• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 268
  • 182
  • 79
  • 26
  • 23
  • 19
  • 12
  • 6
  • 6
  • 5
  • 2
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 693
  • 132
  • 118
  • 97
  • 90
  • 81
  • 80
  • 77
  • 75
  • 74
  • 67
  • 64
  • 61
  • 57
  • 49
  • 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.
111

Development of a drug-eluting 3D bioprinted mesh (GlioMesh) for treatment of glioblastoma multiforme

Hosseinzadeh, Reihaneh 30 April 2018 (has links)
Glioblastoma multiforme (GBM) is among the most aggressive and mortal cancers of the central nervous system. Maximal safe surgical resection, followed by radiotherapy accompanied with chemotherapy is the standard of care for GBM patients. Despite this intensive treatment with conventional approaches, the management of GBM remains poor. The infiltrative nature of cancer cells makes the complete tumour removal by surgery virtually impossible. In addition, the blood-brain barrier’s (BBB) lack of permeability limits the number of effective chemotherapy drugs for GBM. Temozolomide (TMZ) is the most widely used chemotherapeutic agent for GBM because of its ability to pass the BBB. However, high systemic doses required to achieve brain therapeutic level, resulting in numerous side effects. The recurrence of GBM is almost inevitable due to the aforementioned shortcomings of conventional methods of treatment. Therefore, a great deal of effort has been focused on the development of new treatment methods capable of providing a high concentration of chemotherapy drug at the tumour site. Microspheres made from biodegradable polymers hold great potential to keep the chemotherapeutic agent intact within the carrier and locally deliver the drug over an extended period. However, the encapsulation of amphiphilic drug molecules such as TMZ within poly (d, l-lactide-co-glycolide) (PLGA) microspheres with conventional emulsion methods, oil-in-water (o/w), water-in-oil-in-water (w/o/w), is a major challenge. The extremely low encapsulation efficiencies obtained for TMZ-loaded PLGA microspheres using the aforementioned techniques (<7%) hampers the ability to scale up this process. Additionally, the injected microspheres to the tumour site tend to dislocate due to the cerebral flow which reduces the effectiveness of this localized drug delivery strategy. This study has focused on the development of a 3D bioprinted hydrogel-based mesh containing TMZ-loaded PLGA microspheres with high encapsulation efficiency (GlioMesh). To accomplish this, oil-in-oil (o/o) emulsion solvent evaporation technique was used to prepare PLGA microspheres loaded with TMZ. The poor solubility of TMZ in the external oil phase, liquid paraffin, resulted in obtaining encapsulation efficiencies as high as 61%. We then used the 3D bioprinting technology to embed TMZ-loaded PLGA microspheres into an alginate mesh. This provides the advantage of immobilizing the microspheres at the tumour site. Additionally, the flexibility and porosity of 3D bioprinted mesh allow for easy implantation and nutrients transportation to the brain tissue. The incorporation of polymeric microspheres within alginate fibres led to achieving an extended release of TMZ over 50 days. The functionality of GlioMesh in inducing cell cytotoxicity was evaluated by performing in vitro cell viability tests on U87 human glioblastoma cells. Higher cytotoxic effects were observed in the case of treatment with GlioMesh compared to the free drug because of the sustained release properties of our mesh. These data suggest that GlioMesh holds great promise to be used as an implant in the treatment of GBM. / Graduate / 2019-04-19
112

Validating Transgenic Farmington Viruses for the Treatment of Glioblastoma Multiforme

