• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 123
  • 31
  • 21
  • 15
  • 12
  • 9
  • 9
  • 7
  • 3
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 282
  • 282
  • 33
  • 29
  • 28
  • 24
  • 24
  • 23
  • 23
  • 20
  • 18
  • 18
  • 17
  • 16
  • 16
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
61

Validation and early qualification of pancreatic fat deposition as an imaging biomarker of pancreatic cancer risk

Coe, Peter January 2016 (has links)
Introduction: Pancreatic cancer is the 10th most common cause of cancer in the United Kingdom (UK) yet the 5th most common cause of cancer related death. Although excess adiposity, measured as body mass index (BMI), is a risk factor for the development of pancreatic cancer the increase in relative risk is modest. Animal models suggest that the intra-organ deposition of lipids may be more specific to disease risk than anthropometric measurements. There is therefore a need to develop non-invasive methods to quantify intra-pancreatic fat deposition as a potential biomarker for pancreatic cancer predisposition. Cancer Research UK (CRUK) sets out clear guidelines for biomarker discovery and development. Potential biomarkers must go through a process of discovery and assay development followed by qualification. Methods Three streams of research: (i) Stage-one of the PanORAMA project. Assessment of accuracy through comparison of CS-MR and MRS quantified intra-pancreatic fat with histologically quantified intra-pancreatic fat in 12 patients undergoing pancreatic surgery. (ii) Stage-two of the PanORAMA study. Assessment of precision (reproducibility) and comparison with other anthropometric markers of excess adiposity in healthy volunteers (n=15). Refinement of MRS protocols and repeated assessment of precision in healthy volunteers (n=10). (iii) The Breast Risk Reduction Intermittent Dietary Evaluation 2 (BRRIDE-2) trial. Comparison of the effects of Intermittent Energy Restriction (IER) with Daily Energy Restriction (DER) on intra-pancreatic and intra-hepatic fat stores and metabolic markers of disease risk (n=26). Results (i) CS-MR and MRS had agreement with histological assessment of intra-pancreatic fat, but correlations were only moderate to good (rho 0.672 and 0.781 respectively). (ii) CS-MR, and after refinement, MRS, have clinically acceptable precision. This study tested this principle in intra-pancreatic fat in healthy volunteers with a range of intra-pancreatic fat consistent with the literature on the healthy population. (iii) I found no differences in reduction in intra-hepatic or intra-pancreatic fat when comparing IER with DER. Overall, I found that significant reductions (mean: 6.5%) in both of these ectopic fat stores could be achieved with eight-weeks of dietary intervention. Discussion More recent hypotheses on the link between excess adiposity and cancer have focused on the importance of within organ local ectopic fat as an abnormal micro-environment favouring cancer development and progression. Importantly, this hypothesis explains the specificity of epidemiological associations between excess adiposity and cancer risk. The observations that within a given individual, in the presence of short-term weight reduction, there are differential changes in local within organ fats – hepatic fat and pancreatic fat – support the specificity hypothesis. This thesis has put us in position to scale-up and explore the importance of intra-organ fats using non-invasive imaging techniques.
62

Efficacy and Mechanism of Action of a Novel Class of Antic-Cancer Drugs

Teran, Claudia January 2016 (has links)
The incidence of cancer worldwide has increased over the years, and gastrointestinal cancers (G.I.) are amongst the most common forms of cancer. Nevertheless, there is still no curative treatments for this group of tumors. Nucleoside analogues are widely used in cancer treatment. The prevailing compounds are Gemcitabine (used for pancreatic cancer and other carcinomas), 5-Fluorouracil (used in breast, colon, and other cancers), Cytarabine and Clofarabine (used in leukemias). Gemcitabine, the current standard of care for various forms of solid tumors, has a limited efficacy against pancreatic cancer. The objective of this project was the development of effective drugs against pancreatic cancer. We focused on a novel class of nucleoside analogues designed to bypass the most common cellular road blocks and resistance mechanisms. After an extensive screen for cell killing activity, two lead molecules were exclusively studied: LCB2151 and LCB2132. These two molecules showed high efficacy in killing human cancer cells from three different human G.I. cell lines: BxPC3 and Capan-2, two pancreatic cell lines representative of K-Ras positive and negative tumors, as well as the liver cell line HepG2. LCB2151 showed high efficacy in killing Gemcitabine-resistant cancer cells, and a low toxicity in normal cells. Interestingly, results show that these prodrugs can efficiently bypass key resistance mechanisms developed by cancer cells. The results obtained in this project are promising and could pave the way for a more effective treatment of pancreatic cancer.
63

Understanding the Role of the Receptor for Advanced Glycation End-Products (Rage) in Pancreatic Cancer

Swami, Priyanka January 2019 (has links)
Expression of the Receptor for Advanced Glycation End Products (RAGE) and is upregulated in a several cancers. Based on published studies, we hypothesized that RAGE, when overexpressed in pancreatic cancer cells, will promote cell proliferation and migration. To study the role of RAGE in pancreatic cancer, we selected the human pancreatic cancer cell-line PANC-1, and stably transfected the cells with full length RAGE to generate model cell-lines that overexpress RAGE. We obtained two cell-lines PANC-1 FLR2 and PANC-1 FLR3 and examined the influence of RAGE on cellular properties. A significant increase in proliferation but a reduction in migratory abilities of PANC-1 FLR2 and PANC-1 FLR3 cells was observed. The increase in proliferation and reduction in migration was reverted upon knockdown of RAGE in PANC-1 FLR2 cells with siRNA specific for RAGE. The reduction in migration was supported by the reduced levels of vimentin and several integrins in RAGE transfected cells. Furthermore, we observed a downregulation in FAK, AKT, ERK1/2 and NF-κB activity. Growing evidence supports that RAGE is essential for pancreatic cancer progression. It has also been shown that RAGE facilitates pancreatic tumor cell survival by enhancing autophagy and inhibiting apoptosis. The goal of our study was to determine the effect of RAGE inhibition during gemcitabine chemotherapy on the growth of pancreatic tumor. Hence, we investigated the effect of RAGE inhibitors and their combination with gemcitabine in an orthotopic mouse model of pancreatic cancer using mouse pancreatic cancer cell-line KPC 5508. We used two RAGE inhibitors, an anti-RAGE monoclonal antibody (IgG2A11) and a small molecule RAGE inhibitor (FPS-ZM1). We observed a significant reduction in tumor weights of the mice treated with the combination of IgG2A11 and gemcitabine as compared to gemcitabine alone treated mice. The reduction in tumor growth was accompanied with increase in p62 levels (marker of autophagy) and increase in levels of cleaved PARP (marker of apoptosis). We also observed reduction in HMGB1 and phosphorylation levels of ERK1/2 in tumors from the group treated with the combination as compared to the gemcitabine alone treated group. / North Dakota State University. College of Health Professions / NIH Grant # P20 GM109024 from the National Institute of General Medicine
64

The SYK tyrosine kinase suppresses autolysosome biogenesis via activation of mTORC1 in pancreatic cancer cell lines

Hua, Kevin Lee 07 October 2019 (has links)
Spleen tyrosine kinase (SYK) regulates mitogenic signaling, inflammatory responses and cell fate in a number of diverse cell types. KRAS is a proto-oncogene that controls cell growth and proliferation through several mitogenic pathways. In pancreatic cancer, KRAS is frequently mutated, resulting in constitutive activation in 90% of pancreatic cancer cell lines. We previously showed that SYK is highly expressed in a subset of KRAS-mutant pancreatic ductal adenocarcinoma (PDAC) cell lines. We demonstrated that SYK kinase inhibition with PRT062607 (SYKi) causes decreased cell proliferation of PDAC cell lines. Furthermore, combined SYKi and MEK inhibitor (MEKi) treatment promotes additive effects on suppression of PDAC cell proliferation and clonogenic growth. Mechanistically, SYK activates the mTORC1 kinase complex as shown by reduced phosphorylation of ribosomal S6 protein and its upstream kinase p70 S6 kinase (p70S6K) following SYKi treatment in PDAC cell lines. SYK-mediated mTORC1 activation occurs independently of MEK/ERK and PI3K/AKT effector signaling pathways. The mTORC1 complex suppresses lysosome biogenesis and macroautophagy (autophagy). Consequently, mTORC1 suppression via SYK inhibition or shRNA-mediated depletion causes accumulation of autolysosomes. These effects are mediated by the enhanced nuclear localization of MITF, a key transcriptional regulator of genes involved in lysosome biogenesis and autophagy pathway activation. In summary, SYK positively regulates mTORC1 activation in a subset of PDAC cell lines to suppress hyperactivation of autophagy. These findings open new avenues for further exploration of SYK as a critical regulator of the autophagy pathway in KRAS/mTORC1-dependent PDAC, and how this may be exploited for therapeutic benefit.
65

IMPROVING SPECTRAL ANALYSIS WITH THE APPLICATION OF MACHINE LEARNING: STUDY OF LASER-INDUCED BREAKDOWN SPECTROSCOPY (LIBS) AND RAMAN SPECTROSCOPY WITH CLASSIFICATION AND CLUSTERING TECHNIQUES.

Mandrell, Christopher 01 May 2020 (has links)
AN ABSTRACT OF THE THESIS OFChristopher T. Mandrell, for the Master of Science degree in Physics, presented on April 8, 2020, at Southern Illinois University Carbondale.TITLE: IMPROVING SPECTRAL ANALYSIS WITH THE APPLICATION OF MACHINE LEARNING: STUDY OF LASER-INDUCED BREAKDOWN SPECTROSCOPY (LIBS) AND RAMAN SPECTROSCOPY WITH CLASSIFICATION AND CLUSTERING TECHNIQUESMAJOR PROFESSOR: Dr. Poopalasingam SivakumarAtomic and molecular spectroscopy, in the form of LIBS emissions and Raman scattering, respectively, are tools that provide a vast amount of information with little to no sample preparation. For this reason, these techniques are finding their way into a wide range of fields. However, each spectrum is notoriously complicated to analyze, with many complex interactions at play. Machine learning is the result of work on artificial intelligence. It provides tools to train a computer to look for connections in complex data sets that would likely be missed, or not even looked for, by other analytical methods. The combination of highly informative yet complex data with an analysis that is specifically designed to probe highly complex data for meaningful information is a logical step in the analysis of these spectra. Here we apply statistical analysis and classification algorithms to Raman spectra of pancreatic cancer cells and clustering algorithms to LIBS spectra of Mars Curiosity Rover simulants and Raman spectra of Mars Perseverance Rover simulants. We report here high accuracy in the classification of different types of pancreatic cancer cells, and informative clustering of the two Mars rovers’ simulant data.
66

Changing the Pancreatic Cancer Treatment Paradigm: Developing Clostridium novyi as an Intravenously Injectable Solid-State Tumor Therapeutic

Dailey, Kaitlin Marie January 2020 (has links)
The development of a drug able to distinguish between tumor and host cells has been long sought, but the solid tumor microenvironment (TME) confounds many current therapeutics. Solid tumors present several challenges for oncotherapeutics, primarily, (1) aberrant vascularization, resulting in hypoxia, necrosis, abnormally high pH, and (2) tumor immune suppression. Oncolytic microbes are drawn to this microenvironment by an innate ability to selectively penetrate, colonize, and eradicate solid tumors as well as reactivate tumor associated immune components. To consider oncolytic bacteria deployment into this microenvironment, Chapter 1 dives into the background of oncolytic microbes. A discussion of the oncolytic bacterial field state, identifying Clostridium novyi? as a promising species, and details genetic engineering techniques to develop customized bacteria. Despite the promise of C.novyi in preclinical/clinical trials when administered intratumorally, the genetic and biochemical uniqueness of C.novyi necessitated the development of new methodologies to facilitate more widespread acceptance. Chapter 2 reports the development of methods that facilitate experimental work and therapeutic translation of C.novyi, including the ability to work with this obligate micro-anaerobe aerobically on the benchtop. While methods development is a necessary step in the clinical translation of C.novyi so too is choosing the correct model of the TME within which to test a potential anti-cancer therapy. While the typical solid TME includes both phenotypic and genotypic heterogeneity, the methods used to model this disease state often do not reflect this complexity. This simplistic approach may have contributed to stagnant five-year survival rates over the past four decades. Nevertheless, simplistic models are a necessary first step in clinical translation. Chapter 3 explores the impact of cancer cell lines co-cultured with C. novyi to establish the efficacy of this oncolytic bacteria in a monolayer culture. Chapter 4 extends this analysis adding not only a level of complexity by using an in vivo model, but also using CRISPR/Cas9 to modify the genome of C.novyi to encode a tumor targeting peptide, RGD, for expression within the spore coat. The combination of these studies indicates that C. novyi is uniquely poised to accomplish the long sought after selective tumor localization via intravenous delivery.
67

Tocotrienols Inhibit AKT and ERK Activation and Suppress Pancreatic Cancer Cell Proliferation by Suppressing the ErbB2 Pathway

Shin-Kang, Sonyo, Ramsauer, Victoria P., Lightner, Janet, Chakraborty, Kanishka, Stone, William, Campbell, Sharon, Reddy, Shrikanth A.G., Krishnan, Koyamangalath 15 September 2011 (has links)
Tocotrienols are members of the vitamin E family but, unlike tocopherols, possess an unsaturated isoprenoid side chain that confers superior anti-cancer properties. The ability of tocotrienols to selectively inhibit the HMG-CoA reductase pathway through posttranslational degradation and to suppress the activity of transcription factor NF-κB could be the basis for some of these properties. Our studies indicate that γ- and δ-tocotrienols have potent antiproliferative activity in pancreatic cancer cells (Panc-28, MIA PaCa-2, Panc-1, and BxPC-3). Indeed both tocotrienols induced cell death (> 50%) by the MTT cell viability assay in all four pancreatic cancer cell lines. We also examined the effects of the tocotrienols on the AKT and the Ras/Raf/MEK/ERK signaling pathways by Western blotting analysis. γ- and δ-tocotrienol treatment of cells reduced the activation of ERK MAP kinase and that of its downstream mediator RSK (ribosomal protein S6 kinase) in addition to suppressing the activation of protein kinase AKT. Suppression of activation of AKT by γ-tocotrienol led to downregulation of p-GSK-3β and upregulation accompanied by nuclear translocation of Foxo3. These effects were mediated by the downregulation of Her2/ErbB2 at the messenger level. Tocotrienols but not tocopherols were able to induce the observed effects. Our results suggest that the tocotrienol isoforms of vitamin E can induce apoptosis in pancreatic cancer cells through the suppression of vital cell survival and proliferative signaling pathways such as those mediated by the PI3-kinase/AKT and ERK/MAP kinases via downregulation of Her2/ErbB2 expression. The molecular components for this mechanism are not completely elucidated and need further investigation.
68

Dissertation: Sociodemographics and Pancreatic Cancer Survival Rate

Lewis, Sylvester 01 January 2018 (has links)
Pancreatic carcinoma or pancreatic cancer (PaCa) is an insidious disease with a prognosis of 6- to 12-month survival time for a late stage diagnosis. This problem has become crucial given that no study to date had been able to establish a definitive association between independent factors (other than a few diseases) and the survival rate of pancreatic cancer. The purpose of this quantitative, cross-sectional study was to determine whether an association exists between the independent, sociodemographic variables (marital status, age, education, income, and employment) and the outcome variable of survival rate. The social cognitive theory was the framework that provided the blueprint throughout the development of this study and helped guide the analysis of the secondary data, which was procured from the surveillance, epidemiology, and end results program. The sample of 56,166 participants was collected from 2009 to 2013 and Cox proportional hazard was used to analyze the data and arrive at the answers to the research hypotheses. A Cox proportional hazard model was used to analyze whether an association existed between each of the independent variables and the outcome variable. The analysis showed significant association between age, education, income, and employment and survival rate. It was not the same for marital status. These findings could stimulate social change by allowing stakeholders and other policy makers to become aware of the role that sociodemographic factors can play in health care. In addition, a need exists for effective research to be undertaken in the prevention and intervention of this disease. This could then lead to private and public health innovations and procedures to benefit patients with PaCa.
69

Targeting Protein Arginine Methyltransferase 5 as a Novel Therapeutic Approach in Pancreatic & Colorectal Cancer

Prabhu, Lakshmi Milind 12 1900 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Pancreatic ductal adenocarcinoma (PDAC) and colorectal cancer (CRC) are among the most commonly diagnosed forms of cancer in the United States. Due to their widespread prevalence and high mortality rate, it is vital to develop effective therapeutic drugs to combat these deadly diseases. In both PDAC and CRC, the multifunctional factor nuclear factor kappa B (NF-kB), a central coordinator of cellular immune responses, is activated abnormally, leading to tumorigenesis and cancer progression. Therefore, controlling NF-kB activity is critical in the treatment of these cancers. In a previous study, we identified a new mechanism by which NF-kB activity is regulated by an epigenetic enzyme known as protein arginine methyltransferase 5 (PRMT5). We showed that overexpression of PRMT5 not only activated NF-kB, but also significantly promoted several characteristics associated with cancer, including increased cell proliferation, migration, and anchorage-independent growth in both PDAC and CRC cells. Moreover, in order to examine the therapeutic potential of PRMT5 in these cancers, we adapted the state-of-the-art AlphaLISA technique into a high throughput screen (HTS) platform to screen for PRMT5 inhibitors. As a result, we successfully identified the small molecule PR5-LL-CM01 as our lead hit. Further validation experiments confirmed that PR5-LL-CM01 is a potent and specific PRMT5 inhibitor that exhibits significant anti-tumor efficacy in both in vitro and in vivo models of PDAC and CRC. Additionally, in a second screen, we discovered two natural compounds, P1608K04 and P1618J22, that can also function as the PRMT5 inhibitors. These findings further highlight the robustness of the PRMT5- specific AlphaLISA HTS technique. To conclude, we describe here for the first time a novel role of PRMT5 as a tumor-promoting factor in PDAC and CRC through NF-kB activation. By successfully developing and applying an innovative AlphaLISA HTS technique, we discovered PR5-LL-CM01, P1608K04, and P1618J22 as novel PRMT5 inhibitors, with PR5-LL-CM01 showing the strongest potency in both PDAC and CRC models. Therefore, we demonstrated that PRMT5 is a promising therapeutic target in PDAC and CRC, and the novel PRMT5 inhibitor PR5-LL-CM01 could serve as a promising basis for new drug development in PDAC and CRC.
70

Loss of TIP30 Accelerates Pancreatic Cancer Progression and Metastasis

Imasuen Williams, Imade E. 07 1900 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Pancreatic ductal adenocarcinoma (PDAC) is currently the fourth leading cause of cancer-related death in the United States, and is characterized by key driver mutations (e.g. KRAS, TP53, CDKN2A, and SMAD4), elevated expression of growth factors such as TGF-βs and the EGF receptor (EGFR), a markedly desmoplastic stroma, and a propensity to develop multi-organ metastases and chemoresistance. Consistent with its aggressive nature, the 5-year survival rate for PDAC is 8-9%, which demonstrates an urgent need to develop novel therapies. High expression levels of microRNA-10b (miR-10b) in PDAC tissues are associated with decreased patient survival and earlier appearance of metastatic disease following neoadjuvant chemoradiotherapy. miR-10b downregulates the expression of transcription coactivator Tat-Interacting Protein 30 (TIP30) by targeting its 3’UTR. TIP30 has multiple reported functions. TIP30 suppresses tumor formation and metastasis, forms a complex that regulates EGFR trafficking and degradation, and transcriptionally upregulates pro-apoptotic genes. Alterations in TIP30 have been reported in multiple human cancers, including pancreatic cancer. We hypothesized that Tip30-deficiency accelerates PDAC progression and metastasis in a murine model of PDAC. To test this hypothesis, we crossed mice with oncogenic Kras (KC) localized to the pancreas epithelium, with Tip30-deficient mice (K30C). We compared PDAC histopathology between Tip30-heterozygous (K30+/-C) and Tip30-null (K30-/-C) mice. Tip30-heterozygosity accelerated PDAC-lesion-associated pancreatic cancer cell (PCC) pulmonary seeding. By contrast, total loss of Tip30 enhanced PCC micrometastatic seeding to the liver and hepatic metastasis. K30+/-C mice also presented with an early, increased penetrance of lung lesions and lung adenocarcinoma; and PCCs isolated from K30+/-C pancreata exhibited increased EGFR protein levels. These findings suggest that TIP30 deficiency can have a dose-dependent effect on organotropic metastasis and EGFR levels in PCCs. Future studies will delineate the molecular consequences of TIP30 loss in PDAC and contribute to a broader understanding of pancreatic cancer metastasis. / 2020-08-05

Page generated in 0.0504 seconds