40 research outputs found

    Characterization of inter- and intratumoral heterogeneity and the differential immune microenvironment during malignant progression in meningiomas

    Get PDF
    Meningiomas are thought to arise from the arachnoid cells of the leptomeninx and make up the most common primary intracranial tumor in adults. They are usually benign, however in about 20 % of cases, tumors present with an aggressive phenotype and higher risk of recurrence. Risk stratification thereby remains challenging especially for NF2-mutated meningiomas, which make up about two thirds of all cases, as they can occur at the full spectrum of WHO grades in meningioma from 1 to 3. Recently, molecular profiling has gained importance for prognosis in meningioma with several classification systems that have been established mostly based on the DNA methylation of the tumors. However, the DNA methylation-based classification has not been extensively linked to phenotypic traits of the tumor. Nor have meningiomas been investigated regarding intratumoral subpopulations that may exist in parallel and may have different characteristics, especially regarding the stage of progression and ability to recur. In addition, the role of the immune microenvironment in meningiomas is poorly understood, despite the identification of an immune-enriched meningioma subgroup with beneficial outcome in two independent DNA methylation classification systems. In this dissertation, I investigated the consistency of subgroups initially defined on epigenomic level across molecular levels by comparison to transcriptomic and proteomic data. Further, I leveraged single nuclei transcriptomic profiling to dissect intertumoral differences in the expression profile specific to the tumor cell population depending on the tumor subgroup, and to investigate the abundance and phenotype of intratumoral tumor cell subpopulations across samples. Similarly, I analyzed the single nuclei transcriptomic data to characterize tumor-infiltrating immune cells with respect to their abundance and activation status. I furthermore correlated the differences in immune infiltration with the progression-free survival of patients by deconvoluting DNA methylation array data according to their cellular composition. These analyses underlined the coherence of epigenomic meningioma subgroups across transcriptome and proteome. Moreover, I identified six tumor cell subpopulations that were defined by distinct expression profiles und could be identified across samples at varying abundancies depending on the stage of progression. Similarly, I observed profound differences in infiltrating immune cells between tumor subgroups, with a significant enrichment of tumor-associated macrophages in a benign subgroup of NF2-mutated meningiomas as compared to more progressed tumors. In parallel to their abundancy, macrophages changed in activation between benign and malignant cases from an anti- to a pro-tumorigenic phenotype. The evaluation of progression-free survival data revealed a positive correlation to the proportion of infiltrating immune cells as estimated from epigenomic profiles. Altogether, these results highlight the role of multi-level molecular profiling for tumor grading in a paradigmatic, epidemiologically relevant tumor type. They further indicate an important role of tumor-infiltrating macrophages during meningioma progression with possible consequences for risk prediction as well as therapeutic targets in meningioma

    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

    Artificial Intelligence in Brain Tumour Surgery—An Emerging Paradigm

    Get PDF
    Artificial intelligence (AI) platforms have the potential to cause a paradigm shift in brain tumour surgery. Brain tumour surgery augmented with AI can result in safer and more effective treatment. In this review article, we explore the current and future role of AI in patients undergoing brain tumour surgery, including aiding diagnosis, optimising the surgical plan, providing support during the operation, and better predicting the prognosis. Finally, we discuss barriers to the successful clinical implementation, the ethical concerns, and we provide our perspective on how the field could be advanced

    Machine Learning Models for Deciphering Regulatory Mechanisms and Morphological Variations in Cancer

    Get PDF
    The exponential growth of multi-omics biological datasets is resulting in an emerging paradigm shift in fundamental biological research. In recent years, imaging and transcriptomics datasets are increasingly incorporated into biological studies, pushing biology further into the domain of data-intensive-sciences. New approaches and tools from statistics, computer science, and data engineering are profoundly influencing biological research. Harnessing this ever-growing deluge of multi-omics biological data requires the development of novel and creative computational approaches. In parallel, fundamental research in data sciences and Artificial Intelligence (AI) has advanced tremendously, allowing the scientific community to generate a massive amount of knowledge from data. Advances in Deep Learning (DL), in particular, are transforming many branches of engineering, science, and technology. Several of these methodologies have already been adapted for harnessing biological datasets; however, there is still a need to further adapt and tailor these techniques to new and emerging technologies. In this dissertation, we present computational algorithms and tools that we have developed to study gene-regulation and cellular morphology in cancer. The models and platforms that we have developed are general and widely applicable to several problems relating to dysregulation of gene expression in diseases. Our pipelines and software packages are disseminated in public repositories for larger scientific community use. This dissertation is organized in three main projects. In the first project, we present Causal Inference Engine (CIE), an integrated platform for the identification and interpretation of active regulators of transcriptional response. The platform offers visualization tools and pathway enrichment analysis to map predicted regulators to Reactome pathways. We provide a parallelized R-package for fast and flexible directional enrichment analysis to run the inference on custom regulatory networks. Next, we designed and developed MODEX, a fully automated text-mining system to extract and annotate causal regulatory interaction between Transcription Factors (TFs) and genes from the biomedical literature. MODEX uses putative TF-gene interactions derived from high-throughput ChIP-Seq or other experiments and seeks to collect evidence and meta-data in the biomedical literature to validate and annotate the interactions. MODEX is a complementary platform to CIE that provides auxiliary information on CIE inferred interactions by mining the literature. In the second project, we present a Convolutional Neural Network (CNN) classifier to perform a pan-cancer analysis of tumor morphology, and predict mutations in key genes. The main challenges were to determine morphological features underlying a genetic status and assess whether these features were common in other cancer types. We trained an Inception-v3 based model to predict TP53 mutation in five cancer types with the highest rate of TP53 mutations. We also performed a cross-classification analysis to assess shared morphological features across multiple cancer types. Further, we applied a similar methodology to classify HER2 status in breast cancer and predict response to treatment in HER2 positive samples. For this study, our training slides were manually annotated by expert pathologists to highlight Regions of Interest (ROIs) associated with HER2+/- tumor microenvironment. Our results indicated that there are strong morphological features associated with each tumor type. Moreover, our predictions highly agree with manual annotations in the test set, indicating the feasibility of our approach in devising an image-based diagnostic tool for HER2 status and treatment response prediction. We have validated our model using samples from an independent cohort, which demonstrates the generalizability of our approach. Finally, in the third project, we present an approach to use spatial transcriptomics data to predict spatially-resolved active gene regulatory mechanisms in tissues. Using spatial transcriptomics, we identified tissue regions with differentially expressed genes and applied our CIE methodology to predict active TFs that can potentially regulate the marker genes in the region. This project bridged the gap between inference of active regulators using molecular data and morphological studies using images. The results demonstrate a significant local pattern in TF activity across the tissue, indicating differential spatial-regulation in tissues. The results suggest that the integrative analysis of spatial transcriptomics data with CIE can capture discriminant features and identify localized TF-target links in the tissue

    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

    Advanced Computational Methods for Oncological Image Analysis

    Get PDF
    [Cancer is the second most common cause of death worldwide and encompasses highly variable clinical and biological scenarios. Some of the current clinical challenges are (i) early diagnosis of the disease and (ii) precision medicine, which allows for treatments targeted to specific clinical cases. The ultimate goal is to optimize the clinical workflow by combining accurate diagnosis with the most suitable therapies. Toward this, large-scale machine learning research can define associations among clinical, imaging, and multi-omics studies, making it possible to provide reliable diagnostic and prognostic biomarkers for precision oncology. Such reliable computer-assisted methods (i.e., artificial intelligence) together with clinicians’ unique knowledge can be used to properly handle typical issues in evaluation/quantification procedures (i.e., operator dependence and time-consuming tasks). These technical advances can significantly improve result repeatability in disease diagnosis and guide toward appropriate cancer care. Indeed, the need to apply machine learning and computational intelligence techniques has steadily increased to effectively perform image processing operations—such as segmentation, co-registration, classification, and dimensionality reduction—and multi-omics data integration.
    corecore