Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
2.
NPJ Digit Med ; 5(1): 71, 2022 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-35676445

RESUMEN

Prostate cancer is the most frequent cancer in men and a leading cause of cancer death. Determining a patient's optimal therapy is a challenge, where oncologists must select a therapy with the highest likelihood of success and the lowest likelihood of toxicity. International standards for prognostication rely on non-specific and semi-quantitative tools, commonly leading to over- and under-treatment. Tissue-based molecular biomarkers have attempted to address this, but most have limited validation in prospective randomized trials and expensive processing costs, posing substantial barriers to widespread adoption. There remains a significant need for accurate and scalable tools to support therapy personalization. Here we demonstrate prostate cancer therapy personalization by predicting long-term, clinically relevant outcomes using a multimodal deep learning architecture and train models using clinical data and digital histopathology from prostate biopsies. We train and validate models using five phase III randomized trials conducted across hundreds of clinical centers. Histopathological data was available for 5654 of 7764 randomized patients (71%) with a median follow-up of 11.4 years. Compared to the most common risk-stratification tool-risk groups developed by the National Cancer Center Network (NCCN)-our models have superior discriminatory performance across all endpoints, ranging from 9.2% to 14.6% relative improvement in a held-out validation set. This artificial intelligence-based tool improves prognostication over standard tools and allows oncologists to computationally predict the likeliest outcomes of specific patients to determine optimal treatment. Outfitted with digital scanners and internet access, any clinic could offer such capabilities, enabling global access to therapy personalization.

3.
PLoS One ; 16(7): e0252384, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34214101

RESUMEN

Early prediction of patient mortality risks during a pandemic can decrease mortality by assuring efficient resource allocation and treatment planning. This study aimed to develop and compare prognosis prediction machine learning models based on invasive laboratory and noninvasive clinical and demographic data from patients' day of admission. Three Support Vector Machine (SVM) models were developed and compared using invasive, non-invasive, and both groups. The results suggested that non-invasive features could provide mortality predictions that are similar to the invasive and roughly on par with the joint model. Feature inspection results from SVM-RFE and sparsity analysis displayed that, compared with the invasive model, the non-invasive model can provide better performances with a fewer number of features, pointing to the presence of high predictive information contents in several non-invasive features, including SPO2, age, and cardiovascular disorders. Furthermore, while the invasive model was able to provide better mortality predictions for the imminent future, non-invasive features displayed better performance for more distant expiration intervals. Early mortality prediction using non-invasive models can give us insights as to where and with whom to intervene. Combined with novel technologies, such as wireless wearable devices, these models can create powerful frameworks for various medical assignments and patient triage.


Asunto(s)
COVID-19/mortalidad , Pandemias , SARS-CoV-2 , Máquina de Vectores de Soporte , Adulto , Anciano , Anciano de 80 o más Años , Comorbilidad , Registros Electrónicos de Salud , Femenino , Predicción , Humanos , Masculino , Persona de Mediana Edad , Modelos Teóricos , Riesgo , Índice de Severidad de la Enfermedad , Evaluación de Síntomas , Triaje , Adulto Joven
4.
Sci Rep ; 11(1): 8366, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863957

RESUMEN

The reliability of machine learning models can be compromised when trained on low quality data. Many large-scale medical imaging datasets contain low quality labels extracted from sources such as medical reports. Moreover, images within a dataset may have heterogeneous quality due to artifacts and biases arising from equipment or measurement errors. Therefore, algorithms that can automatically identify low quality data are highly desired. In this study, we used data Shapley, a data valuation metric, to quantify the value of training data to the performance of a pneumonia detection algorithm in a large chest X-ray dataset. We characterized the effectiveness of data Shapley in identifying low quality versus valuable data for pneumonia detection. We found that removing training data with high Shapley values decreased the pneumonia detection performance, whereas removing data with low Shapley values improved the model performance. Furthermore, there were more mislabeled examples in low Shapley value data and more true pneumonia cases in high Shapley value data. Our results suggest that low Shapley value indicates mislabeled or poor quality images, whereas high Shapley value indicates data that are valuable for pneumonia detection. Our method can serve as a framework for using data Shapley to denoise large-scale medical imaging datasets.


