29 research outputs found

    Current state of pediatric neuro-oncology imaging, challenges and future directions

    Get PDF
    Imaging plays a central role in neuro-oncology including primary diagnosis, treatment planning, and surveillance of tumors. The emergence of quantitative imaging and radiomics provided an uprecedented opportunity to compile mineable databases that can be utilized in a variety of applications. In this review, we aim to summarize the current state of conventional and advanced imaging techniques, standardization efforts, fast protocols, contrast and sedation in pediatric neuro-oncologic imaging, radiomics-radiogenomics, multi-omics and molecular imaging approaches. We will also address the existing challenges and discuss future directions

    Quantification of Multi-Parametric Magnetic Resonance Imaging Based on Radiomics Analysis for Differentiation of Benign and Malignant Lesions of Prostate

    Get PDF
    Background: The most common cancer (non-cutaneous) malignancy among men is prostate cancer. Management of prostate cancer, including staging and treatment, playing an important role in decreasing mortality rates. Among all current diagnostic tools, multiparametric MRI (mp-MRI) has shown high potential in localizing and staging prostate cancer. Quantification of mp-MRI helps to decrease the dependency of diagnosis on readers’ opinions. Objective: The aim of this research is to set a method based on quantification of mp-MRI images for discrimination between benign and malignant prostatic lesions with fusion-guided MR imaging/transrectal ultrasonography biopsy as a pathology validation reference. Material and Methods: It is an analytical research that 27 patients underwent the mp-MRI examination, including T1- and T2- weighted and diffusion weighted imaging (DWI). Quantification was done by calculating radiomic features from mp-MRI images. Receiver-operating-characteristic curve was done for each feature to evaluate the discriminatory capacity and linear discriminant analysis (LDA) and leave-one-out cross-validation for feature filtering to estimate the sensitivity, specificity and accuracy of the benign and malignant lesion differentiation process is used. Results: An accuracy, sensitivity and specificity of 92.6%, 95.2% and 83.3%, respectively, were achieved from a subset of radiomics features obtained from T2-weighted images and apparent diffusion coefficient (ADC) maps for distinguishing benign and malignant prostate lesions.  Conclusion: Quantification of mp-MRI (T2-weighted images and ADC-maps) based on radiomics feature has potential to distinguish benign with appropriate accuracy from malignant prostate lesions. This technique is helpful in preventing needless biopsies in patients and provides an assisted diagnosis for classifications of prostate lesions

    Radiomic Analysis of Multi-parametric MR Images (MRI) for Classification of Parotid Tumors

    Get PDF
    Background: Characterization of parotid tumors before surgery using multi-parametric magnetic resonance imaging (MRI) scans can support clinical decision making about the best-suited therapeutic strategy for each patient. Objective: This study aims to differentiate benign from malignant parotid tumors through radiomics analysis of multi-parametric MR images, incorporating T2-w images with ADC-map and parametric maps generated from Dynamic Contrast Enhanced MRI (DCE-MRI).Material and Methods: MRI scans of 31 patients with histopathologically-confirmed parotid gland tumors (23 benign, 8 malignant) were included in this retrospective study. For DCE-MRI, semi-quantitative analysis, Tofts pharmacokinetic (PK) modeling, and five-parameter sigmoid modeling were performed and parametric maps were generated. For each patient, borders of the tumors were delineated on whole tumor slices of T2-w image, ADC-map, and the late-enhancement dynamic series of DCE-MRI, creating regions-of-interest (ROIs). Radiomic analysis was performed for the specified ROIs. Results: Among the DCE-MRI-derived parametric maps, wash-in rate (WIR) and PK-derived Ktrans parameters surpassed the accuracy of other parameters based on support vector machine (SVM) classifier. Radiomics analysis of ADC-map outperformed the T2-w and DCE-MRI techniques using the simpler classifier, suggestive of its inherently high sensitivity and specificity. Radiomics analysis of the combination of T2-w image, ADC-map, and DCE-MRI parametric maps resulted in accuracy of 100% with both classifiers with fewer numbers of selected texture features than individual images.  Conclusion: In conclusion, radiomics analysis is a reliable quantitative approach for discrimination of parotid tumors and can be employed as a computer-aided approach for pre-operative diagnosis and treatment planning of the patients

    Radio-Pathomic Approaches in Pediatric Neurooncology: Opportunities and Challenges

    Get PDF
    With medical software platforms moving to cloud environments with scalable storage and computing, the translation of predictive artificial intelligence (AI) models to aid in clinical decision-making and facilitate personalized medicine for cancer patients is becoming a reality. Medical imaging, namely radiologic and histologic images, has immense analytical potential in neuro-oncology, and models utilizing integrated radiomic and pathomic data may yield a synergistic effect and provide a new modality for precision medicine. At the same time, the ability to harness multi-modal data is met with challenges in aggregating data across medical departments and institutions, as well as significant complexity in modeling the phenotypic and genotypic heterogeneity of pediatric brain tumors. In this paper, we review recent pathomic and integrated pathomic, radiomic, and genomic studies with clinical applications. We discuss current challenges limiting translational research on pediatric brain tumors and outline technical and analytical solutions. Overall, we propose that to empower the potential residing in radio-pathomics, systemic changes in cross-discipline data management and end-to-end software platforms to handle multi-modal data sets are needed, in addition to embracing modern AI-powered approaches. These changes can improve the performance of predictive models, and ultimately the ability to advance brain cancer treatments and patient outcomes through the development of such models

    Training and Comparison of nnU-Net and DeepMedic Methods for Autosegmentation of Pediatric Brain Tumors

    Full text link
    Brain tumors are the most common solid tumors and the leading cause of cancer-related death among children. Tumor segmentation is essential in surgical and treatment planning, and response assessment and monitoring. However, manual segmentation is time-consuming and has high inter-operator variability, underscoring the need for more efficient methods. We compared two deep learning-based 3D segmentation models, DeepMedic and nnU-Net, after training with pediatric-specific multi-institutional brain tumor data using based on multi-parametric MRI scans.Multi-parametric preoperative MRI scans of 339 pediatric patients (n=293 internal and n=46 external cohorts) with a variety of tumor subtypes, were preprocessed and manually segmented into four tumor subregions, i.e., enhancing tumor (ET), non-enhancing tumor (NET), cystic components (CC), and peritumoral edema (ED). After training, performance of the two models on internal and external test sets was evaluated using Dice scores, sensitivity, and Hausdorff distance with reference to ground truth manual segmentations. Dice score for nnU-Net internal test sets was (mean +/- SD (median)) 0.9+/-0.07 (0.94) for WT, 0.77+/-0.29 for ET, 0.66+/-0.32 for NET, 0.71+/-0.33 for CC, and 0.71+/-0.40 for ED, respectively. For DeepMedic the Dice scores were 0.82+/-0.16 for WT, 0.66+/-0.32 for ET, 0.48+/-0.27, for NET, 0.48+/-0.36 for CC, and 0.19+/-0.33 for ED, respectively. Dice scores were significantly higher for nnU-Net (p<=0.01). External validation of the trained nnU-Net model on the multi-institutional BraTS-PEDs 2023 dataset revealed high generalization capability in segmentation of whole tumor and tumor core with Dice scores of 0.87+/-0.13 (0.91) and 0.83+/-0.18 (0.89), respectively. Pediatric-specific data trained nnU-Net model is superior to DeepMedic for whole tumor and subregion segmentation of pediatric brain tumors

    Unsupervised Machine Learning Using K-Means Identifies Radiomic Subgroups of Pediatric Low-Grade Gliomas That Correlate With Key Molecular Markers

    Get PDF
    Introduction: Despite advancements in molecular and histopathologic characterization of pediatric low-grade gliomas (pLGGs), there remains significant phenotypic heterogeneity among tumors with similar categorizations. We hypothesized that an unsupervised machine learning approach based on radiomic features may reveal distinct pLGG imaging subtypes. Methods: Multi-parametric MR images (T1 pre- and post-contrast, T2, and T2 FLAIR) from 157 patients with pLGGs were collected and 881 quantitative radiomic features were extracted from tumorous region. Clustering was performed using K-means after applying principal component analysis (PCA) for feature dimensionality reduction. Molecular and demographic data was obtained from the PedCBioportal and compared between imaging subtypes. Results: K-means identified three distinct imaging-based subtypes. Subtypes differed in mutational frequencies of BRAF (p \u3c 0.05) as well as the gene expression of BRAF (p\u3c0.05). It was also found that age (p \u3c 0.05), tumor location (p \u3c 0.01), and tumor histology (p \u3c 0.0001) differed significantly between the imaging subtypes. Conclusion: In this exploratory work, it was found that clustering of pLGGs based on radiomic features identifies distinct, imaging-based subtypes that correlate with important molecular markers and demographic details. This finding supports the notion that incorporation of radiomic data could augment our ability to better characterize pLGGs

    The Brain Tumor Segmentation (BraTS) Challenge 2023: Brain MR Image Synthesis for Tumor Segmentation (BraSyn)

    Full text link
    Automated brain tumor segmentation methods have become well-established and reached performance levels offering clear clinical utility. These methods typically rely on four input magnetic resonance imaging (MRI) modalities: T1-weighted images with and without contrast enhancement, T2-weighted images, and FLAIR images. However, some sequences are often missing in clinical practice due to time constraints or image artifacts, such as patient motion. Consequently, the ability to substitute missing modalities and gain segmentation performance is highly desirable and necessary for the broader adoption of these algorithms in the clinical routine. In this work, we present the establishment of the Brain MR Image Synthesis Benchmark (BraSyn) in conjunction with the Medical Image Computing and Computer-Assisted Intervention (MICCAI) 2023. The primary objective of this challenge is to evaluate image synthesis methods that can realistically generate missing MRI modalities when multiple available images are provided. The ultimate aim is to facilitate automated brain tumor segmentation pipelines. The image dataset used in the benchmark is diverse and multi-modal, created through collaboration with various hospitals and research institutions.Comment: Technical report of BraSy

    The Brain Tumor Segmentation (BraTS) Challenge 2023: Focus on Pediatrics (CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs)

    Full text link
    Pediatric tumors of the central nervous system are the most common cause of cancer-related death in children. The five-year survival rate for high-grade gliomas in children is less than 20\%. Due to their rarity, the diagnosis of these entities is often delayed, their treatment is mainly based on historic treatment concepts, and clinical trials require multi-institutional collaborations. The MICCAI Brain Tumor Segmentation (BraTS) Challenge is a landmark community benchmark event with a successful history of 12 years of resource creation for the segmentation and analysis of adult glioma. Here we present the CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs 2023 challenge, which represents the first BraTS challenge focused on pediatric brain tumors with data acquired across multiple international consortia dedicated to pediatric neuro-oncology and clinical trials. The BraTS-PEDs 2023 challenge focuses on benchmarking the development of volumentric segmentation algorithms for pediatric brain glioma through standardized quantitative performance evaluation metrics utilized across the BraTS 2023 cluster of challenges. Models gaining knowledge from the BraTS-PEDs multi-parametric structural MRI (mpMRI) training data will be evaluated on separate validation and unseen test mpMRI dataof high-grade pediatric glioma. The CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs 2023 challenge brings together clinicians and AI/imaging scientists to lead to faster development of automated segmentation techniques that could benefit clinical trials, and ultimately the care of children with brain tumors

    A Multi-Institutional Meningioma MRI Dataset for Automated Multi-Sequence Image Segmentation

    Get PDF
    Meningiomas are the most common primary intracranial tumors and can be associated with significant morbidity and mortality. Radiologists, neurosurgeons, neuro-oncologists, and radiation oncologists rely on brain MRI for diagnosis, treatment planning, and longitudinal treatment monitoring. However, automated, objective, and quantitative tools for non-invasive assessment of meningiomas on multi-sequence MR images are not available. Here we present the BraTS Pre-operative Meningioma Dataset, as the largest multi-institutional expert annotated multilabel meningioma multi-sequence MR image dataset to date. This dataset includes 1,141 multi-sequence MR images from six sites, each with four structural MRI sequences (T2-, T2/FLAIR-, pre-contrast T1-, and post-contrast T1-weighted) accompanied by expert manually refined segmentations of three distinct meningioma sub-compartments: enhancing tumor, non-enhancing tumor, and surrounding non-enhancing T2/FLAIR hyperintensity. Basic demographic data are provided including age at time of initial imaging, sex, and CNS WHO grade. The goal of releasing this dataset is to facilitate the development of automated computational methods for meningioma segmentation and expedite their incorporation into clinical practice, ultimately targeting improvement in the care of meningioma patients
    corecore