Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Pathol ; 194(5): 721-734, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38320631

RESUMO

Histopathology is the reference standard for pathology diagnosis, and has evolved with the digitization of glass slides [ie, whole slide images (WSIs)]. While trained histopathologists are able to diagnose diseases by examining WSIs visually, this process is time consuming and prone to variability. To address these issues, artificial intelligence models are being developed to generate slide-level representations of WSIs, summarizing the entire slide as a single vector. This enables various computational pathology applications, including interslide search, multimodal training, and slide-level classification. Achieving expressive and robust slide-level representations hinges on patch feature extraction and aggregation steps. This study proposed an additional binary patch grouping (BPG) step, a plugin that can be integrated into various slide-level representation pipelines, to enhance the quality of slide-level representation in bone marrow histopathology. BPG excludes patches with less clinical relevance through minimal interaction with the pathologist; a one-time human intervention for the entire process. This study further investigated domain-general versus domain-specific feature extraction models based on convolution and attention and examined two different feature aggregation methods, with and without BPG, showing BPG's generalizability. The results showed that using BPG boosts the performance of WSI retrieval (mean average precision at 10) by 4% and improves WSI classification (weighted-F1) by 5% compared to not using BPG. Additionally, domain-general large models and parameterized pooling produced the best-quality slide-level representations.


Assuntos
Inteligência Artificial , Medula Óssea , Humanos , Suplementos Nutricionais , Patologistas
2.
Comput Biol Med ; 166: 107530, 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37837726

RESUMO

One of the goals of AI-based computational pathology is to generate compact representations of whole slide images (WSIs) that capture the essential information needed for diagnosis. While such approaches have been applied to histopathology, few applications have been reported in cytology. Bone marrow aspirate cytology is the basis for key clinical decisions in hematology. However, visual inspection of aspirate specimens is a tedious and complex process subject to variation in interpretation, and hematopathology expertise is scarce. The ability to generate a compact representation of an aspirate specimen may form the basis for clinical decision-support tools in hematology. In this study, we leverage our previously published end-to-end AI-based system for counting and classifying cells from bone marrow aspirate WSIs, which enables the direct use of individual cells as inputs rather than WSI patches. We then construct bags of individual cell features from each WSI, and apply multiple instance learning to extract their vector representations. To evaluate the quality of our representations, we conducted WSI retrieval and classification tasks. Our results show that we achieved a mAP@10 of 0.58 ±0.02 in WSI-level image retrieval, surpassing the random-retrieval baseline of 0.39 ±0.1. Furthermore, we predicted five diagnostic labels for individual aspirate WSIs with a weighted-average F1 score of 0.57 ±0.03 using a k-nearest-neighbors (k-NN) model, outperforming guessing using empirical class prior probabilities (0.26 ±0.02). We present the first example of exploring trainable mechanisms to generate compact, slide-level representations in bone marrow cytology with deep learning. This method has the potential to summarize complex semantic information in WSIs toward improved diagnostics in hematology, and may eventually support AI-assisted computational pathology approaches.

3.
J Pathol Inform ; 14: 100334, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37732298

RESUMO

Deep models for cell detection have demonstrated utility in bone marrow cytology, showing impressive results in terms of accuracy and computational efficiency. However, these models have yet to be implemented in the clinical diagnostic workflow. Additionally, the metrics used to evaluate cell detection models are not necessarily aligned with clinical goals and targets. In order to address these issues, we introduce novel, automatically generated visual summaries of bone marrow aspirate specimens called cell projection plots (CPPs). Encompassing relevant biological patterns such as neutrophil maturation, CPPs provide a compact summary of bone marrow aspirate cytology. To gauge clinical relevance, CPPs were inspected by 3 hematopathologists, who decided whether corresponding diagnostic synopses matched with generated CPPs. Pathologists were able to match CPPs to the correct synopsis with a matching degree of 85%. Our finding suggests CPPs can represent clinically relevant information from bone marrow aspirate specimens and may be used to efficiently summarize bone marrow cytology to pathologists. CPPs could be a step toward human-centered implementation of artificial intelligence (AI) in hematopathology, and a basis for a diagnostic-support tool for digital pathology workflows.

4.
bioRxiv ; 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37645835

RESUMO

The circulation of cerebrospinal fluid (CSF) is essential for maintaining brain homeostasis and clearance, and impairments in its flow can lead to various brain disorders. Recent studies have shown that CSF circulation can be interrogated using low b-value diffusion magnetic resonance imaging (low-b dMRI). Nevertheless, the spatial organization of intracranial CSF flow dynamics remains largely elusive. Here, we developed a whole-brain voxel-based analysis framework, termed CSF pseudo-diffusion spatial statistics (CΨSS), to examine CSF mean pseudo-diffusivity (MΨ), a measure of CSF flow magnitude derived from low-b dMRI. We showed that intracranial CSF MΨ demonstrates characteristic covariance patterns by employing seed-based correlation analysis. Importantly, we applied non-negative matrix factorization analysis to further elucidate the covariance patterns of CSF MΨ in a hypothesis-free, data-driven way. We identified distinct CSF spaces that consistently displayed unique pseudo-diffusion characteristics across multiple imaging datasets. Our study revealed that age, sex, brain atrophy, ventricular anatomy, and cerebral perfusion differentially influence MΨ across these CSF spaces. Notably, individuals with anomalous CSF flow patterns displayed incidental findings on multimodal neuroradiological examinations. Our work sets forth a new paradigm to study CSF flow, with potential applications in clinical settings.

5.
Diagn Pathol ; 18(1): 67, 2023 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-37198691

RESUMO

BACKGROUND: Deep learning models applied to healthcare applications including digital pathology have been increasing their scope and importance in recent years. Many of these models have been trained on The Cancer Genome Atlas (TCGA) atlas of digital images, or use it as a validation source. One crucial factor that seems to have been widely ignored is the internal bias that originates from the institutions that contributed WSIs to the TCGA dataset, and its effects on models trained on this dataset. METHODS: 8,579 paraffin-embedded, hematoxylin and eosin stained, digital slides were selected from the TCGA dataset. More than 140 medical institutions (acquisition sites) contributed to this dataset. Two deep neural networks (DenseNet121 and KimiaNet were used to extract deep features at 20× magnification. DenseNet was pre-trained on non-medical objects. KimiaNet has the same structure but trained for cancer type classification on TCGA images. The extracted deep features were later used to detect each slide's acquisition site, and also for slide representation in image search. RESULTS: DenseNet's deep features could distinguish acquisition sites with 70% accuracy whereas KimiaNet's deep features could reveal acquisition sites with more than 86% accuracy. These findings suggest that there are acquisition site specific patterns that could be picked up by deep neural networks. It has also been shown that these medically irrelevant patterns can interfere with other applications of deep learning in digital pathology, namely image search. This study shows that there are acquisition site specific patterns that can be used to identify tissue acquisition sites without any explicit training. Furthermore, it was observed that a model trained for cancer subtype classification has exploited such medically irrelevant patterns to classify cancer types. Digital scanner configuration and noise, tissue stain variation and artifacts, and source site patient demographics are among factors that likely account for the observed bias. Therefore, researchers should be cautious of such bias when using histopathology datasets for developing and training deep networks.


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Redes Neurais de Computação , Corantes , Hematoxilina , Amarelo de Eosina-(YS)
6.
Int J Lab Hematol ; 45 Suppl 2: 87-94, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37257440

RESUMO

An increasing number of machine learning applications are being developed and applied to digital pathology, including hematopathology. The goal of these modern computerized tools is often to support diagnostic workflows by extracting and summarizing information from multiple data sources, including digital images of human tissue. Hematopathology is inherently multimodal and can serve as an ideal case study for machine learning applications. However, hematopathology also poses unique challenges compared to other pathology subspecialities when applying machine learning approaches. By modeling the pathologist workflow and thinking process, machine learning algorithms may be designed to address practical and tangible problems in hematopathology. In this article, we discuss the current trends in machine learning in hematopathology. We review currently available machine learning enabled medical devices supporting hematopathology workflows. We then explore current machine learning research trends of the field with a focus on bone marrow cytology and histopathology, and how adoption of new machine learning tools may be enabled through the transition to digital pathology.


Assuntos
Algoritmos , Aprendizado de Máquina , Humanos , Patologistas , Fluxo de Trabalho
7.
Sci Rep ; 12(1): 19994, 2022 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-36411301

RESUMO

Appearing traces of bias in deep networks is a serious reliability issue which can play a significant role in ethics and generalization related concerns. Recent studies report that the deep features extracted from the histopathology images of The Cancer Genome Atlas (TCGA), the largest publicly available archive, are surprisingly able to accurately classify the whole slide images (WSIs) based on their acquisition site while these features are extracted to primarily discriminate cancer types. This is clear evidence that the utilized Deep Neural Networks (DNNs) unexpectedly detect the specific patterns of the source site, i.e, the hospital of origin, rather than histomorphologic patterns, a biased behavior resulting in degraded trust and generalization. This observation motivated us to propose a method to alleviate the destructive impact of hospital bias through a novel feature selection process. To this effect, we have proposed an evolutionary strategy to select a small set of optimal features to not only accurately represent the histological patterns of tissue samples but also to eliminate the features contributing to internal bias toward the institution. The defined objective function for an optimal subset selection of features is to minimize the accuracy of the model to classify the source institutions which is basically defined as a bias indicator. By the conducted experiments, the selected features extracted by the state-of-the-art network trained on TCGA images (i.e., the KimiaNet), considerably decreased the institutional bias, while improving the quality of features to discriminate the cancer types. In addition, the selected features could significantly improve the results of external validation compared to the entire set of features which has been negatively affected by bias. The proposed scheme is a model-independent approach which can be employed when it is possible to define a bias indicator as a participating objective in a feature selection process; even with unknown bias sources.


Assuntos
Redes Neurais de Computação , Reprodutibilidade dos Testes
8.
Artif Intell Med ; 132: 102368, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36207081

RESUMO

Despite the recent progress in Deep Neural Networks (DNNs) to characterize histopathology images, compactly representing a gigapixel whole-slide image (WSI) via salient features to enable computational pathology is still an urgent need and a significant challenge. In this paper, we propose a novel WSI characterization approach to represent, search and classify biopsy specimens using a compact feature vector (CFV) extracted from a multitude of deep feature vectors. Since the non-optimal design and training of deep networks may result in many irrelevant and redundant features and also cause computational bottlenecks, we proposed a low-cost stochastic method to optimize the output of pre-trained deep networks using evolutionary algorithms to generate a very small set of features to accurately represent each tissue/biopsy. The performance of the proposed method has been assessed using WSIs from the publicly available TCGA image data. In addition to acquiring a very compact representation (i.e., 11,000 times smaller than the initial set of features), the optimized features achieved 93% classification accuracy resulting in 11% improvement compared to the published benchmarks. The experimental results reveal that the proposed method can reliably select salient features of the biopsy sample. Furthermore, the proposed approach holds the potential to immensely facilitate the adoption of digital pathology by enabling a new generation of WSI representation for efficient storage and more user-friendly visualization.


Assuntos
Algoritmos , Redes Neurais de Computação
9.
Neurophotonics ; 9(4): 041410, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35769720

RESUMO

Significance: The firefly enzyme luciferase has been used in a wide range of biological assays, including bioluminescence imaging of adenosine triphosphate (ATP). The biosensor Syn-ATP utilizes subcellular targeting of luciferase to nerve terminals for optical measurement of ATP in this compartment. Manual analysis of Syn-ATP signals is challenging due to signal heterogeneity and cellular motion in long imaging sessions. Here, we have leveraged machine learning tools to develop a method for analysis of bioluminescence images. Aim: Our goal was to create a semiautomated pipeline for analysis of bioluminescence imaging to improve measurements of ATP content in nerve terminals. Approach: We developed an image analysis pipeline that applies machine learning toolkits to distinguish neurons from background signals and excludes neural cell bodies, while also incorporating user input. Results: Side-by-side comparison of manual and semiautomated image analysis demonstrated that the latter improves precision and accuracy of ATP measurements. Conclusions: Our method streamlines data analysis and reduces user-introduced bias, thus enhancing the reproducibility and reliability of quantitative ATP imaging in nerve terminals.

10.
Commun Med (Lond) ; 2: 45, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35603269

RESUMO

Background: Bone marrow cytology is required to make a hematological diagnosis, influencing critical clinical decision points in hematology. However, bone marrow cytology is tedious, limited to experienced reference centers and associated with inter-observer variability. This may lead to a delayed or incorrect diagnosis, leaving an unmet need for innovative supporting technologies. Methods: We develop an end-to-end deep learning-based system for automated bone marrow cytology. Starting with a bone marrow aspirate digital whole slide image, our system rapidly and automatically detects suitable regions for cytology, and subsequently identifies and classifies all bone marrow cells in each region. This collective cytomorphological information is captured in a representation called Histogram of Cell Types (HCT) quantifying bone marrow cell class probability distribution and acting as a cytological patient fingerprint. Results: Our system achieves high accuracy in region detection (0.97 accuracy and 0.99 ROC AUC), and cell detection and cell classification (0.75 mean average precision, 0.78 average F1-score, Log-average miss rate of 0.31). Conclusions: HCT has potential to eventually support more efficient and accurate diagnosis in hematology, supporting AI-enabled computational pathology.

11.
Am J Pathol ; 191(12): 2172-2183, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34508689

RESUMO

Although deep learning networks applied to digital images have shown impressive results for many pathology-related tasks, their black-box approach and limitation in terms of interpretability are significant obstacles for their widespread clinical utility. This study investigates the visualization of deep features (DFs) to characterize two lung cancer subtypes, adenocarcinoma and squamous cell carcinoma. It demonstrates that a subset of DFs, called prominent DFs, can accurately distinguish these two cancer subtypes. Visualization of such individual DFs allows for a better understanding of histopathologic patterns at both the whole-slide and patch levels, and discrimination of these cancer types. These DFs were visualized at the whole slide image level through DF-specific heatmaps and at tissue patch level through the generation of activation maps. In addition, these prominent DFs can distinguish carcinomas of organs other than the lung. This framework may serve as a platform for evaluating the interpretability of any deep network for diagnostic decision making.


Assuntos
Adenocarcinoma de Pulmão/diagnóstico , Carcinoma de Células Escamosas/diagnóstico , Aprendizado Profundo , Neoplasias Pulmonares/diagnóstico , Adenocarcinoma de Pulmão/patologia , Carcinoma de Células Escamosas/patologia , Conjuntos de Dados como Assunto , Diagnóstico Diferencial , Estudos de Viabilidade , Feminino , Humanos , Interpretação de Imagem Assistida por Computador/métodos , Processamento de Imagem Assistida por Computador/métodos , Neoplasias Pulmonares/patologia , Masculino , Redes Neurais de Computação , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
12.
Annu Int Conf IEEE Eng Med Biol Soc ; 2020: 2186-2189, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-33018440

RESUMO

Chest radiography has become the modality of choice for diagnosing pneumonia. However, analyzing chest X-ray images may be tedious, time-consuming and requiring expert knowledge that might not be available in less-developed regions. therefore, computer-aided diagnosis systems are needed. Recently, many classification systems based on deep learning have been proposed. Despite their success, the high development cost for deep networks is still a hurdle for deployment. Deep transfer learning (or simply transfer learning) has the merit of reducing the development cost by borrowing architectures from trained models followed by slight fine-tuning of some layers. Nevertheless, whether deep transfer learning is effective over training from scratch in the medical setting remains a research question for many applications. In this work, we investigate the use of deep transfer learning to classify pneumonia among chest X-ray images. Experimental results demonstrated that, with slight fine-tuning, deep transfer learning brings performance advantage over training from scratch. Three models, ResNet-50, Inception V3 and DensetNet121, were trained separately through transfer learning and from scratch. The former can achieve a 4.1% to 52.5% larger area under the curve (AUC) than those obtained by the latter, suggesting the effectiveness of deep transfer learning for classifying pneumonia in chest X-ray images.


Assuntos
Aprendizado Profundo , Pneumonia , Diagnóstico por Computador , Humanos , Pneumonia/diagnóstico por imagem , Radiografia , Raios X
13.
Annu Int Conf IEEE Eng Med Biol Soc ; 2020: 5308-5311, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-33019182

RESUMO

In this paper, we introduce a new dataset for cancer research containing somatic mutation states of 536 genes of the Cancer Gene Census (CGC). We used somatic mutation information from the Cancer Genome Atlas (TCGA) projects to create this dataset. As preliminary investigations, we employed machine learning techniques, including k-Nearest Neighbors, Decision Tree, Random Forest, and Artificial Neural Networks (ANNs) to evaluate the potential of these somatic mutations for classification of cancer types. We compared our models on accuracy, precision, recall, and F1-score. We observed that ANNs outperformed the other models with F1-score of 0.36 and overall classification accuracy of 40%, and precision ranging from 12% to 92% for different cancer types. The 40% accuracy is significantly higher than random guessing which would have resulted in 3% overall classification accuracy. Although the model has relatively low overall accuracy, it has an average classification specificity of 98%. The ANN achieved high precision scores (> 0.7) for 5 of the 33 cancer types. The introduced dataset can be used for research on TCGA data, such as survival analysis, histopathology image analysis and content-based image retrieval. The dataset is available online for download: https://kimialab.uwaterloo.ca/kimia/.


Assuntos
Neoplasias , Redes Neurais de Computação , Humanos , Aprendizado de Máquina , Mutação , Neoplasias/genética , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...