1,443 research outputs found

    INTEGRATIVE ANALYSIS OF OMICS DATA IN ADULT GLIOMA AND OTHER TCGA CANCERS TO GUIDE PRECISION MEDICINE

    Get PDF
    Transcriptomic profiling and gene expression signatures have been widely applied as effective approaches for enhancing the molecular classification, diagnosis, prognosis or prediction of therapeutic response towards personalized therapy for cancer patients. Thanks to modern genome-wide profiling technology, scientists are able to build engines leveraging massive genomic variations and integrating with clinical data to identify “at risk” individuals for the sake of prevention, diagnosis and therapeutic interventions. In my graduate work for my Ph.D. thesis, I have investigated genomic sequencing data mining to comprehensively characterise molecular classifications and aberrant genomic events associated with clinical prognosis and treatment response, through applying high-dimensional omics genomic data to promote the understanding of gene signatures and somatic molecular alterations contributing to cancer progression and clinical outcomes. Following this motivation, my dissertation has been focused on the following three topics in translational genomics. 1) Characterization of transcriptomic plasticity and its association with the tumor microenvironment in glioblastoma (GBM). I have integrated transcriptomic, genomic, protein and clinical data to increase the accuracy of GBM classification, and identify the association between the GBM mesenchymal subtype and reduced tumorpurity, accompanied with increased presence of tumor-associated microglia. Then I have tackled the sole source of microglial as intrinsic tumor bulk but not their corresponding neurosphere cells through both transcriptional and protein level analysis using a panel of sphere-forming glioma cultures and their parent GBM samples.FurthermoreI have demonstrated my hypothesis through longitudinal analysis of paired primary and recurrent GBM samples that the phenotypic alterations of GBM subtypes are not due to intrinsic proneural-to-mesenchymal transition in tumor cells, rather it is intertwined with increased level of microglia upon disease recurrence. Collectively I have elucidated the critical role of tumor microenvironment (Microglia and macrophages from central nervous system) contributing to the intra-tumor heterogeneity and accurate classification of GBM patients based on transcriptomic profiling, which will not only significantly impact on clinical perspective but also pave the way for preclinical cancer research. 2) Identification of prognostic gene signatures that stratify adult diffuse glioma patientsharboring1p/19q co-deletions. I have compared multiple statistical methods and derived a gene signature significantly associated with survival by applying a machine learning algorithm. Then I have identified inflammatory response and acetylation activity that associated with malignant progression of 1p/19q co-deleted glioma. In addition, I showed this signature translates to other types of adult diffuse glioma, suggesting its universality in the pathobiology of other subset gliomas. My efforts on integrative data analysis of this highly curated data set usingoptimizedstatistical models will reflect the pending update to WHO classification system oftumorsin the central nervous system (CNS). 3) Comprehensive characterization of somatic fusion transcripts in Pan-Cancers. I have identified a panel of novel fusion transcripts across all of TCGA cancer types through transcriptomic profiling. Then I have predicted fusion proteins with kinase activity and hub function of pathway network based on the annotation of genetically mobile domains and functional domain architectures. I have evaluated a panel of in -frame gene fusions as potential driver mutations based on network fusion centrality hypothesis. I have also characterised the emerging complexity of genetic architecture in fusion transcripts through integrating genomic structure and somatic variants and delineating the distinct genomic patterns of fusion events across different cancer types. Overall my exploration of the pathogenetic impact and clinical relevance of candidate gene fusions have provided fundamental insights into the management of a subset of cancer patients by predicting the oncogenic signalling and specific drug targets encoded by these fusion genes. Taken together, the translational genomic research I have conducted during my Ph.D. study will shed new light on precision medicine and contribute to the cancer research community. The novel classification concept, gene signature and fusion transcripts I have identified will address several hotly debated issues in translational genomics, such as complex interactions between tumor bulks and their adjacent microenvironments, prognostic markers for clinical diagnostics and personalized therapy, distinct patterns of genomic structure alterations and oncogenic events in different cancer types, therefore facilitating our understanding of genomic alterations and moving us towards the development of precision medicine

    Computational Hybrid Systems for Identifying Prognostic Gene Markers of Lung Cancer

    Get PDF
    Lung cancer is the most fatal cancer around the world. Current lung cancer prognosis and treatment is based on tumor stage population statistics and could not reliably assess the risk for developing recurrence in individual patients. Biomarkers enable treatment options to be tailored to individual patients based on their tumor molecular characteristics. To date, there is no clinically applied molecular prognostic model for lung cancer. Statistics and feature selection methods identify gene candidates by ranking the association between gene expression and disease outcome, but do not account for the interactions among genes. Computational network methods could model interactions, but have not been used for gene selection due to computational inefficiency. Moreover, the curse of dimensionality in human genome data imposes more computational challenges to these methods.;We proposed two hybrid systems for the identification of prognostic gene signatures for lung cancer using gene expressions measured with DNA microarray. The first hybrid system combined t-tests, Statistical Analysis of Microarray (SAM), and Relief feature selections in multiple gene filtering layers. This combinatorial system identified a 12-gene signature with better prognostic performance than published signatures in treatment selection for stage I and II patients (log-rank P\u3c0.04, Kaplan-Meier analyses). The 12-gene signature is a more significant prognostic factor (hazard ratio=4.19, 95% CI: [2.08, 8.46], P\u3c0.00006) than other clinical covariates. The signature genes were found to be involved in tumorigenesis in functional pathway analyses.;The second proposed system employed a novel computational network model, i.e., implication networks based on prediction logic. This network-based system utilizes gene coexpression networks and concurrent coregulation with signaling pathways for biomarker identification. The first application of the system modeled disease-mediated genome-wide coexpression networks. The entire genomic space were extensively explored and 21 gene signatures were discovered with better prognostic performance than all published signatures in stage I patients not receiving chemotherapy (hazard ratio\u3e1, CPE\u3e0.5, P \u3c 0.05). These signatures could potentially be used for selecting patients for adjuvant chemotherapy. The second application of the system modeled the smoking-mediated coexpression networks and identified a smoking-associated 7-gene signature. The 7-gene signature generated significant prognostication specific to smoking lung cancer patients (log-rank P\u3c0.05, Kaplan-Meier analyses), with implications in diagnostic screening of lung cancer risk in smokers (overall accuracy=74%, P\u3c0.006). The coexpression patterns derived from the implication networks in both applications were successfully validated with molecular interactions reported in the literature (FDR\u3c0.1).;Our studies demonstrated that hybrid systems with multiple gene selection layers outperform traditional methods. Moreover, implication networks could efficiently model genome-scale disease-mediated coexpression networks and crosstalk with signaling pathways, leading to the identification of clinically important gene signatures

    Data mining of gene arrays for biomarkers of survival in ovarian cancer

    Get PDF
    The expected five-year survival rate from a stage III ovarian cancer diagnosis is a mere 22%; this applies to the 7000 new cases diagnosed yearly in the UK. Stratification of patients with this heterogeneous disease, based on active molecular pathways, would aid a targeted treatment improving the prognosis for many cases. While hundreds of genes have been associated with ovarian cancer, few have yet been verified by peer research for clinical significance. Here, a meta-analysis approach was applied to two care fully selected gene expression microarray datasets. Artificial neural networks, Cox univariate survival analyses and T-tests identified genes whose expression was consistently and significantly associated with patient survival. The rigor of this experimental design increases confidence in the genes found to be of interest. A list of 56 genes were distilled from a potential 37,000 to be significantly related to survival in both datasets with a FDR of 1.39859 × 10−11, the identities of which both verify genes already implicated with this disease and provide novel genes and pathways to pursue. Further investigation and validation of these may lead to clinical insights and have potential to predict a patient’s response to treatment or be used as a novel target for therapy

    The Era of Radiogenomics in Precision Medicine: An Emerging Approach to Support Diagnosis, Treatment Decisions, and Prognostication in Oncology

    Get PDF
    With the rapid development of new technologies, including artificial intelligence and genome sequencing, radiogenomics has emerged as a state-of-the-art science in the field of individualized medicine. Radiogenomics combines a large volume of quantitative data extracted from medical images with individual genomic phenotypes and constructs a prediction model through deep learning to stratify patients, guide therapeutic strategies, and evaluate clinical outcomes. Recent studies of various types of tumors demonstrate the predictive value of radiogenomics. And some of the issues in the radiogenomic analysis and the solutions from prior works are presented. Although the workflow criteria and international agreed guidelines for statistical methods need to be confirmed, radiogenomics represents a repeatable and cost-effective approach for the detection of continuous changes and is a promising surrogate for invasive interventions. Therefore, radiogenomics could facilitate computer-aided diagnosis, treatment, and prediction of the prognosis in patients with tumors in the routine clinical setting. Here, we summarize the integrated process of radiogenomics and introduce the crucial strategies and statistical algorithms involved in current studies

    Integrative Transcriptomic Analysis of Long Intergenic Non-Coding RNAs in Cancer.

    Get PDF
    Ph.D. Thesis. University of Hawaiʻi at Mānoa 2017

    Genet-CNV: Boolean Implication Networks for Modeling Genome-Wide Co-occurrence of DNA Copy Number Variations

    Get PDF
    Lung cancer is the leading cause of cancer-related death in the world. Lung cancer can be categorized as non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). NSCLC makes up about 80% to 85% of lung cancer cases diagnosed, whereas SCLC is responsible for 10% to 15% of the cases. It remains a challenge for physicians to identify patients who shall benefit from chemotherapy. In such a scenario, identifying genes that can facilitate therapeutic target discoveries and better understanding disease mechanisms and their regulation in different stages of lung cancer, remains an important topic of research. In this thesis, we develop a computational framework for modelling molecular gene interaction networks, called Genet-CNV, to analyse gene interactions based on DNA Copy Number Variations (CNV). DNA copy number variation is a phenomenon in which sections of the genome are repeated and the number of repeats in the genome varies between individuals in the human population. These variations can be used to study the activity of genes in cancerous cells, compared with that of the normal population. Genet-CNV uses Boolean implication networks to investigate genome-wide DNA CNV to identify relationships called rules, that could potentially lead to the identification of genes of significant biological interest. Boolean implication networks are probabilistic graphical models that express the relationship between two variables terms of six implication rules that can describe if the genes are co-amplified, co-deleted or differentially amplified and deleted. Genet-CNV is run on three publicly available NSCLC genomic datasets. We further evaluate the results obtained with Genet-CNV by comparing them with the benchmark dataset, The Molecular Signatures Database (MSigDB). We identified several genes of interest that are present in survival, apoptosis, proliferation and immunologic pathways. The relationships obtained from this analysis can be tested for biological validations, or to confirm experimental results, thus facilitating the identification of genes playing a significant role in the causation and progress of NSCLC

    Pancreatic Cancer - Early Detection, Prognostic Factors, and Treatment

    Get PDF
    Background: Pancreatic cancer is the fourth leading cause of cancer-related death. Only about 6% of patients are alive 5 years after diagnosis. One reason for this low survival rate is that most patients are diagnosed at a late stage, when the tumor has spread to surrounding tissues or distant organs. Less than 20% of cases are diagnosed at an early stage that allows them to undergo potentially curative surgery. However, even for patients with a tumor that has been surgically removed, local and systemic recurrence is common and the median survival is only 17-23 months. This underscores the importance to identify factors that can predict postresection survival. With technical advances and centralization of care, pancreatic surgery has become a safe procedure. The future optimal treatment for pancreatic cancer is dependent on increased understanding of tumor biology and development of individualized and systemic treatment. Previous experimental studies have reported that mucins, especially the MUC4 mucin, may confer resistance to the chemotherapeutic agent gemcitabine and may serve as targets for the development of novel types of intervention. Aim: The aim of the thesis was to investigate strategies to improve management of pancreatic cancer, with special reference to early detection, prognostic factors, and treatment. Methods: In paper I, 27 prospectively collected serum samples from resectable pancreatic cancer (n=9), benign pancreatic disease (n=9), and healthy controls (n=9) were analyzed by high definition mass spectrometry (HDMSE). In paper II, an artificial neural network (ANN) model was constructed on 84 pancreatic cancer patients undergoing surgical resection. In paper III, we investigated the effects of transition from a low- to a high volume-center for pancreaticoduodenectomy in 221 patients. In paper IV, the grade of concordance in terms of MUC4 expression was examined in 17 tissue sections from primary pancreatic cancer and matched lymph node metastases. In paper V, pancreatic xenograft tumors were generated in 15 immunodeficient mice by subcutaneous injection of MUC4+ human pancreatic cancer cell lines; Capan-1, HPAF-II, or CD18/HPAF. In paper VI, a 76-member combined epigenetics and phosphatase small-molecule inhibitor library was screened against Capan-1 (MUC4+) and Panc-1 (MUC4-) cells, followed by high content screening of protein expression. Results/Conclusion: 134 differentially expressed serum proteins were identified, of which 40 proteins showed a significant up-regulation in the pancreatic cancer group. Pancreatic disease link associations could be made for BAZ2A, CDK13, DAPK1, DST, EXOSC3, INHBE, KAT2B, KIF20B, SMC1B, and SPAG5, by pathway network linkages to p53, the most frequently altered tumor suppressor in pancreatic cancer (I). An ANN survival model was developed, identifying 7 risk factors. The C-index for the model was 0.79, and it performed significantly better than the Cox regression (II). We experienced improved surgical results for pancreaticoduodenectomy after the transition to a high-volume center (≄25 procedures/year), including decreased operative duration, blood loss, hemorrhagic complications, reoperations, and hospital stay. There was also a tendency toward reduced operative mortality, from 4% to 0% (III). MUC4 positivity was detected in most primary pancreatic cancer tissues, as well as in matched metastatic lymph nodes (15/17 vs. 14/17), with a high concordance level (82%) (IV). The tumor incidence was 100% in the xenograft model. The median MUC4 count was found to be highest in Capan-1 tumors. α-SMA and collagen extent were also highest in Capan-1 tumors (V). Apicidin (a histone deacetylase inhibitor) had potent antiproliferative activity against Capan-1 cells and significantly reduced the expression of MUC4 and its transcription factor HNF4α. The combined treatment of apicidin and gemcitabine synergistically inhibited growth of Capan-1 cells (VI)

    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