196 research outputs found

    Automatic classification of cancer tumors using image annotations and ontologies

    Get PDF
    Information about cancer stage in a patient is crucial when clinicians assess treatment progress. Determining cancer stage is a process that takes into account the description, location, characteristics and possible metastasis of cancerous tumors in a patient. It should follow classification standards, such as TNM Classification of Malignant Tumors. However, in clinical practice, the implementation of this process can be tedious and error-prone and create uncertainty. In order to alleviate these problems, we intend to assist radiologists by providing a second opinion in the evaluation of cancer stage in patients. For doing this, SemanticWeb technologies, such as ontologies and reasoning, will be used to automatically classify cancer stages. This classification will use semantic annotations, made by radiologists (using the ePAD tool) and stored in the AIM format, and rules of an ontology representing the TNM standard. The whole process will be validated through a proof of concept with users from the Radiology Dept. of the Stanford University.National Council for Scientific and Technological Development - CNPqCAPE

    Artificial intelligence in cancer imaging: Clinical challenges and applications

    Get PDF
    Judgement, as one of the core tenets of medicine, relies upon the integration of multilayered data with nuanced decision making. Cancer offers a unique context for medical decisions given not only its variegated forms with evolution of disease but also the need to take into account the individual condition of patients, their ability to receive treatment, and their responses to treatment. Challenges remain in the accurate detection, characterization, and monitoring of cancers despite improved technologies. Radiographic assessment of disease most commonly relies upon visual evaluations, the interpretations of which may be augmented by advanced computational analyses. In particular, artificial intelligence (AI) promises to make great strides in the qualitative interpretation of cancer imaging by expert clinicians, including volumetric delineation of tumors over time, extrapolation of the tumor genotype and biological course from its radiographic phenotype, prediction of clinical outcome, and assessment of the impact of disease and treatment on adjacent organs. AI may automate processes in the initial interpretation of images and shift the clinical workflow of radiographic detection, management decisions on whether or not to administer an intervention, and subsequent observation to a yet to be envisioned paradigm. Here, the authors review the current state of AI as applied to medical imaging of cancer and describe advances in 4 tumor types (lung, brain, breast, and prostate) to illustrate how common clinical problems are being addressed. Although most studies evaluating AI applications in oncology to date have not been vigorously validated for reproducibility and generalizability, the results do highlight increasingly concerted efforts in pushing AI technology to clinical use and to impact future directions in cancer care

    AI in Medical Imaging Informatics: Current Challenges and Future Directions

    Get PDF
    This paper reviews state-of-the-art research solutions across the spectrum of medical imaging informatics, discusses clinical translation, and provides future directions for advancing clinical practice. More specifically, it summarizes advances in medical imaging acquisition technologies for different modalities, highlighting the necessity for efficient medical data management strategies in the context of AI in big healthcare data analytics. It then provides a synopsis of contemporary and emerging algorithmic methods for disease classification and organ/ tissue segmentation, focusing on AI and deep learning architectures that have already become the de facto approach. The clinical benefits of in-silico modelling advances linked with evolving 3D reconstruction and visualization applications are further documented. Concluding, integrative analytics approaches driven by associate research branches highlighted in this study promise to revolutionize imaging informatics as known today across the healthcare continuum for both radiology and digital pathology applications. The latter, is projected to enable informed, more accurate diagnosis, timely prognosis, and effective treatment planning, underpinning precision medicine

    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

    Deep Domain Adaptation Learning Framework for Associating Image Features to Tumour Gene Profile

    Get PDF
    While medical imaging and general pathology are routine in cancer diagnosis, genetic sequencing is not always assessable due to the strong phenotypic and genetic heterogeneity of human cancers. Image-genomics integrates medical imaging and genetics to provide a complementary approach to optimise cancer diagnosis by associating tumour imaging traits with clinical data and has demonstrated its potential in identifying imaging surrogates for tumour biomarkers. However, existing image-genomics research has focused on quantifying tumour visual traits according to human understanding, which may not be optimal across different cancer types. The challenge hence lies in the extraction of optimised imaging representations in an objective data-driven manner. Such an approach requires large volumes of annotated image data that are difficult to acquire. We propose a deep domain adaptation learning framework for associating image features to tumour genetic information, exploiting the ability of domain adaptation technique to learn relevant image features from close knowledge domains. Our proposed framework leverages the current state-of-the-art in image object recognition to provide image features to encode subtle variations of tumour phenotypic characteristics with domain adaptation techniques. The proposed framework was evaluated with current state-of-the-art in: (i) tumour histopathology image classification and; (ii) image-genomics associations. The proposed framework demonstrated improved accuracy of tumour classification, as well as providing additional data-derived representations of tumour phenotypic characteristics that exhibit strong image-genomics association. This thesis advances and indicates the potential of image-genomics research to reveal additional imaging surrogates to genetic biomarkers, which has the potential to facilitate cancer diagnosis

    The use of machine learning/deep learning in PET/CT interpretation to aid in outcome prediction in lymphoma

    Get PDF
    Lymphoma is a haematopoietic malignancy consisting of two broad categories: Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL). These categories can be further split into subtypes with classical HL (cHL) and diffuse large B cell lymphoma (DLBCL) being the commonest subtypes. The gold standard imaging modality for staging and response assessment for cHL and DLBCL is 2-deoxy-2-[fluorine-18]fluoro-D-glucose (FDG) positron emission tomography/computed tomography (PET/CT), with patients having a worse prognosis if they do not demonstrate complete metabolic response (CMR). However, approximately 15% of patients will relapse even after CMR. Therefore, being able to identify patients who are likely to relapse it may be possible to stratify treatment early to improve patient outcomes. The aim of this project is to develop and test image derived predictive models based on the baseline PET/CT to risk stratify patients pre-treatment

    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

    Computational Pathology: A Survey Review and The Way Forward

    Full text link
    Computational Pathology CPath is an interdisciplinary science that augments developments of computational approaches to analyze and model medical histopathology images. The main objective for CPath is to develop infrastructure and workflows of digital diagnostics as an assistive CAD system for clinical pathology, facilitating transformational changes in the diagnosis and treatment of cancer that are mainly address by CPath tools. With evergrowing developments in deep learning and computer vision algorithms, and the ease of the data flow from digital pathology, currently CPath is witnessing a paradigm shift. Despite the sheer volume of engineering and scientific works being introduced for cancer image analysis, there is still a considerable gap of adopting and integrating these algorithms in clinical practice. This raises a significant question regarding the direction and trends that are undertaken in CPath. In this article we provide a comprehensive review of more than 800 papers to address the challenges faced in problem design all-the-way to the application and implementation viewpoints. We have catalogued each paper into a model-card by examining the key works and challenges faced to layout the current landscape in CPath. We hope this helps the community to locate relevant works and facilitate understanding of the field's future directions. In a nutshell, we oversee the CPath developments in cycle of stages which are required to be cohesively linked together to address the challenges associated with such multidisciplinary science. We overview this cycle from different perspectives of data-centric, model-centric, and application-centric problems. We finally sketch remaining challenges and provide directions for future technical developments and clinical integration of CPath (https://github.com/AtlasAnalyticsLab/CPath_Survey).Comment: Accepted in Elsevier Journal of Pathology Informatics (JPI) 202

    A graph-based approach for the retrieval of multi-modality medical images

    Get PDF
    Medical imaging has revolutionised modern medicine and is now an integral aspect of diagnosis and patient monitoring. The development of new imaging devices for a wide variety of clinical cases has spurred an increase in the data volume acquired in hospitals. These large data collections offer opportunities for search-based applications in evidence-based diagnosis, education, and biomedical research. However, conventional search methods that operate upon manual annotations are not feasible for this data volume. Content-based image retrieval (CBIR) is an image search technique that uses automatically derived visual features as search criteria and has demonstrable clinical benefits. However, very few studies have investigated the CBIR of multi-modality medical images, which are making a monumental impact in healthcare, e.g., combined positron emission tomography and computed tomography (PET-CT) for cancer diagnosis. In this thesis, we propose a new graph-based method for the CBIR of multi-modality medical images. We derive a graph representation that emphasises the spatial relationships between modalities by structurally constraining the graph based on image features, e.g., spatial proximity of tumours and organs. We also introduce a graph similarity calculation algorithm that prioritises the relationships between tumours and related organs. To enable effective human interpretation of retrieved multi-modality images, we also present a user interface that displays graph abstractions alongside complex multi-modality images. Our results demonstrated that our method achieved a high precision when retrieving images on the basis of tumour location within organs. The evaluation of our proposed UI design by user surveys revealed that it improved the ability of users to interpret and understand the similarity between retrieved PET-CT images. The work in this thesis advances the state-of-the-art by enabling a novel approach for the retrieval of multi-modality medical images

    Automatic Population of Structured Reports from Narrative Pathology Reports

    Get PDF
    There are a number of advantages for the use of structured pathology reports: they can ensure the accuracy and completeness of pathology reporting; it is easier for the referring doctors to glean pertinent information from them. The goal of this thesis is to extract pertinent information from free-text pathology reports and automatically populate structured reports for cancer diseases and identify the commonalities and differences in processing principles to obtain maximum accuracy. Three pathology corpora were annotated with entities and relationships between the entities in this study, namely the melanoma corpus, the colorectal cancer corpus and the lymphoma corpus. A supervised machine-learning based-approach, utilising conditional random fields learners, was developed to recognise medical entities from the corpora. By feature engineering, the best feature configurations were attained, which boosted the F-scores significantly from 4.2% to 6.8% on the training sets. Without proper negation and uncertainty detection, the quality of the structured reports will be diminished. The negation and uncertainty detection modules were built to handle this problem. The modules obtained overall F-scores ranging from 76.6% to 91.0% on the test sets. A relation extraction system was presented to extract four relations from the lymphoma corpus. The system achieved very good performance on the training set, with 100% F-score obtained by the rule-based module and 97.2% F-score attained by the support vector machines classifier. Rule-based approaches were used to generate the structured outputs and populate them to predefined templates. The rule-based system attained over 97% F-scores on the training sets. A pipeline system was implemented with an assembly of all the components described above. It achieved promising results in the end-to-end evaluations, with 86.5%, 84.2% and 78.9% F-scores on the melanoma, colorectal cancer and lymphoma test sets respectively
    • 

    corecore