Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 262(3): 271-288, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38230434

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Biomarcadores de Tumor/genética , Pronóstico , Fenotipo , Reino Unido , Microambiente Tumoral
2.
Histopathology ; 84(6): 915-923, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38433289

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Patólogos , Linfocitos Infiltrantes de Tumor , Inteligencia Artificial , Pronóstico
3.
J Pathol ; 261(4): 378-384, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37794720

RESUMEN

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.


Asunto(s)
Linfocitos Infiltrantes de Tumor , Patólogos , Estados Unidos , Humanos , United States Food and Drug Administration , Linfocitos Infiltrantes de Tumor/patología , Reino Unido
4.
J Pathol ; 260(5): 514-532, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37608771

RESUMEN

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.


Asunto(s)
Neoplasias del Colon , Humanos , Biomarcadores , Benchmarking , Linfocitos Infiltrantes de Tumor , Análisis Espacial , Microambiente Tumoral
5.
J Pathol ; 260(5): 498-513, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37608772

RESUMEN

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.


Asunto(s)
Neoplasias Mamarias Animales , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Linfocitos Infiltrantes de Tumor , Biomarcadores , Aprendizaje Automático
6.
Am J Pathol ; 190(7): 1491-1504, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32277893

RESUMEN

Quantitative assessment of spatial relations between tumor and tumor-infiltrating lymphocytes (TIL) is increasingly important in both basic science and clinical aspects of breast cancer research. We have developed and evaluated convolutional neural network analysis pipelines to generate combined maps of cancer regions and TILs in routine diagnostic breast cancer whole slide tissue images. The combined maps provide insight about the structural patterns and spatial distribution of lymphocytic infiltrates and facilitate improved quantification of TILs. Both tumor and TIL analyses were evaluated by using three convolutional neural network networks (34-layer ResNet, 16-layer VGG, and Inception v4); the results compared favorably with those obtained by using the best published methods. We have produced open-source tools and a public data set consisting of tumor/TIL maps for 1090 invasive breast cancer images from The Cancer Genome Atlas. The maps can be downloaded for further downstream analyses.


Asunto(s)
Neoplasias de la Mama/patología , Aprendizaje Profundo , Linfocitos Infiltrantes de Tumor/patología , Neoplasias de la Mama/inmunología , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Programa de VERF
7.
J Magn Reson Imaging ; 51(2): 341-354, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31041822

RESUMEN

Clinical practice in radiology and pathology requires professional expertise and many years of training to visually evaluate and interpret abnormal phenotypic features in medical images and tissue sections to generate diagnoses that guide patient management and treatment. Recent advances in digital image analysis methods and machine learning have led to significant interest in extracting additional information from medical and digital whole-slide images in radiology and pathology, respectively. This has led to significant interest and research in radiomics and pathomics to correlate phenotypic features of disease with image analytics in order to identify image-based biomarkers. The expanding role of big data in radiology and pathology parallels the development and role of immunohistochemistry (IHC) in the daily practice of pathology. IHC methods were initially developed to provide additional information to help classify tumors and then transformed into an indispensable tool to guide treatment in many types of cancer. IHC markers are used in daily practice to identify specific types of cells and highlight their distributions in tissues in order to distinguish benign from neoplastic cells, determine tumor origin, subclassify neoplasms, and support and confirm diagnoses. In this regard, radiomics, pathomics, and IHC methods are very similar since they enable the extraction of image-based features to characterize various properties of diseases. Due to the dramatic advancements in recent radiomics research, we provide a brief overview of the role of established and emerging IHC biomarkers in various tumor types that have been correlated with radiologic biomarkers to improve diagnostic accuracy, predict prognosis, guide patient management, and select treatment strategies. Level of Evidence: 5 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2020;51:341-354.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Biomarcadores , Humanos , Inmunohistoquímica , Radiografía
8.
Pattern Recognit ; 86: 188-200, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30631215

RESUMEN

We propose a sparse Convolutional Autoencoder (CAE) for simultaneous nucleus detection and feature extraction in histopathology tissue images. Our CAE detects and encodes nuclei in image patches in tissue images into sparse feature maps that encode both the location and appearance of nuclei. A primary contribution of our work is the development of an unsupervised detection network by using the characteristics of histopathology image patches. The pretrained nucleus detection and feature extraction modules in our CAE can be fine-tuned for supervised learning in an end-to-end fashion. We evaluate our method on four datasets and achieve state-of-the-art results. In addition, we are able to achieve comparable performance with only 5% of the fully- supervised annotation cost.

9.
AJR Am J Roentgenol ; 204(1): 128-39, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25539248

RESUMEN

OBJECTIVE: The purpose of this article is to review the imaging features of necrotizing fasciitis and its potential mimics. Key imaging features are emphasized to enable accurate and efficient interpretation of variables that are essential in appropriate management. CONCLUSION: Necrotizing fasciitis is a medical emergency with potential lethal outcome. Dissecting gas along fascial planes in the absence of penetrating trauma (including iatrogenic) is essentially pathognomonic. However, the lack of soft-tissue emphysema does not exclude the diagnosis. Mimics of necrotizing fasciitis include nonnecrotizing fasciitis (eosinophilic, paraneoplastic, inflammatory (lupus myofasciitis, Churg-Strauss, nodular, or proliferative), myositis, neoplasm, myonecrosis, inflammatory myopathy, and compartment syndrome. Necrotizing fasciitis is a clinical diagnosis, and imaging can reveal nonspecific or negative findings (particularly during the early course of disease). One should be familiar with salient clinical and imaging findings of necrotizing fasciitis to facilitate a more rapid and accurate diagnosis and be aware that its diagnosis necessitates immediate discussion with the referring physician.


Asunto(s)
Fascitis Necrotizante/diagnóstico , Inflamación/diagnóstico , Enfermedades Musculares/diagnóstico , Neoplasias/diagnóstico , Tomografía Computarizada por Rayos X/métodos , Adulto , Anciano , Diagnóstico Diferencial , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Persona de Mediana Edad , Adulto Joven
10.
IEEE Trans Med Imaging ; 43(7): 2599-2609, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38381642

RESUMEN

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.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Procesamiento de Imagen Asistido por Computador/métodos , Humanos , Algoritmos , Aprendizaje Automático no Supervisado , Aprendizaje Profundo , Animales , Bases de Datos Factuales , Redes Neurales de la Computación
11.
Med Image Anal ; 93: 103070, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38176354

RESUMEN

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.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Aprendizaje Automático , Patología , Humanos , Fenotipo , Patología/métodos
12.
NPJ Precis Oncol ; 8(1): 9, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38200147

RESUMEN

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.

13.
Artículo en Inglés | MEDLINE | ID: mdl-38741683

RESUMEN

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.

14.
Comput Methods Programs Biomed ; 239: 107631, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37271050

RESUMEN

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.


Asunto(s)
Neoplasias , Redes Neurales de la Computación , Humanos , Algoritmos , Programas Informáticos , Técnicas Histológicas
15.
Proc Mach Learn Res ; 227: 74-94, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38817539

RESUMEN

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.

16.
Cancers (Basel) ; 14(9)2022 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-35565277

RESUMEN

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.

17.
J Med Imaging (Bellingham) ; 9(4): 047501, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35911208

RESUMEN

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.

18.
J Pathol Inform ; 13: 5, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35136672

RESUMEN

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.

19.
JPEN J Parenter Enteral Nutr ; 45(1): 208-211, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33085780

RESUMEN

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.


Asunto(s)
Automonitorización de la Glucosa Sanguínea/métodos , Glucemia/metabolismo , COVID-19/complicaciones , Hiperglucemia/tratamiento farmacológico , Insulina/administración & dosificación , Nutrición Parenteral/efectos adversos , Glucemia/análisis , COVID-19/diagnóstico , Enfermedad Crítica/terapia , Humanos , Hiperglucemia/sangre , Hiperglucemia/diagnóstico , Masculino , Persona de Mediana Edad , Pandemias , SARS-CoV-2
20.
Appl Immunohistochem Mol Morphol ; 29(2): 144-151, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32554975

RESUMEN

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.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Regulación Neoplásica de la Expresión Génica , Queratina-17/biosíntesis , Proteínas de Neoplasias/biosíntesis , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/mortalidad , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA