Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Rheumatology (Oxford) ; 63(1): 103-110, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-37074923

ABSTRACT

OBJECTIVE: Stratifying the risk of death in SSc-related interstitial lung disease (SSc-ILD) is a challenging issue. The extent of lung fibrosis on high-resolution CT (HRCT) is often assessed by a visual semiquantitative method that lacks reliability. We aimed to assess the potential prognostic value of a deep-learning-based algorithm enabling automated quantification of ILD on HRCT in patients with SSc. METHODS: We correlated the extent of ILD with the occurrence of death during follow-up, and evaluated the additional value of ILD extent in predicting death based on a prognostic model including well-known risk factors in SSc. RESULTS: We included 318 patients with SSc, among whom 196 had ILD; the median follow-up was 94 months (interquartile range 73-111). The mortality rate was 1.6% at 2 years and 26.3% at 10 years. For each 1% increase in the baseline ILD extent (up to 30% of the lung), the risk of death at 10 years was increased by 4% (hazard ratio 1.04, 95% CI 1.01, 1.07, P = 0.004). We constructed a risk prediction model that showed good discrimination for 10-year mortality (c index 0.789). Adding the automated quantification of ILD significantly improved the model for 10-year survival prediction (P = 0.007). Its discrimination was only marginally improved, but it improved prediction of 2-year mortality (difference in time-dependent area under the curve 0.043, 95% CI 0.002, 0.084, P = 0.040). CONCLUSION: The deep-learning-based, computer-aided quantification of ILD extent on HRCT provides an effective tool for risk stratification in SSc. It might help identify patients at short-term risk of death.


Subject(s)
Lung Diseases, Interstitial , Scleroderma, Systemic , Humans , Prognosis , Reproducibility of Results , Vital Capacity , Lung Diseases, Interstitial/diagnostic imaging , Lung Diseases, Interstitial/etiology , Lung Diseases, Interstitial/epidemiology , Lung , Scleroderma, Systemic/complications , Scleroderma, Systemic/diagnostic imaging , Tomography, X-Ray Computed
2.
Nat Med ; 29(8): 2110-2120, 2023 08.
Article in English | MEDLINE | ID: mdl-37488289

ABSTRACT

The mechanisms of action of and resistance to trastuzumab deruxtecan (T-DXd), an anti-HER2-drug conjugate for breast cancer treatment, remain unclear. The phase 2 DAISY trial evaluated the efficacy of T-DXd in patients with HER2-overexpressing (n = 72, cohort 1), HER2-low (n = 74, cohort 2) and HER2 non-expressing (n = 40, cohort 3) metastatic breast cancer. In the full analysis set population (n = 177), the confirmed objective response rate (primary endpoint) was 70.6% (95% confidence interval (CI) 58.3-81) in cohort 1, 37.5% (95% CI 26.4-49.7) in cohort 2 and 29.7% (95% CI 15.9-47) in cohort 3. The primary endpoint was met in cohorts 1 and 2. Secondary endpoints included safety. No new safety signals were observed. During treatment, HER2-expressing tumors (n = 4) presented strong T-DXd staining. Conversely, HER2 immunohistochemistry 0 samples (n = 3) presented no or very few T-DXd staining (Pearson correlation coefficient r = 0.75, P = 0.053). Among patients with HER2 immunohistochemistry 0 metastatic breast cancer, 5 of 14 (35.7%, 95% CI 12.8-64.9) with ERBB2 expression below the median presented a confirmed objective response as compared to 3 of 10 (30%, 95% CI 6.7-65.2) with ERBB2 expression above the median. Although HER2 expression is a determinant of T-DXd efficacy, our study suggests that additional mechanisms may also be involved. (ClinicalTrials.gov identifier NCT04132960 .).


Subject(s)
Breast Neoplasms , Immunoconjugates , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Trastuzumab/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Camptothecin/therapeutic use
3.
Cancers (Basel) ; 15(12)2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37370717

ABSTRACT

Valvular Heart Disease (VHD) is a known late complication of radiotherapy for childhood cancer (CC), and identifying high-risk survivors correctly remains a challenge. This paper focuses on the distribution of the radiation dose absorbed by heart tissues. We propose that a dosiomics signature could provide insight into the spatial characteristics of the heart dose associated with a VHD, beyond the already-established risk induced by high doses. We analyzed data from the 7670 survivors of the French Childhood Cancer Survivors' Study (FCCSS), 3902 of whom were treated with radiotherapy. In all, 63 (1.6%) survivors that had been treated with radiotherapy experienced a VHD, and 57 of them had heterogeneous heart doses. From the heart-dose distribution of each survivor, we extracted 93 first-order and spatial dosiomics features. We trained random forest algorithms adapted for imbalanced classification and evaluated their predictive performance compared to the performance of standard mean heart dose (MHD)-based models. Sensitivity analyses were also conducted for sub-populations of survivors with spatially heterogeneous heart doses. Our results suggest that MHD and dosiomics-based models performed equally well globally in our cohort and that, when considering the sub-population having received a spatially heterogeneous dose distribution, the predictive capability of the models is significantly improved by the use of the dosiomics features. If these findings are further validated, the dosiomics signature may be incorporated into machine learning algorithms for radiation-induced VHD risk assessment and, in turn, into the personalized refinement of follow-up guidelines.

4.
Jpn J Radiol ; 41(3): 235-244, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36350524

ABSTRACT

Artificial intelligence (AI) has been a very active research topic over the last years and thoracic imaging has particularly benefited from the development of AI and in particular deep learning. We have now entered a phase of adopting AI into clinical practice. The objective of this article was to review the current applications and perspectives of AI in thoracic oncology. For pulmonary nodule detection, computer-aided detection (CADe) tools have been commercially available since the early 2000s. The more recent rise of deep learning and the availability of large annotated lung nodule datasets have allowed the development of new CADe tools with fewer false-positive results per examination. Classical machine learning and deep-learning methods were also used for pulmonary nodule segmentation allowing nodule volumetry and pulmonary nodule characterization. For pulmonary nodule characterization, radiomics and deep-learning approaches were used. Data from the National Lung Cancer Screening Trial (NLST) allowed the development of several computer-aided diagnostic (CADx) tools for diagnosing lung cancer on chest computed tomography. Finally, AI has been used as a means to perform virtual biopsies and to predict response to treatment or survival. Thus, many detection, characterization and stratification tools have been proposed, some of which are commercially available.


Subject(s)
Deep Learning , Lung Neoplasms , Humans , Artificial Intelligence , Lung Neoplasms/pathology , Early Detection of Cancer , Tomography, X-Ray Computed
5.
IEEE Trans Med Imaging ; 42(3): 697-712, 2023 03.
Article in English | MEDLINE | ID: mdl-36264729

ABSTRACT

Image registration is a fundamental medical image analysis task, and a wide variety of approaches have been proposed. However, only a few studies have comprehensively compared medical image registration approaches on a wide range of clinically relevant tasks. This limits the development of registration methods, the adoption of research advances into practice, and a fair benchmark across competing approaches. The Learn2Reg challenge addresses these limitations by providing a multi-task medical image registration data set for comprehensive characterisation of deformable registration algorithms. A continuous evaluation will be possible at https://learn2reg.grand-challenge.org. Learn2Reg covers a wide range of anatomies (brain, abdomen, and thorax), modalities (ultrasound, CT, MR), availability of annotations, as well as intra- and inter-patient registration evaluation. We established an easily accessible framework for training and validation of 3D registration methods, which enabled the compilation of results of over 65 individual method submissions from more than 20 unique teams. We used a complementary set of metrics, including robustness, accuracy, plausibility, and runtime, enabling unique insight into the current state-of-the-art of medical image registration. This paper describes datasets, tasks, evaluation methods and results of the challenge, as well as results of further analysis of transferability to new datasets, the importance of label supervision, and resulting bias. While no single approach worked best across all tasks, many methodological aspects could be identified that push the performance of medical image registration to new state-of-the-art performance. Furthermore, we demystified the common belief that conventional registration methods have to be much slower than deep-learning-based methods.


