Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Med Image Anal ; 94: 103149, 2024 May.
Article in English | MEDLINE | ID: mdl-38574542

ABSTRACT

The variation in histologic staining between different medical centers is one of the most profound challenges in the field of computer-aided diagnosis. The appearance disparity of pathological whole slide images causes algorithms to become less reliable, which in turn impedes the wide-spread applicability of downstream tasks like cancer diagnosis. Furthermore, different stainings lead to biases in the training which in case of domain shifts negatively affect the test performance. Therefore, in this paper we propose MultiStain-CycleGAN, a multi-domain approach to stain normalization based on CycleGAN. Our modifications to CycleGAN allow us to normalize images of different origins without retraining or using different models. We perform an extensive evaluation of our method using various metrics and compare it to commonly used methods that are multi-domain capable. First, we evaluate how well our method fools a domain classifier that tries to assign a medical center to an image. Then, we test our normalization on the tumor classification performance of a downstream classifier. Furthermore, we evaluate the image quality of the normalized images using the Structural similarity index and the ability to reduce the domain shift using the Fréchet inception distance. We show that our method proves to be multi-domain capable, provides a very high image quality among the compared methods, and can most reliably fool the domain classifier while keeping the tumor classifier performance high. By reducing the domain influence, biases in the data can be removed on the one hand and the origin of the whole slide image can be disguised on the other, thus enhancing patient data privacy.


Subject(s)
Coloring Agents , Neoplasms , Humans , Coloring Agents/chemistry , Staining and Labeling , Algorithms , Diagnosis, Computer-Assisted , Image Processing, Computer-Assisted/methods
3.
Lab Invest ; 104(6): 102049, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38513977

ABSTRACT

Although pathological tissue analysis is typically performed on single 2-dimensional (2D) histologic reference slides, 3-dimensional (3D) reconstruction from a sequence of histologic sections could provide novel opportunities for spatial analysis of the extracted tissue. In this review, we analyze recent works published after 2018 and report information on the extracted tissue types, the section thickness, and the number of sections used for reconstruction. By analyzing the technological requirements for 3D reconstruction, we observe that software tools exist, both free and commercial, which include the functionality to perform 3D reconstruction from a sequence of histologic images. Through the analysis of the most recent works, we provide an overview of the workflows and tools that are currently used for 3D reconstruction from histologic sections and address points for future work, such as a missing common file format or computer-aided analysis of the reconstructed model.

4.
Nat Commun ; 15(1): 524, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38225244

ABSTRACT

Artificial intelligence (AI) systems have been shown to help dermatologists diagnose melanoma more accurately, however they lack transparency, hindering user acceptance. Explainable AI (XAI) methods can help to increase transparency, yet often lack precise, domain-specific explanations. Moreover, the impact of XAI methods on dermatologists' decisions has not yet been evaluated. Building upon previous research, we introduce an XAI system that provides precise and domain-specific explanations alongside its differential diagnoses of melanomas and nevi. Through a three-phase study, we assess its impact on dermatologists' diagnostic accuracy, diagnostic confidence, and trust in the XAI-support. Our results show strong alignment between XAI and dermatologist explanations. We also show that dermatologists' confidence in their diagnoses, and their trust in the support system significantly increase with XAI compared to conventional AI. This study highlights dermatologists' willingness to adopt such XAI systems, promoting future use in the clinic.


Subject(s)
Melanoma , Trust , Humans , Artificial Intelligence , Dermatologists , Melanoma/diagnosis , Diagnosis, Differential
5.
PLoS One ; 19(1): e0297146, 2024.
Article in English | MEDLINE | ID: mdl-38241314

ABSTRACT

Pathologists routinely use immunohistochemical (IHC)-stained tissue slides against MelanA in addition to hematoxylin and eosin (H&E)-stained slides to improve their accuracy in diagnosing melanomas. The use of diagnostic Deep Learning (DL)-based support systems for automated examination of tissue morphology and cellular composition has been well studied in standard H&E-stained tissue slides. In contrast, there are few studies that analyze IHC slides using DL. Therefore, we investigated the separate and joint performance of ResNets trained on MelanA and corresponding H&E-stained slides. The MelanA classifier achieved an area under receiver operating characteristics curve (AUROC) of 0.82 and 0.74 on out of distribution (OOD)-datasets, similar to the H&E-based benchmark classification of 0.81 and 0.75, respectively. A combined classifier using MelanA and H&E achieved AUROCs of 0.85 and 0.81 on the OOD datasets. DL MelanA-based assistance systems show the same performance as the benchmark H&E classification and may be improved by multi stain classification to assist pathologists in their clinical routine.


Subject(s)
Deep Learning , Melanoma , Humans , Melanoma/diagnosis , Immunohistochemistry , MART-1 Antigen , ROC Curve
6.
NPJ Precis Oncol ; 7(1): 98, 2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37752266

ABSTRACT

Studies have shown that colorectal cancer prognosis can be predicted by deep learning-based analysis of histological tissue sections of the primary tumor. So far, this has been achieved using a binary prediction. Survival curves might contain more detailed information and thus enable a more fine-grained risk prediction. Therefore, we established survival curve-based CRC survival predictors and benchmarked them against standard binary survival predictors, comparing their performance extensively on the clinical high and low risk subsets of one internal and three external cohorts. Survival curve-based risk prediction achieved a very similar risk stratification to binary risk prediction for this task. Exchanging other components of the pipeline, namely input tissue and feature extractor, had largely identical effects on model performance independently of the type of risk prediction. An ensemble of all survival curve-based models exhibited a more robust performance, as did a similar ensemble based on binary risk prediction. Patients could be further stratified within clinical risk groups. However, performance still varied across cohorts, indicating limited generalization of all investigated image analysis pipelines, whereas models using clinical data performed robustly on all cohorts.

7.
Med Image Anal ; 89: 102914, 2023 10.
Article in English | MEDLINE | ID: mdl-37544085

ABSTRACT

In the past years, deep learning has seen an increase in usage in the domain of histopathological applications. However, while these approaches have shown great potential, in high-risk environments deep learning models need to be able to judge their uncertainty and be able to reject inputs when there is a significant chance of misclassification. In this work, we conduct a rigorous evaluation of the most commonly used uncertainty and robustness methods for the classification of Whole Slide Images, with a focus on the task of selective classification, where the model should reject the classification in situations in which it is uncertain. We conduct our experiments on tile-level under the aspects of domain shift and label noise, as well as on slide-level. In our experiments, we compare Deep Ensembles, Monte-Carlo Dropout, Stochastic Variational Inference, Test-Time Data Augmentation as well as ensembles of the latter approaches. We observe that ensembles of methods generally lead to better uncertainty estimates as well as an increased robustness towards domain shifts and label noise, while contrary to results from classical computer vision benchmarks no systematic gain of the other methods can be shown. Across methods, a rejection of the most uncertain samples reliably leads to a significant increase in classification accuracy on both in-distribution as well as out-of-distribution data. Furthermore, we conduct experiments comparing these methods under varying conditions of label noise. Lastly, we publish our code framework to facilitate further research on uncertainty estimation on histopathological data.


Subject(s)
Benchmarking , Humans , Uncertainty , Probability
8.
Eur J Cancer ; 183: 131-138, 2023 04.
Article in English | MEDLINE | ID: mdl-36854237

ABSTRACT

BACKGROUND: In machine learning, multimodal classifiers can provide more generalised performance than unimodal classifiers. In clinical practice, physicians usually also rely on a range of information from different examinations for diagnosis. In this study, we used BRAF mutation status prediction in melanoma as a model system to analyse the contribution of different data types in a combined classifier because BRAF status can be determined accurately by sequencing as the current gold standard, thus nearly eliminating label noise. METHODS: We trained a deep learning-based classifier by combining individually trained random forests of image, clinical and methylation data to predict BRAF-V600 mutation status in primary and metastatic melanomas of The Cancer Genome Atlas cohort. RESULTS: With our multimodal approach, we achieved an area under the receiver operating characteristic curve of 0.80, whereas the individual classifiers yielded areas under the receiver operating characteristic curve of 0.63 (histopathologic image data), 0.66 (clinical data) and 0.66 (methylation data) on an independent data set. CONCLUSIONS: Our combined approach can predict BRAF status to some extent by identifying BRAF-V600 specific patterns at the histologic, clinical and epigenetic levels. The multimodal classifiers have improved generalisability in predicting BRAF mutation status.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Proto-Oncogene Proteins B-raf/genetics , Melanoma/pathology , Skin Neoplasms/pathology , Mutation , Epigenesis, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...