Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Nat Protoc ; 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39285224

RESUMO

Hematoxylin- and eosin-stained whole-slide images (WSIs) are the foundation of diagnosis of cancer. In recent years, development of deep learning-based methods in computational pathology has enabled the prediction of biomarkers directly from WSIs. However, accurately linking tissue phenotype to biomarkers at scale remains a crucial challenge for democratizing complex biomarkers in precision oncology. This protocol describes a practical workflow for solid tumor associative modeling in pathology (STAMP), enabling prediction of biomarkers directly from WSIs by using deep learning. The STAMP workflow is biomarker agnostic and allows for genetic and clinicopathologic tabular data to be included as an additional input, together with histopathology images. The protocol consists of five main stages that have been successfully applied to various research problems: formal problem definition, data preprocessing, modeling, evaluation and clinical translation. The STAMP workflow differentiates itself through its focus on serving as a collaborative framework that can be used by clinicians and engineers alike for setting up research projects in the field of computational pathology. As an example task, we applied STAMP to the prediction of microsatellite instability (MSI) status in colorectal cancer, showing accurate performance for the identification of tumors high in MSI. Moreover, we provide an open-source code base, which has been deployed at several hospitals across the globe to set up computational pathology workflows. The STAMP workflow requires one workday of hands-on computational execution and basic command line knowledge.

3.
Radiology ; 312(1): e232304, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-39012249

RESUMO

Background The level of background parenchymal enhancement (BPE) at breast MRI provides predictive and prognostic information and can have diagnostic implications. However, there is a lack of standardization regarding BPE assessment. Purpose To investigate how well results of quantitative BPE assessment methods correlate among themselves and with assessments made by radiologists experienced in breast MRI. Materials and Methods In this pseudoprospective analysis of 5773 breast MRI examinations from 3207 patients (mean age, 60 years ± 10 [SD]), the level of BPE was prospectively categorized according to the Breast Imaging Reporting and Data System by radiologists experienced in breast MRI. For automated extraction of BPE, fibroglandular tissue (FGT) was segmented in an automated pipeline. Four different published methods for automated quantitative BPE extractions were used: two methods (A and B) based on enhancement intensity and two methods (C and D) based on the volume of enhanced FGT. The results from all methods were correlated, and agreement was investigated in comparison with the respective radiologist-based categorization. For surrogate validation of BPE assessment, how accurately the methods distinguished premenopausal women with (n = 50) versus without (n = 896) antihormonal treatment was determined. Results Intensity-based methods (A and B) exhibited a correlation with radiologist-based categorization of 0.56 ± 0.01 and 0.55 ± 0.01, respectively, and volume-based methods (C and D) had a correlation of 0.52 ± 0.01 and 0.50 ± 0.01 (P < .001). There were notable correlation differences (P < .001) between the BPE determined with the four methods. Among the four quantitation methods, method D offered the highest accuracy for distinguishing women with versus without antihormonal therapy (P = .01). Conclusion Results of different methods for quantitative BPE assessment agree only moderately among themselves or with visual categories reported by experienced radiologists; intensity-based methods correlate more closely with radiologists' ratings than volume-based methods. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Mann in this issue.


Assuntos
Neoplasias da Mama , Mama , Imageamento por Ressonância Magnética , Humanos , Feminino , Pessoa de Meia-Idade , Imageamento por Ressonância Magnética/métodos , Neoplasias da Mama/diagnóstico por imagem , Mama/diagnóstico por imagem , Interpretação de Imagem Assistida por Computador/métodos , Adulto , Estudos Prospectivos , Aumento da Imagem/métodos , Idoso , Reprodutibilidade dos Testes , Estudos Retrospectivos
4.
Clin Oral Investig ; 28(7): 381, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38886242

RESUMO

OBJECTIVES: Tooth extraction is one of the most frequently performed medical procedures. The indication is based on the combination of clinical and radiological examination and individual patient parameters and should be made with great care. However, determining whether a tooth should be extracted is not always a straightforward decision. Moreover, visual and cognitive pitfalls in the analysis of radiographs may lead to incorrect decisions. Artificial intelligence (AI) could be used as a decision support tool to provide a score of tooth extractability. MATERIAL AND METHODS: Using 26,956 single teeth images from 1,184 panoramic radiographs (PANs), we trained a ResNet50 network to classify teeth as either extraction-worthy or preservable. For this purpose, teeth were cropped with different margins from PANs and annotated. The usefulness of the AI-based classification as well that of dentists was evaluated on a test dataset. In addition, the explainability of the best AI model was visualized via a class activation mapping using CAMERAS. RESULTS: The ROC-AUC for the best AI model to discriminate teeth worthy of preservation was 0.901 with 2% margin on dental images. In contrast, the average ROC-AUC for dentists was only 0.797. With a 19.1% tooth extractions prevalence, the AI model's PR-AUC was 0.749, while the dentist evaluation only reached 0.589. CONCLUSION: AI models outperform dentists/specialists in predicting tooth extraction based solely on X-ray images, while the AI performance improves with increasing contextual information. CLINICAL RELEVANCE: AI could help monitor at-risk teeth and reduce errors in indications for extractions.


Assuntos
Inteligência Artificial , Radiografia Panorâmica , Extração Dentária , Humanos , Odontólogos , Feminino , Masculino , Adulto
5.
Sci Rep ; 14(1): 10594, 2024 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-38719953

RESUMO

Colorectal liver metastases (CRLM) are the predominant factor limiting survival in patients with colorectal cancer and liver resection with complete tumor removal is the best treatment option for these patients. This study examines the predictive ability of three-dimensional lung volumetry (3DLV) based on preoperative computerized tomography (CT), to predict postoperative pulmonary complications in patients undergoing major liver resection for CRLM. Patients undergoing major curative liver resection for CRLM between 2010 and 2021 with a preoperative CT scan of the thorax within 6 weeks of surgery, were included. Total lung volume (TLV) was calculated using volumetry software 3D-Slicer version 4.11.20210226 including Chest Imaging Platform extension ( http://www.slicer.org ). The area under the curve (AUC) of a receiver-operating characteristic analysis was used to define a cut-off value of TLV, for predicting the occurrence of postoperative respiratory complications. Differences between patients with TLV below and above the cut-off were examined with Chi-square or Fisher's exact test and Mann-Whitney U tests and logistic regression was used to determine independent risk factors for the development of respiratory complications. A total of 123 patients were included, of which 35 (29%) developed respiratory complications. A predictive ability of TLV regarding respiratory complications was shown (AUC 0.62, p = 0.036) and a cut-off value of 4500 cm3 was defined. Patients with TLV < 4500 cm3 were shown to suffer from significantly higher rates of respiratory complications (44% vs. 21%, p = 0.007) compared to the rest. Logistic regression analysis identified TLV < 4500 cm3 as an independent predictor for the occurrence of respiratory complications (odds ratio 3.777, 95% confidence intervals 1.488-9.588, p = 0.005). Preoperative 3DLV is a viable technique for prediction of postoperative pulmonary complications in patients undergoing major liver resection for CRLM. More studies in larger cohorts are necessary to further evaluate this technique.


Assuntos
Neoplasias Colorretais , Hepatectomia , Neoplasias Hepáticas , Complicações Pós-Operatórias , Tomografia Computadorizada por Raios X , Humanos , Feminino , Masculino , Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Pessoa de Meia-Idade , Neoplasias Hepáticas/cirurgia , Neoplasias Hepáticas/secundário , Idoso , Hepatectomia/efeitos adversos , Hepatectomia/métodos , Complicações Pós-Operatórias/etiologia , Pulmão/patologia , Pulmão/diagnóstico por imagem , Pulmão/cirurgia , Estudos Retrospectivos , Imageamento Tridimensional , Medidas de Volume Pulmonar , Fatores de Risco , Período Pré-Operatório
6.
Nat Rev Gastroenterol Hepatol ; 21(8): 585-599, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38627537

RESUMO

Liver cancer has high incidence and mortality globally. Artificial intelligence (AI) has advanced rapidly, influencing cancer care. AI systems are already approved for clinical use in some tumour types (for example, colorectal cancer screening). Crucially, research demonstrates that AI can analyse histopathology, radiology and natural language in liver cancer, and can replace manual tasks and access hidden information in routinely available clinical data. However, for liver cancer, few of these applications have translated into large-scale clinical trials or clinically approved products. Here, we advocate for the incorporation of AI in all stages of liver cancer management. We present a taxonomy of AI approaches in liver cancer, highlighting areas with academic and commercial potential, and outline a policy for AI-based liver cancer management, including interdisciplinary training of researchers, clinicians and patients. The potential of AI in liver cancer is immense, but effort is required to ensure that AI can fulfil expectations.


Assuntos
Inteligência Artificial , Neoplasias Hepáticas , Humanos , Neoplasias Hepáticas/terapia , Pesquisa Biomédica
7.
Comput Biol Med ; 175: 108410, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38678938

RESUMO

Latent diffusion models (LDMs) have emerged as a state-of-the-art image generation method, outperforming previous Generative Adversarial Networks (GANs) in terms of training stability and image quality. In computational pathology, generative models are valuable for data sharing and data augmentation. However, the impact of LDM-generated images on histopathology tasks compared to traditional GANs has not been systematically studied. We trained three LDMs and a styleGAN2 model on histology tiles from nine colorectal cancer (CRC) tissue classes. The LDMs include 1) a fine-tuned version of stable diffusion v1.4, 2) a Kullback-Leibler (KL)-autoencoder (KLF8-DM), and 3) a vector quantized (VQ)-autoencoder deploying LDM (VQF8-DM). We assessed image quality through expert ratings, dimensional reduction methods, distribution similarity measures, and their impact on training a multiclass tissue classifier. Additionally, we investigated image memorization in the KLF8-DM and styleGAN2 models. All models provided a high image quality, with the KLF8-DM achieving the best Frechet Inception Distance (FID) and expert rating scores for complex tissue classes. For simpler classes, the VQF8-DM and styleGAN2 models performed better. Image memorization was negligible for both styleGAN2 and KLF8-DM models. Classifiers trained on a mix of KLF8-DM generated and real images achieved a 4% improvement in overall classification accuracy, highlighting the usefulness of these images for dataset augmentation. Our systematic study of generative methods showed that KLF8-DM produces the highest quality images with negligible image memorization. The higher classifier performance in the generatively augmented dataset suggests that this augmentation technique can be employed to enhance histopathology classifiers for various tasks.


Assuntos
Neoplasias Colorretais , Humanos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/diagnóstico por imagem , Interpretação de Imagem Assistida por Computador/métodos , Processamento de Imagem Assistida por Computador/métodos , Algoritmos
8.
Eur Radiol Exp ; 8(1): 53, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38689178

RESUMO

BACKGROUND: To compare denoising diffusion probabilistic models (DDPM) and generative adversarial networks (GAN) for recovering contrast-enhanced breast magnetic resonance imaging (MRI) subtraction images from virtual low-dose subtraction images. METHODS: Retrospective, ethically approved study. DDPM- and GAN-reconstructed single-slice subtraction images of 50 breasts with enhancing lesions were compared to original ones at three dose levels (25%, 10%, 5%) using quantitative measures and radiologic evaluations. Two radiologists stated their preference based on the reconstruction quality and scored the lesion conspicuity as compared to the original, blinded to the model. Fifty lesion-free maximum intensity projections were evaluated for the presence of false-positives. Results were compared between models and dose levels, using generalized linear mixed models. RESULTS: At 5% dose, both radiologists preferred the GAN-generated images, whereas at 25% dose, both radiologists preferred the DDPM-generated images. Median lesion conspicuity scores did not differ between GAN and DDPM at 25% dose (5 versus 5, p = 1.000) and 10% dose (4 versus 4, p = 1.000). At 5% dose, both readers assigned higher conspicuity to the GAN than to the DDPM (3 versus 2, p = 0.007). In the lesion-free examinations, DDPM and GAN showed no differences in the false-positive rate at 5% (15% versus 22%), 10% (10% versus 6%), and 25% (6% versus 4%) (p = 1.000). CONCLUSIONS: Both GAN and DDPM yielded promising results in low-dose image reconstruction. However, neither of them showed superior results over the other model for all dose levels and evaluation metrics. Further development is needed to counteract false-positives. RELEVANCE STATEMENT: For MRI-based breast cancer screening, reducing the contrast agent dose is desirable. Diffusion probabilistic models and generative adversarial networks were capable of retrospectively enhancing the signal of low-dose images. Hence, they may supplement imaging with reduced doses in the future. KEY POINTS: • Deep learning may help recover signal in low-dose contrast-enhanced breast MRI. • Two models (DDPM and GAN) were trained at different dose levels. • Radiologists preferred DDPM at 25%, and GAN images at 5% dose. • Lesion conspicuity between DDPM and GAN was similar, except at 5% dose. • GAN and DDPM yield promising results in low-dose image reconstruction.


Assuntos
Neoplasias da Mama , Meios de Contraste , Imageamento por Ressonância Magnética , Humanos , Feminino , Estudos Retrospectivos , Meios de Contraste/administração & dosagem , Neoplasias da Mama/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Pessoa de Meia-Idade , Modelos Estatísticos , Adulto , Idoso
9.
Diagnostics (Basel) ; 14(5)2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38472955

RESUMO

Increased attention has been given to MRI in radiation-free screening for malignant nodules in recent years. Our objective was to compare the performance of human readers and radiomic feature analysis based on stand-alone and complementary CT and MRI imaging in classifying pulmonary nodules. This single-center study comprises patients with CT findings of pulmonary nodules who underwent additional lung MRI and whose nodules were classified as benign/malignant by resection. For radiomic features analysis, 2D segmentation was performed for each lung nodule on axial CT, T2-weighted (T2w), and diffusion (DWI) images. The 105 extracted features were reduced by iterative backward selection. The performance of radiomics and human readers was compared by calculating accuracy with Clopper-Pearson confidence intervals. Fifty patients (mean age 63 +/- 10 years) with 66 pulmonary nodules (40 malignant) were evaluated. ACC values for radiomic features analysis vs. radiologists based on CT alone (0.68; 95%CI: 0.56, 0.79 vs. 0.59; 95%CI: 0.46, 0.71), T2w alone (0.65; 95%CI: 0.52, 0.77 vs. 0.68; 95%CI: 0.54, 0.78), DWI alone (0.61; 95%CI:0.48, 0.72 vs. 0.73; 95%CI: 0.60, 0.83), combined T2w/DWI (0.73; 95%CI: 0.60, 0.83 vs. 0.70; 95%CI: 0.57, 0.80), and combined CT/T2w/DWI (0.83; 95%CI: 0.72, 0.91 vs. 0.64; 95%CI: 0.51, 0.75) were calculated. This study is the first to show that by combining quantitative image information from CT, T2w, and DWI datasets, pulmonary nodule assessment through radiomics analysis is superior to using one modality alone, even exceeding human readers' performance.

10.
NPJ Precis Oncol ; 8(1): 72, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38519519

RESUMO

The technological progress in artificial intelligence (AI) has massively accelerated since 2022, with far-reaching implications for oncology and cancer research. Large language models (LLMs) now perform at human-level competency in text processing. Notably, both text and image processing networks are increasingly based on transformer neural networks. This convergence enables the development of multimodal AI models that take diverse types of data as an input simultaneously, marking a qualitative shift from specialized niche models which were prevalent in the 2010s. This editorial summarizes these developments, which are expected to impact precision oncology in the coming years.

