the tumour microenvironment, including the potential interplay between various cell types. However, there are significant challenges to widespread integration of these technologies in daily research and clinical practice. This review addresses the challenges and potential solutions within a structured framework of action from a regulatory and clinical trial perspective. New developments within the field of immunophenotyping using multiplexed tissue imaging platforms and associated digital pathology are also described, with a specific focus on translational implications across different subtypes of cancer. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Breast Neoplasms , Humans , Female , Biomarkers, Tumor/genetics , Prognosis , Phenotype , United Kingdom , Tumor Microenvironment
4.
NPJ Precis Oncol ; 8(1): 9, 2024 Jan 10.
Article En | MEDLINE | ID: mdl-38200147

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.

5.
Med Image Anal ; 93: 103070, 2024 Apr.
Article En | MEDLINE | ID: mdl-38176354

We propose DiRL, a Diversity-inducing Representation Learning technique for histopathology imaging. Self-supervised learning (SSL) techniques, such as contrastive and non-contrastive approaches, have been shown to learn rich and effective representations of digitized tissue samples with limited pathologist supervision. Our analysis of vanilla SSL-pretrained models' attention distribution reveals an insightful observation: sparsity in attention, i.e, models tends to localize most of their attention to some prominent patterns in the image. Although attention sparsity can be beneficial in natural images due to these prominent patterns being the object of interest itself, this can be sub-optimal in digital pathology; this is because, unlike natural images, digital pathology scans are not object-centric, but rather a complex phenotype of various spatially intermixed biological components. Inadequate diversification of attention in these complex images could result in crucial information loss. To address this, we leverage cell segmentation to densely extract multiple histopathology-specific representations, and then propose a prior-guided dense pretext task, designed to match the multiple corresponding representations between the views. Through this, the model learns to attend to various components more closely and evenly, thus inducing adequate diversification in attention for capturing context-rich representations. Through quantitative and qualitative analysis on multiple tasks across cancer types, we demonstrate the efficacy of our method and observe that the attention is more globally distributed.


Image Processing, Computer-Assisted , Machine Learning , Pathology , Humans , Phenotype , Pathology/methods
6.
J Pathol ; 261(4): 378-384, 2023 12.
Article En | MEDLINE | ID: mdl-37794720

Quantifying tumor-infiltrating lymphocytes (TILs) in breast cancer tumors is a challenging task for pathologists. With the advent of whole slide imaging that digitizes glass slides, it is possible to apply computational models to quantify TILs for pathologists. Development of computational models requires significant time, expertise, consensus, and investment. To reduce this burden, we are preparing a dataset for developers to validate their models and a proposal to the Medical Device Development Tool (MDDT) program in the Center for Devices and Radiological Health of the U.S. Food and Drug Administration (FDA). If the FDA qualifies the dataset for its submitted context of use, model developers can use it in a regulatory submission within the qualified context of use without additional documentation. Our dataset aims at reducing the regulatory burden placed on developers of models that estimate the density of TILs and will allow head-to-head comparison of multiple computational models on the same data. In this paper, we discuss the MDDT preparation and submission process, including the feedback we received from our initial interactions with the FDA and propose how a qualified MDDT validation dataset could be a mechanism for open, fair, and consistent measures of computational model performance. Our experiences will help the community understand what the FDA considers relevant and appropriate (from the perspective of the submitter), at the early stages of the MDDT submission process, for validating stromal TIL density estimation models and other potential computational models. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.


Lymphocytes, Tumor-Infiltrating , Pathologists , United States , Humans , United States Food and Drug Administration , Lymphocytes, Tumor-Infiltrating/pathology , United Kingdom
7.
J Pathol ; 260(5): 514-532, 2023 08.
Article En | MEDLINE | ID: mdl-37608771

Modern histologic imaging platforms coupled with machine learning methods have provided new opportunities to map the spatial distribution of immune cells in the tumor microenvironment. However, there exists no standardized method for describing or analyzing spatial immune cell data, and most reported spatial analyses are rudimentary. In this review, we provide an overview of two approaches for reporting and analyzing spatial data (raster versus vector-based). We then provide a compendium of spatial immune cell metrics that have been reported in the literature, summarizing prognostic associations in the context of a variety of cancers. We conclude by discussing two well-described clinical biomarkers, the breast cancer stromal tumor infiltrating lymphocytes score and the colon cancer Immunoscore, and describe investigative opportunities to improve clinical utility of these spatial biomarkers. © 2023 The Pathological Society of Great Britain and Ireland.


Colonic Neoplasms , Humans , Biomarkers , Benchmarking , Lymphocytes, Tumor-Infiltrating , Spatial Analysis , Tumor Microenvironment
8.
J Pathol ; 260(5): 498-513, 2023 08.
Article En | MEDLINE | ID: mdl-37608772

The clinical significance of the tumor-immune interaction in breast cancer is now established, and tumor-infiltrating lymphocytes (TILs) have emerged as predictive and prognostic biomarkers for patients with triple-negative (estrogen receptor, progesterone receptor, and HER2-negative) breast cancer and HER2-positive breast cancer. How computational assessments of TILs might complement manual TIL assessment in trial and daily practices is currently debated. Recent efforts to use machine learning (ML) to automatically evaluate TILs have shown promising results. We review state-of-the-art approaches and identify pitfalls and challenges of automated TIL evaluation by studying the root cause of ML discordances in comparison to manual TIL quantification. We categorize our findings into four main topics: (1) technical slide issues, (2) ML and image analysis aspects, (3) data challenges, and (4) validation issues. The main reason for discordant assessments is the inclusion of false-positive areas or cells identified by performance on certain tissue patterns or design choices in the computational implementation. To aid the adoption of ML for TIL assessment, we provide an in-depth discussion of ML and image analysis, including validation issues that need to be considered before reliable computational reporting of TILs can be incorporated into the trial and routine clinical management of patients with triple-negative breast cancer. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Mammary Neoplasms, Animal , Triple Negative Breast Neoplasms , Humans , Animals , Lymphocytes, Tumor-Infiltrating , Biomarkers , Machine Learning
9.
Comput Methods Programs Biomed ; 239: 107631, 2023 Sep.
Article En | MEDLINE | ID: mdl-37271050

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.


Neoplasms , Neural Networks, Computer , Humans , Algorithms , Software , Histological Techniques
10.
Article En | MEDLINE | ID: mdl-38741683

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.

11.
Proc Mach Learn Res ; 227: 74-94, 2023 Jul.
Article En | MEDLINE | ID: mdl-38817539

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.

12.
J Med Imaging (Bellingham) ; 9(4): 047501, 2022 Jul.
Article En | MEDLINE | ID: mdl-35911208

Purpose: Validation of artificial intelligence (AI) algorithms in digital pathology with a reference standard is necessary before widespread clinical use, but few examples focus on creating a reference standard based on pathologist annotations. This work assesses the results of a pilot study that collects density estimates of stromal tumor-infiltrating lymphocytes (sTILs) in breast cancer biopsy specimens. This work will inform the creation of a validation dataset for the evaluation of AI algorithms fit for a regulatory purpose. Approach: Collaborators and crowdsourced pathologists contributed glass slides, digital images, and annotations. Here, "annotations" refer to any marks, segmentations, measurements, or labels a pathologist adds to a report, image, region of interest (ROI), or biological feature. Pathologists estimated sTILs density in 640 ROIs from hematoxylin and eosin stained slides of 64 patients via two modalities: an optical light microscope and two digital image viewing platforms. Results: The pilot study generated 7373 sTILs density estimates from 29 pathologists. Analysis of annotations found the variability of density estimates per ROI increases with the mean; the root mean square differences were 4.46, 14.25, and 26.25 as the mean density ranged from 0% to 10%, 11% to 40%, and 41% to 100%, respectively. The pilot study informs three areas of improvement for future work: technical workflows, annotation platforms, and agreement analysis methods. Upgrades to the workflows and platforms will improve operability and increase annotation speed and consistency. Conclusions: Exploratory data analysis demonstrates the need to develop new statistical approaches for agreement. The pilot study dataset and analysis methods are publicly available to allow community feedback. The development and results of the validation dataset will be publicly available to serve as an instructive tool that can be replicated by developers and researchers.

13.
Cancers (Basel) ; 14(9)2022 Apr 26.
Article En | MEDLINE | ID: mdl-35565277

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.

14.
J Pathol Inform ; 13: 5, 2022.
Article En | MEDLINE | ID: mdl-35136672

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.

15.
J Pathol Inform ; 12: 45, 2021.
Article En | MEDLINE | ID: mdl-34881099

PURPOSE: Validating artificial intelligence algorithms for clinical use in medical images is a challenging endeavor due to a lack of standard reference data (ground truth). This topic typically occupies a small portion of the discussion in research papers since most of the efforts are focused on developing novel algorithms. In this work, we present a collaboration to create a validation dataset of pathologist annotations for algorithms that process whole slide images. We focus on data collection and evaluation of algorithm performance in the context of estimating the density of stromal tumor-infiltrating lymphocytes (sTILs) in breast cancer. METHODS: We digitized 64 glass slides of hematoxylin- and eosin-stained invasive ductal carcinoma core biopsies prepared at a single clinical site. A collaborating pathologist selected 10 regions of interest (ROIs) per slide for evaluation. We created training materials and workflows to crowdsource pathologist image annotations on two modes: an optical microscope and two digital platforms. The microscope platform allows the same ROIs to be evaluated in both modes. The workflows collect the ROI type, a decision on whether the ROI is appropriate for estimating the density of sTILs, and if appropriate, the sTIL density value for that ROI. RESULTS: In total, 19 pathologists made 1645 ROI evaluations during a data collection event and the following 2 weeks. The pilot study yielded an abundant number of cases with nominal sTIL infiltration. Furthermore, we found that the sTIL densities are correlated within a case, and there is notable pathologist variability. Consequently, we outline plans to improve our ROI and case sampling methods. We also outline statistical methods to account for ROI correlations within a case and pathologist variability when validating an algorithm. CONCLUSION: We have built workflows for efficient data collection and tested them in a pilot study. As we prepare for pivotal studies, we will investigate methods to use the dataset as an external validation tool for algorithms. We will also consider what it will take for the dataset to be fit for a regulatory purpose: study size, patient population, and pathologist training and qualifications. To this end, we will elicit feedback from the Food and Drug Administration via the Medical Device Development Tool program and from the broader digital pathology and AI community. Ultimately, we intend to share the dataset, statistical methods, and lessons learned.

16.
Sci Rep ; 11(1): 2809, 2021 02 02.
Article En | MEDLINE | ID: mdl-33531581

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.


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
17.
J Diabetes Sci Technol ; 15(3): 607-614, 2021 05.
Article En | MEDLINE | ID: mdl-33435706

OBJECTIVE: We assessed the clinical utility and accuracy of real-time continuous glucose monitoring (rtCGM) (Dexcom G6) in managing diabetes patients with severe COVID-19 infection following admission to the intensive care unit (ICU). METHODS: We present retrospective analysis of masked rtCGM in 30 patients with severe COVID-19. rtCGM was used during the first 24 hours for comparison with arterial-line point of care (POC) values, where clinicians utilized rtCGM data to adjust insulin therapy in patients if rtCGM values were within 20% of point-of-care (POC) values during the masked period. An investigator-developed survey was administered to assess nursing staff (n = 66) perceptions regarding the use of rtCGM in the ICU. RESULTS: rtCGM data were used to adjust insulin therapy in 30 patients. Discordance between rtCGM and POC glucose values were observed in 11 patients but the differences were not considered clinically significant. Mean sensor glucose decreased from 235.7 ± 42.1 mg/dL (13.1 ± 2.1 mmol/L) to 202.7 ± 37.6 mg/dL (11.1 ± 2.1 mmol/L) with rtCGM management. Improvements in mean sensor glucose were observed in 77% of patients (n = 23) with concomitant reductions in daily POC measurements in 50% of patients (n = 15) with rtCGM management. The majority (63%) of nurses reported that rtCGM was helpful for improving care for patients with diabetes patients during the COVID-19 pandemic, and 49% indicated that rtCGM reduced their use of personal protective equipment (PPE). CONCLUSIONS: Our findings provide a strong rationale to increase clinician awareness for the adoption and implementation of rtCGM systems in the ICU. Additional studies are needed to further understand the utility of rtCGM in critically ill patients and other clinical care settings.


Attitude of Health Personnel , Blood Glucose/metabolism , COVID-19/therapy , Diabetes Mellitus, Type 2/diagnosis , Health Knowledge, Attitudes, Practice , Intensive Care Units , Nursing Staff, Hospital , Remote Sensing Technology , Biomarkers/blood , Blood Glucose/drug effects , COVID-19/diagnosis , Critical Care Nursing , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Patient Admission , Predictive Value of Tests , Prognosis , Remote Sensing Technology/instrumentation , Reproducibility of Results , Retrospective Studies , Time Factors
18.
Appl Immunohistochem Mol Morphol ; 29(2): 144-151, 2021 02 01.
Article En | MEDLINE | ID: mdl-32554975

The major roles of keratin 17 (K17) as a prognostic biomarker have been highlighted in a range of human malignancies. However, its relevance to esophageal squamous cell carcinoma (ESCC) remains unexplored. In this study, the relationship between K17 expression and clinicopathologic parameters and survival were determined by RNA sequencing (RNA-Seq) in 90 ESCCs and by immunohistochemistry (IHC) in 68 ESCCs. K17 expression was significantly higher in ESCC than in paired normal tissues at both the messenger RNA and protein levels. K17 messenger RNA and staining by IHC were significantly correlated with aggressive characteristics, including advanced clinical stage, invasion depth, and lymph node metastases; and were predictive of poor prognosis in advanced disease patients. Furthermore, K17 expression was detected by IHC in high-grade premalignant lesions of the esophageal mucosa, suggesting that K17 could also be a biomarker of dysplasia of the esophageal mucosa. Overall, this study established that K17 is a negative prognostic biomarker for the most common subtype of esophageal cancer.


Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic , Keratin-17/biosynthesis , Neoplasm Proteins/biosynthesis , Aged , Aged, 80 and over , Disease-Free Survival , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Predictive Value of Tests , Retrospective Studies , Survival Rate
19.
Front Oncol ; 11: 806603, 2021.
Article En | MEDLINE | ID: mdl-35251953

The role of tumor infiltrating lymphocytes (TILs) as a biomarker to predict disease progression and clinical outcomes has generated tremendous interest in translational cancer research. We present an updated and enhanced deep learning workflow to classify 50x50 um tiled image patches (100x100 pixels at 20x magnification) as TIL positive or negative based on the presence of 2 or more TILs in gigapixel whole slide images (WSIs) from the Cancer Genome Atlas (TCGA). This workflow generates TIL maps to study the abundance and spatial distribution of TILs in 23 different types of cancer. We trained three state-of-the-art, popular convolutional neural network (CNN) architectures (namely VGG16, Inception-V4, and ResNet-34) with a large volume of training data, which combined manual annotations from pathologists (strong annotations) and computer-generated labels from our previously reported first-generation TIL model for 13 cancer types (model-generated annotations). Specifically, this training dataset contains TIL positive and negative patches from cancers in additional organ sites and curated data to help improve algorithmic performance by decreasing known false positives and false negatives. Our new TIL workflow also incorporates automated thresholding to convert model predictions into binary classifications to generate TIL maps. The new TIL models all achieve better performance with improvements of up to 13% in accuracy and 15% in F-score. We report these new TIL models and a curated dataset of TIL maps, referred to as TIL-Maps-23, for 7983 WSIs spanning 23 types of cancer with complex and diverse visual appearances, which will be publicly available along with the code to evaluate performance. Code Available at: https://github.com/ShahiraAbousamra/til_classification.

20.
JPEN J Parenter Enteral Nutr ; 45(1): 208-211, 2021 01.
Article En | MEDLINE | ID: mdl-33085780

Many patients admitted to the intensive care unit (ICU) are acutely malnourished and often require aggressive and early nutrition support with parenteral nutrition (PN). However, PN-induced hyperglycemia is a predictor of hospital mortality and is associated with increased length of stay. Elevated blood glucose in hospitalized patients with coronavirus disease 2019 (COVID-19) is also associated with increased mortality. Real-time continuous glucose monitoring (rtCGM) is primarily used in the outpatient setting, but there is rapidly growing interest in its applicability to help treat dysglycemia in critically ill patients, especially during the ongoing COVID-19 pandemic. We assessed the use of rtCGM data (Dexcom G6) in a 58-year-old male admitted to the ICU for severe COVID-19 infection, who developed PN-induced hyperglycemia with markedly elevated total daily insulin requirements as high as 128 units. rtCGM was used to safely titrate insulin infusion and monitor glucose levels. No episodes of hypoglycemia were observed, despite an extremely aggressive insulin regimen. This case demonstrates the potential utility of rtCGM in the critical care setting and highlights its potential to help conserve personal protective equipment and minimize unnecessary staff exposure in the setting of COVID-19.


Blood Glucose Self-Monitoring/methods , Blood Glucose/metabolism , COVID-19/complications , Hyperglycemia/drug therapy , Insulin/administration & dosage , Parenteral Nutrition/adverse effects , Blood Glucose/analysis , COVID-19/diagnosis , Critical Illness/therapy , Humans , Hyperglycemia/blood , Hyperglycemia/diagnosis , Male , Middle Aged , Pandemics , SARS-CoV-2
...