105 research outputs found

    Quality of Radiomic Features in Glioblastoma Multiforme: Impact of Semi-Automated Tumor Segmentation Software.

    Get PDF
    ObjectiveThe purpose of this study was to evaluate the reliability and quality of radiomic features in glioblastoma multiforme (GBM) derived from tumor volumes obtained with semi-automated tumor segmentation software.Materials and methodsMR images of 45 GBM patients (29 males, 16 females) were downloaded from The Cancer Imaging Archive, in which post-contrast T1-weighted imaging and fluid-attenuated inversion recovery MR sequences were used. Two raters independently segmented the tumors using two semi-automated segmentation tools (TumorPrism3D and 3D Slicer). Regions of interest corresponding to contrast-enhancing lesion, necrotic portions, and non-enhancing T2 high signal intensity component were segmented for each tumor. A total of 180 imaging features were extracted, and their quality was evaluated in terms of stability, normalized dynamic range (NDR), and redundancy, using intra-class correlation coefficients, cluster consensus, and Rand Statistic.ResultsOur study results showed that most of the radiomic features in GBM were highly stable. Over 90% of 180 features showed good stability (intra-class correlation coefficient [ICC] ≥ 0.8), whereas only 7 features were of poor stability (ICC < 0.5). Most first order statistics and morphometric features showed moderate-to-high NDR (4 > NDR ≥1), while above 35% of the texture features showed poor NDR (< 1). Features were shown to cluster into only 5 groups, indicating that they were highly redundant.ConclusionThe use of semi-automated software tools provided sufficiently reliable tumor segmentation and feature stability; thus helping to overcome the inherent inter-rater and intra-rater variability of user intervention. However, certain aspects of feature quality, including NDR and redundancy, need to be assessed for determination of representative signature features before further development of radiomics

    Robustness of pet radiomics features: Impact of co-registration with mri

    Get PDF
    Radiomics holds great promise in the field of cancer management. However, the clinical application of radiomics has been hampered by uncertainty about the robustness of the features extracted from the images. Previous studies have reported that radiomics features are sensitive to changes in voxel size resampling and interpolation, image perturbation, or slice thickness. This study aims to observe the variability of positron emission tomography (PET) radiomics features under the impact of co-registration with magnetic resonance imaging (MRI) using the difference percentage coefficient, and the Spearman’s correlation coefficient for three groups of images: (i) original PET, (ii) PET after co-registration with T1-weighted MRI and (iii) PET after co-registration with FLAIR MRI. Specifically, seventeen patients with brain cancers undergoing [11C]-Methionine PET were considered. Successively, PET images were co-registered with MRI sequences and 107 features were extracted for each mentioned group of images. The variability analysis revealed that shape features, first-order features and two subgroups of higher-order features possessed a good robustness, unlike the remaining groups of features, which showed large differences in the difference percentage coeffi-cient. Furthermore, using the Spearman’s correlation coefficient, approximately 40% of the selected features differed from the three mentioned groups of images. This is an important consideration for users conducting radiomics studies with image co-registration constraints to avoid errors in cancer diagnosis, prognosis, and clinical outcome prediction

    Medical Image Analytics (Radiomics) with Machine/Deeping Learning for Outcome Modeling in Radiation Oncology

    Full text link
    Image-based quantitative analysis (radiomics) has gained great attention recently. Radiomics possesses promising potentials to be applied in the clinical practice of radiotherapy and to provide personalized healthcare for cancer patients. However, there are several challenges along the way that this thesis will attempt to address. Specifically, this thesis focuses on the investigation of repeatability and reproducibility of radiomics features, the development of new machine/deep learning models, and combining these for robust outcomes modeling and their applications in radiotherapy. Radiomics features suffer from robustness issues when applied to outcome modeling problems, especially in head and neck computed tomography (CT) images. These images tend to contain streak artifacts due to patients’ dental implants. To investigate the influence of artifacts for radiomics modeling performance, we firstly developed an automatic artifact detection algorithm using gradient-based hand-crafted features. Then, comparing the radiomics models trained on ‘clean’ and ‘contaminated’ datasets. The second project focused on using hand-crafted radiomics features and conventional machine learning methods for the prediction of overall response and progression-free survival for Y90 treated liver cancer patients. By identifying robust features and embedding prior knowledge in the engineered radiomics features and using bootstrapped LASSO to select robust features, we trained imaging and dose based models for the desired clinical endpoints, highlighting the complementary nature of this information in Y90 outcomes prediction. Combining hand-crafted and machine learnt features can take advantage of both expert domain knowledge and advanced data-driven approaches (e.g., deep learning). Thus, we proposed a new variational autoencoder network framework that modeled radiomics features, clinical factors, and raw CT images for the prediction of intrahepatic recurrence-free and overall survival for hepatocellular carcinoma (HCC) patients in this third project. The proposed approach was compared with widely used Cox proportional hazard model for survival analysis. Our proposed methods achieved significant improvement in terms of the prediction using the c-index metric highlighting the value of advanced modeling techniques in learning from limited and heterogeneous information in actuarial prediction of outcomes. Advances in stereotactic radiation therapy (SBRT) has led to excellent local tumor control with limited toxicities for HCC patients, but intrahepatic recurrence still remains prevalent. As an extension of the third project, we not only hope to predict the time to intrahepatic recurrence, but also the location where the tumor might recur. This will be clinically beneficial for better intervention and optimizing decision making during the process of radiotherapy treatment planning. To address this challenging task, firstly, we proposed an unsupervised registration neural network to register atlas CT to patient simulation CT and obtain the liver’s Couinaud segments for the entire patient cohort. Secondly, a new attention convolutional neural network has been applied to utilize multimodality images (CT, MR and 3D dose distribution) for the prediction of high-risk segments. The results showed much improved efficiency for obtaining segments compared with conventional registration methods and the prediction performance showed promising accuracy for anticipating the recurrence location as well. Overall, this thesis contributed new methods and techniques to improve the utilization of radiomics for personalized radiotherapy. These contributions included new algorithm for detecting artifacts, a joint model of dose with image heterogeneity, combining hand-crafted features with machine learnt features for actuarial radiomics modeling, and a novel approach for predicting location of treatment failure.PHDApplied PhysicsUniversity of Michigan, Horace H. Rackham School of Graduate Studieshttp://deepblue.lib.umich.edu/bitstream/2027.42/163092/1/liswei_1.pd

    Robustness of PET Radiomics Features: Impact of Co-Registration with MRI

    Get PDF
    Radiomics holds great promise in the field of cancer management. However, the clinical application of radiomics has been hampered by uncertainty about the robustness of the features extracted from the images. Previous studies have reported that radiomics features are sensitive to changes in voxel size resampling and interpolation, image perturbation, or slice thickness. This study aims to observe the variability of positron emission tomography (PET) radiomics features under the impact of co-registration with magnetic resonance imaging (MRI) using the difference percentage coefficient, and the Spearman’s correlation coefficient for three groups of images: (i) original PET, (ii) PET after co-registration with T1-weighted MRI and (iii) PET after co-registration with FLAIR MRI. Specifically, seventeen patients with brain cancers undergoing [11C]-Methionine PET were considered. Successively, PET images were co-registered with MRI sequences and 107 features were extracted for each mentioned group of images. The variability analysis revealed that shape features, first-order features and two subgroups of higher-order features possessed a good robustness, unlike the remaining groups of features, which showed large differences in the difference percentage coefficient. Furthermore, using the Spearman’s correlation coefficient, approximately 40% of the selected features differed from the three mentioned groups of images. This is an important consideration for users conducting radiomics studies with image co-registration constraints to avoid errors in cancer diagnosis, prognosis, and clinical outcome prediction

    Artifical intelligence in rectal cancer

    Get PDF

    Identification of cancer hallmarks in patients with non-metastatic colon cancer after surgical resection

    Get PDF
    Colon cancer is one of the most common cancers in the world, and the therapeutic workflow is dependent on the TNM staging system and the presence of clinical risk factors. However, in the case of patients with non-metastatic disease, evaluating the benefit of adjuvant chemotherapy is a clinical challenge. Radiomics could be seen as a non-invasive novel imaging biomarker able to outline tumor phenotype and to predict patient prognosis by analyzing preoperative medical images. Radiomics might provide decisional support for oncologists with the goal to reduce the number of arbitrary decisions in the emerging era of personalized medicine. To date, much evidence highlights the strengths of radiomics in cancer workup, but several aspects limit the use of radiomics methods as routine. The study aimed to develop a radiomic model able to identify high-risk colon cancer by analyzing pre-operative CT scans. The study population comprised 148 patients: 108 with non-metastatic colon cancer were retrospectively enrolled from January 2015 to June 2020, and 40 patients were used as the external validation cohort. The population was divided into two groups—High-risk and No-risk—following the presence of at least one high-risk clinical factor. All patients had baseline CT scans, and 3D cancer segmentation was performed on the portal phase by two expert radiologists using open-source software (3DSlicer v4.10.2). Among the 107 radiomic features extracted, stable features were selected to evaluate the inter-class correlation (ICC) (cut-off ICC > 0.8). Stable features were compared between the two groups (T-test or Mann–Whitney), and the significant features were selected for univariate and multivariate logistic regression to build a predictive radiomic model. The radiomic model was then validated with an external cohort. In total, 58/108 were classified as High-risk and 50/108 as No-risk. A total of 35 radiomic features were stable (0.81 ≤ ICC <  0.92). Among these, 28 features were significantly different between the two groups (p < 0.05), and only 9 features were selected to build the radiomic model. The radiomic model yielded an AUC of 0.73 in the internal cohort and 0.75 in the external cohort. In conclusion, the radiomic model could be seen as a performant, non-invasive imaging tool to properly stratify colon cancers with high-risk diseas

    QUANTITATIVE IMAGING FOR PRECISION MEDICINE IN HEAD AND NECK CANCER PATIENTS

    Get PDF
    The purpose of this work was to determine if prediction models using quantitative imaging measures in head and neck squamous cell carcinoma (HNSCC) patients could be improved when noise due to imaging was reduced. This was investigated separately for salivary gland function using dynamic contrast enhanced magnetic resonance imaging (DCE-MRI), overall survival using computed tomography (CT)-based radiomics, and overall survival using positron emission tomography (PET)-based radiomics. From DCE-MRI, where T1-weighted images are serially acquired after injection of contrast, quantitative measures of diffusion can be obtained from the series of images. Radiomics is the study of the relationship of voxels to one another providing measures of texture from the area of interest. Quantitative information obtained from imaging could help in radiation treatment planning by providing quantifiable spatial information with computational models for assigning dose to regions to improve patient outcome, both survival and quality of life. By reducing the noise within the quantitative data, the prediction accuracy could improve to move this type of work closer to clinical practice. For each imaging modality sources of noise that could impact the patient analysis were identified, quantified, and if possible minimized during the patient analysis. In MRI, a large potential source of uncertainty was the image registration. To evaluate this, both physical and synthetic phantoms were used, which showed that registration of MR images was high, with all root mean square errors below 3 mm. Then, 15 HNSCC patients with pre-, mid-, and post-treatment DCE-MRI scans were evaluated. However, differences in algorithm output were found to be a large source of noise as different algorithms could not consistently rank patients as above or below the median for quantitative metrics from DCE-MRI. Therefore, further analysis using this modality was not pursued. In CT, a large potential source of noise that could impact patient analysis was the inter-scanner variability. To investigate this a controlled protocol was designed and used to image, along with the local head and chest protocols, a radiomics phantom on 100 CT scanners. This demonstrated that the inter-scanner variability could be reduced by over 50% using a controlled protocol compared to local protocols. Additionally, it was shown that the reconstruction parameters impact feature values while most acquisition parameters do not, therefore, most of this benefit can be achieved using a radiomics reconstruction with no additional dose to the patient. Then to evaluate this impact in patient studies, 726 HNSCC patients with CT images were used to create and test a Cox proportional hazards model for overall survival. Those patients with the same imaging protocol were subset and a new Cox proportional hazards model was created and tested in order to determine if the reduction in noise due to controlling the imaging protocol translated into improved prediction. However, noise between patient populations from different institutions was shown to be larger than the reduction in noise due to a controlled imaging protocol. In PET, a large potential source of noise that could impact patient analysis was the imaging protocol. A phantom scanned on three different scanners and vendors demonstrated that on a single vendor, imaging parameter choices did not affect radiomics feature values, but inter-scanner variances could be large. Then, 686 HNSCC patients with PET images were used to create and test a Cox proportional hazards model for overall survival. Those patients with the same imaging protocol were subset and a new Cox proportional hazards model was created and tested in order to determine if the reduction in noise due to controlling the imaging protocol on a vendor translated into improved prediction. However, no predictive radiomics signature could be determined for any subset of the patient cohort that resulted in significant stratification of patients into high and low risk. This study demonstrated that the imaging variability could be quantified and controlled for in each modality. However, for each modality there were larger sources of noise identified that did not allow for improvement in prediction modeling of salivary gland function or overall survival using quantitative imaging metrics for MRI, CT, or PET
    • …
    corecore