11.
Commun Med (Lond) ; 4(1): 46, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38486100

RESUMO

BACKGROUND: Artificial intelligence (AI) models are increasingly used in the medical domain. However, as medical data is highly sensitive, special precautions to ensure its protection are required. The gold standard for privacy preservation is the introduction of differential privacy (DP) to model training. Prior work indicates that DP has negative implications on model accuracy and fairness, which are unacceptable in medicine and represent a main barrier to the widespread use of privacy-preserving techniques. In this work, we evaluated the effect of privacy-preserving training of AI models regarding accuracy and fairness compared to non-private training. METHODS: We used two datasets: (1) A large dataset (N = 193,311) of high quality clinical chest radiographs, and (2) a dataset (N = 1625) of 3D abdominal computed tomography (CT) images, with the task of classifying the presence of pancreatic ductal adenocarcinoma (PDAC). Both were retrospectively collected and manually labeled by experienced radiologists. We then compared non-private deep convolutional neural networks (CNNs) and privacy-preserving (DP) models with respect to privacy-utility trade-offs measured as area under the receiver operating characteristic curve (AUROC), and privacy-fairness trade-offs, measured as Pearson's r or Statistical Parity Difference. RESULTS: We find that, while the privacy-preserving training yields lower accuracy, it largely does not amplify discrimination against age, sex or co-morbidity. However, we find an indication that difficult diagnoses and subgroups suffer stronger performance hits in private training. CONCLUSIONS: Our study shows that - under the challenging realistic circumstances of a real-life clinical dataset - the privacy-preserving training of diagnostic deep learning models is possible with excellent diagnostic accuracy and fairness.


Artificial intelligence (AI), in which computers can learn to do tasks that normally require human intelligence, is particularly useful in medical imaging. However, AI should be used in a way that preserves patient privacy. We explored the balance between maintaining patient data privacy and AI performance in medical imaging. We use an approach called differential privacy to protect the privacy of patients' images. We show that, although training AI with differential privacy leads to a slight decrease in accuracy, it does not substantially increase bias against different age groups, genders, or patients with multiple health conditions. However, we notice that AI faces more challenges in accurately diagnosing complex cases and specific subgroups when trained under these privacy constraints. These findings highlight the importance of designing AI systems that are both privacy-conscious and capable of reliable diagnoses across patient groups.

12.
Nat Commun ; 15(1): 1603, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38383555

RESUMO

A knowledge gap persists between machine learning (ML) developers (e.g., data scientists) and practitioners (e.g., clinicians), hampering the full utilization of ML for clinical data analysis. We investigated the potential of the ChatGPT Advanced Data Analysis (ADA), an extension of GPT-4, to bridge this gap and perform ML analyses efficiently. Real-world clinical datasets and study details from large trials across various medical specialties were presented to ChatGPT ADA without specific guidance. ChatGPT ADA autonomously developed state-of-the-art ML models based on the original study's training data to predict clinical outcomes such as cancer development, cancer progression, disease complications, or biomarkers such as pathogenic gene sequences. Following the re-implementation and optimization of the published models, the head-to-head comparison of the ChatGPT ADA-crafted ML models and their respective manually crafted counterparts revealed no significant differences in traditional performance metrics (p ≥ 0.072). Strikingly, the ChatGPT ADA-crafted ML models often outperformed their counterparts. In conclusion, ChatGPT ADA offers a promising avenue to democratize ML in medicine by simplifying complex data analyses, yet should enhance, not replace, specialized training and resources, to promote broader applications in medical research and practice.


Assuntos
Algoritmos , Neoplasias , Humanos , Benchmarking , Idioma , Aprendizado de Máquina
13.
Arch Gynecol Obstet ; 309(4): 1543-1549, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37975899

RESUMO

PURPOSE: The market and application possibilities for artificial intelligence are currently growing at high speed and are increasingly finding their way into gynecology. While the medical side is highly represented in the current literature, the patient's perspective is still lagging behind. Therefore, the aim of this study was to evaluate the recommendations of ChatGPT regarding patient inquiries about the possible therapy of gynecological leading symptoms in a palliative situation by experts. METHODS: Case vignettes were constructed for 10 common concomitant symptoms in gynecologic oncology tumors in a palliative setting, and patient queries regarding therapy of these symptoms were generated as prompts for ChatGPT. Five experts in palliative care and gynecologic oncology evaluated the responses with respect to guideline adherence and applicability and identified advantages and disadvantages. RESULTS: The overall rating of ChatGPT responses averaged 4.1 (5 = strongly agree; 1 = strongly disagree). The experts saw an average guideline conformity of the therapy recommendations with a value of 4.0. ChatGPT sometimes omits relevant therapies and does not provide an individual assessment of the suggested therapies, but does indicate that a physician consultation is additionally necessary. CONCLUSIONS: Language models, such as ChatGPT, can provide valid and largely guideline-compliant therapy recommendations in their freely available and thus in principle accessible version for our patients. For a complete therapy recommendation, an evaluation of the therapies, their individual adjustment as well as a filtering of possible wrong recommendations, a medical expert's opinion remains indispensable.