Asunto(s)
Algoritmos , Diagnóstico por Imagen/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Aprendizaje Automático , Redes Neurales de la Computación , Neumonía/diagnóstico , Radiografía Torácica/métodos , Conjuntos de Datos como Asunto , Humanos
5.
Nature ; 580(7802): 252-256, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32269341

RESUMEN

Accurate assessment of cardiac function is crucial for the diagnosis of cardiovascular disease1, screening for cardiotoxicity2 and decisions regarding the clinical management of patients with a critical illness3. However, human assessment of cardiac function focuses on a limited sampling of cardiac cycles and has considerable inter-observer variability despite years of training4,5. Here, to overcome this challenge, we present a video-based deep learning algorithm-EchoNet-Dynamic-that surpasses the performance of human experts in the critical tasks of segmenting the left ventricle, estimating ejection fraction and assessing cardiomyopathy. Trained on echocardiogram videos, our model accurately segments the left ventricle with a Dice similarity coefficient of 0.92, predicts ejection fraction with a mean absolute error of 4.1% and reliably classifies heart failure with reduced ejection fraction (area under the curve of 0.97). In an external dataset from another healthcare system, EchoNet-Dynamic predicts the ejection fraction with a mean absolute error of 6.0% and classifies heart failure with reduced ejection fraction with an area under the curve of 0.96. Prospective evaluation with repeated human measurements confirms that the model has variance that is comparable to or less than that of human experts. By leveraging information across multiple cardiac cycles, our model can rapidly identify subtle changes in ejection fraction, is more reproducible than human evaluation and lays the foundation for precise diagnosis of cardiovascular disease in real time. As a resource to promote further innovation, we also make publicly available a large dataset of 10,030 annotated echocardiogram videos.


Asunto(s)
Aprendizaje Profundo , Cardiopatías/diagnóstico , Cardiopatías/fisiopatología , Corazón/fisiología , Corazón/fisiopatología , Modelos Cardiovasculares , Grabación en Video , Fibrilación Atrial , Conjuntos de Datos como Asunto , Ecocardiografía , Insuficiencia Cardíaca/fisiopatología , Hospitales , Humanos , Estudios Prospectivos , Reproducibilidad de los Resultados , Función Ventricular Izquierda/fisiología
6.
NPJ Digit Med ; 3: 10, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31993508

RESUMEN

Echocardiography uses ultrasound technology to capture high temporal and spatial resolution images of the heart and surrounding structures, and is the most common imaging modality in cardiovascular medicine. Using convolutional neural networks on a large new dataset, we show that deep learning applied to echocardiography can identify local cardiac structures, estimate cardiac function, and predict systemic phenotypes that modify cardiovascular risk but not readily identifiable to human interpretation. Our deep learning model, EchoNet, accurately identified the presence of pacemaker leads (AUC = 0.89), enlarged left atrium (AUC = 0.86), left ventricular hypertrophy (AUC = 0.75), left ventricular end systolic and diastolic volumes ( R 2 = 0.74 and R 2 = 0.70), and ejection fraction ( R 2 = 0.50), as well as predicted systemic phenotypes of age ( R 2 = 0.46), sex (AUC = 0.88), weight ( R 2 = 0.56), and height ( R 2 = 0.33). Interpretation analysis validates that EchoNet shows appropriate attention to key cardiac structures when performing human-explainable tasks and highlights hypothesis-generating regions of interest when predicting systemic phenotypes difficult for human interpretation. Machine learning on echocardiography images can streamline repetitive tasks in the clinical workflow, provide preliminary interpretation in areas with insufficient qualified cardiologists, and predict phenotypes challenging for human evaluation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA