Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
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.
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
3.
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
4.
J Pathol ; 244(5): 512-524, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29288495

RESUMEN

The Cancer Genome Atlas (TCGA) represents one of several international consortia dedicated to performing comprehensive genomic and epigenomic analyses of selected tumour types to advance our understanding of disease and provide an open-access resource for worldwide cancer research. Thirty-three tumour types (selected by histology or tissue of origin, to include both common and rare diseases), comprising >11 000 specimens, were subjected to DNA sequencing, copy number and methylation analysis, and transcriptomic, proteomic and histological evaluation. Each cancer type was analysed individually to identify tissue-specific alterations, and make correlations across different molecular platforms. The final dataset was then normalized and combined for the PanCancer Initiative, which seeks to identify commonalities across different cancer types or cells of origin/lineage, or within anatomically or morphologically related groups. An important resource generated along with the rich molecular studies is an extensive digital pathology slide archive, composed of frozen section tissue directly related to the tissues analysed as part of TCGA, and representative formalin-fixed paraffin-embedded, haematoxylin and eosin (H&E)-stained diagnostic slides. These H&E image resources have primarily been used to verify diagnoses and histological subtypes with some limited extraction of standard pathological variables such as mitotic activity, grade, and lymphocytic infiltrates. Largely overlooked is the richness of these scanned images for more sophisticated feature extraction approaches coupled with machine learning, and ultimately correlation with molecular features and clinical endpoints. Here, we document initial attempts to exploit TCGA imaging archives, and describe some of the tools, and the rapidly evolving image analysis/feature extraction landscape. Our hope is to inform, and ultimately inspire and challenge, the pathology and cancer research communities to exploit these imaging resources so that the full potential of this integral platform of TCGA can be used to complement and enhance the insightful integrated analyses from the genomic and epigenomic platforms. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Biomarcadores de Tumor/genética , Genómica/métodos , Neoplasias/genética , Neoplasias/patología , Patología Molecular/métodos , Bases de Datos Genéticas , Epigénesis Genética , Predisposición Genética a la Enfermedad , Genoma Humano , Humanos , Interpretación de Imagen Asistida por Computador , Neoplasias/terapia , Fenotipo , Valor Predictivo de las Pruebas
5.
Med Image Anal ; 95: 103162, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38593644

RESUMEN

Active Learning (AL) has the potential to solve a major problem of digital pathology: the efficient acquisition of labeled data for machine learning algorithms. However, existing AL methods often struggle in realistic settings with artifacts, ambiguities, and class imbalances, as commonly seen in the medical field. The lack of precise uncertainty estimations leads to the acquisition of images with a low informative value. To address these challenges, we propose Focused Active Learning (FocAL), which combines a Bayesian Neural Network with Out-of-Distribution detection to estimate different uncertainties for the acquisition function. Specifically, the weighted epistemic uncertainty accounts for the class imbalance, aleatoric uncertainty for ambiguous images, and an OoD score for artifacts. We perform extensive experiments to validate our method on MNIST and the real-world Panda dataset for the classification of prostate cancer. The results confirm that other AL methods are 'distracted' by ambiguities and artifacts which harm the performance. FocAL effectively focuses on the most informative images, avoiding ambiguities and artifacts during acquisition. For both experiments, FocAL outperforms existing AL approaches, reaching a Cohen's kappa of 0.764 with only 0.69% of the labeled Panda data.


Asunto(s)
Neoplasias de la Próstata , Humanos , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Masculino , Aprendizaje Automático , Teorema de Bayes , Algoritmos , Interpretación de Imagen Asistida por Computador/métodos , Artefactos , Redes Neurales de la Computación
6.
Artículo en Inglés | MEDLINE | ID: mdl-31997849

RESUMEN

Histologic assessment of stromal tumor infiltrating lymphocytes (sTIL) as a surrogate of the host immune response has been shown to be prognostic and potentially chemo-predictive in triple-negative and HER2-positive breast cancers. The current practice of manual assessment is prone to intra- and inter-observer variability. Furthermore, the inter-play of sTILs, tumor cells, other microenvironment mediators, their spatial relationships, quantity, and other image-based features have yet to be determined exhaustively and systemically. Towards analysis of these aspects, we developed a deep learning based method for joint region-level and nucleus-level segmentation and classification of breast cancer H&E tissue whole slide images. Our proposed method simultaneously identifies tumor, fibroblast, and lymphocyte nuclei, along with key histologic region compartments including tumor and stroma. We also show how the resultant segmentation masks can be combined with seeding approaches to yield accurate nucleus classifications. Furthermore, we outline a simple workflow for calibrating computational scores to human scores for consistency. The pipeline identifies key compartments with high accuracy (Dice= overall: 0.78, tumor: 0.83, and fibroblasts: 0.77). ROC AUC for nucleus classification is high at 0.89 (micro-average), 0.89 (lymphocytes), 0.90 (tumor), and 0.78 (fibroblasts). Spearman correlation between computational sTIL and pathologist consensus is high (R=0.73, p<0.001) and is higher than inter-pathologist correlation (R=0.66, p<0.001). Both manual and computational sTIL scores successfully stratify patients by clinical progression outcomes.

7.
Proc IEEE Int Conf Big Data ; 2015: 928-935, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27796014

RESUMEN

Recent advances in microscopy imaging and genomics have created an explosion of patient data in the pathology domain. Whole-slide images (WSIs) of tissues can now capture disease processes as they unfold in high resolution, recording the visual cues that have been the basis of pathologic diagnosis for over a century. Each WSI contains billions of pixels and up to a million or more microanatomic objects whose appearances hold important prognostic information. Computational image analysis enables the mining of massive WSI datasets to extract quantitative morphologic features describing the visual qualities of patient tissues. When combined with genomic and clinical variables, this quantitative information provides scientists and clinicians with insights into disease biology and patient outcomes. To facilitate interaction with this rich resource, we have developed a web-based machine-learning framework that enables users to rapidly build classifiers using an intuitive active learning process that minimizes data labeling effort. In this paper we describe the architecture and design of this system, and demonstrate its effectiveness through quantification of glioma brain tumors.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA