605 research outputs found

    The 5th International Conference on Biomedical Engineering and Biotechnology (ICBEB 2016)

    Get PDF

    Quantification of tumour heterogenity in MRI

    Get PDF
    Cancer is the leading cause of death that touches us all, either directly or indirectly. It is estimated that the number of newly diagnosed cases in the Netherlands will increase to 123,000 by the year 2020. General Dutch statistics are similar to those in the UK, i.e. over the last ten years, the age-standardised incidence rate1 has stabilised at around 355 females and 415 males per 100,000. Figure 1 shows the cancer incidence per gender. In the UK, the rise in lifetime risk of cancer is more than one in three and depends on many factors, including age, lifestyle and genetic makeup

    ๊ต๋ชจ์„ธํฌ์ข… ํ™˜์ž์—์„œ์˜ ์—ญ๋™์  ์กฐ์˜์ฆ๊ฐ• ์ž๊ธฐ๊ณต๋ช…์˜์ƒ์˜ ๋ผ๋””์˜ค๋ฏน์Šค ์ ์ˆ˜๋ฅผ ์ด์šฉํ•œ IDH ๋Œ์—ฐ๋ณ€์ด ์ƒํƒœ ๋…๋ฆฝ์  ๊ณ ์œ„ํ—˜๊ตฐ ์˜ˆ์ธก ์—ฐ๊ตฌ

    Get PDF
    ํ•™์œ„๋…ผ๋ฌธ(๋ฐ•์‚ฌ) -- ์„œ์šธ๋Œ€ํ•™๊ต๋Œ€ํ•™์› : ์˜๊ณผ๋Œ€ํ•™ ์˜ํ•™๊ณผ, 2021.8. ์ตœ์Šนํ™.Objective To develop a radiomics risk score based on dynamic contrast-enhanced (DCE) MRI for the prediction high-risk group in glioblastoma patients. Materials and Methods One hundred fifty patients (92 men (61.3%); mean age, 60.5 ยฑ 13.5 years) with glioblastoma who underwent a preoperative MRI were enrolled in the study. Six hundred forty-two radiomic features were extracted from Ktrans, Vp and Ve maps of DCE MRI, where regions of interest were based on both non-enhancing T2 hyperintense areas and T1-weighted contrast-enhancing areas. Using feature selection algorithms, significant radiomic features were selected. Subsequently, a radiomics risk score was developed using a weighted combination of the selected features in the discovery set (n = 105) and validated in the validation set (n = 45) by investigating the difference in prognosis between โ€œradiomics risk scoreโ€ groups. Finally, a multivariate Cox-regression for 1-year progression-free survival was performed using the radiomics risk score and clinical variables. Results Sixteen radiomic features obtained from non-enhancing T2 hyperintense areas were selected out of 642 features. The radiomics risk score stratified high- and low-risk groups in both the discovery and validation set in log rank test (both p < 0.001). The radiomics risk score increased the risk of progression in glioblastoma patients, independently of IDH-mutation status (HR = 3.56, p = 0.004; HR = 0.34, p = 0.022, respectively). Conclusion We developed and assessed the โ€œradiomics risk scoreโ€ from the features of DCE MRI based on non-enhancing T2 hyperintense areas for risk stratification of progression at 1 year in glioblastoma patients, which was independent of IDH mutational status.๋ชฉ์ : ๋ณธ ์—ฐ๊ตฌ์˜ ๋ชฉ์ ์€ ๊ต๋ชจ์„ธํฌ์ข… ํ™˜์ž์˜ ๊ณ ์œ„ํ—˜๊ตฐ์„ ์˜ˆ์ธกํ•˜๊ธฐ ์œ„ํ•ด ์„œ ์—ญ๋™์  ์กฐ์˜์ฆ๊ฐ• ์ž๊ธฐ๊ณต๋ช…์˜์ƒ ๊ธฐ๋ฐ˜์˜ ๋ผ๋””์˜ค๋ฏน์Šค ์ ์ˆ˜๋ฅผ ๊ฐœ๋ฐœํ•˜๋Š” ๊ฒƒ์ด๋‹ค. ๋ฐฉ๋ฒ•: ๋ณธ ์—ฐ๊ตฌ์—๋Š” ์ˆ˜์ˆ  ์ „ DCE MRI๋ฅผ ์‹œํ–‰๋ฐ›์€ ๊ต๋ชจ์„ธํฌ์ข… ํ™˜์ž 150 ๋ช… (๋‚จ์„ฑ 92 ๋ช… (61.3 %), ํ‰๊ท  ์—ฐ๋ น 60.5 ยฑ 13.5 ์„ธ)์ด ํฌํ•จ๋˜์—ˆ๋‹ค. DCE MRI์˜ Ktrans, Vp ๋ฐ Ve ์ง€๋„ ๊ฐ๊ฐ์—์„œ 640 ๊ฐœ์˜ radiomics ์ง€ํ‘œ๊ฐ€ ์ถ”์ถœ๋˜์—ˆ์œผ๋ฉฐ, ์ด๋ฅผ ์œ„ํ•˜์—ฌ ์ข…์–‘ ๋ถ€์œ„๋Š” ์กฐ์˜์ฆ๊ฐ• T1WI ์™€ FLAIR ์˜์ƒ์„ ์ด์šฉํ•˜์—ฌ segmentation ํ•˜์˜€๋‹ค. ์ง€ํ‘œ ์„ ํƒ ์•Œ๊ณ ๋ฆฌ์ฆ˜์„ ์‚ฌ์šฉํ•˜์—ฌ 642 ๊ฐœ ์ง€ํ‘œ ์ค‘ ์˜ˆํ›„ ์˜ˆ์ธก์— ํŠน์ด์ ์ธ radiomics ์ง€ํ‘œ๋ฅผ ์„ ํƒํ–ˆ๋‹ค. ๋‹ค์Œ์œผ๋กœ, discovery set (n = 105)์—์„œ ์„ ํƒ๋œ ์ง€ํ‘œ์˜ ๊ฐ€์ค‘์น˜ ์กฐํ•ฉ์„ ์‚ฌ์šฉํ•˜์—ฌ radiomics risk score๋ฅผ ๊ฐœ๋ฐœํ•˜๊ณ  radiomics risk score์— ๊ธฐ๋ฐ˜ํ•˜์—ฌ ๊ณ ์œ„ํ—˜ ๋ฐ ์ €์œ„ํ—˜ ๊ทธ๋ฃน ๊ฐ„์˜ ์˜ˆํ›„ ์ฐจ์ด๋ฅผ ์กฐ์‚ฌํ•˜์—ฌ validation set (n = 45)์—์„œ ๊ฒ€์ฆํ•˜์˜€๋‹ค. ๋งˆ์ง€๋ง‰์œผ๋กœ, 1๋…„ ๋ฌด์ง„ํ–‰ ์ƒ์กด์œจ ๋ถ„์„์„ ์œ„ํ•œ ๋‹ค๋ณ€๋Ÿ‰ Cox- ํšŒ๊ท€ ๊ธฐ๋ฒ•์„ ์‚ฌ์šฉํ•˜์—ฌ ์ž„์ƒ ๋ณ€์ˆ˜์™€ ํ•จ๊ป˜ radiomics risk score์˜ ์˜ˆํ›„ ์˜ˆ์ธก๋ ฅ์„ ํ‰๊ฐ€ํ•˜์˜€๋‹ค. ๊ฒฐ๊ณผ: ๋น„์กฐ์˜์ฆ๊ฐ• T2 ๊ณ ์‹ ํ˜ธ ์˜์—ญ์—์„œ ์–ป์€ 16 ๊ฐ€์ง€ radiomics ์ง€ํ‘œ๊ฐ€ 642๊ฐœ ์ง€ํ‘œ ์ค‘ ์„ ํƒ๋˜์—ˆ๋‹ค. ์ด ๋‘๊ฐ€์ง€ ์ง€ํ‘œ๋ฅผ ์ด์šฉํ•˜์—ฌ Radiomics risk score๋ฅผ ๋งŒ๋“ค์—ˆ์œผ๋ฉฐ, ์ด๋ฅผ ์ด์šฉํ•˜์˜€์„ ๋•Œ, ๋กœ๊ทธ ์ˆœ์œ„ ํ…Œ์ŠคํŠธ์—์„œ discovery ๋ฐ test set์—์„œ ๊ณ ์œ„ํ—˜๊ตฐ๊ณผ ์ € ์œ„ํ—˜๊ตฐ์„ ์œ ์˜๋ฏธํ•˜๊ฒŒ ๊ตฌ๋ถ„ํ•  ์ˆ˜ ์žˆ์—ˆ๋‹ค (p<0.001). Radiomics risk score๋Š” isocitrate dehydrogenase (IDH) ๋Œ์—ฐ๋ณ€์ด์™€ ๋…๋ฆฝ์ ์ธ ์˜ˆํ›„ ์˜ˆ์ธก์ธ์ž์˜€๋‹ค (Hazard ratio (HR) = 3.56 (p = 0.004)). ๊ฒฐ๋ก : ๊ต๋ชจ์„ธํฌ์ข… ํ™˜์ž์—์„œ 1๋…„ ๋ฌด์ง„ํ–‰ ์ƒ์กด์œจ ์˜ˆ์ธก์— ์žˆ์–ด ๋น„์กฐ์˜์ฆ๊ฐ• T2 ๊ณ ์‹ ํ˜ธ ์˜์—ญ์—์„œ์˜ DCE MRI ๊ธฐ๋ฐ˜์˜ radiomics risk score ๊ฐ€ ์šฐ์ˆ˜ํ•œ ์„ฑ์ ์„ ๋ณด์˜€์œผ๋ฉฐ, ํ–ฅํ›„ ์ด๋ฅผ ์ด์šฉํ•œ ์ž„์ƒ ์ด์šฉ ๊ฐ€๋Šฅ์„ฑ์ด ๊ธฐ๋Œ€๋œ๋‹ค.Introduction 4 Materials and methods 14 Results 23 Discussion 27 References 34 Tables 52 Figures 58 Supplementary materials 71 Abstract in Korean 90๋ฐ•

    Computational Imaging Biomarkers For Precision Medicine: Characterizing Heterogeneity In Breast Cancer

    Get PDF
    In the United States, 1 in 8 women are diagnosed with breast cancer. Breast tumor heterogeneity is well-established, with intratumor heterogeneity manifesting spatially and temporally. Increased heterogeneity is associated with adverse clinical outcomes. Current critical disease treatment decisions are made on the basis of biomarkers acquired from tissue samples, largely under sampling the heterogeneous disease burden. In order to drive precision medicine treatment strategies for cancer, personalized biomarkers are needed to truly characterize intratumor heterogeneity. Medical imaging can provide anon-invasive, whole tumor sampling of disease burden at the time of diagnosis and allows for longitudinal monitoring of disease progression. The studies outlined in this thesis introduce analytical tools developed through computer vision, bioinformatics, and machine learning and use diagnostic and longitudinal clinical images of breast cancer to develop computational imaging biomarkers characterizing intratumor heterogeneity. Intrinsic imaging phenotypes of spatial heterogeneity, identified in dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) images at the time of diagnosis, were identified and validated, demonstrating improved prognostic value over conventional histopathologic biomarkers when predicting 10-year recurrence free survival. Intrinsic phenotypes of longitudinal change in spatial heterogeneity in response to neoadjuvant treatment, identified in DCE-MRI were identified and leveraged as prognostic and predictive biomarkers, demonstrating augmented prognostic value when added to conventional histopathologic and personalized molecular biomarkers. To better characterize 4-D spatial and temporal heterogeneity, illuminated through dynamic positron emission tomography imaging, a novel 4-D segmentation algorithm was developed to identify spatially constrained, functionally discrete intratumor sub-regions. Quantifying the identified sub-regions through a novel imaging signature demonstrated the prognostic value of characterizing intratumor heterogeneity when predicting recurrence free survival, demonstrating prognostic improvement over established histopathologic biomarkers and conventional kinetic model derived parameters. Collectively, the studies in this thesis demonstrate the value of leveraging computational imaging biomarkers to characterize intratumor heterogeneity. Such biomarkers have the potential to be utilized towards precision medicine for cancer care

    Novel Semi-Supervised Learning Models to Balance Data Inclusivity and Usability in Healthcare Applications

    Get PDF
    abstract: Semi-supervised learning (SSL) is sub-field of statistical machine learning that is useful for problems that involve having only a few labeled instances with predictor (X) and target (Y) information, and abundance of unlabeled instances that only have predictor (X) information. SSL harnesses the target information available in the limited labeled data, as well as the information in the abundant unlabeled data to build strong predictive models. However, not all the included information is useful. For example, some features may correspond to noise and including them will hurt the predictive model performance. Additionally, some instances may not be as relevant to model building and their inclusion will increase training time and potentially hurt the model performance. The objective of this research is to develop novel SSL models to balance data inclusivity and usability. My dissertation research focuses on applications of SSL in healthcare, driven by problems in brain cancer radiomics, migraine imaging, and Parkinsonโ€™s Disease telemonitoring. The first topic introduces an integration of machine learning (ML) and a mechanistic model (PI) to develop an SSL model applied to predicting cell density of glioblastoma brain cancer using multi-parametric medical images. The proposed ML-PI hybrid model integrates imaging information from unbiopsied regions of the brain as well as underlying biological knowledge from the mechanistic model to predict spatial tumor density in the brain. The second topic develops a multi-modality imaging-based diagnostic decision support system (MMI-DDS). MMI-DDS consists of modality-wise principal components analysis to incorporate imaging features at different aggregation levels (e.g., voxel-wise, connectivity-based, etc.), a constrained particle swarm optimization (cPSO) feature selection algorithm, and a clinical utility engine that utilizes inverse operators on chosen principal components for white-box classification models. The final topic develops a new SSL regression model with integrated feature and instance selection called s2SSL (with โ€œs2โ€ referring to selection in two different ways: feature and instance). s2SSL integrates cPSO feature selection and graph-based instance selection to simultaneously choose the optimal features and instances and build accurate models for continuous prediction. s2SSL was applied to smartphone-based telemonitoring of Parkinsonโ€™s Disease patients.Dissertation/ThesisDoctoral Dissertation Industrial Engineering 201

    Metabolomics of Therapy Response in Preclinical Glioblastoma : a Multi-Slice MRSI-Based Volumetric Analysis for Noninvasive Assessment of Temozolomide Treatment

    Get PDF
    Glioblastoma (GBM) is the most common aggressive primary brain tumor in adults, with a short survival time even after aggressive therapy. Non-invasive surrogate biomarkers of therapy response may be relevant for improving patient survival. Previous work produced such biomarkers in preclinical GBM using semi-supervised source extraction and single-slice Magnetic Resonance Spectroscopic Imaging (MRSI). Nevertheless, GBMs are heterogeneous and single-slice studies could prevent obtaining relevant information. The purpose of this work was to evaluate whether a multi-slice MRSI approach, acquiring consecutive grids across the tumor, is feasible for preclinical models and may produce additional insight into therapy response. Nosological images were analyzed pixel-by-pixel and a relative responding volume, the Tumor Responding Index (TRI), was defined to quantify response. Heterogeneous response levels were observed and treated animals were ascribed to three arbitrary predefined groups: high response (HR, n = 2), TRI = 68.2 ยฑ 2.8%, intermediate response (IR, n = 6), TRI = 41.1 ยฑ 4.2% and low response (LR, n = 2), TRI = 13.4 ยฑ 14.3%, producing therapy response categorization which had not been fully registered in single-slice studies. Results agreed with the multi-slice approach being feasible and producing an inverse correlation between TRI and Ki67 immunostaining. Additionally, ca. 7-day oscillations of TRI were observed, suggesting that host immune system activation in response to treatment could contribute to the responding patterns detected

    Radiomics in Glioblastoma: Current Status and Challenges Facing Clinical Implementation

    Get PDF
    Radiomics analysis has had remarkable progress along with advances in medical imaging, most notability in central nervous system malignancies. Radiomics refers to the extraction of a large number of quantitative features that describe the intensity, texture and geometrical characteristics attributed to the tumor radiographic data. These features have been used to build predictive models for diagnosis, prognosis, and therapeutic response. Such models are being combined with clinical, biological, genetics and proteomic features to enhance reproducibility. Broadly, the four steps necessary for radiomic analysis are: (1) image acquisition, (2) segmentation or labeling, (3) feature extraction, and (4) statistical analysis. Major methodological challenges remain prior to clinical implementation. Essential steps include: adoption of an optimized standard imaging process, establishing a common criterion for performing segmentation, fully automated extraction of radiomic features without redundancy, and robust statistical modeling validated in the prospective setting. This review walks through these steps in detail, as it pertains to high grade gliomas. The impact on precision medicine will be discussed, as well as the challenges facing clinical implementation of radiomic in the current management of glioblastoma

    Role of machine learning in early diagnosis of kidney diseases.

    Get PDF
    Machine learning (ML) and deep learning (DL) approaches have been used as indispensable tools in modern artificial intelligence-based computer-aided diagnostic (AIbased CAD) systems that can provide non-invasive, early, and accurate diagnosis of a given medical condition. These AI-based CAD systems have proven themselves to be reproducible and have the generalization ability to diagnose new unseen cases with several diseases and medical conditions in different organs (e.g., kidneys, prostate, brain, liver, lung, breast, and bladder). In this dissertation, we will focus on the role of such AI-based CAD systems in early diagnosis of two kidney diseases, namely: acute rejection (AR) post kidney transplantation and renal cancer (RC). A new renal computer-assisted diagnostic (Renal-CAD) system was developed to precisely diagnose AR post kidney transplantation at an early stage. The developed Renal-CAD system perform the following main steps: (1) auto-segmentation of the renal allograft from surrounding tissues from diffusion weighted magnetic resonance imaging (DW-MRI) and blood oxygen level-dependent MRI (BOLD-MRI), (2) extraction of image markers, namely: voxel-wise apparent diffusion coefficients (ADCs) are calculated from DW-MRI scans at 11 different low and high b-values and then represented as cumulative distribution functions (CDFs) and extraction of the transverse relaxation rate (R2*) values from the segmented kidneys using BOLD-MRI scans at different echotimes, (3) integration of multimodal image markers with the associated clinical biomarkers, serum creatinine (SCr) and creatinine clearance (CrCl), and (4) diagnosing renal allograft status as nonrejection (NR) or AR by utilizing these integrated biomarkers and the developed deep learning classification model built on stacked auto-encoders (SAEs). Using a leaveone- subject-out cross-validation approach along with SAEs on a total of 30 patients with transplanted kidney (AR = 10 and NR = 20), the Renal-CAD system demonstrated 93.3% accuracy, 90.0% sensitivity, and 95.0% specificity in differentiating AR from NR. Robustness of the Renal-CAD system was also confirmed by the area under the curve value of 0.92. Using a stratified 10-fold cross-validation approach, the Renal-CAD system demonstrated its reproduciblity and robustness with a diagnostic accuracy of 86.7%, sensitivity of 80.0%, specificity of 90.0%, and AUC of 0.88. In addition, a new renal cancer CAD (RC-CAD) system for precise diagnosis of RC at an early stage was developed, which incorporates the following main steps: (1) estimating the morphological features by applying a new parametric spherical harmonic technique, (2) extracting appearance-based features, namely: first order textural features are calculated and second order textural features are extracted after constructing the graylevel co-occurrence matrix (GLCM), (3) estimating the functional features by constructing wash-in/wash-out slopes to quantify the enhancement variations across different contrast enhanced computed tomography (CE-CT) phases, (4) integrating all the aforementioned features and modeling a two-stage multilayer perceptron artificial neural network (MLPANN) classifier to classify the renal tumor as benign or malignant and identify the malignancy subtype. On a total of 140 RC patients (malignant = 70 patients (ccRCC = 40 and nccRCC = 30) and benign angiomyolipoma tumors = 70), the developed RC-CAD system was validated using a leave-one-subject-out cross-validation approach. The developed RC-CAD system achieved a sensitivity of 95.3% ยฑ 2.0%, a specificity of 99.9% ยฑ 0.4%, and Dice similarity coefficient of 0.98 ยฑ 0.01 in differentiating malignant from benign renal tumors, as well as an overall accuracy of 89.6% ยฑ 5.0% in the sub-typing of RCC. The diagnostic abilities of the developed RC-CAD system were further validated using a randomly stratified 10-fold cross-validation approach. The results obtained using the proposed MLP-ANN classification model outperformed other machine learning classifiers (e.g., support vector machine, random forests, and relational functional gradient boosting) as well as other different approaches from the literature. In summary, machine and deep learning approaches have shown potential abilities to be utilized to build AI-based CAD systems. This is evidenced by the promising diagnostic performance obtained by both Renal-CAD and RC-CAD systems. For the Renal- CAD, the integration of functional markers extracted from multimodal MRIs with clinical biomarkers using SAEs classification model, potentially improved the final diagnostic results evidenced by high accuracy, sensitivity, and specificity. The developed Renal-CAD demonstrated high feasibility and efficacy for early, accurate, and non-invasive identification of AR. For the RC-CAD, integrating morphological, textural, and functional features extracted from CE-CT images using a MLP-ANN classification model eventually enhanced the final results in terms of accuracy, sensitivity, and specificity, making the proposed RC-CAD a reliable noninvasive diagnostic tool for RC. The early and accurate diagnosis of AR or RC will help physicians to provide early intervention with the appropriate treatment plan to prolong the life span of the diseased kidney, increase the survival chance of the patient, and thus improve the healthcare outcome in the U.S. and worldwide
    • โ€ฆ
    corecore