Rowe, Katelynn January 2015 (has links)
Glioblastoma is the most common primary brain tumour in adults. Despite the aggressive standard of care currently used, median patient survival following treatment is only 14 months. Innovative treatment options are needed for these patients. Recently, oncolytic viruses have emerged as promising immunotherapies for the treatment of solid tumours. Preliminary work in our lab has demonstrated that Farmington virus, a novel brain-safe oncolytic rhabdovirus, can be engineered to encode a tumour-associated antigen (TAA) to prime and boost antigen-specific adaptive immune responses. Since other rhabdoviruses share this boosting capacity, a heterologous rhabdovirus prime/boost regimen can be designed to combine two powerful oncolytics and a robust anti-TAA adaptive immune response. We evaluated Farmington’s ability to vaccinate against a self- glioblastoma antigen and two foreign glioblastoma-associated antigens. Farmington was able to vaccinate against the foreign antigens, leading to efficacy in prophylactic and therapeutic glioblastoma models. Additionally, treatment with heterologous rhabdoviruses demonstrated efficacy in an aggressive murine mammary carcinoma model. Herein, we demonstrate promising preliminary results for a novel glioblastoma therapeutic approach. Le glioblastome est la tumeur primaire la plus fréquente chez l’adulte. La survie moyenne des patients n’excède pas 14 mois malgré une prise en charge thérapeutique agressive. Par conséquent, la mise au point de traitements innovants et efficaces est une nécessité pour ces patients. Des avancées récentes ont mise en évidence l’intérêt des virus oncolytiques dans le traitement des tumeurs solides. Des travaux préliminaires réalisés au sein de notre laboratoire ont, en effet, démontré que le virus Farmington pouvait être modifié afin d’exprimer un antigène associé aux tumeurs (AAT), pour initier et potentialiser une réponse immunitaire adaptative spécifique. D’autres rhabdovirus possèdent des capacités de potentialisation immunitaire similaires et peuvent être utilisés en association avec le virus Farmington modifié pour amorcer et amplifier la réponse immunitaire oncolytique de l’hôte. Le but de ce projet était d’évaluer le potentiel du virus Farmington comme vaccin contre des antigènes tumoraux d’origine endogène ou exogène associés au glioblastome. Nos résultats ont montré que le virus Farmington a la capacité d’induire une réponse immunitaire prophylactique et thérapeutique contre les antigènes tumoraux exogènes dans des modèles de glioblastome. De plus, l’utilisation de rhabdovirus hétérologues s’est aussi révélée efficace pour le traitement de carcinome mammaire agressif chez la souris. Cette étude préliminaire apporte des résultats prometteurs pour le développement de nouvelles approches thérapeutiques efficaces dans le traitement du glioblastome.
113

Correlação clínico-topográfica em glioblastomas multiformes nas síndromes motoras: significados fisiopatológicos

de Cássia Guimarães Lucena, Rita January 2005 (has links)
Made available in DSpace on 2014-06-12T23:03:12Z (GMT). No. of bitstreams: 2 arquivo8791_1.pdf: 2283735 bytes, checksum: 1046687aa31d4b605b161ab475c9b576 (MD5) license.txt: 1748 bytes, checksum: 8a4605be74aa9ea9d79846c1fba20a33 (MD5) Previous issue date: 2005 / O Glioblastoma Multiforme (GBM) é o tumor glial com maior grau de malignidade. Acomete principalmente os hemisférios cerebrais apresentando sintomas e sinais focais ou gerais, relacionados ao tamanho, localização e taxa de crescimento tumoral. Objetivo: Analisar a relação do déficit motor com a topografia do GBM. Método: Foram estudados 43 casos de GBM, referidos quanto à idade, sexo, localização e a síndrome motora. Resultados: o tumor predominou em adultos (média de 55 anos), sexo masculino (55,82%), localização frontal (aproximadamente 40%). A hemiparesia prevaleceu como distúrbio motor, somente não ocorrendo em 2 casos de lesão frontal, 2 temporais, 1 parietal, 1 occipital e 1 fronto-temporal. Conclusão: Os achados clínico-topográficos favorecem os efeitos infiltrativos (lesões extensas) como responsáveis pela síndrome motora em detrimento aos efeitos compressivos (lesões localizadas)
114

The Diverse Roles of Non-muscle Myosin II in Tumorigenesis

Picariello, Hannah Stubbs 28 August 2019 (has links)
No description available.
115

Recapitulating Brain Tumor Microenvironment with In Vitro Engineered Models

Cui, Yixiao January 2020 (has links)
No description available.
116

Evaluation préclinique de l'impact des facteurs HAF et HIF-2 sur la croissance des glioblastomes et leur réponse à la radiothérapie / Preclinical evaluation of the impact of HAF and HIF-2 on glioblastoma growth and response to radiotherapy

Lambert, Gaelle 11 December 2018 (has links)
L’hypoxie tumorale est l’une des principales causes de l’agressivité des glioblastomes (GB). Plusieurs études attestent de l’implication de l’isoforme HIF-1α (hypoxia inducible factor-1α) dans la progression de ces tumeurs et dans leur résistance à la radiothérapie (RT). Plus récemment, il a été établi que l’isoforme HIF-2α régule la capacité tumorigénique des cellules souches de GB (CSG). Cependant, le rôle de ce facteur dans la croissance des cellules de GB différenciées et leur réponse à la RT est moins documenté. Dans ce contexte, l’objectif de ce travail a été de renforcer ces connaissances à l’échelle préclinique en utilisant deux approches d’ARN interférence (ARNi) pour moduler l’expression de HIF-2 : cibler directement HIF-2α ou cibler HAF (hypoxia associated factor), un facteur impliqué dans le switch de HIF-1α vers HIF-2α. Les résultats obtenus sur un modèle orthotopique de cellules humaines de GB (U251-MG) différenciées montrent que l’invalidation de HAF conduit à un fort ralentissement de la croissance de ces tumeurs mais indépendamment de HIF-1α ou HIF-2α. L’effet de l’invalidation de HIF-2α serait, quant à lui, dépendant de l’environnement tumoral. En effet, la diminution d’expression de HIF-2α dans les cellules U251 ne modifie pas la croissance tumorale dans un modèle de greffe sous-cutanée, alors que celle-ci favorise la croissance tumorale lorsque les cellules de GB sont implantées en intracérébral. Par comparaison aux tumeurs contrôles, ces tumeurs sont plus invasives et mieux perfusées. In vitro, l’inhibition de l’expression de HIF-2α n’a aucun effet sur la survie des cellules U251 alors qu’elle diminue la mort apoptotique de ces cellules exposées aux rayons X.L’ensemble des données présentées dans cette étude suggère que HAF et HIF-2α pourraient réguler la capacité tumorigénique des cellules de GB différenciées, tout comme observé pour les CSG. En outre, ces résultats soulignent la nécessité de prendre en compte le microenvironnement cellulaire afin de mieux comprendre le comportement de la tumeur dans son environnement hypoxique. / Hypoxia is one of the main causes of glioblastoma (GB) aggressiveness. Various studies attest on the involvement of the HIF-1α isoform (hypoxia inducible factor-1α) in the progression of these tumors and in their resistance to radiation therapy (RT). More recently, it was established that the HIF-2α isoform regulates the tumorigenic capacity of GB stem cells (GSC). However, the role of this factor in the growth of differentiated GB cells and their response to RT is less documented. In this context, the goal of this work was to strengthen this knowledge at the preclinical level by using two RNA interference (RNAi) strategies to modulate the expression of HIF-2: one directly targets HIF-2α, the other one targets HAF (hypoxia associated factor), a factor involved in the switch of HIF-1α to HIF-2α. Our results obtained on an orthotopic model of differentiated human GB (U251-MG) cells showed that the invalidation of HAF leads to a strong slowdown in tumor growth but independently of HIF-1α or HIF-2α. On the other hand, the effect of HIF-2α silencing seems dependent on the tumor environment. Indeed, the extinction ofHIF-2α expression in U251 cells does not modify tumor growth in a subcutaneous model, whereas it promotes tumor growth when GB cells are intracerebrally grafted. Compared to control tumors, these tumors are more invasive and highly perfused. In vitro, the inhibition of HIF-2α expression has no effect on GB cell survival whereas decreasing the X-rays induced apoptotic death.Collectively, these data suggest that HAF and HIF-2α could regulate the tumorigenic capacity of differentiated GB cells, like it does in CSGs. In addition, these results highlight the need to take into account the cellular microenvironment to better understand the behavior of the tumor in its hypoxic environment.
117

Spatial and genomic analysis of the glioblastoma tumor microenvironment

Chen, Andrew January 2020 (has links)
Glioblastoma (GBM) is an aggressive brain cancer with devastating outcomes and few effective treatments. Although immunotherapy has shown promise in treating a variety of cancers, it is still unclear if and how it can be effectively used in GBM. Elucidating this will require a better understanding of the mechanistic role of immune cells and their interactions in the GBM tumor microenvironment. This thesis utilizes recent technological developments in cancer genomics and imaging to study the mechanisms underlying immunotherapy and the tumor microenvironment. First, we will provide background on our current understanding of GBM, its immune microenvironment, as well as modern sequencing and imaging methods. Second, we will present a longitudinal study of GBM patients before and after treatment with PD-1 immunotherapy. Only a small fraction of GBM patients respond to this type of therapy, so we perform genomic, transcriptomic, and spatial analyses to compare the molecular features of these rare responders versus non-responders. We show that clinical response to PD-1 immunotherapy in GBM is associated with specific molecular alterations and immune infiltration profiles that reflect the tumor’s clonal evolution during treatment. The most common infiltrating immune cells in GBM are macrophages, which are implicated in a wide variety of pro-tumor and anti-tumor roles. We then focus on this specific immune population by analyzing single-cell expression data from GBM tumors. We identify a novel macrophage subpopulation characterized by expression of the scavenger receptor MARCO, which drives tumor progression in GBM and is altered over the course of PD-1 immunotherapy. Next, we demonstrate that the methods we have developed for GBM are applicable to understanding the tumor microenvironments of other cancers as well. We analyze a cohort of melanoma cases to show that transcriptomic and imaging features can be combined to create a biomarker that stratifies patients into different risk groups. Finally, while most of the image analysis described so far has utilized histopathology, we include two appendices where we demonstrate new ways to process and analyze Magnetic Resonance Imaging (MRI) in GBM.
118

Lymfom centrálního nervového systému v obraze magnetické rezonance. / Magnetic resonance imaging of central nervous system lymphoma.

