Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Artif Intell ; 7: 1408843, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39118787

RESUMEN

Cancer research encompasses data across various scales, modalities, and resolutions, from screening and diagnostic imaging to digitized histopathology slides to various types of molecular data and clinical records. The integration of these diverse data types for personalized cancer care and predictive modeling holds the promise of enhancing the accuracy and reliability of cancer screening, diagnosis, and treatment. Traditional analytical methods, which often focus on isolated or unimodal information, fall short of capturing the complex and heterogeneous nature of cancer data. The advent of deep neural networks has spurred the development of sophisticated multimodal data fusion techniques capable of extracting and synthesizing information from disparate sources. Among these, Graph Neural Networks (GNNs) and Transformers have emerged as powerful tools for multimodal learning, demonstrating significant success. This review presents the foundational principles of multimodal learning including oncology data modalities, taxonomy of multimodal learning, and fusion strategies. We delve into the recent advancements in GNNs and Transformers for the fusion of multimodal data in oncology, spotlighting key studies and their pivotal findings. We discuss the unique challenges of multimodal learning, such as data heterogeneity and integration complexities, alongside the opportunities it presents for a more nuanced and comprehensive understanding of cancer. Finally, we present some of the latest comprehensive multimodal pan-cancer data sources. By surveying the landscape of multimodal data integration in oncology, our goal is to underline the transformative potential of multimodal GNNs and Transformers. Through technological advancements and the methodological innovations presented in this review, we aim to chart a course for future research in this promising field. This review may be the first that highlights the current state of multimodal modeling applications in cancer using GNNs and transformers, presents comprehensive multimodal oncology data sources, and sets the stage for multimodal evolution, encouraging further exploration and development in personalized cancer care.

2.
Sensors (Basel) ; 24(5)2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38475170

RESUMEN

The advancements in data acquisition, storage, and processing techniques have resulted in the rapid growth of heterogeneous medical data. Integrating radiological scans, histopathology images, and molecular information with clinical data is essential for developing a holistic understanding of the disease and optimizing treatment. The need for integrating data from multiple sources is further pronounced in complex diseases such as cancer for enabling precision medicine and personalized treatments. This work proposes Multimodal Integration of Oncology Data System (MINDS)-a flexible, scalable, and cost-effective metadata framework for efficiently fusing disparate data from public sources such as the Cancer Research Data Commons (CRDC) into an interconnected, patient-centric framework. MINDS consolidates over 41,000 cases from across repositories while achieving a high compression ratio relative to the 3.78 PB source data size. It offers sub-5-s query response times for interactive exploration. MINDS offers an interface for exploring relationships across data types and building cohorts for developing large-scale multimodal machine learning models. By harmonizing multimodal data, MINDS aims to potentially empower researchers with greater analytical ability to uncover diagnostic and prognostic insights and enable evidence-based personalized care. MINDS tracks granular end-to-end data provenance, ensuring reproducibility and transparency. The cloud-native architecture of MINDS can handle exponential data growth in a secure, cost-optimized manner while ensuring substantial storage optimization, replication avoidance, and dynamic access capabilities. Auto-scaling, access controls, and other mechanisms guarantee pipelines' scalability and security. MINDS overcomes the limitations of existing biomedical data silos via an interoperable metadata-driven approach that represents a pivotal step toward the future of oncology data integration.


Asunto(s)
Neoplasias , Humanos , Reproducibilidad de los Resultados
3.
Lab Invest ; 103(11): 100255, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37757969

RESUMEN

Digital pathology has transformed the traditional pathology practice of analyzing tissue under a microscope into a computer vision workflow. Whole-slide imaging allows pathologists to view and analyze microscopic images on a computer monitor, enabling computational pathology. By leveraging artificial intelligence (AI) and machine learning (ML), computational pathology has emerged as a promising field in recent years. Recently, task-specific AI/ML (eg, convolutional neural networks) has risen to the forefront, achieving above-human performance in many image-processing and computer vision tasks. The performance of task-specific AI/ML models depends on the availability of many annotated training datasets, which presents a rate-limiting factor for AI/ML development in pathology. Task-specific AI/ML models cannot benefit from multimodal data and lack generalization, eg, the AI models often struggle to generalize to new datasets or unseen variations in image acquisition, staining techniques, or tissue types. The 2020s are witnessing the rise of foundation models and generative AI. A foundation model is a large AI model trained using sizable data, which is later adapted (or fine-tuned) to perform different tasks using a modest amount of task-specific annotated data. These AI models provide in-context learning, can self-correct mistakes, and promptly adjust to user feedback. In this review, we provide a brief overview of recent advances in computational pathology enabled by task-specific AI, their challenges and limitations, and then introduce various foundation models. We propose to create a pathology-specific generative AI based on multimodal foundation models and present its potentially transformative role in digital pathology. We describe different use cases, delineating how it could serve as an expert companion of pathologists and help them efficiently and objectively perform routine laboratory tasks, including quantifying image analysis, generating pathology reports, diagnosis, and prognosis. We also outline the potential role that foundation models and generative AI can play in standardizing the pathology laboratory workflow, education, and training.


Asunto(s)
Inteligencia Artificial , Aprendizaje Automático , Patología , Humanos , Procesamiento de Imagen Asistido por Computador , Redes Neurales de la Computación , Patólogos , Patología/tendencias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA