87 research outputs found

    I2ECR: Integrated and Intelligent Environment for Clinical Research

    Get PDF
    Clinical trials are designed to produce new knowledge about a certain disease, drug or treatment. During these studies, a huge amount of data is collected about participants, therapies, clinical procedures, outcomes, adverse events and so on. A multicenter, randomized, phase III clinical trial in Hematology enrolls up to hundreds of subjects and evaluates post-treatment outcomes on stratified sub- groups of subjects for a period of many years. Therefore, data collection in clinical trials is becoming complex, with huge amount of clinical and biological variables. Outside the medical field, data warehouses (DWs) are widely employed. A Data Ware-house is a “collection of integrated, subject-oriented databases designed to support the decision-making process”. To verify whether DWs might be useful for data quality and association analysis, a team of biomedical engineers, clinicians, biologists and statisticians developed the “I2ECR” project. I2ECR is an Integrated and Intelligent Environment for Clinical Research where clinical and omics data stand together for clinical use (reporting) and for generation of new clinical knowledge. I2ECR has been built from the “MCL0208” phase III, prospective, clinical trial, sponsored by the Fondazione Italiana Linfomi (FIL); this is actually a translational study, accounting for many clinical data, along with several clinical prognostic indexes (e.g. MIPI - Mantle International Prognostic Index), pathological information, treatment and outcome data, biological assessments of disease (MRD - Minimal Residue Disease), as well as many biological, ancillary studies, such as Mutational Analysis, Gene Expression Profiling (GEP) and Pharmacogenomics. In this trial forty-eight Italian medical centers were actively involved, for a total of 300 enrolled subjects. Therefore, I2ECR main objectives are: • to propose an integration project on clinical and molecular data quality concepts. The application of a clear row-data analysis as well as clinical trial monitoring strategies to implement a digital platform where clinical, biological and “omics” data are imported from different sources and well-integrated in a data- ware-house • to be a dynamic repository of data congruency quality rules. I2ECR allows to monitor, in a semi-automatic manner, the quality of data, in relation to the clinical data imported from eCRFs (electronic Case Report Forms) and from biologic and mutational datasets internally edited by local laboratories. Therefore, I2ECR will be able to detect missing data and mistakes derived from non-conventional data- entry activities by centers. • to provide to clinical stake-holders a platform from where they can easily design statistical and data mining analysis. The term Data Mining (DM) identifies a set of tools to searching for hidden patterns of interest in large and multivariate datasets. The applications of DM techniques in the medical field range from outcome prediction and patient classification to genomic medicine and molecular biology. I2ECR allows to clinical stake-holders to propose innovative methods of supervised and unsupervised feature extraction, data classification and statistical analysis on heterogeneous datasets associated to the MCL0208 clinical trial. Although MCL0208 study is the first example of data-population of I2ECR, the environment will be able to import data from clinical studies designed for other onco-hematologic diseases, too

    Multimodal Data Fusion and Quantitative Analysis for Medical Applications

    Get PDF
    Medical big data is not only enormous in its size, but also heterogeneous and complex in its data structure, which makes conventional systems or algorithms difficult to process. These heterogeneous medical data include imaging data (e.g., Positron Emission Tomography (PET), Computerized Tomography (CT), Magnetic Resonance Imaging (MRI)), and non-imaging data (e.g., laboratory biomarkers, electronic medical records, and hand-written doctor notes). Multimodal data fusion is an emerging vital field to address this urgent challenge, aiming to process and analyze the complex, diverse and heterogeneous multimodal data. The fusion algorithms bring great potential in medical data analysis, by 1) taking advantage of complementary information from different sources (such as functional-structural complementarity of PET/CT images) and 2) exploiting consensus information that reflects the intrinsic essence (such as the genetic essence underlying medical imaging and clinical symptoms). Thus, multimodal data fusion benefits a wide range of quantitative medical applications, including personalized patient care, more optimal medical operation plan, and preventive public health. Though there has been extensive research on computational approaches for multimodal fusion, there are three major challenges of multimodal data fusion in quantitative medical applications, which are summarized as feature-level fusion, information-level fusion and knowledge-level fusion: • Feature-level fusion. The first challenge is to mine multimodal biomarkers from high-dimensional small-sample multimodal medical datasets, which hinders the effective discovery of informative multimodal biomarkers. Specifically, efficient dimension reduction algorithms are required to alleviate "curse of dimensionality" problem and address the criteria for discovering interpretable, relevant, non-redundant and generalizable multimodal biomarkers. • Information-level fusion. The second challenge is to exploit and interpret inter-modal and intra-modal information for precise clinical decisions. Although radiomics and multi-branch deep learning have been used for implicit information fusion guided with supervision of the labels, there is a lack of methods to explicitly explore inter-modal relationships in medical applications. Unsupervised multimodal learning is able to mine inter-modal relationship as well as reduce the usage of labor-intensive data and explore potential undiscovered biomarkers; however, mining discriminative information without label supervision is an upcoming challenge. Furthermore, the interpretation of complex non-linear cross-modal associations, especially in deep multimodal learning, is another critical challenge in information-level fusion, which hinders the exploration of multimodal interaction in disease mechanism. • Knowledge-level fusion. The third challenge is quantitative knowledge distillation from multi-focus regions on medical imaging. Although characterizing imaging features from single lesions using either feature engineering or deep learning methods have been investigated in recent years, both methods neglect the importance of inter-region spatial relationships. Thus, a topological profiling tool for multi-focus regions is in high demand, which is yet missing in current feature engineering and deep learning methods. Furthermore, incorporating domain knowledge with distilled knowledge from multi-focus regions is another challenge in knowledge-level fusion. To address the three challenges in multimodal data fusion, this thesis provides a multi-level fusion framework for multimodal biomarker mining, multimodal deep learning, and knowledge distillation from multi-focus regions. Specifically, our major contributions in this thesis include: • To address the challenges in feature-level fusion, we propose an Integrative Multimodal Biomarker Mining framework to select interpretable, relevant, non-redundant and generalizable multimodal biomarkers from high-dimensional small-sample imaging and non-imaging data for diagnostic and prognostic applications. The feature selection criteria including representativeness, robustness, discriminability, and non-redundancy are exploited by consensus clustering, Wilcoxon filter, sequential forward selection, and correlation analysis, respectively. SHapley Additive exPlanations (SHAP) method and nomogram are employed to further enhance feature interpretability in machine learning models. • To address the challenges in information-level fusion, we propose an Interpretable Deep Correlational Fusion framework, based on canonical correlation analysis (CCA) for 1) cohesive multimodal fusion of medical imaging and non-imaging data, and 2) interpretation of complex non-linear cross-modal associations. Specifically, two novel loss functions are proposed to optimize the discovery of informative multimodal representations in both supervised and unsupervised deep learning, by jointly learning inter-modal consensus and intra-modal discriminative information. An interpretation module is proposed to decipher the complex non-linear cross-modal association by leveraging interpretation methods in both deep learning and multimodal consensus learning. • To address the challenges in knowledge-level fusion, we proposed a Dynamic Topological Analysis framework, based on persistent homology, for knowledge distillation from inter-connected multi-focus regions in medical imaging and incorporation of domain knowledge. Different from conventional feature engineering and deep learning, our DTA framework is able to explicitly quantify inter-region topological relationships, including global-level geometric structure and community-level clusters. K-simplex Community Graph is proposed to construct the dynamic community graph for representing community-level multi-scale graph structure. The constructed dynamic graph is subsequently tracked with a novel Decomposed Persistence algorithm. Domain knowledge is incorporated into the Adaptive Community Profile, summarizing the tracked multi-scale community topology with additional customizable clinically important factors

    Complexity Reduction in Image-Based Breast Cancer Care

    Get PDF
    The diversity of malignancies of the breast requires personalized diagnostic and therapeutic decision making in a complex situation. This thesis contributes in three clinical areas: (1) For clinical diagnostic image evaluation, computer-aided detection and diagnosis of mass and non-mass lesions in breast MRI is developed. 4D texture features characterize mass lesions. For non-mass lesions, a combined detection/characterisation method utilizes the bilateral symmetry of the breast s contrast agent uptake. (2) To improve clinical workflows, a breast MRI reading paradigm is proposed, exemplified by a breast MRI reading workstation prototype. Instead of mouse and keyboard, it is operated using multi-touch gestures. The concept is extended to mammography screening, introducing efficient navigation aids. (3) Contributions to finite element modeling of breast tissue deformations tackle two clinical problems: surgery planning and the prediction of the breast deformation in a MRI biopsy device

    Brainlesion: Glioma, Multiple Sclerosis, Stroke and Traumatic Brain Injuries

    Get PDF
    This two-volume set LNCS 12962 and 12963 constitutes the thoroughly refereed proceedings of the 7th International MICCAI Brainlesion Workshop, BrainLes 2021, as well as the RSNA-ASNR-MICCAI Brain Tumor Segmentation (BraTS) Challenge, the Federated Tumor Segmentation (FeTS) Challenge, the Cross-Modality Domain Adaptation (CrossMoDA) Challenge, and the challenge on Quantification of Uncertainties in Biomedical Image Quantification (QUBIQ). These were held jointly at the 23rd Medical Image Computing for Computer Assisted Intervention Conference, MICCAI 2020, in September 2021. The 91 revised papers presented in these volumes were selected form 151 submissions. Due to COVID-19 pandemic the conference was held virtually. This is an open access book

    Quantitative imaging in radiation oncology

    Get PDF
    Artificially intelligent eyes, built on machine and deep learning technologies, can empower our capability of analysing patients’ images. By revealing information invisible at our eyes, we can build decision aids that help our clinicians to provide more effective treatment, while reducing side effects. The power of these decision aids is to be based on patient tumour biologically unique properties, referred to as biomarkers. To fully translate this technology into the clinic we need to overcome barriers related to the reliability of image-derived biomarkers, trustiness in AI algorithms and privacy-related issues that hamper the validation of the biomarkers. This thesis developed methodologies to solve the presented issues, defining a road map for the responsible usage of quantitative imaging into the clinic as decision support system for better patient care

    Irish Machine Vision and Image Processing Conference Proceedings 2017

    Get PDF
    • …
    corecore