Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Sci Rep ; 12(1): 12762, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35882891

RESUMO

The use of multicentric data is becoming essential for developing generalizable radiomic signatures. In particular, Magnetic Resonance Imaging (MRI) data used in brain oncology are often heterogeneous in terms of scanners and acquisitions, which significantly impact quantitative radiomic features. Various methods have been proposed to decrease dependency, including methods acting directly on MR images, i.e., based on the application of several preprocessing steps before feature extraction or the ComBat method, which harmonizes radiomic features themselves. The ComBat method used for radiomics may be misleading and presents some limitations, such as the need to know the labels associated with the "batch effect". In addition, a statistically representative sample is required and the applicability of a signature whose batch label is not present in the train set is not possible. This work aimed to compare a priori and a posteriori radiomic harmonization methods and propose a code adaptation to be machine learning compatible. Furthermore, we have developed AutoComBat, which aims to automatically determine the batch labels, using either MRI metadata or quality metrics as inputs of the proposed constrained clustering. A heterogeneous dataset consisting of high and low-grade gliomas coming from eight different centers was considered. The different methods were compared based on their ability to decrease relative standard deviation of radiomic features extracted from white matter and on their performance on a classification task using different machine learning models. ComBat and AutoComBat using image-derived quality metrics as inputs for batch assignment and preprocessing methods presented promising results on white matter harmonization, but with no clear consensus for all MR images. Preprocessing showed the best results on the T1w-gd images for the grading task. For T2w-flair, AutoComBat, using either metadata plus quality metrics or metadata alone as inputs, performs better than the conventional ComBat, highlighting its potential for data harmonization. Our results are MRI weighting, feature class and task dependent and require further investigations on other datasets.


Assuntos
Neoplasias Encefálicas , Glioma , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Neoplasias Encefálicas/diagnóstico por imagem , Glioma/diagnóstico por imagem , Glioma/patologia , Humanos , Aprendizado de Máquina , Imageamento por Ressonância Magnética/métodos
2.
IEEE/ACM Trans Comput Biol Bioinform ; 19(6): 3317-3331, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34714749

RESUMO

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.


Assuntos
Algoritmos , Neoplasias , Humanos , Análise por Conglomerados , Genômica , Reconhecimento Automatizado de Padrão/métodos , Neoplasias/genética , Perfilação da Expressão Gênica/métodos
3.
Eur Respir J ; 2021 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795038

RESUMO

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.

4.
Int J Radiat Oncol Biol Phys ; 110(4): 947-956, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33609591

RESUMO

PURPOSE: Patients with cancer are presumed to be more vulnerable to COVID-19. We evaluated a screening strategy combining chest computed tomography (CT) and reverse-transcription polymerase chain reaction (RT-PCR) for patients treated with radiation therapy at our cancer center located in a COVID-19 French hotspot during the first wave of the pandemic. METHODS AND MATERIALS: Chest CT images were proposed during radiation therapy CT simulation. Images were reviewed by an expert radiologist according to the COVID-19 Reporting and Data System classification. Nasal swabs with RT-PCR assay were initially proposed in cases of suspicious imaging or clinical context and were eventually integrated into the systematic screening. A dedicated radiation therapy workflow was proposed for COVID-19 patients to limit the risk of contamination. RESULTS: From March 18, 2020 to May 1, 2020, 480 patients were screened by chest CT, and 313 patients had both chest CT and RT-PCR (65%). The cumulative incidence of COVID-19 was 5.4% (95% confidence interval [CI], 3.6-7.8; 26 of 480 patients). Diagnosis of COVID-19 was made before radiation therapy for 22 patients (84.6%) and during RT for 4 patients (15.3%). Chest CT directly aided the diagnosis of 7 cases in which the initial RT-PCR was negative or not feasible, out of a total of 480 patients (1.5%) and 517 chest CT acquisitions. Four patients with COVID-19 at the time of the chest CT screening had a false negative CT. Sensitivity and specificity of chest CT screening in patients with both RT-PCR and chest CT testing were estimated at 0.82 (95% CI, 0.60-0.95) and 0.98 (95% CI, 0.96-0.99), respectively. Adaptation of the radiation therapy treatment was made for all patients, with 7 postponed treatments (median: 5 days; interquartile range, 1.5-14.8). CONCLUSIONS: The benefit of systematic use of chest CT screening during CT simulation for patients undergoing radiation therapy during the COVID-19 pandemic seemed limited.


Assuntos
Teste de Ácido Nucleico para COVID-19 , COVID-19/diagnóstico , Tomografia Computadorizada Multidetectores , Neoplasias/radioterapia , Adolescente , Adulto , Idoso , COVID-19/complicações , COVID-19/diagnóstico por imagem , COVID-19/epidemiologia , Institutos de Câncer , Criança , Intervalos de Confiança , Feminino , França/epidemiologia , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Neoplasias/complicações , Radiografia Torácica/métodos , Estudos Retrospectivos , Sensibilidade e Especificidade , Tomografia Computadorizada Espiral , Adulto Jovem
5.
J Immunother Cancer ; 8(2)2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33188037

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Radioterapia (Especialidade)/métodos , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Microambiente Tumoral
6.
Sci Rep ; 10(1): 12340, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32704007

RESUMO

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.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Encéfalo/diagnóstico por imagem , Glioma/diagnóstico por imagem , Imageamento por Ressonância Magnética/normas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
7.
Int J Radiat Oncol Biol Phys ; 108(3): 813-823, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32417412

RESUMO

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.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Aprendizado Profundo , Imageamento por Ressonância Magnética/métodos , Tomografia Computadorizada por Raios X/métodos , Encéfalo/diagnóstico por imagem , Neoplasias Encefálicas/radioterapia , Meios de Contraste , Humanos , Imageamento por Ressonância Magnética/normas , Redes Neurais de Computação , Radiometria , Radioterapia/normas , Estudos Retrospectivos , Crânio/diagnóstico por imagem
8.
Front Comput Neurosci ; 14: 17, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32265680

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA