Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cancer Radiother ; 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39138047

ABSTRACT

Over the last decades, the use of artificial intelligence, machine learning and deep learning in medical fields has skyrocketed. Well known for their results in segmentation, motion management and posttreatment outcome tasks, investigations of machine learning and deep learning models as fast dose calculation or quality assurance tools have been present since 2000. The main motivation for this increasing research and interest in artificial intelligence, machine learning and deep learning is the enhancement of treatment workflows, specifically dosimetry and quality assurance accuracy and time points, which remain important time-consuming aspects of clinical patient management. Since 2014, the evolution of models and architectures for dose calculation has been related to innovations and interest in the theory of information research with pronounced improvements in architecture design. The use of knowledge-based approaches to patient-specific methods has also considerably improved the accuracy of dose predictions. This paper covers the state of all known deep learning architectures and models applied to external radiotherapy with a description of each architecture, followed by a discussion on the performance and future of deep learning predictive models in external radiotherapy.

2.
J Fr Ophtalmol ; 47(7): 104242, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39013268

ABSTRACT

In the last decade, artificial intelligence (AI) has significantly impacted ophthalmology, particularly in managing corneal diseases, a major reversible cause of blindness. This review explores AI's transformative role in the corneal subspecialty, which has adopted advanced technology for superior clinical judgment, early diagnosis, and personalized therapy. While AI's role in anterior segment diseases is less documented compared to glaucoma and retinal pathologies, this review highlights its integration into corneal diagnostics through imaging techniques like slit-lamp biomicroscopy, anterior segment optical coherence tomography (AS-OCT), and in vivo confocal biomicroscopy. AI has been pivotal in refining decision-making and prognosis for conditions such as keratoconus, infectious keratitis, and dystrophies. Multi-disease deep learning neural networks (MDDNs) have shown diagnostic ability in classifying corneal diseases using AS-OCT images, achieving notable metrics like an AUC of 0.910. AI's progress over two decades has significantly improved the accuracy of diagnosing conditions like keratoconus and microbial keratitis. For instance, AI has achieved a 90.7% accuracy rate in classifying bacterial and fungal keratitis and an AUC of 0.910 in differentiating various corneal diseases. Convolutional neural networks (CNNs) have enhanced the analysis of color-coded corneal maps, yielding up to 99.3% diagnostic accuracy for keratoconus. Deep learning algorithms have also shown robust performance in detecting fungal hyphae on in vivo confocal microscopy, with precise quantification of hyphal density. AI models combining tomography scans and visual acuity have demonstrated up to 97% accuracy in keratoconus staging according to the Amsler-Krumeich classification. However, the review acknowledges the limitations of current AI models, including their reliance on binary classification, which may not capture the complexity of real-world clinical presentations with multiple coexisting disorders. Challenges also include dependency on data quality, diverse imaging protocols, and integrating multimodal images for a generalized AI diagnosis. The need for interpretability in AI models is emphasized to foster trust and applicability in clinical settings. Looking ahead, AI has the potential to unravel the intricate mechanisms behind corneal pathologies, reduce healthcare's carbon footprint, and revolutionize diagnostic and management paradigms. Ethical and regulatory considerations will accompany AI's clinical adoption, marking an era where AI not only assists but augments ophthalmic care.

3.
Cancer Radiother ; 28(2): 208-217, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38519291

ABSTRACT

In recent years, with the development of artificial intelligence, deep learning has been gradually applied to clinical treatment and research. It has also found its way into the applications in radiotherapy, a crucial method for cancer treatment. This study summarizes the commonly used and latest deep learning algorithms (including transformer, and diffusion models), introduces the workflow of different radiotherapy, and illustrates the application of different algorithms in different radiotherapy modules, as well as the defects and challenges of deep learning in the field of radiotherapy, so as to provide some help for the development of automatic radiotherapy for cancer.


Subject(s)
Deep Learning , Neoplasms , Humans , Artificial Intelligence , Neoplasms/radiotherapy , Algorithms , Radiotherapy Planning, Computer-Assisted/methods
4.
Cancer Radiother ; 27(8): 705-711, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37932182

ABSTRACT

PURPOSE: The purpose of this study was to develop a model for predicting chemoradiation response in non-small cell lung cancer (NSCLC) patients by integrating radiomics and deep-learning features and combined intra- and peritumoral regions with pre-treated CT images. MATERIALS AND METHODS: This study enrolled 462 patients with NSCLC who received chemoradiation. On the basis of pretreated CT images, we developed three models to compare the prediction of chemoradiation: intratumoral, peritumoral and combined regions. To further illustrate each model, we established different feature integration methods: a) radiomics model with 1500 features; b) deep learning model with a multiple instance learning algorithm; c) integrated model by integrating radiomic and deep learning features. For radiomics and integrated models, support vector machine and the least absolute shrinkage and selection operator were used to extract and select features. Transfer learning and max pooling algorithms were used to identify high informative features in deep learning models. We applied ten-fold cross validation in model training and testing. RESULTS: The best area under the curve (AUC) of intratumoral, peritumoral and combined models were 0.89 (95% CI, 0.74-0.93), 0.86 (95% CI, 0.75-0.92) and 0.92 (95% CI, 0.81-0.95), respectively. It indicated the importance of the peritumoral region for treatment response prediction and should be used in combination with the intratumoral region. Integrated models gave better results than models with radiomics and deep learning features alone in all regions of interest and radiomics models outperformed deep learning models in any comparative models. CONCLUSIONS: The model that integrate radiomic and deep learning features and combined intra- and peritumoral regions provide valuable information in predicting treatment response of chemoradiation. It can help oncologists customize personalized clinical treatment plans for NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Deep Learning , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/therapy , Algorithms , Area Under Curve , Retrospective Studies
5.
J Fr Ophtalmol ; 46(7): 706-711, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37537126

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the performance of ChatGPT, a cutting-edge artificial intelligence (AI) language model developed by OpenAI, in successfully completing the French language version of the European Board of Ophthalmology (EBO) examination and to assess its potential role in medical education and knowledge assessment. METHODS: ChatGPT, based on the GPT-4 architecture, was exposed to a series of EBO examination questions in French, covering various aspects of ophthalmology. The AI's performance was evaluated by comparing its responses with the correct answers provided by ophthalmology experts. Additionally, the study assessed the time taken by ChatGPT to answer each question as a measure of efficiency. RESULTS: ChatGPT achieved a 91% success rate on the EBO examination, demonstrating a high level of competency in ophthalmology knowledge and application. The AI provided correct answers across all question categories, indicating a strong understanding of basic sciences, clinical knowledge, and clinical management. The AI model also answered the questions rapidly, taking only a fraction of the time needed by human test-takers. CONCLUSION: ChatGPT's performance on the French language version of the EBO examination demonstrates its potential to be a valuable tool in medical education and knowledge assessment. Further research is needed to explore optimal ways to implement AI language models in medical education and to address the associated ethical and practical concerns.


Subject(s)
Artificial Intelligence , Ophthalmology , Humans , Language
6.
Cancer Radiother ; 27(6-7): 542-547, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37481344

ABSTRACT

Over the last decades, the refinement of radiation therapy techniques has been associated with an increasing interest for individualized radiation therapy with the aim of increasing or maintaining tumor control and reducing radiation toxicity. Developments in artificial intelligence (AI), particularly machine learning and deep learning, in imaging sciences, including nuclear medecine, have led to significant enthusiasm for the concept of "rapid learning health system". AI combined with radiomics applied to (18F)-fluorodeoxyglucose positron emission tomography/computed tomography ([18F]-FDG PET/CT) offers a unique opportunity for the development of predictive models that can help stratify each patient's risk and guide treatment decisions for optimal outcomes and quality of life of patients treated with radiation therapy. Here we present an overview of the current contribution of AI and radiomics-based machine learning models applied to (18F)-FDG PET/CT in the management of cancer treated by radiation therapy.


Subject(s)
Positron Emission Tomography Computed Tomography , Radiation Oncology , Humans , Fluorodeoxyglucose F18 , Artificial Intelligence , Quality of Life
7.
Cancer Radiother ; 27(5): 398-406, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37482464

ABSTRACT

PURPOSE: This study aims to perform a comprehensive systematic review of deep learning (DL) models in predicting RT-induced toxicity. MATERIALS AND METHODS: A literature review was performed following the PRISMA guidelines. Systematic searches were performed in PubMed, Scopus, Cochrane and Embase databases from the earliest record up to September 2022. Related studies on deep learning models for radiotherapy toxicity prediction were selected based on predefined PICOS criteria. RESULTS: Fourteen studies of radiotherapy-treated patients on different types of cancer [prostate (n=2), HNC (n=4), liver (n=2), lung (n=4), cervical (n=1), and oesophagus (n=1)] were eligible for inclusion in the systematic review. Information regarding patient characteristics and model development was summarized. Several approaches, such as ensemble learning, data augmentation, and transfer learning, that were utilized by selected studies were discussed. CONCLUSION: Deep learning techniques are able to produce a consistent performance for toxicity prediction. Future research using large and diverse datasets and standardization of the study methodologies are required to improve the consistency of the research output.


Subject(s)
Deep Learning , Neoplasms , Male , Humans
8.
Rev Mal Respir ; 40(4): 308-313, 2023 Apr.
Article in French | MEDLINE | ID: mdl-36894376

ABSTRACT

Artificial intelligence (AI) is a growing field that has the potential to transform many areas of society, including healthcare. For a physician, it is important to understand the basics of AI and its potential applications in medicine. AI refers to the development of computer systems capable of performing tasks that typically require human intelligence, such as pattern recognition, learning from data, and decision-making. This technology can be used to analyze large amounts of patient data and to identify trends and patterns that can be difficult for human physicians to detect. This can help doctors to manage their workload more efficiently and provide better care for their patients. All in all, AI has the potential to dramatically improve the practice of medicine and improve patient outcomes. In this work, the definition and the key principles of AI are outlined, with particular focus on the field of machine learning, which has been undergoing considerable development in medicine, providing clinicians with in-depth understanding of the principles underlying the new technologies ensuring improved health care.


Subject(s)
Artificial Intelligence , Physicians , Humans , Machine Learning , Delivery of Health Care , Workload
9.
Cancer Radiother ; 26(8): 1008-1015, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35803861

ABSTRACT

PURPOSE: Deep learning (DL) techniques are widely used in medical imaging and in particular for segmentation. Indeed, manual segmentation of organs at risk (OARs) is time-consuming and suffers from inter- and intra-observer segmentation variability. Image segmentation using DL has given very promising results. In this work, we present and compare the results of segmentation of OARs and a clinical target volume (CTV) in thoracic CT images using three DL models. MATERIALS AND METHODS: We used CT images of 52 patients with breast cancer from a public dataset. Automatic segmentation of the lungs, the heart and a CTV was performed using three models based on the U-Net architecture. Three metrics were used to quantify and compare the segmentation results obtained with these models: the Dice similarity coefficient (DSC), the Jaccard coefficient (J) and the Hausdorff distance (HD). RESULTS: The obtained values of DSC, J and HD were presented for each segmented organ and for the three models. Examples of automatic segmentation were presented and compared to the corresponding ground truth delineations. Our values were also compared to recent results obtained by other authors. CONCLUSION: The performance of three DL models was evaluated for the delineation of the lungs, the heart and a CTV. This study showed clearly that these 2D models based on the U-Net architecture can be used to delineate organs in CT images with a good performance compared to other models. Generally, the three models present similar performances. Using a dataset with more CT images, the three models should give better results.


Subject(s)
Deep Learning , Humans , Image Processing, Computer-Assisted , Tomography, X-Ray Computed/methods , Organs at Risk , Observer Variation
10.
J Fr Ophtalmol ; 45(2): 216-232, 2022 Feb.
Article in French | MEDLINE | ID: mdl-34991909

ABSTRACT

In recent years, research in artificial intelligence (AI) has experienced an unprecedented surge in the field of ophthalmology, in particular glaucoma. The diagnosis and follow-up of glaucoma is complex and relies on a body of clinical evidence and ancillary tests. This large amount of information from structural and functional testing of the optic nerve and macula makes glaucoma a particularly appropriate field for the application of AI. In this paper, we will review work using AI in the field of glaucoma, whether for screening, diagnosis or detection of progression. Many AI strategies have shown promising results for glaucoma detection using fundus photography, optical coherence tomography, or automated perimetry. The combination of these imaging modalities increases the performance of AI algorithms, with results comparable to those of humans. We will discuss potential applications as well as obstacles and limitations to the deployment and validation of such models. While there is no doubt that AI has the potential to revolutionize glaucoma management and screening, research in the coming years will need to address unavoidable questions regarding the clinical significance of such results and the explicability of the predictions.


Subject(s)
Artificial Intelligence , Glaucoma , Algorithms , Diagnostic Techniques, Ophthalmological , Glaucoma/diagnosis , Humans , Visual Field Tests
11.
Cancer Radiother ; 26(3): 494-501, 2022 May.
Article in English | MEDLINE | ID: mdl-34711488

ABSTRACT

The technological advancement heralded the arrival of precision radiotherapy (RT), thereby increasing the therapeutic ratio and decreasing the side effects from treatment. Contour of target volumes (TV) and organs at risk (OARs) in RT is a complicated process. In recent years, automatic contouring of TV and OARs has rapidly developed due to the advances in deep learning (DL). This technology has the potential to save time and to reduce intra- or inter-observer variability. In this paper, the authors provide an overview of RT, introduce the concept of DL, summarize the data characteristics of the included literature, summarize the possible challenges for DL in the future, and discuss the possible research directions.


Subject(s)
Deep Learning , Radiation Oncology , Humans , Observer Variation , Organs at Risk , Radiotherapy Planning, Computer-Assisted
12.
Bull Cancer ; 108(11S): 11S35-11S45, 2021 Dec.
Article in French | MEDLINE | ID: mdl-34969514

ABSTRACT

HER2 is an important prognostic and predictive biomarker in breast cancer. Its detection makes it possible to define which patients will benefit from a targeted treatment. While assessment of HER2 status by immunohistochemistry in positive vs negative categories is well implemented and reproducible, the introduction of a new "HER2-low" category could raise some concerns about its scoring and reproducibility. We herein described the current HER2 testing methods and the application of innovative machine learning techniques to improve these determinations, as well as the main challenges and opportunities related to the implementation of digital pathology in the up-and-coming AI era.


Subject(s)
Artificial Intelligence , Breast Neoplasms/chemistry , Breast Neoplasms/diagnosis , Receptor, ErbB-2/analysis , Algorithms , Breast Neoplasms/genetics , Diagnosis, Computer-Assisted , Female , Gene Amplification , Genes, erbB-2 , Humans , Machine Learning , Practice Guidelines as Topic , Reproducibility of Results
13.
Cancer Radiother ; 25(6-7): 607-616, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34389243

ABSTRACT

Deep-learning (DL)-based auto-contouring solutions have recently been proposed as a convincing alternative to decrease workload of target volumes and organs-at-risk (OAR) delineation in radiotherapy planning and improve inter-observer consistency. However, there is minimal literature of clinical implementations of such algorithms in a clinical routine. In this paper we first present an update of the state-of-the-art of DL-based solutions. We then summarize recent recommendations proposed by the European society for radiotherapy and oncology (ESTRO) to be followed before any clinical implementation of artificial intelligence-based solutions in clinic. The last section describes the methodology carried out by three French radiation oncology departments to deploy CE-marked commercial solutions. Based on the information collected, a majority of OAR are retained by the centers among those proposed by the manufacturers, validating the usefulness of DL-based models to decrease clinicians' workload. Target volumes, with the exception of lymph node areas in breast, head and neck and pelvic regions, whole breast, breast wall, prostate and seminal vesicles, are not available in the three commercial solutions at this time. No implemented workflows are currently available to continuously improve the models, but these can be adapted/retrained in some solutions during the commissioning phase to best fit local practices. In reported experiences, automatic workflows were implemented to limit human interactions and make the workflow more fluid. Recommendations published by the ESTRO group will be of importance for guiding physicists in the clinical implementation of patient specific and regular quality assurances.


Subject(s)
Deep Learning , Neoplasms/diagnostic imaging , Organs at Risk/diagnostic imaging , Radiation Oncology/methods , Radiotherapy Planning, Computer-Assisted/methods , Europe , Humans , Neoplasms/radiotherapy , Practice Guidelines as Topic , Radiotherapy, Image-Guided/methods , Societies, Medical , Workload
14.
Praxis (Bern 1994) ; 110(1): 48-53, 2021 Jan.
Article in German | MEDLINE | ID: mdl-33406927

ABSTRACT

Artificial Intelligence in Radiology - Definition, Potential and Challenges Abstract. Artificial Intelligence (AI) is omnipresent. It has neatly permeated our daily life, even if we are not always fully aware of its ubiquitous presence. The healthcare sector in particular is experiencing a revolution which will change our daily routine considerably in the near future. Due to its advanced digitization and its historical technical affinity radiology is especially prone to these developments. But what exactly is AI and what makes AI so potent that established medical disciplines such as radiology worry about their future job perspectives? What are the assets of AI in radiology today - and what are the major challenges? This review article tries to give some answers to these questions.


Subject(s)
Artificial Intelligence , Radiology , Forecasting , Humans , Machine Learning
15.
Genome ; 64(4): 416-425, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33091314

ABSTRACT

Precision medicine is an emerging approach to clinical research and patient care that focuses on understanding and treating disease by integrating multi-modal or multi-omics data from an individual to make patient-tailored decisions. With the large and complex datasets generated using precision medicine diagnostic approaches, novel techniques to process and understand these complex data were needed. At the same time, computer science has progressed rapidly to develop techniques that enable the storage, processing, and analysis of these complex datasets, a feat that traditional statistics and early computing technologies could not accomplish. Machine learning, a branch of artificial intelligence, is a computer science methodology that aims to identify complex patterns in data that can be used to make predictions or classifications on new unseen data or for advanced exploratory data analysis. Machine learning analysis of precision medicine's multi-modal data allows for broad analysis of large datasets and ultimately a greater understanding of human health and disease. This review focuses on machine learning utilization for precision medicine's "big data", in the context of genetics, genomics, and beyond.


Subject(s)
Genomics/methods , Machine Learning , Precision Medicine/methods , Artificial Intelligence , Humans
16.
Cancer Radiother ; 24(5): 403-410, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32265157

ABSTRACT

PURPOSE: Radiomics are a set of methods used to leverage medical imaging and extract quantitative features that can characterize a patient's phenotype. All modalities can be used with several different software packages. Specific informatics methods can then be used to create meaningful predictive models. In this review, we will explain the major steps of a radiomics analysis pipeline and then present the studies published in the context of radiation therapy. METHODS: A literature review was performed on Medline using the search engine PubMed. The search strategy included the search terms "radiotherapy", "radiation oncology" and "radiomics". The search was conducted in July 2019 and reference lists of selected articles were hand searched for relevance to this review. RESULTS: A typical radiomics workflow always includes five steps: imaging and segmenting, data curation and preparation, feature extraction, exploration and selection and finally modeling. In radiation oncology, radiomics studies have been published to explore different clinical outcome in lung (n=5), head and neck (n=5), esophageal (n=3), rectal (n=3), pancreatic (n=2) cancer and brain metastases (n=2). The quality of these retrospective studies is heterogeneous and their results have not been translated to the clinic. CONCLUSION: Radiomics has a great potential to predict clinical outcome and better personalize treatment. But the field is still young and constantly evolving. Improvement in bias reduction techniques and multicenter studies will hopefully allow more robust and generalizable models.


Subject(s)
Diagnostic Imaging/methods , Neoplasms/diagnostic imaging , Neoplasms/radiotherapy , Radiation Oncologists , Radiotherapy Planning, Computer-Assisted/methods , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/secondary , Data Analysis , Data Curation/methods , Deep Learning , Esophageal Neoplasms/diagnostic imaging , Head and Neck Neoplasms/diagnostic imaging , Humans , Lung Neoplasms/diagnostic imaging , Pancreatic Neoplasms/diagnostic imaging , Phenotype , Radiotherapy/methods , Rectal Neoplasms/diagnostic imaging , Reproducibility of Results , Retrospective Studies
17.
Ann Pathol ; 39(2): 130-136, 2019 Apr.
Article in French | MEDLINE | ID: mdl-30772062

ABSTRACT

Histopathology is the fundamental tool of pathology used for more than a century to establish the final diagnosis of lung cancer. In addition, the phenotypic data contained in the histological images reflects the overall effect of molecular alterations on the behavior of cancer cells and provides a practical visual reading of the aggressiveness of the disease. However, the human evaluation of the histological images is sometimes subjective and may lack reproducibility. Therefore, computational analysis of histological imaging using so-called "artificial intelligence" (AI) approaches has recently received considerable attention to improve this diagnostic accuracy. Thus, computational analysis of lung cancer images has recently been evaluated for the optimization of histological or cytological classification, prognostic prediction or genomic profile of patients with lung cancer. This rapidly growing field constantly demonstrates great power in the field of computing medical imaging by producing highly accurate detection, segmentation or recognition tasks. However, there are still several challenges or issues to be addressed in order to successfully succeed the actual transfer into clinical routine. The objective of this review is to emphasize recent applications of AI in pulmonary cancer pathology, but also to clarify the advantages and limitations of this approach, as well as the perspectives to be implemented for a potential transfer into clinical routine.


Subject(s)
Artificial Intelligence , Lung Neoplasms/pathology , Humans , Pathology, Clinical/methods
SELECTION OF CITATIONS
SEARCH DETAIL