Assuntos
Neoplasias dos Genitais Femininos , Ginecologia , Humanos , Feminino , Inteligência Artificial , Neoplasias dos Genitais Femininos/tratamento farmacológico , Cooperação do Paciente , Fidelidade a Diretrizes
14.
Lancet Digit Health ; 6(1): e33-e43, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38123254

RESUMO

BACKGROUND: Precise prognosis prediction in patients with colorectal cancer (ie, forecasting survival) is pivotal for individualised treatment and care. Histopathological tissue slides of colorectal cancer specimens contain rich prognostically relevant information. However, existing studies do not have multicentre external validation with real-world sample processing protocols, and algorithms are not yet widely used in clinical routine. METHODS: In this retrospective, multicentre study, we collected tissue samples from four groups of patients with resected colorectal cancer from Australia, Germany, and the USA. We developed and externally validated a deep learning-based prognostic-stratification system for automatic prediction of overall and cancer-specific survival in patients with resected colorectal cancer. We used the model-predicted risk scores to stratify patients into different risk groups and compared survival outcomes between these groups. Additionally, we evaluated the prognostic value of these risk groups after adjusting for established prognostic variables. FINDINGS: We trained and validated our model on a total of 4428 patients. We found that patients could be divided into high-risk and low-risk groups on the basis of the deep learning-based risk score. On the internal test set, the group with a high-risk score had a worse prognosis than the group with a low-risk score, as reflected by a hazard ratio (HR) of 4·50 (95% CI 3·33-6·09) for overall survival and 8·35 (5·06-13·78) for disease-specific survival (DSS). We found consistent performance across three large external test sets. In a test set of 1395 patients, the high-risk group had a lower DSS than the low-risk group, with an HR of 3·08 (2·44-3·89). In two additional test sets, the HRs for DSS were 2·23 (1·23-4·04) and 3·07 (1·78-5·3). We showed that the prognostic value of the deep learning-based risk score is independent of established clinical risk factors. INTERPRETATION: Our findings indicate that attention-based self-supervised deep learning can robustly offer a prognosis on clinical outcomes in patients with colorectal cancer, generalising across different populations and serving as a potentially new prognostic tool in clinical decision making for colorectal cancer management. We release all source codes and trained models under an open-source licence, allowing other researchers to reuse and build upon our work. FUNDING: The German Federal Ministry of Health, the Max-Eder-Programme of German Cancer Aid, the German Federal Ministry of Education and Research, the German Academic Exchange Service, and the EU.


Assuntos
Neoplasias Colorretais , Aprendizado Profundo , Humanos , Estudos Retrospectivos , Prognóstico , Fatores de Risco , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/patologia
15.
Med Image Anal ; 92: 103059, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38104402

RESUMO

Artificial intelligence (AI) has a multitude of applications in cancer research and oncology. However, the training of AI systems is impeded by the limited availability of large datasets due to data protection requirements and other regulatory obstacles. Federated and swarm learning represent possible solutions to this problem by collaboratively training AI models while avoiding data transfer. However, in these decentralized methods, weight updates are still transferred to the aggregation server for merging the models. This leaves the possibility for a breach of data privacy, for example by model inversion or membership inference attacks by untrusted servers. Somewhat-homomorphically-encrypted federated learning (SHEFL) is a solution to this problem because only encrypted weights are transferred, and model updates are performed in the encrypted space. Here, we demonstrate the first successful implementation of SHEFL in a range of clinically relevant tasks in cancer image analysis on multicentric datasets in radiology and histopathology. We show that SHEFL enables the training of AI models which outperform locally trained models and perform on par with models which are centrally trained. In the future, SHEFL can enable multiple institutions to co-train AI models without forsaking data governance and without ever transmitting any decryptable data to untrusted servers.


Assuntos
Neoplasias , Radiologia , Humanos , Inteligência Artificial , Aprendizagem , Neoplasias/diagnóstico por imagem , Processamento de Imagem Assistida por Computador
16.
J Pathol ; 262(3): 310-319, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38098169

RESUMO

Deep learning applied to whole-slide histopathology images (WSIs) has the potential to enhance precision oncology and alleviate the workload of experts. However, developing these models necessitates large amounts of data with ground truth labels, which can be both time-consuming and expensive to obtain. Pathology reports are typically unstructured or poorly structured texts, and efforts to implement structured reporting templates have been unsuccessful, as these efforts lead to perceived extra workload. In this study, we hypothesised that large language models (LLMs), such as the generative pre-trained transformer 4 (GPT-4), can extract structured data from unstructured plain language reports using a zero-shot approach without requiring any re-training. We tested this hypothesis by utilising GPT-4 to extract information from histopathological reports, focusing on two extensive sets of pathology reports for colorectal cancer and glioblastoma. We found a high concordance between LLM-generated structured data and human-generated structured data. Consequently, LLMs could potentially be employed routinely to extract ground truth data for machine learning from unstructured pathology reports in the future. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Assuntos
Glioblastoma , Medicina de Precisão , Humanos , Aprendizado de Máquina , Reino Unido
17.
Radiologie (Heidelb) ; 64(4): 295-303, 2024 Apr.
Artigo em Alemão | MEDLINE | ID: mdl-38158404

RESUMO

Magnetic resonance imaging (MRI) is the clinical method of choice for cartilage imaging in the context of degenerative and nondegenerative joint diseases. The MRI-based definitions of osteoarthritis rely on the detection of osteophytes, cartilage pathologies, bone marrow edema and meniscal lesions but currently a scientific consensus is lacking. In the clinical routine proton density-weighted, fat-suppressed 2D turbo spin echo sequences with echo times of 30-40 ms are predominantly used, which are sufficiently sensitive and specific for the assessment of cartilage. The additionally acquired T1-weighted sequences are primarily used for evaluating other intra-articular and periarticular structures. Diagnostically relevant artifacts include magic angle and chemical shift artifacts, which can lead to artificial signal enhancement in cartilage or incorrect representations of the subchondral lamina and its thickness. Although scientifically validated, high-resolution 3D gradient echo sequences (for cartilage segmentation) and compositional MR sequences (for quantification of physical tissue parameters) are currently reserved for scientific research questions. The future integration of artificial intelligence techniques in areas such as image reconstruction (to reduce scan times while maintaining image quality), image analysis (for automated identification of cartilage defects), and image postprocessing (for automated segmentation of cartilage in terms of volume and thickness) will significantly improve the diagnostic workflow and advance the field further.


Assuntos
Doenças das Cartilagens , Cartilagem Articular , Osteoartrite do Joelho , Humanos , Osteoartrite do Joelho/patologia , Cartilagem Articular/patologia , Inteligência Artificial , Doenças das Cartilagens/patologia , Imageamento por Ressonância Magnética/métodos
18.
Eur J Cancer ; 194: 113335, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37862795

RESUMO

AIM: Gastric cancer (GC) is a tumour entity with highly variant outcomes. Lymph node metastasis is a prognostically adverse biomarker. We hypothesised that GC primary tissue contains information that is predictive of lymph node status and patient prognosis and that this information can be extracted using deep learning (DL). METHODS: Using three patient cohorts comprising 1146 patients, we trained and validated a DL system to predict lymph node status directly from haematoxylin and eosin-stained GC tissue sections. We investigated the concordance between the DL-based prediction from the primary tumour slides (aiN score) and the histopathological lymph node status (pN). Furthermore, we assessed the prognostic value of the aiN score alone and when combined with the pN status. RESULTS: The aiN score predicted the pN status reaching area under the receiver operating characteristic curves of 0.71 in the training cohort and 0.69 and 0.65 in the two test cohorts. In a multivariate Cox analysis, the aiN score was an independent predictor of patient survival with hazard ratios of 1.5 in the training cohort and of 1.3 and 2.2 in the two test cohorts. A combination of the aiN score and the pN status prognostically stratified patients by survival with p-values <0.05 in logrank tests. CONCLUSION: GC primary tumour tissue contains additional prognostic information that is accessible using the aiN score. In combination with the pN status, this can be used for personalised management of GC patients after prospective validation.


Assuntos
Aprendizado Profundo , Neoplasias Gástricas , Humanos , Estudos Retrospectivos , Neoplasias Gástricas/patologia , Linfonodos/patologia , Prognóstico
19.
Cancer Cell ; 41(9): 1650-1661.e4, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37652006

RESUMO

Deep learning (DL) can accelerate the prediction of prognostic biomarkers from routine pathology slides in colorectal cancer (CRC). However, current approaches rely on convolutional neural networks (CNNs) and have mostly been validated on small patient cohorts. Here, we develop a new transformer-based pipeline for end-to-end biomarker prediction from pathology slides by combining a pre-trained transformer encoder with a transformer network for patch aggregation. Our transformer-based approach substantially improves the performance, generalizability, data efficiency, and interpretability as compared with current state-of-the-art algorithms. After training and evaluating on a large multicenter cohort of over 13,000 patients from 16 colorectal cancer cohorts, we achieve a sensitivity of 0.99 with a negative predictive value of over 0.99 for prediction of microsatellite instability (MSI) on surgical resection specimens. We demonstrate that resection specimen-only training reaches clinical-grade performance on endoscopic biopsy tissue, solving a long-standing diagnostic problem.


Assuntos
Algoritmos , Neoplasias Colorretais , Humanos , Biomarcadores , Biópsia , Instabilidade de Microssatélites , Neoplasias Colorretais/genética
20.
Radiology ; 307(5): e222223, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37278629

RESUMO

Background Deep learning (DL) models can potentially improve prognostication of rectal cancer but have not been systematically assessed. Purpose To develop and validate an MRI DL model for predicting survival in patients with rectal cancer based on segmented tumor volumes from pretreatment T2-weighted MRI scans. Materials and Methods DL models were trained and validated on retrospectively collected MRI scans of patients with rectal cancer diagnosed between August 2003 and April 2021 at two centers. Patients were excluded from the study if there were concurrent malignant neoplasms, prior anticancer treatment, incomplete course of neoadjuvant therapy, or no radical surgery performed. The Harrell C-index was used to determine the best model, which was applied to internal and external test sets. Patients were stratified into high- and low-risk groups based on a fixed cutoff calculated in the training set. A multimodal model was also assessed, which used DL model-computed risk score and pretreatment carcinoembryonic antigen level as input. Results The training set included 507 patients (median age, 56 years [IQR, 46-64 years]; 355 men). In the validation set (n = 218; median age, 55 years [IQR, 47-63 years]; 144 men), the best algorithm reached a C-index of 0.82 for overall survival. The best model reached hazard ratios of 3.0 (95% CI: 1.0, 9.0) in the high-risk group in the internal test set (n = 112; median age, 60 years [IQR, 52-70 years]; 76 men) and 2.3 (95% CI: 1.0, 5.4) in the external test set (n = 58; median age, 57 years [IQR, 50-67 years]; 38 men). The multimodal model further improved the performance, with a C-index of 0.86 and 0.67 for the validation and external test set, respectively. Conclusion A DL model based on preoperative MRI was able to predict survival of patients with rectal cancer. The model could be used as a preoperative risk stratification tool. Published under a CC BY 4.0 license. Supplemental material is available for this article. See also the editorial by Langs in this issue.


Assuntos
Aprendizado Profundo , Neoplasias Retais , Masculino , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Neoplasias Retais/diagnóstico por imagem , Neoplasias Retais/terapia , Imageamento por Ressonância Magnética , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA