2,363 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 ā€œat riskā€ 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

    Curation-free biomodules mechanisms in prostate cancer predict recurrent disease

    Full text link
    Abstract Motivation Gene expression-based prostate cancer gene signatures of poor prognosis are hampered by lack of gene feature reproducibility and a lack of understandability of their function. Molecular pathway-level mechanisms are intrinsically more stable and more robust than an individual gene. The Functional Analysis of Individual Microarray Expression (FAIME) we developed allows distinctive sample-level pathway measurements with utility for correlation with continuous phenotypes (e.g. survival). Further, we and others have previously demonstrated that pathway-level classifiers can be as accurate as gene-level classifiers using curated genesets that may implicitly comprise ascertainment biases (e.g. KEGG, GO). Here, we hypothesized that transformation of individual prostate cancer patient gene expression to pathway-level mechanisms derived from automated high throughput analyses of genomic datasets may also permit personalized pathway analysis and improve prognosis of recurrent disease. Results Via FAIME, three independent prostate gene expression arrays with both normal and tumor samples were transformed into two distinct types of molecular pathway mechanisms: (i) the curated Gene Ontology (GO) and (ii) dynamic expression activity networks of cancer (Cancer Modules). FAIME-derived mechanisms for tumorigenesis were then identified and compared. Curated GO and computationally generated "Cancer Module" mechanisms overlap significantly and are enriched for known oncogenic deregulations and highlight potential areas of investigation. We further show in two independent datasets that these pathway-level tumorigenesis mechanisms can identify men who are more likely to develop recurrent prostate cancer (log-rank_p = 0.019). Conclusion Curation-free biomodules classification derived from congruent gene expression activation breaks from the paradigm of recapitulating the known curated pathway mechanism universe.http://deepblue.lib.umich.edu/bitstream/2027.42/112452/1/12920_2013_Article_384.pd

    Advantages of genomic complexity: bioinformatics opportunities in microRNA cancer signatures

    Get PDF
    MicroRNAs, small non-coding RNAs, may act as tumor suppressors or oncogenes, and each regulate their own transcription and that of hundreds of genes, often in a tissue-dependent manner. This creates a tightly interwoven network regulating and underlying oncogenesis and cancer biology. Although protein-coding gene signatures and single protein pathway markers have proliferated over the past decade, routine adoption of the former has been hampered by interpretability, reproducibility, and dimensionality, whereas the single moleculeā€“phenotype reductionism of the latter is often overly simplistic to account for complex phenotypes. MicroRNA-derived biomarkers offer a powerful alternative; they have both the flexibility of gene expression signature classifiers and the desirable mechanistic transparency of single protein biomarkers. Furthermore, several advances have recently demonstrated the robust detection of microRNAs from various biofluids, thus providing an additional opportunity for obtaining bioinformatically derived biomarkers to accelerate the identification of individual patients for personalized therapy

    EPMA position paper in cancer:current overview and future perspectives

    Get PDF
    At present, a radical shift in cancer treatment is occurring in terms of predictive, preventive, and personalized medicine (PPPM). Individual patients will participate in more aspects of their healthcare. During the development of PPPM, many rapid, specific, and sensitive new methods for earlier detection of cancer will result in more efficient management of the patient and hence a better quality of life. Coordination of the various activities among different healthcare professionals in primary, secondary, and tertiary care requires well-defined competencies, implementation of training and educational programs, sharing of data, and harmonized guidelines. In this position paper, the current knowledge to understand cancer predisposition and risk factors, the cellular biology of cancer, predictive markers and treatment outcome, the improvement in technologies in screening and diagnosis, and provision of better drug development solutions are discussed in the context of a better implementation of personalized medicine. Recognition of the major risk factors for cancer initiation is the key for preventive strategies (EPMA J. 4(1):6, 2013). Of interest, cancer predisposing syndromes in particular the monogenic subtypes that lead to cancer progression are well defined and one should focus on implementation strategies to identify individuals at risk to allow preventive measures and early screening/diagnosis. Implementation of such measures is disturbed by improper use of the data, with breach of data protection as one of the risks to be heavily controlled. Population screening requires in depth cost-benefit analysis to justify healthcare costs, and the parameters screened should provide information that allow an actionable and deliverable solution, for better healthcare provision

    Integration of breast cancer gene signatures based on graph centrality

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Various gene-expression signatures for breast cancer are available for the prediction of clinical outcome. However due to small overlap between different signatures, it is challenging to integrate existing disjoint signatures to provide a unified insight on the association between gene expression and clinical outcome.</p> <p>Results</p> <p>In this paper, we propose a method to integrate different breast cancer gene signatures by using graph centrality in a context-constrained protein interaction network (PIN). The context-constrained PIN for breast cancer is built by integrating complete PIN and various gene signatures reported in literatures. Then, we use graph centralities to quantify the importance of genes to breast cancer. Finally, we get reliable gene signatures that are consisted by the genes with high graph centrality. The genes which are well-known breast cancer genes, such as TP53 and BRCA1, are ranked extremely high in our results. Compared with previous results by functional enrichment analysis, graph centralities, especially the eigenvector centrality and subgraph centrality, based gene signatures are more tightly related to breast cancer. We validate these signatures on genome-wide microarray dataset and found strong association between the expression of these signature genes and pathologic parameters.</p> <p>Conclusions</p> <p>In summary, graph centralities provide a novel way to connect different cancer signatures and to understand the mechanism of relationship between gene expression and clinical outcome of breast cancer. Moreover, this method is not only can be used on breast cancer, but also can be used on other gene expression related diseases and drug studies.</p

    Modeling Cancer Progression via Pathway Dependencies

    Get PDF
    Cancer is a heterogeneous disease often requiring a complexity of alterations to drive a normal cell to a malignancy and ultimately to a metastatic state. Certain genetic perturbations have been implicated for initiation and progression. However, to a great extent, underlying mechanisms often remain elusive. These genetic perturbations are most likely reflected by the altered expression of sets of genes or pathways, rather than individual genes, thus creating a need for models of deregulation of pathways to help provide an understanding of the mechanisms of tumorigenesis. We introduce an integrative hierarchical analysis of tumor progression that discovers which a priori defined pathways are relevant either throughout or in particular steps of progression. Pathway interaction networks are inferred for these relevant pathways over the steps in progression. This is followed by the refinement of the relevant pathways to those genes most differentially expressed in particular disease stages. The final analysis infers a gene interaction network for these refined pathways. We apply this approach to model progression in prostate cancer and melanoma, resulting in a deeper understanding of the mechanisms of tumorigenesis. Our analysis supports previous findings for the deregulation of several pathways involved in cell cycle control and proliferation in both cancer types. A novel finding of our analysis is a connection between ErbB4 and primary prostate cancer

    Molecular Mechanisms of Therapeutic Resistance in Cancer.

    Full text link
    Development of therapeutic resistance limits the efficacy of current cancer treatment. Understanding the molecular basis for therapeutic resistance should facilitate the identification of actionable targets and development of new combination therapies for cancer patients. Yet the understanding of therapeutic resistance still remains incomplete. In this thesis, clinically relevant mouse models coupled with systematic genomic and imaging technologies are used to identify mechanisms driving resistance, which also formulate novel therapeutic paradigms for patients with drug-resistant tumors. In the first study, a genetically engineered mouse model of ovarian endometrioid adenocarcinoma (OEA) was utilized in combination with molecular imaging to understand mechanisms of chemoresistance in OEA. It was demonstrated that AKT signaling pathway was activated upon chemotherapy (cisplatin) administration, which protected cells from apoptosis and thereby leading to the development of resistance. In support of this observation, inhibition of AKT activity improved the efficacy of chemotherapy by enhanced induction of apoptosis. A second study was undertaken to develop a new understanding of the mechanistic basis for therapeutic resistance in glioblastoma using a patient derived xenograft model. An integrated transcriptome analysis revealed that chemoradioresistance was associated with an increased expression of genes involved in the mesenchymal and stem cell phenotype as well as a decreased expression of genes involved in cell death. TGF-Ī² signaling was identified to be central to each of the mesenchymal/stem phenotype and therefore a critical player in modulating therapeutic resistance. In support, treatment with a TGF-Ī² inhibitor partially restored the sensitivity to therapy in TMZ/IR resistant tumors. Overall, this thesis demonstrated the importance of the AKT and TGF-Ī² signaling pathways in therapeutic resistance in a subset of ovarian cancer and glioblastoma patients, which provides clinical guidance for applying new combination therapies. It also demonstrates the concept that the combination of clinically relevant mouse models, molecular imaging and systematic genomic analysis can be used to derive novel insights into the dynamic signaling processes involved with gain of resistance. Future studies are needed to investigate if targeting these resistance mechanisms delays or prevents the development of resistance in treatment-naĆÆve patients.PHDCellular and Molecular BiologyUniversity of Michigan, Horace H. Rackham School of Graduate Studieshttp://deepblue.lib.umich.edu/bitstream/2027.42/111378/1/hanxiaow_1.pd

    Metabolomic Profiling Reveals a Role for Androgen in Activating Amino Acid Metabolism and Methylation in Prostate Cancer Cells

    Get PDF
    Prostate cancer is the second leading cause of cancer related death in American men. Development and progression of clinically localized prostate cancer is highly dependent on androgen signaling. Metastatic tumors are initially responsive to anti-androgen therapy, however become resistant to this regimen upon progression. Genomic and proteomic studies have implicated a role for androgen in regulating metabolic processes in prostate cancer. However, there have been no metabolomic profiling studies conducted thus far that have examined androgen-regulated biochemical processes in prostate cancer. Here, we have used unbiased metabolomic profiling coupled with enrichment-based bioprocess mapping to obtain insights into the biochemical alterations mediated by androgen in prostate cancer cell lines. Our findings indicate that androgen exposure results in elevation of amino acid metabolism and alteration of methylation potential in prostate cancer cells. Further, metabolic phenotyping studies confirm higher flux through pathways associated with amino acid metabolism in prostate cancer cells treated with androgen. These findings provide insight into the potential biochemical processes regulated by androgen signaling in prostate cancer. Clinically, if validated, these pathways could be exploited to develop therapeutic strategies that supplement current androgen ablative treatments while the observed androgen-regulated metabolic signatures could be employed as biomarkers that presage the development of castrate-resistant prostate cancer

    Discovering cancer-associated transcripts by RNA sequencing

    Full text link
    High-throughput sequencing of poly-adenylated RNA (RNA-Seq) in human cancers shows remarkable potential to identify uncharacterized aspects of tumor biology, including gene fusions with therapeutic significance and disease markers such as long non-coding RNA (lncRNA) species. However, the analysis of RNA-Seq data places unprecedented demands upon computational infrastructures and algorithms, requiring novel bioinformatics approaches. To meet these demands, we present two new open-source software packages - ChimeraScan and AssemblyLine - designed to detect gene fusion events and novel lncRNAs, respectively. RNA-Seq studies utilizing ChimeraScan led to discoveries of new families of recurrent gene fusions in breast cancers and solitary fibrous tumors. Further, ChimeraScan was one of the key components of the repertoire of computational tools utilized in data analysis for MI-ONCOSEQ, a clinical sequencing initiative to identify potentially informative and actionable mutations in cancer patientsā€™ tumors. AssemblyLine, by contrast, reassembles RNA sequencing data into full-length transcripts ab initio. In head-to-head analyses AssemblyLine compared favorably to existing ab initio approaches and unveiled abundant novel lncRNAs, including antisense and intronic lncRNAs disregarded by previous studies. Moreover, we used AssemblyLine to define the prostate cancer transcriptome from a large patient cohort and discovered myriad lncRNAs, including 121 prostate cancer-associated transcripts (PCATs) that could potentially serve as novel disease markers. Functional studies of two PCATs - PCAT-1 and SChLAP1 - revealed cancer-promoting roles for these lncRNAs. PCAT1, a lncRNA expressed from chromosome 8q24, promotes cell proliferation and represses the tumor suppressor BRCA2. SChLAP1, located in a chromosome 2q31 ā€˜gene desertā€™, independently predicts poor patient outcomes, including metastasis and cancer-specific mortality. Mechanistically, SChLAP1 antagonizes the genome-wide localization and regulatory functions of the SWI/SNF chromatin-modifying complex. Collectively, this work demonstrates the utility of ChimeraScan and AssemblyLine as open-source bioinformatics tools. Our applications of ChimeraScan and AssemblyLine led to the discovery of new classes of recurrent and clinically informative gene fusions, and established a prominent role for lncRNAs in coordinating aggressive prostate cancer, respectively. We expect that the methods and findings described herein will establish a precedent for RNA-Seq-based studies in cancer biology and assist the research community at large in making similar discoveries.PHDBioinformaticsUniversity of Michigan, Horace H. Rackham School of Graduate Studieshttp://deepblue.lib.umich.edu/bitstream/2027.42/120814/1/mkiyer_1.pd
    • ā€¦
    corecore