Matching Items (13)
152885-Thumbnail Image.png
Description
Patients with malignant brain tumors have a median survival of approximately 15 months following diagnosis, regardless of currently available treatments which include surgery followed by radiation and chemotherapy. Improvement in the survival of brain cancer patients requires the design of new therapeutic modalities that take advantage of common phenotypes. One

Patients with malignant brain tumors have a median survival of approximately 15 months following diagnosis, regardless of currently available treatments which include surgery followed by radiation and chemotherapy. Improvement in the survival of brain cancer patients requires the design of new therapeutic modalities that take advantage of common phenotypes. One such phenotype is the metabolic dysregulation that is a hallmark of cancer cells. It has therefore been postulated that one approach to treating brain tumors may be by metabolic alteration such as that which occurs through the use of the ketogenic diet (KD). The KD is high-fat, low-carbohydrate diet that induces ketosis and has been utilized for the non-pharmacologic treatment of refractory epilepsy. It has been shown that this metabolic therapy enhances survival and potentiates standard therapy in mouse models of malignant gliomas, yet the anti-tumor mechanisms are not fully understood.

The current study reports that KetoCal® (KC; 4:1 fat:protein/carbohydrates), fed ad libitum, alters hypoxia, angiogenic, and inflammatory pathways in a mouse model of glioma. Tumors from animals maintained on KC showed reduced expression of the hypoxia marker carbonic anhydrase 9 (CA IX), a reduction in hypoxia inducible factor 1-alpha (HIF-1α) and decreased activation of nuclear factor kappa B (NF-κB). Animals maintained on KC also showed a reduction in expression of vascular endothelial growth factor receptor 2 (VEGFR2) and decreased microvasculature in their tumors. Further, peritumoral edema was significantly reduced in animals fed the KC and protein analysis showed significantly altered expression of the tight junction protein zona occludens-1 (ZO-1) and the water channeling protein aquaporin-4 (AQP4), both of which have been implicated in malignant processes in glioma, including the formation of peritumoral edema in patients. Taken together the data suggests that KC alters multiple processes involved in malignant progression of gliomas. A greater understanding of the effects of the ketogenic diet as an adjuvant therapy will allow for a more rational approach to its clinical use.
ContributorsWoolf, Eric C (Author) / Scheck, Adrienne C (Thesis advisor) / Lake, Douglas F (Committee member) / LaBaer, Joshua (Committee member) / Arizona State University (Publisher)
Created2014
137354-Thumbnail Image.png
Description
The objective of the research presented here was to validate the use of kinetic models for the analysis of the dynamic behavior of a contrast agent in tumor tissue and evaluate the utility of such models in determining kinetic properties - in particular perfusion and molecular binding uptake associated with

The objective of the research presented here was to validate the use of kinetic models for the analysis of the dynamic behavior of a contrast agent in tumor tissue and evaluate the utility of such models in determining kinetic properties - in particular perfusion and molecular binding uptake associated with tissue hypoxia - of the imaged tissue, from concentration data acquired with dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) procedure. Data from two separate DCE-MRI experiments, performed in the past, using a standard contrast agent and a hypoxia-binding agent respectively, were analyzed. The results of the analysis demonstrated that the models used may provide novel characterization of the tumor tissue properties. Future research will work to further characterize the physical significance of the estimated parameters, particularly to provide quantitative oxygenation data for the imaged tissue.
ContributorsMartin, Jonathan Michael (Author) / Kodibagkar, Vikram (Thesis director) / Rege, Kaushal (Committee member) / Barrett, The Honors College (Contributor) / Chemical Engineering Program (Contributor) / School of Mathematical and Statistical Sciences (Contributor)
Created2013-12
137469-Thumbnail Image.png
Description
Oxygen delivery is crucial for the development of healthy, functional tissue. Low tissue oxygenation, or hypoxia, is a characteristic that is common in many tumors. Hypoxia contributes to tumor malignancy and can reduce the success of chemotherapy and radiation treatment. There is a current need to noninvasively measure tumor oxygenation

Oxygen delivery is crucial for the development of healthy, functional tissue. Low tissue oxygenation, or hypoxia, is a characteristic that is common in many tumors. Hypoxia contributes to tumor malignancy and can reduce the success of chemotherapy and radiation treatment. There is a current need to noninvasively measure tumor oxygenation or pO2 in patients to determine a personalized treatment method. This project focuses on creating and characterizing nanoemulsions using a pO2 reporter molecule hexamethyldisiloxane (HMDSO) and its longer chain variants as well as assessing their cytotoxicity. We also explored creating multi-modal (MRI/Fluorescence) nanoemulsions.
ContributorsGrucky, Marian Louise (Author) / Kodibagkar, Vikram (Thesis director) / Rege, Kaushal (Committee member) / Stabenfeldt, Sarah (Committee member) / Barrett, The Honors College (Contributor) / Harrington Bioengineering Program (Contributor)
Created2013-05
137057-Thumbnail Image.png
Description
Osteosarcoma (OS) is the most prevalent primary tumor of bone in the pediatric age group [1]. The long-term cancer free survival has improved in patients with localized cancer; however, less than 20% of patients diagnosed with metastatic disease survive without relapse [2]. While these findings emphasize the urgent need for

Osteosarcoma (OS) is the most prevalent primary tumor of bone in the pediatric age group [1]. The long-term cancer free survival has improved in patients with localized cancer; however, less than 20% of patients diagnosed with metastatic disease survive without relapse [2]. While these findings emphasize the urgent need for new therapeutic agents, the lack of understanding of the factors and the tumor microenvironment that lead to therapy resistance in OS has significantly hampered progress towards improved prognosis. Recent clinical reports have shown a negative correlation between tumor hypoxia and overall survival in OS patients [4]. In addition to the up-regulation of hypoxia inducible factors (HIFs), it has been shown that hypoxia can trigger an adaptive response such as the unfolded protein response (UPR) that allows tumor cells to avoid therapy-induced death [3,4,7,10].
Using in vitro experimental models of both SAOS-2 (non-metastatic) and 143-b (metastatic) osteosarcoma cell lines and Western blot analysis, we have demonstrated that basal levels of molecular chaperone BiP (Binding immunoglobulin protein, or GRP-78) and peIF2α (phospho-eukaryotic initiation factor 2 alpha), both markers of the UPR, were higher in SAOS-2 than 143-b cells. We also show that both these markers were further up-regulated upon exposure to hypoxia, as evidenced by the increase in banding intensity in both SAOS-2 and 143-b cells. Furthermore, analysis of another UPR marker, ATF6 (activating transcription factor 6) showed that basal levels of active nuclear ATF6 were slightly higher in SAOS-2 cells than in 143-b cells. However, unlike the other UPR markers these levels were significantly reduced upon exposure to hypoxia (0.1% O2). In addition to hypoxia, treatment with Cisplatin also had similar effects on the expression of aforementioned UPR markers: BiP and peIF2α. We found that the 143-b OS cells were more sensitive to the Cisplatin treatment than the SAOS-2 OS cells, and thus more prone to cell-mediated death.
Our findings shed light on the unknown mechanisms underlying chemotherapeutic drug resistance in osteosarcoma patients. Our research may lead to novel therapies that seek out and destroy the chemoresistant OS cells within the hypoxia core of tumors, thereby preventing survival and metastasis, and ultimately improving the chances of survival amongst OS patients.
ContributorsFaraj, Janine Jean (Author) / Chandler, Douglas (Thesis director) / Sertil, Aparna (Committee member) / Sweazea, Karen (Committee member) / Barrett, The Honors College (Contributor) / School of Life Sciences (Contributor) / School of Historical, Philosophical and Religious Studies (Contributor)
Created2014-05
135102-Thumbnail Image.png
Description
Magnetic resonance imaging (MRI) of changes in metabolic activity in tumors and metabolic abnormalities can provide a window to understanding the complex behavior of malignant tumors. Both diagnostics and treatment options can be improved through the further comprehension of the processes that contribute to tumor malignancy and growth. By detecting

Magnetic resonance imaging (MRI) of changes in metabolic activity in tumors and metabolic abnormalities can provide a window to understanding the complex behavior of malignant tumors. Both diagnostics and treatment options can be improved through the further comprehension of the processes that contribute to tumor malignancy and growth. By detecting and disturbing this activity through personalized treatments, it is the hope to provide better diagnostics and care to patients. Experimenting with multicellular tumor spheroids (MCTS) allows for a rapid, inexpensive and convenient solution to studying multiple in vitro tumors. High quality magnetic resonance images of small samples, such as spheroid, however, are difficult to achieve with current radio frequency coils. In addition, in order for the information provided by these scans to accurately represent the interactions and metabolic activity in vivo, there is a need for a perfused vascular network. A perfused vascular network has the potential to improve metabolic realism and particle transport within a tumor spheroid. By creating a more life-like cancer model and allowing the progressive imaging of metabolic functions of such small samples, a better, more efficient mode of studying metabolic activity in cancer can be created and research efforts can expand. The progress described in this paper attempts to address both of these current shortcomings of metabolic cancer research and offers potential solutions, while acknowledging the potential of future work to improve cancer research with MCTS.
ContributorsTobey, John Paul (Author) / Kodibagkar, Vikram (Thesis director) / Sadleir, Rosalind (Committee member) / Barrett, The Honors College (Contributor)
Created2016-12
155581-Thumbnail Image.png
Description
A tumor is a heterogeneous combination of proliferating tumor cells, infiltrating immune cells and stromal components along with a variety of associated host tissue cells, collectively termed the tumor microenvironment (TME). The constituents of the TME and their interaction with the host organ shape and define the properties of tumors

A tumor is a heterogeneous combination of proliferating tumor cells, infiltrating immune cells and stromal components along with a variety of associated host tissue cells, collectively termed the tumor microenvironment (TME). The constituents of the TME and their interaction with the host organ shape and define the properties of tumors and contribute towards the acquisition of hallmark traits such as hypoxia. Hypoxia imparts resistance to cancer from chemotherapy and radiotherapy due to the decreased production of reactive oxygen species and also promotes angiogenesis, malignant progression and metastasis. It also provides a powerful physiological stimulus that can be exploited as a tumor-specific condition, allowing for the rational design of anticancer hypoxia-activated pro-drugs (HAP). Accurate evaluation of tumor oxygenation in response to therapeutics interventions at various stages of growth should provide a better understanding of tumor response to therapy, potentially allowing therapy to be tailored to individual characteristics. The primary goal of this research was to investigate the utility of prospective identification of hypoxic tumors, by two different Magnetic Resonance Imaging (MRI) based oximetry approaches, in successful treatment with hypoxia activated therapy. In the present study, I report the utility of these two techniques 1) PISTOL (Proton Imaging of Siloxanes to map Tissue Oxygenation Levels) and 2) use of a hypoxia binding T1 contrast agent GdDO3NI in reporting the modulations of hypoxia pre and post hypoxia activated therapies in pre-clinical models of cancer. I have performed these studies in non-small cell lung cancer (NSCLC) and epidermoid carcinoma (NCI-H1975 and A431 cell lines, respectively) as well as in patient derived xenograft models of NSCLC. Both the oximetry techniques have the potential to differentiate between normoxic and hypoxic regions of the tumor and reveal both baseline heterogeneity and differential response to therapeutic intervention. The response of the tumor models to therapeutic interventions indicates that, in conjunction with pO2, other factors such as tumor perfusion (essential for delivering HAPs) and relative expression of nitroreductases (essential for activating HAPs) may play an important role. The long term goal of the proposed research is the clinical translation of both the MRI techniques and aiding the design and development of personalized therapy (e.g. patient stratification for novel hypoxia activated pro-drugs) particularly for cancer.
ContributorsAgarwal, Shubhangi (Author) / Kodibagkar, Vikram D (Thesis advisor) / Inge, Landon J (Committee member) / Nikkhah, Mehdi (Committee member) / Pagel, Mark D. (Committee member) / Sadleir, Rosalind J (Committee member) / Arizona State University (Publisher)
Created2017
135480-Thumbnail Image.png
Description
Compressed sensing magnetic resonance spectroscopic imaging (MRSI) is a noninvasive and in vivo potential diagnostic technique for cancer imaging. This technique undersamples the distribution of specific cancer biomarkers within an MR image as well as changes in the temporal dimension and subsequently reconstructs the missing data. This technique has been

Compressed sensing magnetic resonance spectroscopic imaging (MRSI) is a noninvasive and in vivo potential diagnostic technique for cancer imaging. This technique undersamples the distribution of specific cancer biomarkers within an MR image as well as changes in the temporal dimension and subsequently reconstructs the missing data. This technique has been shown to retain a high level of fidelity even with an acceleration factor of 5. Currently there exist several different scanner types that each have their separate analytical methods in MATLAB. A graphical user interface (GUI) was created to facilitate a single computing platform for these different scanner types in order to improve the ease and efficiency with which researchers and clinicians interact with this technique. A GUI was successfully created for both prospective and retrospective MRSI data analysis. This GUI retained the original high fidelity of the reconstruction technique and gave the user the ability to load data, load reference images, display intensity maps, display spectra mosaics, generate a mask, display the mask, display kspace and save the corresponding spectra, reconstruction, and mask files. Parallelization of the reconstruction algorithm was explored but implementation was ultimately unsuccessful. Future work could consist of integrating this parallelization method, adding intensity overlay functionality and improving aesthetics.
ContributorsLammers, Luke Michael (Author) / Kodibagkar, Vikram (Thesis director) / Hu, Harry (Committee member) / Harrington Bioengineering Program (Contributor) / Barrett, The Honors College (Contributor)
Created2016-05
Description
Animals are thought to die at high temperatures because proteins and cell membranes lose their structural integrity. Alternatively, a newer hypothesis (the oxygen and capacity limitation of thermal tolerance, or OCLTT) states that death occurs because oxygen supply becomes limited at high temperatures. Consequently, animals exposed to hypoxia are more

Animals are thought to die at high temperatures because proteins and cell membranes lose their structural integrity. Alternatively, a newer hypothesis (the oxygen and capacity limitation of thermal tolerance, or OCLTT) states that death occurs because oxygen supply becomes limited at high temperatures. Consequently, animals exposed to hypoxia are more sensitive to heating than those exposed to normoxia or hyperoxia. We hypothesized that animals raised in hypoxia would acclimate to the low oxygen supply, thereby making them less sensitive to heating. Such acclimation would be expressed as greater heat tolerance and better flight performance in individuals raised at lower oxygen concentrations. We raised flies (Drosophila melanogaster) from eggs to adults under oxygen concentrations ranging from 10% to 31% and measured two aspects of thermal tolerance: 1) the time required for flies to lose motor function at 39.5°C at normoxia (21%), referred to as knock-down time, and 2) flight performance at 37°, 39°, or 41°C and 12%, 21%, or 31% oxygen. Contrary to our prediction, flies from all treatments had the same knock-down time. However, flight performance at hypoxia was greatest for flies raised in hypoxia, but flight performance at normoxia and hyperoxia was greatest for flies raised at hyperoxia. Thus, flight performance acclimated to oxygen supply during development, but heat tolerance did not. Our data does not support the OCLTT hypothesis, but instead supports the beneficial acclimation hypothesis, which proposes that acclimation improves the function of an organism during environmental change.
ContributorsShiehzadegan, Shayan (Co-author) / VadenBrooks, John (Co-author) / Le, Jackie (Co-author) / Smith, Colton (Co-author) / Shiehzadegan, Shima (Co-author) / Angilletta, Michael (Co-author, Thesis director) / VandenBrooks, John (Committee member) / Klok, C. J. (Committee member) / School of Life Sciences (Contributor) / Barrett, The Honors College (Contributor)
Created2016-05
Description

Our cells need constant fuel and oxygen for the body to work properly and maintain cellular function. In high altitudes tissue oxygen levels fall and the body must work against this hypoxic challenge to maintain energetics and limit oxidative stress. Mammals living at high altitudes are challenged to sustain thermogenesis

Our cells need constant fuel and oxygen for the body to work properly and maintain cellular function. In high altitudes tissue oxygen levels fall and the body must work against this hypoxic challenge to maintain energetics and limit oxidative stress. Mammals living at high altitudes are challenged to sustain thermogenesis and aerobic exercise despite reduced amounts of available oxygen. Enhancements in oxidative capacity and oxygen diffusion capacity of skeletal muscle may be necessary to compensate for insufficient oxygen supply in tissues. Hypoxic conditions can cause a switch from aerobic metabolism to anaerobic metabolism. Due to previous research of Graham Scott and colleagues on “Adaptive Modifications of Muscle Phenotype in High-Altitude Deer Mice” and the SMack Lab at Arizona State University, the question of how low atmospheric oxygen levels affects the enzymatic activity in the gastrocnemius muscle of Gelada Monkeys compared to Rhesus Macaque Monkeys was researched. Lactate Dehydrogenase (LDH) activity was measured in the gastrocnemius tissue of 6 Gelada Monkeys (highland) and 6 Rhesus Macaque monkeys (lowland). LDH was expected to be greater in Gelada tissue samples due to heightened anaerobic metabolism in the presence of limited available oxygen in high altitude environments. Results showed higher LDH in Rhesus Macaque samples compared to Gelada samples, but this difference was not statistically significant. Despite nonsignificant data, this experiment is insightful into the effects of Hypoxic adaptation in skeletal muscle enzymatic activity in primates.

ContributorsSalehi, Yasmine (Author) / Snyder-Mackler, Noah (Thesis director) / Trumble, Ben (Committee member) / Barrett, The Honors College (Contributor) / School of Life Sciences (Contributor) / School of Human Evolution & Social Change (Contributor)
Created2023-05
173004-Thumbnail Image.png
Description

In 1861, William John Little published, “On The Influence of Abnormal Parturition, Difficult Labors, Premature Birth, and Asphyxia Neonatorum, on the Mental and Physical Condition of the Child, Especially in Relation to Deformities,” hereafter “Abnormal Parturition,” in the Transactions of the Obstetrical Society of London. In the article, Little discussed

In 1861, William John Little published, “On The Influence of Abnormal Parturition, Difficult Labors, Premature Birth, and Asphyxia Neonatorum, on the Mental and Physical Condition of the Child, Especially in Relation to Deformities,” hereafter “Abnormal Parturition,” in the Transactions of the Obstetrical Society of London. In the article, Little discussed the causes and types of what he refers to as abnormal births, and theorized how those births affect an infant’s likelihood of exhibiting a deformity. Little defined abnormal births as those involving an atypical maternal or fetal presentation, such as a slow birthing process or a fetus exiting the birth canal feet first rather than head first. In his article, Little published one of the first definitional frameworks to describe a condition causing rigidity and stiffness in the limbs that is often associated with birth-related trauma, which was then called Little’s disease, but is modernly known as spastic Cerebral Palsy.

Created2021-05-02