Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Sensors (Basel) ; 23(3)2023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-36772129

RESUMO

Research exploring CycleGAN-based synthetic image generation has recently accelerated in the medical community due to its ability to leverage unpaired images effectively. However, a commonly established drawback of the CycleGAN, the introduction of artifacts in generated images, makes it unreliable for medical imaging use cases. In an attempt to address this, we explore the effect of structure losses on the CycleGAN and propose a generalized frequency-based loss that aims at preserving the content in the frequency domain. We apply this loss to the use-case of cone-beam computed tomography (CBCT) translation to computed tomography (CT)-like quality. Synthetic CT (sCT) images generated from our methods are compared against baseline CycleGAN along with other existing structure losses proposed in the literature. Our methods (MAE: 85.5, MSE: 20433, NMSE: 0.026, PSNR: 30.02, SSIM: 0.935) quantitatively and qualitatively improve over the baseline CycleGAN (MAE: 88.8, MSE: 24244, NMSE: 0.03, PSNR: 29.37, SSIM: 0.935) across all investigated metrics and are more robust than existing methods. Furthermore, no observable artifacts or loss in image quality were observed. Finally, we demonstrated that sCTs generated using our methods have superior performance compared to the original CBCT images on selected downstream tasks.


Assuntos
Tomografia Computadorizada de Feixe Cônico , Processamento de Imagem Assistida por Computador , Processamento de Imagem Assistida por Computador/métodos , Tomografia Computadorizada por Raios X , Artefatos , Benchmarking
2.
Cancers (Basel) ; 14(5)2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35267597

RESUMO

Problem. Image biomarker analysis, also known as radiomics, is a tool for tissue characterization and treatment prognosis that relies on routinely acquired clinical images and delineations. Due to the uncertainty in image acquisition, processing, and segmentation (delineation) protocols, radiomics often lack reproducibility. Radiomics harmonization techniques have been proposed as a solution to reduce these sources of uncertainty and/or their influence on the prognostic model performance. A relevant question is how to estimate the protocol-induced uncertainty of a specific image biomarker, what the effect is on the model performance, and how to optimize the model given the uncertainty. Methods. Two non-small cell lung cancer (NSCLC) cohorts, composed of 421 and 240 patients, respectively, were used for training and testing. Per patient, a Monte Carlo algorithm was used to generate three hundred synthetic contours with a surface dice tolerance measure of less than 1.18 mm with respect to the original GTV. These contours were subsequently used to derive 104 radiomic features, which were ranked on their relative sensitivity to contour perturbation, expressed in the parameter η. The top four (low η) and the bottom four (high η) features were selected for two models based on the Cox proportional hazards model. To investigate the influence of segmentation uncertainty on the prognostic model, we trained and tested the setup in 5000 augmented realizations (using a Monte Carlo sampling method); the log-rank test was used to assess the stratification performance and stability of segmentation uncertainty. Results. Although both low and high η setup showed significant testing set log-rank p-values (p = 0.01) in the original GTV delineations (without segmentation uncertainty introduced), in the model with high uncertainty, to effect ratio, only around 30% of the augmented realizations resulted in model performance with p < 0.05 in the test set. In contrast, the low η setup performed with a log-rank p < 0.05 in 90% of the augmented realizations. Moreover, the high η setup classification was uncertain in its predictions for 50% of the subjects in the testing set (for 80% agreement rate), whereas the low η setup was uncertain only in 10% of the cases. Discussion. Estimating image biomarker model performance based only on the original GTV segmentation, without considering segmentation, uncertainty may be deceiving. The model might result in a significant stratification performance, but can be unstable for delineation variations, which are inherent to manual segmentation. Simulating segmentation uncertainty using the method described allows for more stable image biomarker estimation, selection, and model development. The segmentation uncertainty estimation method described here is universal and can be extended to estimate other protocol uncertainties (such as image acquisition and pre-processing).

3.
Phys Med Biol ; 66(16)2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34289463

RESUMO

Radiomics is an active area of research in medical image analysis, however poor reproducibility of radiomics has hampered its application in clinical practice. This issue is especially prominent when radiomic features are calculated from noisy images, such as low dose computed tomography (CT) scans. In this article, we investigate the possibility of improving the reproducibility of radiomic features calculated on noisy CTs by using generative models for denoising. Our work concerns two types of generative models-encoder-decoder network (EDN) and conditional generative adversarial network (CGAN). We then compared their performance against a more traditional 'non-local means' denoising algorithm. We added noise to sinograms of full dose CTs to mimic low dose CTs with two levels of noise: low-noise CT and high-noise CT. Models were trained on high-noise CTs and used to denoise low-noise CTs without re-training. We tested the performance of our model in real data, using a dataset of same-day repeated low dose CTs in order to assess the reproducibility of radiomic features in denoised images. EDN and the CGAN achieved similar improvements on the concordance correlation coefficients (CCC) of radiomic features for low-noise images from 0.87 [95%CI, (0.833, 0.901)] to 0.92 [95%CI, (0.909, 0.935)] and for high-noise images from 0.68 [95%CI, (0.617, 0.745)] to 0.92 [95%CI, (0.909, 0.936)], respectively. The EDN and the CGAN improved the test-retest reliability of radiomic features (mean CCC increased from 0.89 [95%CI, (0.881, 0.914)] to 0.94 [95%CI, (0.927, 0.951)]) based on real low dose CTs. These results show that denoising using EDN and CGANs could be used to improve the reproducibility of radiomic features calculated from noisy CTs. Moreover, images at different noise levels can be denoised to improve the reproducibility using the above models without need for re-training, provided the noise intensity is not excessively greater that of the high-noise CTs. To the authors' knowledge, this is the first effort to improve the reproducibility of radiomic features calculated on low dose CT scans by applying generative models.


Assuntos
Algoritmos , Tomografia Computadorizada por Raios X , Simulação por Computador , Processamento de Imagem Assistida por Computador , Reprodutibilidade dos Testes
4.
Med Phys ; 48(3): 1226-1238, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33368399

RESUMO

BACKGROUND: Radiomics refers to the extraction of a large number of image biomarker describing the tumor phenotype displayed in a medical image. Extracted from positron emission tomography (PET) images, radiomics showed diagnostic and prognostic value for several cancer types. However, a large number of radiomic features are nonreproducible or highly correlated with conventional PET metrics. Moreover, radiomic features used in the clinic should yield relevant information about tumor texture. In this study, we propose a framework to identify technical and clinical meaningful features and exemplify our results using a PET non-small cell lung cancer (NSCLC) dataset. MATERIALS AND METHODS: The proposed selection procedure consists of several steps. A priori, we only include features that were found to be reproducible in a multicenter setting. Next, we apply a voxel randomization step to identify features that reflect actual textural information, that is, that yield in 90% of the patient scans a value significantly different from random texture. Finally, the remaining features were correlated with standard PET metrics to further remove redundancy with common PET metrics. The selection procedure was performed for different volume ranges, that is, excluding lesions with smaller volumes in order to assess the effect of tumor size on the results. To exemplify our procedure, the selected features were used to predict 1-yr survival in a dataset of 150 NSCLC patients. A predictive model was built using volume as predictive factor for smaller, and one of the selected features as predictive factor for bigger lesions. The prediction accuracy of the both models were compared with the prediction accuracy of volume. RESULTS: The number of selected features depended on the lesion size included in the analysis. When including the whole dataset, from 19 features reflecting actual texture only two were found to be not strongly correlated with conventional PET metrics. When excluding lesions smaller than 11.49 and 33.10 mL (25 and 50 percentile of the dataset), four out of 27 features and 13 out of 29 features remained after eliminating features highly correlated with standard PET metrics. When excluding lesions smaller than 103.9 mL (75 percentile), 33 out of 53 features remained. For larger lesions, some of these features outperformed volume in terms of classification accuracy (increase of 4-10%). The combination of using volume as predictor for smaller and one of the selected features for larger lesions also improved the accuracy when compared with volume only (increase from 72% to 76%). CONCLUSION: When performing radiomic analysis for smaller lesions, it should be first carefully investigated if a textural feature reflects actual heterogeneity information. Next, verification of the absence of correlation with all conventional PET metrics is essential in order to assess the additional value of radiomic features. Radiomic analysis with lesions larger than 11.4 mL might give additional information to conventional metrics while at the same time reflecting actual tumor texture. Using a combination of volume and one of the selected features for prediction yields promise to increase accuracy and reliability of a radiomic model.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Fluordesoxiglucose F18 , Humanos , Processamento de Imagem Assistida por Computador , Neoplasias Pulmonares/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Reprodutibilidade dos Testes , Tomografia Computadorizada por Raios X
5.
Phys Imaging Radiat Oncol ; 20: 30-33, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34667885

RESUMO

Radiomics is referred to as quantitative imaging of biomarkers used for clinical outcome prognosis or tumor characterization. In order to bridge radiomics and its clinical application, we aimed to build an integrated solution of radiomics extraction with an open-source Picture Archiving and Communication System (PACS). The integrated SQLite4Radiomics software was tested in three different imaging modalities and its performance was benchmarked in lung cancer open datasets RIDER and MMD with median extraction time of 10.7 (percentiles 25-75: 8.9-18.7) seconds per ROI in three different configurations.

6.
Phys Imaging Radiat Oncol ; 20: 69-75, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34816024

RESUMO

PURPOSE: Although quantitative image biomarkers (radiomics) show promising value for cancer diagnosis, prognosis, and treatment assessment, these biomarkers still lack reproducibility. In this systematic review, we aimed to assess the progress in radiomics reproducibility and repeatability in the recent years. METHODS AND MATERIALS: Four hundred fifty-one abstracts were retrieved according to the original PubMed search pattern with the publication dates ranging from 2017/05/01 to 2020/12/01. Each abstract including the keywords was independently screened by four observers. Forty-two full-text articles were selected for further analysis. Patient population data, radiomic feature classes, feature extraction software, image preprocessing, and reproducibility results were extracted from each article. To support the community with a standardized reporting strategy, we propose a specific reporting checklist to evaluate the feasibility to reproduce each study. RESULTS: Many studies continue to under-report essential reproducibility information: all but one clinical and all but two phantom studies missed to report at least one important item reporting image acquisition. The studies included in this review indicate that all radiomic features are sensitive to image acquisition, reconstruction, tumor segmentation, and interpolation. However, the amount of sensitivity is feature dependent, for instance, textural features were, in general, less robust than statistical features. CONCLUSIONS: Radiomics repeatability, reproducibility, and reporting quality can substantially be improved regarding feature extraction software and settings, image preprocessing and acquisition, cutoff values for stable feature selection. Our proposed radiomics reporting checklist can serve to simplify and improve the reporting and, eventually, guarantee the possibility to fully replicate and validate radiomic studies.

7.
Phys Med ; 71: 24-30, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32088562

RESUMO

PURPOSE: Highlighting the risk of biases in radiomics-based models will help improve their quality and increase usage as decision support systems in the clinic. In this study we use machine learning-based methods to identify the presence of volume-confounding effects in radiomics features. Methods 841 radiomics features were extracted from two retrospective publicly available datasets of lung and head neck cancers using open source software. Unsupervised hierarchical clustering and principal component analysis (PCA) identified relations between radiomics and clinical outcomes (overall survival). Bootstrapping techniques with logistic regression verified features' prognostic power and robustness. Results Over 80% of the features had large pairwise correlations. Nearly 30% of the features presented strong correlations with tumor volume. Using volume-independent features for clustering and PCA did not allow risk stratification of patients. Clinical predictors outperformed radiomics features in bootstrapping and logistic regression. Conclusions The adoption of safeguards in radiomics is imperative to improve the quality of radiomics studies. We proposed machine learning (ML) - based methods for robust radiomics signatures development.


Assuntos
Neoplasias Pulmonares/diagnóstico por imagem , Aprendizado de Máquina , Radiometria/métodos , Carcinoma de Células Escamosas de Cabeça e Pescoço/diagnóstico por imagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/radioterapia , Análise por Conglomerados , Bases de Dados Factuais , Sistemas de Apoio a Decisões Clínicas , Feminino , Humanos , Neoplasias Laríngeas/diagnóstico por imagem , Neoplasias Laríngeas/radioterapia , Neoplasias Pulmonares/radioterapia , Masculino , Pessoa de Meia-Idade , Neoplasias Orofaríngeas/diagnóstico por imagem , Neoplasias Orofaríngeas/radioterapia , Análise de Componente Principal , Análise de Regressão , Estudos Retrospectivos , Software , Carcinoma de Células Escamosas de Cabeça e Pescoço/radioterapia , Tomografia Computadorizada por Raios X
8.
Med Phys ; 47(11): 5931-5940, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32521049

RESUMO

PURPOSE: One of the most frequently cited radiomics investigations showed that features automatically extracted from routine clinical images could be used in prognostic modeling. These images have been made publicly accessible via The Cancer Imaging Archive (TCIA). There have been numerous requests for additional explanatory metadata on the following datasets - RIDER, Interobserver, Lung1, and Head-Neck1. To support repeatability, reproducibility, generalizability, and transparency in radiomics research, we publish the subjects' clinical data, extracted radiomics features, and digital imaging and communications in medicine (DICOM) headers of these four datasets with descriptive metadata, in order to be more compliant with findable, accessible, interoperable, and reusable (FAIR) data management principles. ACQUISITION AND VALIDATION METHODS: Overall survival time intervals were updated using a national citizens registry after internal ethics board approval. Spatial offsets of the primary gross tumor volume (GTV) regions of interest (ROIs) associated with the Lung1 CT series were improved on the TCIA. GTV radiomics features were extracted using the open-source Ontology-Guided Radiomics Analysis Workflow (O-RAW). We reshaped the output of O-RAW to map features and extraction settings to the latest version of Radiomics Ontology, so as to be consistent with the Image Biomarker Standardization Initiative (IBSI). Digital imaging and communications in medicine metadata was extracted using a research version of Semantic DICOM (SOHARD, GmbH, Fuerth; Germany). Subjects' clinical data were described with metadata using the Radiation Oncology Ontology. All of the above were published in Resource Descriptor Format (RDF), that is, triples. Example SPARQL queries are shared with the reader to use on the online triples archive, which are intended to illustrate how to exploit this data submission. DATA FORMAT: The accumulated RDF data are publicly accessible through a SPARQL endpoint where the triples are archived. The endpoint is remotely queried through a graph database web application at http://sparql.cancerdata.org. SPARQL queries are intrinsically federated, such that we can efficiently cross-reference clinical, DICOM, and radiomics data within a single query, while being agnostic to the original data format and coding system. The federated queries work in the same way even if the RDF data were partitioned across multiple servers and dispersed physical locations. POTENTIAL APPLICATIONS: The public availability of these data resources is intended to support radiomics features replication, repeatability, and reproducibility studies by the academic community. The example SPARQL queries may be freely used and modified by readers depending on their research question. Data interoperability and reusability are supported by referencing existing public ontologies. The RDF data are readily findable and accessible through the aforementioned link. Scripts used to create the RDF are made available at a code repository linked to this submission: https://gitlab.com/UM-CDS/FAIR-compliant_clinical_radiomics_and_DICOM_metadata.


Assuntos
Metadados , Bases de Dados Factuais , Alemanha , Humanos , Reprodutibilidade dos Testes , Fluxo de Trabalho
9.
Clin Transl Radiat Oncol ; 19: 33-38, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31417963

RESUMO

PURPOSE: Radiomics are quantitative features extracted from medical images. Many radiomic features depend not only on tumor properties, but also on non-tumor related factors such as scanner signal-to-noise ratio (SNR), reconstruction kernel and other image acquisition settings. This causes undesirable value variations in the features and reduces the performance of prediction models. In this paper, we investigate whether we can use phantom measurements to characterize and correct for the scanner SNR dependence. METHODS: We used a phantom with 17 regions of interest (ROI) to investigate the influence of different SNR values. CT scans were acquired with 9 different exposure settings. We developed an additive correction model to reduce scanner SNR influence. RESULTS: Sixty-two of 92 radiomic features showed high variance due to the scanner SNR. Of these 62 features, 47 showed at least a factor 2 significant standard deviation reduction by using the additive correction model. We assessed the clinical relevance of radiomics instability by using a 221 NSCLC patient cohort measured with the same scanner. CONCLUSIONS: Phantom measurements show that roughly two third of the radiomic features depend on the exposure setting of the scanner. The dependence can be modeled and corrected significantly reducing the variation in feature values with at least a factor of 2. More complex models will likely increase the correctability. Scanner SNR correction will result in more reliable radiomics predictions in NSCLC.

10.
Sci Data ; 6(1): 218, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31641134

RESUMO

Prediction modelling with radiomics is a rapidly developing research topic that requires access to vast amounts of imaging data. Methods that work on decentralized data are urgently needed, because of concerns about patient privacy. Previously published computed tomography medical image sets with gross tumour volume (GTV) outlines for non-small cell lung cancer have been updated with extended follow-up. In a previous study, these were referred to as Lung1 (n = 421) and Lung2 (n = 221). The Lung1 dataset is made publicly accessible via The Cancer Imaging Archive (TCIA; https://www.cancerimagingarchive.net ). We performed a decentralized multi-centre study to develop a radiomic signature (hereafter "ZS2019") in one institution and validated the performance in an independent institution, without the need for data exchange and compared this to an analysis where all data was centralized. The performance of ZS2019 for 2-year overall survival validated in distributed radiomics was not statistically different from the centralized validation (AUC 0.61 vs 0.61; p = 0.52). Although slightly different in terms of data and methods, no statistically significant difference in performance was observed between the new signature and previous work (c-index 0.58 vs 0.65; p = 0.37). Our objective was not the development of a new signature with the best performance, but to suggest an approach for distributed radiomics. Therefore, we used a similar method as an earlier study. We foresee that the Lung1 dataset can be further re-used for testing radiomic models and investigating feature reproducibility.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Neoplasias Pulmonares/diagnóstico por imagem , Conjuntos de Dados como Assunto , Humanos , Tomografia Computadorizada por Raios X
11.
Med Phys ; 46(3): 1512-1518, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30629299

RESUMO

PURPOSE: The aim of this paper is to describe a public, open-access, computed tomography (CT) phantom image set acquired at three centers and collected especially for radiomics reproducibility research. The dataset is useful to test radiomic features reproducibility with respect to various parameters, such as acquisition settings, scanners, and reconstruction algorithms. ACQUISITION AND VALIDATION METHODS: Three phantoms were scanned in three independent institutions. Images of the following phantoms were acquired: Catphan 700 and COPDGene Phantom II (Phantom Laboratory, Greenwich, NY, USA), and the Triple modality 3D Abdominal Phantom (CIRS, Norfolk, VA, USA). Data were collected at three Dutch medical centers: MAASTRO Clinic (Maastricht, NL), Radboud University Medical Center (Nijmegen, NL), and University Medical Center Groningen (Groningen, NL) with scanners from two different manufacturers Siemens Healthcare and Philips Healthcare. The following acquisition parameter were varied in the phantom scans: slice thickness, reconstruction kernels, and tube current. DATA FORMAT AND USAGE NOTES: We made the dataset publically available on the Dutch instance of "Extensible Neuroimaging Archive Toolkit-XNAT" (https://xnat.bmia.nl). The dataset is freely available and reusable with attribution (Creative Commons 3.0 license). POTENTIAL APPLICATIONS: Our goal was to provide a findable, open-access, annotated, and reusable CT phantom dataset for radiomics reproducibility studies. Reproducibility testing and harmonization are fundamental requirements for wide generalizability of radiomics-based clinical prediction models. It is highly desirable to include only reproducible features into models, to be more assured of external validity across hitherto unseen contexts. In this view, phantom data from different centers represent a valuable source of information to exclude CT radiomic features that may already be unstable with respect to simplified structures and tightly controlled scan settings. The intended extension of our shared dataset is to include other modalities and phantoms with more realistic lesion simulations.


Assuntos
Bases de Dados Factuais , Imagens de Fantasmas , Doença Pulmonar Obstrutiva Crônica/diagnóstico por imagem , Radiografia Abdominal , Radiografia Torácica , Tomografia Computadorizada por Raios X/métodos , Algoritmos , Humanos , Processamento de Imagem Assistida por Computador/métodos , Estudos Multicêntricos como Assunto , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA