497 research outputs found

    Bioinformatics analysis identifies key genes of prognostic value in lung cancer

    Get PDF
    Lung cancer is the most common human malignancy worldwide and can be divided into different types of carcinomas depending on their pathological features. Advances in medical science and technology have led to the identification of some lung cancer-related marker genes, including EGFR (epidermal growth factor receptor), BRAF (B-Raf proto-oncogene), RAS (RAS proto-oncogene, GTPase) and HER2 (human epidermal growth factor receptor 2). However, the underlying biomarker and key genes associated with different types of lung cancer are still poorly understood. In this study, we analyzed a GEO (Gene Expression Omnibus) dataset and identified 28 upregulated intersection DEGs (different expression genes) and 125 downregulated intersection DEGs among AC (adenocarcinoma), PTC (primary typical carcinoid), PLCC (primary large cell carcinoma), PLCNC (primary large cell lung carcinoma) and PSCLC (primary small cell lung carcinoma). Through PPI (protein-protein interaction) network analysis, we identified 14 genes among the DEGs, namely MFAP4 (microfibril-associated protein 4), PDZD2 (PDZ domain containing 2), FBLN1 (fibulin 1), FBLN5 (fibulin 5), EFEMP1 (EGF containing fibulin extracellular matrix protein 1), KDR (kinase insert domain receptor), S1PR1 (sphingosine-1-phosphate receptor 1), CAV1 (caveolin 1), GRK5 (G protein-coupled receptor kinase 5), EDNRA (endothelin receptor type A), EDNRB (endothelin receptor type B), CALCRL (calcitonin receptor-like receptor), PTGER4 (prostaglandin E receptor 4), and ADRB1 (adrenoceptor beta 1), which were found to be downregulated in different subtypes of lung cancer and associated with poor survival outcomes. In addition, most of the screened DEGs demonstrated good predictive ability in LUAD (lung adenocarcinoma) and LUSC (lung squamous cell carcinoma). Among them, MFAP4 was found to promote cell proliferation while also suppressing cell migration and angiogenesis. In summary, we propose MFAP4, PDZD2, FBLN1, FBLN5, EFEMP1, KDR, S1PR1, CAV1, GRK5, EDNRA, EDNRB, CALCRL, PTGER4 and ADRB1 as potential prognostic markers in lung cancer patients

    SITC cancer immunotherapy resource document: a compass in the land of biomarker discovery.

    Get PDF
    Since the publication of the Society for Immunotherapy of Cancer\u27s (SITC) original cancer immunotherapy biomarkers resource document, there have been remarkable breakthroughs in cancer immunotherapy, in particular the development and approval of immune checkpoint inhibitors, engineered cellular therapies, and tumor vaccines to unleash antitumor immune activity. The most notable feature of these breakthroughs is the achievement of durable clinical responses in some patients, enabling long-term survival. These durable responses have been noted in tumor types that were not previously considered immunotherapy-sensitive, suggesting that all patients with cancer may have the potential to benefit from immunotherapy. However, a persistent challenge in the field is the fact that only a minority of patients respond to immunotherapy, especially those therapies that rely on endogenous immune activation such as checkpoint inhibitors and vaccination due to the complex and heterogeneous immune escape mechanisms which can develop in each patient. Therefore, the development of robust biomarkers for each immunotherapy strategy, enabling rational patient selection and the design of precise combination therapies, is key for the continued success and improvement of immunotherapy. In this document, we summarize and update established biomarkers, guidelines, and regulatory considerations for clinical immune biomarker development, discuss well-known and novel technologies for biomarker discovery and validation, and provide tools and resources that can be used by the biomarker research community to facilitate the continued development of immuno-oncology and aid in the goal of durable responses in all patients

    The Era of Next-Generation Sequencing in Clinical Oncology

    Get PDF

    The Era of Next-Generation Sequencing in Clinical Oncology

    Get PDF

    Network-based methods for biological data integration in precision medicine

    Full text link
    [eng] The vast and continuously increasing volume of available biomedical data produced during the last decades opens new opportunities for large-scale modeling of disease biology, facilitating a more comprehensive and integrative understanding of its processes. Nevertheless, this type of modelling requires highly efficient computational systems capable of dealing with such levels of data volumes. Computational approximations commonly used in machine learning and data analysis, namely dimensionality reduction and network-based approaches, have been developed with the goal of effectively integrating biomedical data. Among these methods, network-based machine learning stands out due to its major advantage in terms of biomedical interpretability. These methodologies provide a highly intuitive framework for the integration and modelling of biological processes. This PhD thesis aims to explore the potential of integration of complementary available biomedical knowledge with patient-specific data to provide novel computational approaches to solve biomedical scenarios characterized by data scarcity. The primary focus is on studying how high-order graph analysis (i.e., community detection in multiplex and multilayer networks) may help elucidate the interplay of different types of data in contexts where statistical power is heavily impacted by small sample sizes, such as rare diseases and precision oncology. The central focus of this thesis is to illustrate how network biology, among the several data integration approaches with the potential to achieve this task, can play a pivotal role in addressing this challenge provided its advantages in molecular interpretability. Through its insights and methodologies, it introduces how network biology, and in particular, models based on multilayer networks, facilitates bringing the vision of precision medicine to these complex scenarios, providing a natural approach for the discovery of new biomedical relationships that overcomes the difficulties for the study of cohorts presenting limited sample sizes (data-scarce scenarios). Delving into the potential of current artificial intelligence (AI) and network biology applications to address data granularity issues in the precision medicine field, this PhD thesis presents pivotal research works, based on multilayer networks, for the analysis of two rare disease scenarios with specific data granularities, effectively overcoming the classical constraints hindering rare disease and precision oncology research. The first research article presents a personalized medicine study of the molecular determinants of severity in congenital myasthenic syndromes (CMS), a group of rare disorders of the neuromuscular junction (NMJ). The analysis of severity in rare diseases, despite its importance, is typically neglected due to data availability. In this study, modelling of biomedical knowledge via multilayer networks allowed understanding the functional implications of individual mutations in the cohort under study, as well as their relationships with the causal mutations of the disease and the different levels of severity observed. Moreover, the study presents experimental evidence of the role of a previously unsuspected gene in NMJ activity, validating the hypothetical role predicted using the newly introduced methodologies. The second research article focuses on the applicability of multilayer networks for gene priorization. Enhancing concepts for the analysis of different data granularities firstly introduced in the previous article, the presented research provides a methodology based on the persistency of network community structures in a range of modularity resolution, effectively providing a new framework for gene priorization for patient stratification. In summary, this PhD thesis presents major advances on the use of multilayer network-based approaches for the application of precision medicine to data-scarce scenarios, exploring the potential of integrating extensive available biomedical knowledge with patient-specific data

    Computational Methods for the Analysis of Genomic Data and Biological Processes

    Get PDF
    In recent decades, new technologies have made remarkable progress in helping to understand biological systems. Rapid advances in genomic profiling techniques such as microarrays or high-performance sequencing have brought new opportunities and challenges in the fields of computational biology and bioinformatics. Such genetic sequencing techniques allow large amounts of data to be produced, whose analysis and cross-integration could provide a complete view of organisms. As a result, it is necessary to develop new techniques and algorithms that carry out an analysis of these data with reliability and efficiency. This Special Issue collected the latest advances in the field of computational methods for the analysis of gene expression data, and, in particular, the modeling of biological processes. Here we present eleven works selected to be published in this Special Issue due to their interest, quality, and originality

    Multi-Modality Automatic Lung Tumor Segmentation Method Using Deep Learning and Radiomics

    Get PDF
    Delineation of the tumor volume is the initial and fundamental step in the radiotherapy planning process. The current clinical practice of manual delineation is time-consuming and suffers from observer variability. This work seeks to develop an effective automatic framework to produce clinically usable lung tumor segmentations. First, to facilitate the development and validation of our methodology, an expansive database of planning CTs, diagnostic PETs, and manual tumor segmentations was curated, and an image registration and preprocessing pipeline was established. Then a deep learning neural network was constructed and optimized to utilize dual-modality PET and CT images for lung tumor segmentation. The feasibility of incorporating radiomics and other mechanisms such as a tumor volume-based stratification scheme for training/validation/testing were investigated to improve the segmentation performance. The proposed methodology was evaluated both quantitatively with similarity metrics and clinically with physician reviews. In addition, external validation with an independent database was also conducted. Our work addressed some of the major limitations that restricted clinical applicability of the existing approaches and produced automatic segmentations that were consistent with the manually contoured ground truth and were highly clinically-acceptable according to both the quantitative and clinical evaluations. Both novel approaches of implementing a tumor volume-based training/validation/ testing stratification strategy as well as incorporating voxel-wise radiomics feature images were shown to improve the segmentation performance. The results showed that the proposed method was effective and robust, producing automatic lung tumor segmentations that could potentially improve both the quality and consistency of manual tumor delineation

    Artificial Intelligence in Oncology Drug Discovery and Development

    Get PDF
    There exists a profound conflict at the heart of oncology drug development. The efficiency of the drug development process is falling, leading to higher costs per approved drug, at the same time personalised medicine is limiting the target market of each new medicine. Even as the global economic burden of cancer increases, the current paradigm in drug development is unsustainable. In this book, we discuss the development of techniques in machine learning for improving the efficiency of oncology drug development and delivering cost-effective precision treatment. We consider how to structure data for drug repurposing and target identification, how to improve clinical trials and how patients may view artificial intelligence

    Bioinformatics and Machine Learning for Cancer Biology

    Get PDF
    Cancer is a leading cause of death worldwide, claiming millions of lives each year. Cancer biology is an essential research field to understand how cancer develops, evolves, and responds to therapy. By taking advantage of a series of “omics” technologies (e.g., genomics, transcriptomics, and epigenomics), computational methods in bioinformatics and machine learning can help scientists and researchers to decipher the complexity of cancer heterogeneity, tumorigenesis, and anticancer drug discovery. Particularly, bioinformatics enables the systematic interrogation and analysis of cancer from various perspectives, including genetics, epigenetics, signaling networks, cellular behavior, clinical manifestation, and epidemiology. Moreover, thanks to the influx of next-generation sequencing (NGS) data in the postgenomic era and multiple landmark cancer-focused projects, such as The Cancer Genome Atlas (TCGA) and Clinical Proteomic Tumor Analysis Consortium (CPTAC), machine learning has a uniquely advantageous role in boosting data-driven cancer research and unraveling novel methods for the prognosis, prediction, and treatment of cancer
    • 

    corecore