Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
medRxiv ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38712033

RESUMEN

Computational analysis of histopathological specimens holds promise in identifying biomarkers, elucidating disease mechanisms, and streamlining clinical diagnosis. However, the application of deep learning techniques in vascular pathology remains underexplored. Here, we present a comprehensive evaluation of deep learning-based approaches to analyze digital whole-slide images of abdominal aortic aneurysm samples from 369 patients from three European centers. Deep learning demonstrated robust performance in predicting inflammatory characteristics, particularly in the adventitia, as well as fibrosis grade and remaining elastic fibers in the tunica media. Overall, this study represents the first comprehensive evaluation of computational pathology in vascular disease and has the potential to contribute to improved understanding of abdominal aortic aneurysm pathophysiology and personalization of treatment strategies, particularly when integrated with radiological phenotypes and clinical outcomes.

2.
Commun Med (Lond) ; 4(1): 71, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38605106

RESUMEN

BACKGROUND: The field of Artificial Intelligence (AI) holds transformative potential in medicine. However, the lack of universal reporting guidelines poses challenges in ensuring the validity and reproducibility of published research studies in this field. METHODS: Based on a systematic review of academic publications and reporting standards demanded by both international consortia and regulatory stakeholders as well as leading journals in the fields of medicine and medical informatics, 26 reporting guidelines published between 2009 and 2023 were included in this analysis. Guidelines were stratified by breadth (general or specific to medical fields), underlying consensus quality, and target research phase (preclinical, translational, clinical) and subsequently analyzed regarding the overlap and variations in guideline items. RESULTS: AI reporting guidelines for medical research vary with respect to the quality of the underlying consensus process, breadth, and target research phase. Some guideline items such as reporting of study design and model performance recur across guidelines, whereas other items are specific to particular fields and research stages. CONCLUSIONS: Our analysis highlights the importance of reporting guidelines in clinical AI research and underscores the need for common standards that address the identified variations and gaps in current guidelines. Overall, this comprehensive overview could help researchers and public stakeholders reinforce quality standards for increased reliability, reproducibility, clinical validity, and public trust in AI research in healthcare. This could facilitate the safe, effective, and ethical translation of AI methods into clinical applications that will ultimately improve patient outcomes.


Artificial Intelligence (AI) refers to computer systems that can perform tasks that normally require human intelligence, like recognizing patterns or making decisions. AI has the potential to transform healthcare, but research on AI in medicine needs clear rules so caregivers and patients can trust it. This study reviews and compares 26 existing guidelines for reporting on AI in medicine. The key differences between these guidelines are their target areas (medicine in general or specific medical fields), the ways they were created, and the research stages they address. While some key items like describing the AI model recurred across guidelines, others were specific to the research area. The analysis shows gaps and variations in current guidelines. Overall, transparent reporting is important, so AI research is reliable, reproducible, trustworthy, and safe for patients. This systematic review of guidelines aims to increase the transparency of AI research, supporting an ethical and safe progression of AI from research into clinical practice.

3.
Comput Biol Med ; 175: 108410, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38678938

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/diagnóstico por imagen , Interpretación de Imagen Asistida por Computador/métodos , Procesamiento de Imagen Asistido por Computador/métodos , Algoritmos
5.
Radiology ; 311(1): e232741, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38625006

RESUMEN

Background Procedural details of mechanical thrombectomy in patients with ischemic stroke are important predictors of clinical outcome and are collected for prospective studies or national stroke registries. To date, these data are collected manually by human readers, a labor-intensive task that is prone to errors. Purpose To evaluate the use of the large language models (LLMs) GPT-4 and GPT-3.5 to extract data from neuroradiology reports on mechanical thrombectomy in patients with ischemic stroke. Materials and Methods This retrospective study included consecutive reports from patients with ischemic stroke who underwent mechanical thrombectomy between November 2022 and September 2023 at institution 1 and between September 2016 and December 2019 at institution 2. A set of 20 reports was used to optimize the prompt, and the ability of the LLMs to extract procedural data from the reports was compared using the McNemar test. Data manually extracted by an interventional neuroradiologist served as the reference standard. Results A total of 100 internal reports from 100 patients (mean age, 74.7 years ± 13.2 [SD]; 53 female) and 30 external reports from 30 patients (mean age, 72.7 years ± 13.5; 18 male) were included. All reports were successfully processed by GPT-4 and GPT-3.5. Of 2800 data entries, 2631 (94.0% [95% CI: 93.0, 94.8]; range per category, 61%-100%) data points were correctly extracted by GPT-4 without the need for further postprocessing. With 1788 of 2800 correct data entries, GPT-3.5 produced fewer correct data entries than did GPT-4 (63.9% [95% CI: 62.0, 65.6]; range per category, 14%-99%; P < .001). For the external reports, GPT-4 extracted 760 of 840 (90.5% [95% CI: 88.3, 92.4]) correct data entries, while GPT-3.5 extracted 539 of 840 (64.2% [95% CI: 60.8, 67.4]; P < .001). Conclusion Compared with GPT-3.5, GPT-4 more frequently extracted correct procedural data from free-text reports on mechanical thrombectomy performed in patients with ischemic stroke. © RSNA, 2024 Supplemental material is available for this article.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Femenino , Masculino , Anciano , Accidente Cerebrovascular Isquémico/diagnóstico por imagen , Accidente Cerebrovascular Isquémico/cirugía , Estudios Retrospectivos , Estudios Prospectivos , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/cirugía , Trombectomía
6.
Artículo en Inglés | MEDLINE | ID: mdl-38627537

RESUMEN

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.

8.
Tumori ; : 3008916241231035, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38606831

RESUMEN

Artificial intelligence (AI) applications in oncology are at the forefront of transforming healthcare during the Fourth Industrial Revolution, driven by the digital data explosion. This review provides an accessible introduction to the field of AI, presenting a concise yet structured overview of the foundations of AI, including expert systems, classical machine learning, and deep learning, along with their contextual application in clinical research and healthcare. We delve into the current applications of AI in oncology, with a particular focus on diagnostic imaging and pathology. Numerous AI tools have already received regulatory approval, and more are under active development, bringing clear benefits but not without challenges. We discuss the importance of data security, the need for transparent and interpretable models, and the ethical considerations that must guide AI development in healthcare. By providing a perspective on the opportunities and challenges, this review aims to inform and guide researchers, clinicians, and policymakers in the adoption of AI in oncology.

9.
NPJ Precis Oncol ; 8(1): 72, 2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38519519

RESUMEN

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.

10.
Commun Med (Lond) ; 4(1): 48, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38491101

RESUMEN

BACKGROUND: The objective of this comprehensive pan-cancer study is to evaluate the potential of deep learning (DL) for molecular profiling of multi-omic biomarkers directly from hematoxylin and eosin (H&E)-stained whole slide images. METHODS: A total of 12,093 DL models predicting 4031 multi-omic biomarkers across 32 cancer types were trained and validated. The study included a broad range of genetic, transcriptomic, and proteomic biomarkers, as well as established prognostic markers, molecular subtypes, and clinical outcomes. RESULTS: Here we show that 50% of the models achieve an area under the curve (AUC) of 0.644 or higher. The observed AUC for 25% of the models is at least 0.719 and exceeds 0.834 for the top 5%. Molecular profiling with image-based histomorphological features is generally considered feasible for most of the investigated biomarkers and across different cancer types. The performance appears to be independent of tumor purity, sample size, and class ratio (prevalence), suggesting a degree of inherent predictability in histomorphology. CONCLUSIONS: The results demonstrate that DL holds promise to predict a wide range of biomarkers across the omics spectrum using only H&E-stained histological slides of solid tumors. This paves the way for accelerating diagnosis and developing more precise treatments for cancer patients.


Molecular profiling tests are used to check cancers for changes in certain genes, proteins, or other molecules. Results of such tests can be used to identify the most effective treatment for cancer patients. Faster and more accessible alternatives to standard tests are needed to improve cancer care. This study investigates whether deep learning (DL), a series of advanced computer techniques, can perform molecular profiling directly from routinely-collected images of tumor specimens used for diagnostic purposes. Over 12,000 DL models were utilized to evaluate thousands of biomarkers using statistical approaches. The results indicate that DL can effectively detect molecular changes in a tumor from these images, for many biomarkers and tumor types. The study shows that DL-based molecular profiling from images is possible. Introducing this type of approach into routine clinical workflows could potentially accelerate treatment decisions and improve outcomes.

11.
Genome Med ; 16(1): 44, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38539231

RESUMEN

Histopathology and genomic profiling are cornerstones of precision oncology and are routinely obtained for patients with cancer. Traditionally, histopathology slides are manually reviewed by highly trained pathologists. Genomic data, on the other hand, is evaluated by engineered computational pipelines. In both applications, the advent of modern artificial intelligence methods, specifically machine learning (ML) and deep learning (DL), have opened up a fundamentally new way of extracting actionable insights from raw data, which could augment and potentially replace some aspects of traditional evaluation workflows. In this review, we summarize current and emerging applications of DL in histopathology and genomics, including basic diagnostic as well as advanced prognostic tasks. Based on a growing body of evidence, we suggest that DL could be the groundwork for a new kind of workflow in oncology and cancer research. However, we also point out that DL models can have biases and other flaws that users in healthcare and research need to know about, and we propose ways to address them.


Asunto(s)
Aprendizaje Profundo , Neoplasias , Humanos , Inteligencia Artificial , Neoplasias/genética , Neoplasias/diagnóstico , Medicina de Precisión/métodos , Genómica/métodos
12.
Eur Radiol Exp ; 8(1): 10, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38326501

RESUMEN

BACKGROUND: Pretraining labeled datasets, like ImageNet, have become a technical standard in advanced medical image analysis. However, the emergence of self-supervised learning (SSL), which leverages unlabeled data to learn robust features, presents an opportunity to bypass the intensive labeling process. In this study, we explored if SSL for pretraining on non-medical images can be applied to chest radiographs and how it compares to supervised pretraining on non-medical images and on medical images. METHODS: We utilized a vision transformer and initialized its weights based on the following: (i) SSL pretraining on non-medical images (DINOv2), (ii) supervised learning (SL) pretraining on non-medical images (ImageNet dataset), and (iii) SL pretraining on chest radiographs from the MIMIC-CXR database, the largest labeled public dataset of chest radiographs to date. We tested our approach on over 800,000 chest radiographs from 6 large global datasets, diagnosing more than 20 different imaging findings. Performance was quantified using the area under the receiver operating characteristic curve and evaluated for statistical significance using bootstrapping. RESULTS: SSL pretraining on non-medical images not only outperformed ImageNet-based pretraining (p < 0.001 for all datasets) but, in certain cases, also exceeded SL on the MIMIC-CXR dataset. Our findings suggest that selecting the right pretraining strategy, especially with SSL, can be pivotal for improving diagnostic accuracy of artificial intelligence in medical imaging. CONCLUSIONS: By demonstrating the promise of SSL in chest radiograph analysis, we underline a transformative shift towards more efficient and accurate AI models in medical imaging. RELEVANCE STATEMENT: Self-supervised learning highlights a paradigm shift towards the enhancement of AI-driven accuracy and efficiency in medical imaging. Given its promise, the broader application of self-supervised learning in medical imaging calls for deeper exploration, particularly in contexts where comprehensive annotated datasets are limited.


Asunto(s)
Inteligencia Artificial , Aprendizaje Profundo , Bases de Datos Factuales
13.
Histopathology ; 84(7): 1139-1153, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38409878

RESUMEN

BACKGROUND: Artificial intelligence (AI) has numerous applications in pathology, supporting diagnosis and prognostication in cancer. However, most AI models are trained on highly selected data, typically one tissue slide per patient. In reality, especially for large surgical resection specimens, dozens of slides can be available for each patient. Manually sorting and labelling whole-slide images (WSIs) is a very time-consuming process, hindering the direct application of AI on the collected tissue samples from large cohorts. In this study we addressed this issue by developing a deep-learning (DL)-based method for automatic curation of large pathology datasets with several slides per patient. METHODS: We collected multiple large multicentric datasets of colorectal cancer histopathological slides from the United Kingdom (FOXTROT, N = 21,384 slides; CR07, N = 7985 slides) and Germany (DACHS, N = 3606 slides). These datasets contained multiple types of tissue slides, including bowel resection specimens, endoscopic biopsies, lymph node resections, immunohistochemistry-stained slides, and tissue microarrays. We developed, trained, and tested a deep convolutional neural network model to predict the type of slide from the slide overview (thumbnail) image. The primary statistical endpoint was the macro-averaged area under the receiver operating curve (AUROCs) for detection of the type of slide. RESULTS: In the primary dataset (FOXTROT), with an AUROC of 0.995 [95% confidence interval [CI]: 0.994-0.996] the algorithm achieved a high classification performance and was able to accurately predict the type of slide from the thumbnail image alone. In the two external test cohorts (CR07, DACHS) AUROCs of 0.982 [95% CI: 0.979-0.985] and 0.875 [95% CI: 0.864-0.887] were observed, which indicates the generalizability of the trained model on unseen datasets. With a confidence threshold of 0.95, the model reached an accuracy of 94.6% (7331 classified cases) in CR07 and 85.1% (2752 classified cases) for the DACHS cohort. CONCLUSION: Our findings show that using the low-resolution thumbnail image is sufficient to accurately classify the type of slide in digital pathology. This can support researchers to make the vast resource of existing pathology archives accessible to modern AI models with only minimal manual annotations.


Asunto(s)
Neoplasias Colorrectales , Aprendizaje Profundo , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/diagnóstico , Redes Neurales de la Computación , Procesamiento de Imagen Asistido por Computador/métodos , Interpretación de Imagen Asistida por Computador/métodos
15.
Nat Commun ; 15(1): 1253, 2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38341402

RESUMEN

Deep Learning (DL) can predict biomarkers from cancer histopathology. Several clinically approved applications use this technology. Most approaches, however, predict categorical labels, whereas biomarkers are often continuous measurements. We hypothesize that regression-based DL outperforms classification-based DL. Therefore, we develop and evaluate a self-supervised attention-based weakly supervised regression method that predicts continuous biomarkers directly from 11,671 images of patients across nine cancer types. We test our method for multiple clinically and biologically relevant biomarkers: homologous recombination deficiency score, a clinically used pan-cancer biomarker, as well as markers of key biological processes in the tumor microenvironment. Using regression significantly enhances the accuracy of biomarker prediction, while also improving the predictions' correspondence to regions of known clinical relevance over classification. In a large cohort of colorectal cancer patients, regression-based prediction scores provide a higher prognostic value than classification-based scores. Our open-source regression approach offers a promising alternative for continuous biomarker analysis in computational pathology.


Asunto(s)
Aprendizaje Profundo , Neoplasias , Humanos , Biomarcadores de Tumor/genética , Tecnología , Microambiente Tumoral
16.
Nat Commun ; 15(1): 1603, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383555

RESUMEN

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.


Asunto(s)
Algoritmos , Neoplasias , Humanos , Benchmarking , Lenguaje , Aprendizaje Automático
17.
BMC Med Genomics ; 17(1): 48, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38317154

RESUMEN

BACKGROUND: Digitized histopathological tissue slides and genomics profiling data are available for many patients with solid tumors. In the last 5 years, Deep Learning (DL) has been broadly used to extract clinically actionable information and biological knowledge from pathology slides and genomic data in cancer. In addition, a number of recent studies have introduced multimodal DL models designed to simultaneously process both images from pathology slides and genomic data as inputs. By comparing patterns from one data modality with those in another, multimodal DL models are capable of achieving higher performance compared to their unimodal counterparts. However, the application of these methodologies across various tumor entities and clinical scenarios lacks consistency. METHODS: Here, we present a systematic survey of the academic literature from 2010 to November 2023, aiming to quantify the application of DL for pathology, genomics, and the combined use of both data types. After filtering 3048 publications, our search identified 534 relevant articles which then were evaluated by basic (diagnosis, grading, subtyping) and advanced (mutation, drug response and survival prediction) application types, publication year and addressed cancer tissue. RESULTS: Our analysis reveals a predominant application of DL in pathology compared to genomics. However, there is a notable surge in DL incorporation within both domains. Furthermore, while DL applied to pathology primarily targets the identification of histology-specific patterns in individual tissues, DL in genomics is more commonly used in a pan-cancer context. Multimodal DL, on the contrary, remains a niche topic, evidenced by a limited number of publications, primarily focusing on prognosis predictions. CONCLUSION: In summary, our quantitative analysis indicates that DL not only has a well-established role in histopathology but is also being successfully integrated into both genomic and multimodal applications. In addition, there is considerable potential in multimodal DL for harnessing further advanced tasks, such as predicting drug response. Nevertheless, this review also underlines the need for further research to bridge the existing gaps in these fields.


Asunto(s)
Aprendizaje Profundo , Neoplasias , Humanos , Neoplasias/genética , Medicina de Precisión , Genómica , Mutación
18.
NPJ Precis Oncol ; 8(1): 23, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38291217

RESUMEN

Until recently the application of artificial intelligence (AI) in precision oncology was confined to activities in drug development and had limited impact on the personalisation of therapy. Now, a number of approaches have been proposed for the personalisation of drug and cell therapies with AI applied to therapy design, planning and delivery at the patient's bedside. Some drug and cell-based therapies are already tuneable to the individual to optimise efficacy, to reduce toxicity, to adapt the dosing regime, to design combination therapy approaches and, preclinically, even to personalise the receptor design of cell therapies. Developments in AI-based healthcare are accelerating through the adoption of foundation models, and generalist medical AI models have been proposed. The application of these approaches in therapy design is already being explored and realistic short-term advances include the application to the personalised design and delivery of drugs and cell therapies. With this pace of development, the limiting step to adoption will likely be the capacity and appropriateness of regulatory frameworks. This article explores emerging concepts and new ideas for the regulation of AI-enabled personalised cancer therapies in the context of existing and in development governance frameworks.

19.
Eur Urol Oncol ; 7(1): 157-159, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37839981
20.
Eur Radiol ; 34(2): 1176-1178, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37580599
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...