Subject(s)
Abdominal Cavity , Deep Learning , Humans , Algorithms , Brain/diagnostic imaging , Abdomen/diagnostic imaging , Image Processing, Computer-Assisted/methods
6.
Cancers (Basel) ; 14(23)2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36497352

ABSTRACT

Simple and accessible prognostic factors are paramount for solid cancer patients experiencing life-threatening complications. The aim of this study is to appraise the impact of functional and nutritional status and skeletal muscle mass in this population. We conducted a retrospective (2007−2020) single-center study by enrolling adult patients with solid cancers requiring unplanned ICU admission. Performance status, body weight, and albumin level were collected at ICU admission and over six months. Skeletal muscle mass was assessed at ICU admission by measuring muscle areas normalized by height (SMI). Four-hundred and sixty-two patients were analyzed, mainly with gastro-intestinal (34.8%) and lung (29.9%) neoplasms. Moreover, 92.8% of men and 67.3% of women were deemed cachectic. In the multivariate analysis, performance status at ICU admission (CSH 1.74 [1.27−2.39], p < 0.001) and the six month increase in albumin level (CSH 0.38 [0.16−0.87], p = 0.02) were independent predictors of ICU mortality. In the subgroup of mechanically ventilated patients, the psoas SMI was independently associated with ICU mortality (CSH 0.82 [0.67−0.98], p = 0.04). Among the 368 ICU-survivors, the performance status at ICU admission (CSH 1.34 [1.14−1.59], p < 0.001) and the six-month weight loss (CSH 1.33 [1.17−2.99], p = 0.01) were associated with a one-year mortality rate. Most cancer patients displayed cachexia at ICU admission. Time courses of nutritional parameters may aid the prediction of short- and long-term outcomes.

8.
IEEE/ACM Trans Comput Biol Bioinform ; 19(6): 3317-3331, 2022.
Article in English | MEDLINE | ID: mdl-34714749

ABSTRACT

Precision medicine is a paradigm shift in healthcare relying heavily on genomics data. However, the complexity of biological interactions, the large number of genes as well as the lack of comparisons on the analysis of data, remain a tremendous bottleneck regarding clinical adoption. In this paper, we introduce a novel, automatic and unsupervised framework to discover low-dimensional gene biomarkers. Our method is based on the LP-Stability algorithm, a high dimensional center-based unsupervised clustering algorithm. It offers modularity as concerns metric functions and scalability, while being able to automatically determine the best number of clusters. Our evaluation includes both mathematical and biological criteria to define a quantitative metric. The recovered signature is applied to a variety of biological tasks, including screening of biological pathways and functions, and characterization relevance on tumor types and subtypes. Quantitative comparisons among different distance metrics, commonly used clustering methods and a referential gene signature used in the literature, confirm state of the art performance of our approach. In particular, our signature, based on 27 genes, reports at least 30 times better mathematical significance (average Dunn's Index) and 25% better biological significance (average Enrichment in Protein-Protein Interaction) than those produced by other referential clustering methods. Finally, our signature reports promising results on distinguishing immune inflammatory and immune desert tumors, while reporting a high balanced accuracy of 92% on tumor types classification and averaged balanced accuracy of 68% on tumor subtypes classification, which represents, respectively 7% and 9% higher performance compared to the referential signature.


Subject(s)
Algorithms , Neoplasms , Humans , Cluster Analysis , Genomics , Pattern Recognition, Automated/methods , Neoplasms/genetics , Gene Expression Profiling/methods
9.
Eur Respir J ; 2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34795038

ABSTRACT

OBJECTIVES: Lumacaftor-ivacaftor is a cystic fibrosis transmembrane conductance regulator (CFTR) modulator known to improve clinical status in people with cystic fibrosis (CF). This study aimed to assess lung structural changes after one year of lumacaftor-ivacaftor treatment, and to use unsupervised machine learning to identify morphological phenotypes of lung disease that are associated with response to lumacaftor-ivacaftor. METHODS: Adolescents and adults with CF from the French multicenter real-world prospective observational study evaluating the first year of treatment with lumacaftor-ivacaftor were included if they had pretherapeutic and follow-up chest computed tomography (CT)-scans available. CT scans were visually scored using a modified Bhalla score. A k-mean clustering method was performed based on 120 radiomics features extracted from unenhanced pretherapeutic chest CT scans. RESULTS: A total of 283 patients were included. The Bhalla score significantly decreased after 1 year of lumacaftor-ivacaftor (-1.40±1.53 points compared with pretherapeutic CT; p<0.001). This finding was related to a significant decrease in mucus plugging (-0.35±0.62 points; p<0.001), bronchial wall thickening (-0.24±0.52 points; p<0.001) and parenchymal consolidations (-0.23±0.51 points; p<0.001). Cluster analysis identified 3 morphological clusters. Patients from cluster C were more likely to experience an increase in percent predicted forced expiratory volume in 1 sec (ppFEV1) ≥5 under lumacaftor-ivacaftor than those in the other clusters (54% of responders versus 32% and 33%; p=0.01). CONCLUSION: One year treatment with lumacaftor-ivacaftor was associated with a significant visual improvement of bronchial disease on chest CT. Radiomics features on pretherapeutic CT scan may help in predicting lung function response under lumacaftor-ivacaftor.

10.
Diagn Interv Imaging ; 102(11): 691-695, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34686464

ABSTRACT

PURPOSE: The purpose of this study was to determine whether a single reconstruction kernel or both high and low frequency kernels should be used for training deep learning models for the segmentation of diffuse lung disease on chest computed tomography (CT). MATERIALS AND METHODS: Two annotated datasets of COVID-19 pneumonia (323,960 slices) and interstitial lung disease (ILD) (4,284 slices) were used. Annotated CT images were used to train a U-Net architecture to segment disease. All CT slices were reconstructed using both a lung kernel (LK) and a mediastinal kernel (MK). Three different trainings, resulting in three different models were compared for each disease: training on LK only, MK only or LK+MK images. Dice similarity scores (DSC) were compared using the Wilcoxon signed-rank test. RESULTS: Models only trained on LK images performed better on LK images than on MK images (median DSC = 0.62 [interquartile range (IQR): 0.54, 0.69] vs. 0.60 [IQR: 0.50, 0.70], P < 0.001 for COVID-19 and median DSC = 0.62 [IQR: 0.56, 0.69] vs. 0.50 [IQR 0.43, 0.57], P < 0.001 for ILD). Similarly, models only trained on MK images performed better on MK images (median DSC = 0.62 [IQR: 0.53, 0.68] vs. 0.54 [IQR: 0.47, 0.63], P < 0.001 for COVID-19 and 0.69 [IQR: 0.61, 0.73] vs. 0.63 [IQR: 0.53, 0.70], P < 0.001 for ILD). Models trained on both kernels performed better or similarly than those trained on only one kernel. For COVID-19, median DSC was 0.67 (IQR: =0.59, 0.73) when applied on LK images and 0.67 (IQR: 0.60, 0.74) when applied on MK images (P < 0.001 for both). For ILD, median DSC was 0.69 (IQR: 0.63, 0.73) when applied on LK images (P = 0.006) and 0.68 (IQR: 0.62, 0.72) when applied on MK images (P > 0.99). CONCLUSION: Reconstruction kernels impact the performance of deep learning-based models for lung disease segmentation. Training on both LK and MK images improves the performance.


Subject(s)
COVID-19 , Deep Learning , Humans , Image Processing, Computer-Assisted , Lung/diagnostic imaging , SARS-CoV-2 , Tomography, X-Ray Computed
11.
Radiology ; 298(1): 189-198, 2021 01.
Article in English | MEDLINE | ID: mdl-33078999

ABSTRACT

Background Longitudinal follow-up of interstitial lung diseases (ILDs) at CT mainly relies on the evaluation of the extent of ILD, without accounting for lung shrinkage. Purpose To develop a deep learning-based method to depict worsening of ILD based on lung shrinkage detection from elastic registration of chest CT scans in patients with systemic sclerosis (SSc). Materials and Methods Patients with SSc evaluated between January 2009 and October 2017 who had undergone at least two unenhanced supine CT scans of the chest and pulmonary function tests (PFTs) performed within 3 months were retrospectively included. Morphologic changes on CT scans were visually assessed by two observers and categorized as showing improvement, stability, or worsening of ILD. Elastic registration between baseline and follow-up CT images was performed to obtain deformation maps of the whole lung. Jacobian determinants calculated from the deformation maps were given as input to a deep learning-based classifier to depict morphologic and functional worsening. For this purpose, the set was randomly split into training, validation, and test sets. Correlations between mean Jacobian values and changes in PFT measurements were evaluated with the Spearman correlation. Results A total of 212 patients (median age, 53 years; interquartile range, 45-62 years; 177 women) were included as follows: 138 for the training set (65%), 34 for the validation set (16%), and 40 for the test set (21%). Jacobian maps demonstrated lung parenchyma shrinkage of the posterior lung bases in patients found to have worsened ILD at visual assessment. The classifier detected morphologic and functional worsening with an accuracy of 80% (32 of 40 patients; 95% confidence interval [CI]: 64%, 91%) and 83% (33 of 40 patients; 95% CI: 67%, 93%), respectively. Jacobian values correlated with changes in forced vital capacity (R = -0.38; 95% CI: -0.25, -0.49; P < .001) and diffusing capacity for carbon monoxide (R = -0.42; 95% CI: -0.27, -0.54; P < .001). Conclusion Elastic registration of CT scans combined with a deep learning classifier aided in the diagnosis of morphologic and functional worsening of interstitial lung disease in patients with systemic sclerosis. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Verschakelen in this issue.


Subject(s)
Deep Learning , Lung Diseases, Interstitial/diagnostic imaging , Radiographic Image Interpretation, Computer-Assisted/methods , Scleroderma, Systemic/diagnostic imaging , Tomography, X-Ray Computed/methods , Female , Humans , Longitudinal Studies , Lung/diagnostic imaging , Male , Middle Aged , Retrospective Studies
12.
Med Image Anal ; 67: 101860, 2021 01.
Article in English | MEDLINE | ID: mdl-33171345

ABSTRACT

Coronavirus disease 2019 (COVID-19) emerged in 2019 and disseminated around the world rapidly. Computed tomography (CT) imaging has been proven to be an important tool for screening, disease quantification and staging. The latter is of extreme importance for organizational anticipation (availability of intensive care unit beds, patient management planning) as well as to accelerate drug development through rapid, reproducible and quantified assessment of treatment response. Even if currently there are no specific guidelines for the staging of the patients, CT together with some clinical and biological biomarkers are used. In this study, we collected a multi-center cohort and we investigated the use of medical imaging and artificial intelligence for disease quantification, staging and outcome prediction. Our approach relies on automatic deep learning-based disease quantification using an ensemble of architectures, and a data-driven consensus for the staging and outcome prediction of the patients fusing imaging biomarkers with clinical and biological attributes. Highly promising results on multiple external/independent evaluation cohorts as well as comparisons with expert human readers demonstrate the potentials of our approach.


Subject(s)
Artificial Intelligence , COVID-19/diagnostic imaging , Pneumonia, Viral/diagnostic imaging , Biomarkers/analysis , Disease Progression , Humans , Neural Networks, Computer , Prognosis , Radiographic Image Interpretation, Computer-Assisted , SARS-CoV-2 , Triage
13.
IEEE J Biomed Health Inform ; 25(6): 2125-2136, 2021 06.
Article in English | MEDLINE | ID: mdl-33206611

ABSTRACT

We investigate the use of recent advances in deep learning and propose an end-to-end trainable multi-instance convolutional neural network within a mixture-of-experts formulation that combines information from two types of data-images and clinical attributes-for the diagnosis of lymphocytosis. The convolutional network learns to extract meaningful features from images of blood cells using an embedding level approach and aggregates them. Moreover, the mixture-of-experts model combines information from these images as well as clinical attributes to form an end-to-end trainable pipeline for diagnosis of lymphocytosis. Our results demonstrate that even the convolutional network by itself is able to discover meaningful associations between the images and the diagnosis, indicating the presence of important unexploited information in the images. The mixture-of-experts formulation is shown to be more robust while maintaining performance via. a repeatability study to assess the effect of variability in data acquisition on the predictions. The proposed methods are compared with different methods from literature based both on conventional handcrafted features and machine learning, and on recent deep learning models based on attention mechanisms. Our method reports a balanced accuracy of [Formula: see text] and outperfroms the handcrafted feature-based and attention-based approaches as well that of biologists which scored [Formula: see text], [Formula: see text] and [Formula: see text] respectively. These results give insights on the potentials of the applicability of the proposed method in clinical practice. Our code and datasets can be found at https://github.com/msahasrabudhe/lymphoMIL.


Subject(s)
Lymphocytosis , Humans , Lymphocytosis/diagnosis , Machine Learning , Neural Networks, Computer
14.
J Immunother Cancer ; 8(2)2020 11.
Article in English | MEDLINE | ID: mdl-33188037

ABSTRACT

BACKGROUND: Combining radiotherapy (RT) with immuno-oncology (IO) therapy (IORT) may enhance IO-induced antitumor response. Quantitative imaging biomarkers can be used to provide prognosis, predict tumor response in a non-invasive fashion and improve patient selection for IORT. A biologically inspired CD8 T-cells-associated radiomics signature has been developed on previous cohorts. We evaluated here whether this CD8 radiomic signature is associated with lesion response, whether it may help to assess disease spatial heterogeneity for predicting outcomes of patients treated with IORT. We also evaluated differences between irradiated and non-irradiated lesions. METHODS: Clinical data from patients with advanced solid tumors in six independent clinical studies of IORT were investigated. Immunotherapy consisted of 4 different drugs (antiprogrammed death-ligand 1 or anticytotoxic T-lymphocyte-associated protein 4 in monotherapy). Most patients received stereotactic RT to one lesion. Irradiated and non-irradiated lesions were delineated from baseline and the first evaluation CT scans. Radiomic features were extracted from contrast-enhanced CT images and the CD8 radiomics signature was applied. A responding lesion was defined by a decrease in lesion size of at least 30%. Dispersion metrices of the radiomics signature were estimated to evaluate the impact of tumor heterogeneity in patient's response. RESULTS: A total of 94 patients involving multiple lesions (100 irradiated and 189 non-irradiated lesions) were considered for a statistical interpretation. Lesions with high CD8 radiomics score at baseline were associated with significantly higher tumor response (area under the receiving operating characteristic curve (AUC)=0.63, p=0.0020). Entropy of the radiomics scores distribution on all lesions was shown to be associated with progression-free survival (HR=1.67, p=0.040), out-of-field abscopal response (AUC=0.70, p=0.014) and overall survival (HR=2.08, p=0.023), which remained significant in a multivariate analysis including clinical and biological variables. CONCLUSIONS: These results enhance the predictive value of the biologically inspired CD8 radiomics score and suggests that tumor heterogeneity should be systematically considered in patients treated with IORT. This CD8 radiomics signature may help select patients who are most likely to benefit from IORT.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/radiotherapy , Radiation Oncology/methods , Aged , Female , Humans , Male , Middle Aged , Prognosis , Tumor Microenvironment
15.
Sci Rep ; 10(1): 12340, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704007

ABSTRACT

Radiomics relies on the extraction of a wide variety of quantitative image-based features to provide decision support. Magnetic resonance imaging (MRI) contributes to the personalization of patient care but suffers from being highly dependent on acquisition and reconstruction parameters. Today, there are no guidelines regarding the optimal pre-processing of MR images in the context of radiomics, which is crucial for the generalization of published image-based signatures. This study aims to assess the impact of three different intensity normalization methods (Nyul, WhiteStripe, Z-Score) typically used in MRI together with two methods for intensity discretization (fixed bin size and fixed bin number). The impact of these methods was evaluated on first- and second-order radiomics features extracted from brain MRI, establishing a unified methodology for future radiomics studies. Two independent MRI datasets were used. The first one (DATASET1) included 20 institutional patients with WHO grade II and III gliomas who underwent post-contrast 3D axial T1-weighted (T1w-gd) and axial T2-weighted fluid attenuation inversion recovery (T2w-flair) sequences on two different MR devices (1.5 T and 3.0 T) with a 1-month delay. Jensen-Shannon divergence was used to compare pairs of intensity histograms before and after normalization. The stability of first-order and second-order features across the two acquisitions was analysed using the concordance correlation coefficient and the intra-class correlation coefficient. The second dataset (DATASET2) was extracted from the public TCIA database and included 108 patients with WHO grade II and III gliomas and 135 patients with WHO grade IV glioblastomas. The impact of normalization and discretization methods was evaluated based on a tumour grade classification task (balanced accuracy measurement) using five well-established machine learning algorithms. Intensity normalization highly improved the robustness of first-order features and the performances of subsequent classification models. For the T1w-gd sequence, the mean balanced accuracy for tumour grade classification was increased from 0.67 (95% CI 0.61-0.73) to 0.82 (95% CI 0.79-0.84, P = .006), 0.79 (95% CI 0.76-0.82, P = .021) and 0.82 (95% CI 0.80-0.85, P = .005), respectively, using the Nyul, WhiteStripe and Z-Score normalization methods compared to no normalization. The relative discretization makes unnecessary the use of intensity normalization for the second-order radiomics features. Even if the bin number for the discretization had a small impact on classification performances, a good compromise was obtained using the 32 bins considering both T1w-gd and T2w-flair sequences. No significant improvements in classification performances were observed using feature selection. A standardized pre-processing pipeline is proposed for the use of radiomics in MRI of brain tumours. For models based on first- and second-order features, we recommend normalizing images with the Z-Score method and adopting an absolute discretization approach. For second-order feature-based signatures, relative discretization can be used without prior normalization. In both cases, 32 bins for discretization are recommended. This study may pave the way for the multicentric development and validation of MR-based radiomics biomarkers.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain/diagnostic imaging , Glioma/diagnostic imaging , Magnetic Resonance Imaging/standards , Female , Humans , Male , Middle Aged
16.
Int J Radiat Oncol Biol Phys ; 108(3): 813-823, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32417412

ABSTRACT

PURPOSE: This study aims to evaluate the impact of key parameters on the pseudo computed tomography (pCT) quality generated from magnetic resonance imaging (MRI) with a 3-dimensional (3D) convolutional neural network. METHODS AND MATERIALS: Four hundred two brain tumor cases were retrieved, yielding associations between 182 computed tomography (CT) and T1-weighted MRI (T1) scans, 180 CT and contrast-enhanced T1-weighted MRI (T1-Gd) scans, and 40 CT, T1, and T1-Gd scans. A 3D CNN was used to map T1 or T1-Gd onto CT scans and evaluate the importance of different components. First, the training set size's influence on testing set accuracy was assessed. Moreover, we evaluated the MRI sequence impact, using T1-only and T1-Gd-only cohorts. We then investigated 4 MRI standardization approaches (histogram-based, zero-mean/unit-variance, white stripe, and no standardization) based on training, validation, and testing cohorts composed of 242, 81, and 79 patients cases, respectively, as well as a bias field correction influence. Finally, 2 networks, namely HighResNet and 3D UNet, were compared to evaluate the architecture's impact on the pCT quality. The mean absolute error, gamma indices, and dose-volume histograms were used as evaluation metrics. RESULTS: Generating models using all the available cases for training led to higher pCT quality. The T1 and T1-Gd models had a maximum difference in gamma index means of 0.07 percentage point. The mean absolute error obtained with white stripe was 78 ± 22 Hounsfield units, which slightly outperformed histogram-based, zero-mean/unit-variance, and no standardization (P < .0001). Regarding the network architectures, 3%/3 mm gamma indices of 99.83% ± 0.19% and 99.74% ± 0.24% were obtained for HighResNet and 3D UNet, respectively. CONCLUSIONS: Our best pCTs were generated using more than 200 samples in the training data set. Training with T1 only and T1-Gd only did not significantly affect performance. Regardless of the preprocessing applied, the dosimetry quality remained equivalent and relevant for potential use in clinical practice.


Subject(s)
Brain Neoplasms/diagnostic imaging , Deep Learning , Magnetic Resonance Imaging/methods , Tomography, X-Ray Computed/methods , Brain/diagnostic imaging , Brain Neoplasms/radiotherapy , Contrast Media , Humans , Magnetic Resonance Imaging/standards , Neural Networks, Computer , Radiometry , Radiotherapy/standards , Retrospective Studies , Skull/diagnostic imaging
17.
Front Comput Neurosci ; 14: 17, 2020.
Article in English | MEDLINE | ID: mdl-32265680

ABSTRACT

Image registration and segmentation are the two most studied problems in medical image analysis. Deep learning algorithms have recently gained a lot of attention due to their success and state-of-the-art results in variety of problems and communities. In this paper, we propose a novel, efficient, and multi-task algorithm that addresses the problems of image registration and brain tumor segmentation jointly. Our method exploits the dependencies between these tasks through a natural coupling of their interdependencies during inference. In particular, the similarity constraints are relaxed within the tumor regions using an efficient and relatively simple formulation. We evaluated the performance of our formulation both quantitatively and qualitatively for registration and segmentation problems on two publicly available datasets (BraTS 2018 and OASIS 3), reporting competitive results with other recent state-of-the-art methods. Moreover, our proposed framework reports significant amelioration (p < 0.005) for the registration performance inside the tumor locations, providing a generic method that does not need any predefined conditions (e.g., absence of abnormalities) about the volumes to be registered. Our implementation is publicly available online at https://github.com/TheoEst/joint_registration_tumor_segmentation.

18.
Radiol Artif Intell ; 2(4): e190006, 2020 Jul.
Article in English | MEDLINE | ID: mdl-33937829

ABSTRACT

PURPOSE: To develop a deep learning algorithm for the automatic assessment of the extent of systemic sclerosis (SSc)-related interstitial lung disease (ILD) on chest CT images. MATERIALS AND METHODS: This retrospective study included 208 patients with SSc (median age, 57 years; 167 women) evaluated between January 2009 and October 2017. A multicomponent deep neural network (AtlasNet) was trained on 6888 fully annotated CT images (80% for training and 20% for validation) from 17 patients with no, mild, or severe lung disease. The model was tested on a dataset of 400 images from another 20 patients, independently partially annotated by three radiologist readers. The ILD contours from the three readers and the deep learning neural network were compared by using the Dice similarity coefficient (DSC). The correlation between disease extent obtained from the deep learning algorithm and that obtained by using pulmonary function tests (PFTs) was then evaluated in the remaining 171 patients and in an external validation dataset of 31 patients based on the analysis of all slices of the chest CT scan. The Spearman rank correlation coefficient (ρ) was calculated to evaluate the correlation between disease extent and PFT results. RESULTS: The median DSCs between the readers and the deep learning ILD contours ranged from 0.74 to 0.75, whereas the median DSCs between contours from radiologists ranged from 0.68 to 0.71. The disease extent obtained from the algorithm, by analyzing the whole CT scan, correlated with the diffusion lung capacity for carbon monoxide, total lung capacity, and forced vital capacity (ρ = -0.76, -0.70, and -0.62, respectively; P < .001 for all) in the dataset for the correlation with PFT results. The disease extents correlated with diffusion lung capacity for carbon monoxide, total lung capacity, and forced vital capacity were ρ = -0.65, -0.70, and -0.57, respectively, in the external validation dataset (P < .001 for all). CONCLUSION: The developed algorithm performed similarly to radiologists for disease-extent contouring, which correlated with pulmonary function to assess CT images from patients with SSc-related ILD.Supplemental material is available for this article.© RSNA, 2020.

19.
Eur J Radiol ; 123: 108774, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31841881

ABSTRACT

Artificial intelligence is a hot topic in medical imaging. The development of deep learning methods and in particular the use of convolutional neural networks (CNNs), have led to substantial performance gain over the classic machine learning techniques. Multiple usages are currently being evaluated, especially for thoracic imaging, such as such as lung nodule evaluation, tuberculosis or pneumonia detection or quantification of diffuse lung diseases. Chest radiography is a near perfect domain for the development of deep learning algorithms for automatic interpretation, requiring large annotated datasets, in view of the high number of procedures and increasing data availability. Current algorithms are able to detect up to 14 common anomalies, when present as isolated findings. Chest computed tomography is another major field of application for artificial intelligence, especially in the perspective of large scale lung cancer screening. It is important for radiologists to apprehend, contribute actively and lead this new era of radiology powered by artificial intelligence. Such a perspective requires understanding new terms and concepts associated with machine learning. The objective of this paper is to provide useful definitions for understanding the methods used and their possibilities, and report current and future developments for thoracic imaging. Prospective validation of AI tools will be required before reaching routine clinical implementation.


Subject(s)
Artificial Intelligence , Radiography, Thoracic/methods , Thorax/diagnostic imaging , Algorithms , Deep Learning , Early Detection of Cancer/methods , Humans , Lung Neoplasms/diagnostic imaging , Machine Learning , Radiologists
20.
Eur Radiol ; 29(3): 1616-1624, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30105410

ABSTRACT

The recent explosion of 'big data' has ushered in a new era of artificial intelligence (AI) algorithms in every sphere of technological activity, including medicine, and in particular radiology. However, the recent success of AI in certain flagship applications has, to some extent, masked decades-long advances in computational technology development for medical image analysis. In this article, we provide an overview of the history of AI methods for radiological image analysis in order to provide a context for the latest developments. We review the functioning, strengths and limitations of more classical methods as well as of the more recent deep learning techniques. We discuss the unique characteristics of medical data and medical science that set medicine apart from other technological domains in order to highlight not only the potential of AI in radiology but also the very real and often overlooked constraints that may limit the applicability of certain AI methods. Finally, we provide a comprehensive perspective on the potential impact of AI on radiology and on how to evaluate it not only from a technical point of view but also from a clinical one, so that patients can ultimately benefit from it. KEY POINTS: • Artificial intelligence (AI) research in medical imaging has a long history • The functioning, strengths and limitations of more classical AI methods is reviewed, together with that of more recent deep learning methods. • A perspective is provided on the potential impact of AI on radiology and on its evaluation from both technical and clinical points of view.


Subject(s)
Artificial Intelligence/trends , Radiographic Image Interpretation, Computer-Assisted/methods , Technology, Radiologic/trends , Algorithms , Deep Learning , Forecasting , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...