Matching Items (5)
Filtering by

Clear all filters

151241-Thumbnail Image.png
Description
Cancer is a disease that affects millions of people worldwide each year. The metastatic progression of cancer is the number one reason for cancer related deaths. Cancer preventions rely on the early identification of tumor cells as well as a detailed understanding of cancer as a whole. Identifying proteins specific

Cancer is a disease that affects millions of people worldwide each year. The metastatic progression of cancer is the number one reason for cancer related deaths. Cancer preventions rely on the early identification of tumor cells as well as a detailed understanding of cancer as a whole. Identifying proteins specific to tumor cells provide an opportunity to develop noninvasive clinical tests and further our understanding of tumor biology. Using liquid chromatography-mass spectrometry (LC-MS/MS) a short peptide was identified in pancreatic cancer patient plasma that was not found in normal samples, and mapped back to QSOX1 protein. Immunohistochemistry was performed probing for QSOX1 in tumor tissue and discovered that QSOX1 is highly over-expressed in pancreatic and breast tumors. QSOX1 is a FAD-dependent sulfhydryl oxidase that is extremely efficient at forming disulfide bonds in nascent proteins. While the enzymology of QSOX1 has been well studied, the tumor biology of QSOX1 has not been studied. To begin to determine the advantage that QSOX1 over-expression provides to tumors, short hairpin RNA (shRNA) were used to reduce the expression of QSOX1 in human tumor cell lines. Following the loss of QSOX1 growth rate, apoptosis, cell cycle and invasive potential were compared between tumor cells transduced with shQSOX1 and control tumor cells. Knock-down of QSOX1 protein suppressed tumor cell growth but had no effect on apoptosis and cell cycle regulation. However, shQSOX1 dramatically inhibited the abilities of both pancreatic and breast tumor cells to invade through Matrigel in a modified Boyden chamber assay. Mechanistically, shQSOX1-transduced tumor cells secreted MMP-2 and -9 that were less active than MMP-2 and -9 from control cells. Taken together, these results suggest that the mechanism of QSOX1-mediated tumor cell invasion is through the post-translational activation of MMPs. This dissertation represents the first in depth study of the role that QSOX1 plays in tumor cell biology.
ContributorsKatchman, Benjamin A (Author) / Lake, Douglas F. (Thesis advisor) / Rawls, Jeffery A (Committee member) / Miller, Laurence J (Committee member) / Chang, Yung (Committee member) / Arizona State University (Publisher)
Created2012
156061-Thumbnail Image.png
Description
The rate of progress in improving survival of patients with solid tumors is slow due to late stage diagnosis and poor tumor characterization processes that fail to effectively reflect the nature of tumor before treatment or the subsequent change in its dynamics because of treatment. Further advancement of targeted therapies

The rate of progress in improving survival of patients with solid tumors is slow due to late stage diagnosis and poor tumor characterization processes that fail to effectively reflect the nature of tumor before treatment or the subsequent change in its dynamics because of treatment. Further advancement of targeted therapies relies on advancements in biomarker research. In the context of solid tumors, bio-specimen samples such as biopsies serve as the main source of biomarkers used in the treatment and monitoring of cancer, even though biopsy samples are susceptible to sampling error and more importantly, are local and offer a narrow temporal scope.

Because of its established role in cancer care and its non-invasive nature imaging offers the potential to complement the findings of cancer biology. Over the past decade, a compelling body of literature has emerged suggesting a more pivotal role for imaging in the diagnosis, prognosis, and monitoring of diseases. These advances have facilitated the rise of an emerging practice known as Radiomics: the extraction and analysis of large numbers of quantitative features from medical images to improve disease characterization and prediction of outcome. It has been suggested that radiomics can contribute to biomarker discovery by detecting imaging traits that are complementary or interchangeable with other markers.

This thesis seeks further advancement of imaging biomarker discovery. This research unfolds over two aims: I) developing a comprehensive methodological pipeline for converting diagnostic imaging data into mineable sources of information, and II) investigating the utility of imaging data in clinical diagnostic applications. Four validation studies were conducted using the radiomics pipeline developed in aim I. These studies had the following goals: (1 distinguishing between benign and malignant head and neck lesions (2) differentiating benign and malignant breast cancers, (3) predicting the status of Human Papillomavirus in head and neck cancers, and (4) predicting neuropsychological performances as they relate to Alzheimer’s disease progression. The long-term objective of this thesis is to improve patient outcome and survival by facilitating incorporation of routine care imaging data into decision making processes.
ContributorsRanjbar, Sara (Author) / Kaufman, David (Thesis advisor) / Mitchell, Joseph R. (Thesis advisor) / Runger, George C. (Committee member) / Arizona State University (Publisher)
Created2017
155725-Thumbnail Image.png
Description
Random forest (RF) is a popular and powerful technique nowadays. It can be used for classification, regression and unsupervised clustering. In its original form introduced by Leo Breiman, RF is used as a predictive model to generate predictions for new observations. Recent researches have proposed several methods based on RF

Random forest (RF) is a popular and powerful technique nowadays. It can be used for classification, regression and unsupervised clustering. In its original form introduced by Leo Breiman, RF is used as a predictive model to generate predictions for new observations. Recent researches have proposed several methods based on RF for feature selection and for generating prediction intervals. However, they are limited in their applicability and accuracy. In this dissertation, RF is applied to build a predictive model for a complex dataset, and used as the basis for two novel methods for biomarker discovery and generating prediction interval.

Firstly, a biodosimetry is developed using RF to determine absorbed radiation dose from gene expression measured from blood samples of potentially exposed individuals. To improve the prediction accuracy of the biodosimetry, day-specific models were built to deal with day interaction effect and a technique of nested modeling was proposed. The nested models can fit this complex data of large variability and non-linear relationships.

Secondly, a panel of biomarkers was selected using a data-driven feature selection method as well as handpick, considering prior knowledge and other constraints. To incorporate domain knowledge, a method called Know-GRRF was developed based on guided regularized RF. This method can incorporate domain knowledge as a penalized term to regulate selection of candidate features in RF. It adds more flexibility to data-driven feature selection and can improve the interpretability of models. Know-GRRF showed significant improvement in cross-species prediction when cross-species correlation was used to guide selection of biomarkers. The method can also compete with existing methods using intrinsic data characteristics as alternative of domain knowledge in simulated datasets.

Lastly, a novel non-parametric method, RFerr, was developed to generate prediction interval using RF regression. This method is widely applicable to any predictive models and was shown to have better coverage and precision than existing methods on the real-world radiation dataset, as well as benchmark and simulated datasets.
ContributorsGuan, Xin (Author) / Liu, Li (Thesis advisor) / Runger, George C. (Thesis advisor) / Dinu, Valentin (Committee member) / Arizona State University (Publisher)
Created2017
153975-Thumbnail Image.png
Description
Breast cancer is the most common cancer and currently the second leading cause of death among women in the United States. Patients’ five-year relative survival rate decreases from 99% to 25% when breast cancer is diagnosed late. Immune checkpoint blockage has shown to be a promising therapy to improve patients’

Breast cancer is the most common cancer and currently the second leading cause of death among women in the United States. Patients’ five-year relative survival rate decreases from 99% to 25% when breast cancer is diagnosed late. Immune checkpoint blockage has shown to be a promising therapy to improve patients’ outcome in many other cancers. However, due to the lack of early diagnosis, the treatment is normally given in the later stages. An early diagnosis system for breast cancer could potentially revolutionize current treatment strategies, improve patients’ outcomes and even eradicate the disease. The current breast cancer diagnostic methods cannot meet this demand. A simple, effective, noninvasive and inexpensive early diagnostic technology is needed. Immunosignature technology leverages the power of the immune system to find cancer early. Antibodies targeting tumor antigens in the blood are probed on a high-throughput random peptide array and generate a specific binding pattern called the immunosignature.

In this dissertation, I propose a scenario for using immunosignature technology to detect breast cancer early and to implement an early treatment strategy by using the PD-L1 immune checkpoint inhibitor. I develop a methodology to describe the early diagnosis and treatment of breast cancer in a FVB/N neuN breast cancer mouse model. By comparing FVB/N neuN transgenic mice and age-matched wild type controls, I have found and validated specific immunosignatures at multiple time points before tumors are palpable. Immunosignatures change along with tumor development. Using a late-stage immunosignature to predict early samples, or vice versa, cannot achieve high prediction performance. By using the immunosignature of early breast cancer, I show that at the time of diagnosis, early treatment with the checkpoint blockade, anti-PD-L1, inhibits tumor growth in FVB/N neuN transgenic mouse model. The mRNA analysis of the PD-L1 level in mice mammary glands suggests that it is more effective to have treatment early.

Novel discoveries are changing understanding of breast cancer and improving strategies in clinical treatment. Researchers and healthcare professionals are actively working in the early diagnosis and early treatment fields. This dissertation provides a step along the road for better diagnosis and treatment of breast cancer.
ContributorsDuan, Hu (Author) / Johnston, Stephen Albert (Thesis advisor) / Hartwell, Leland Harrison (Committee member) / Dinu, Valentin (Committee member) / Chang, Yung (Committee member) / Arizona State University (Publisher)
Created2015
153561-Thumbnail Image.png
Description
Currently in the US, many patients with cancer do not benefit from the population-based screening, due to challenges associated with the existing cancer screening scheme. Blood-based diagnostic assays have the potential to detect diseases in a non-invasive way. Proteins released from small early tumors may only be present intermittently and

Currently in the US, many patients with cancer do not benefit from the population-based screening, due to challenges associated with the existing cancer screening scheme. Blood-based diagnostic assays have the potential to detect diseases in a non-invasive way. Proteins released from small early tumors may only be present intermittently and get diluted to tiny concentrations in the blood, making them difficult to use as biomarkers. However, they can induce autoantibody (AAb) responses, which can amplify the signal and persist in the blood even if the antigen is gone. Circulating autoantibodies is a promising class of molecules that have potential to serve as early detection biomarkers for cancers. This Ph.D thesis aims to screen for autoantibody biomarkers for the early detection of two deadly cancer, basal-like breast cancer and lung adenocarcinoma. First, a method was developed to display proteins in both native and denatured conformation on protein array. This method adopted a novel protein tag technology, called HaloTag, to covalently immobilize proteins on glass slide surface. The covalent attachment allowed these proteins to endure harsh treatment without getting dissociated from slide surface, which enabled the profiling of antibody responses against both conformational and linear epitopes. Next, a plasma screening protocol was optimized to significantly increase signal to noise ratio of protein array based AAb detection. Following this, the AAb responses in basal-like breast cancer were explored using nucleic acid programmable protein arrays (NAPPA) containing 10,000 full-length human proteins in 45 cases and 45 controls. After verification in a large sample set (145 basal-like breast cancer cases / 145 controls / 70 non-basal breast cancer) by ELISA, a 13-AAb classifier was developed to differentiate patients from controls with a sensitivity of 33% at 98% specificity. Similar approach was also applied to the lung cancer study to identify AAbs that distinguished lung cancer patients from computed-tomography positive benign pulmonary nodules (137 lung cancer cases, 127 smoker controls, 170 benign controls). In this study, two panels of AAbs were discovered that showed promising sensitivity and specificity. Six out of eight AAb targets were also found to have elevated mRNA level in lung adenocarcinoma patients using TCGA data. These projects as a whole provide novel insights on the association between AAbs and cancer, as well as general B cell antigenicity against self-proteins.
ContributorsWang, Jie (Author) / LaBaer, Joshua (Thesis advisor) / Anderson, Karen S (Committee member) / Lake, Douglas F (Committee member) / Chang, Yung (Committee member) / Arizona State University (Publisher)
Created2015