Koubská, Eva January 2020 (has links)
Background: The aim of this study was to describe the morphological signs of the central nervous system lymphoma (CNSL) in magnetic resonance imaging (MRI). We compared morphological characteristics of primary CNSL (PCNSL) and secondary CNSL (SCNSL) and also of PCNSL and glioblastoma (GBM). Methods: We included 64 patients with PCNSL (ten of them were immunocompromised), 21 patients with SCNSL and 54 patients with GBM. The diagnosis was confirmed histologically in all patients. We evaluated morphological signs on the first MRI examination. Additionally, in patients with PCNSL, we evaluated the development of the disease on follow-up examination before histological confirmation of the diagnosis, if available. Results: In most patients with PCNSL (62.5%) the tumor was localized supratentorially and presented as multiple lesions (53.1%) or as a diffuse infiltrative lesion (23.4%). In 87.5% of the patients the lesions reached the brain surface. Infiltration of ependyma was seen in 56.3%, infiltration of meninges in 39.1% and infiltration of cranial nerves in 48.5% of patients. Restriction of diffusion in some part of the tumor was apparent in nearly all patients (97.6%) with PCNSL. After administration of contrast media, marked enhancement was usually seen. In immunocompetent patients, homogenous...
119

A Case History of Glioma Progression

Ohgaki, Hiroko, Watanabe, Kunihiko, Peraud, Aurelia, Biernat, Wojciech, Von Deimling, Andreas, Yasargil, M. Gazi, Yonekawa, Yasuhiro, Kleihues, Paul 01 May 1999 (has links)
Low-grade diffuse astrocytomas have an intrinsic tendency for malignant progression but the factors determining the kinetics of this process are still poorly understood. We report here the case of a male patient who developed a fibrillary astrocytoma at the age of 33 years and who underwent six surgical interventions over a period of 17 years without radiotherapy or chemotherapy. The first three biopsies spanned a period of 11 years and led to the diagnosis of low-grade, diffuse astrocytoma (WHO grade II), with a growth fraction (MIB-1 labeling index) of 2.3-3.7%. The fourth to sixth biopsies showed histological features of anaplastic astrocytoma (WHO grade III), with growth fractions between 5.0 and 10.5%. The fraction of gemistocytic neoplastic astrocytes also increased, from 0.3% in the first biopsy to 17.5% in the last biopsy and preceded the increase in proliferative activity and transition to anaplastic astrocytoma. The fraction of tumor cells immunoreactive to BCL-2 increased from 0.3% to 8.2%. A p53 mutation in codon 273 (CGT→TGT, Arg→Cys) was identified in the first biopsy and persisted throughout the course of the disease. However, the fraction of cells with p53 protein accumulation increased significantly during progression, from 3.2% in the first biopsy to 13.7% in the last. The absence of additional genetic alterations (PTEN mutations, loss of chromosome 10 and 19q) may be responsible for the slow progression and lack of glioblastoma features even after a 17-year disease duration.
120

Characterization and Therapeutic Targeting of Surface Markers in Glioblastoma Pre-Clinical Models

SAVAGE, NEIL January 2023 (has links)
Glioblastoma (GBM) remains the most aggressive primary brain tumor in adults. Since 2005, Standard of Care (SoC) consists of surgical resection followed by radiation and adjuvant chemotherapy with temozolomide. Treatment failure is attributed to intratumoral heterogeneity with populations capable of mechanisms to repair damaged DNA. Given the lack of progress to improve patient outcomes, the current work encompasses how multi-omic approaches can be utilized to uncover novel biology in GBM and develop precision medicines to exploit these cancer specific phenomena. Using patient derived GBM samples I first used the surface marker CD133 to interrogate glioblastoma stem cells, a subpopulation of cells identified to withstand conventional therapies and lead to tumor relapse. I used a genome-wide CRISPR-Cas9 library to conduct an unbiased loss-of-function phenotypic screen to identify regulators of CD133. I then validated SOX2 as a direct transcription factor to PROM1 encoding CD133. These findings further show the untapped potential of CRISPR to uncover novel biology to directly apply to broader fields of stem cells and cancer biology. Next, I combed GBM data sets at transcriptomic and proteomic levels to identify understudied proteins as potential targets for immunotherapies. Glycoprotein nonmetastatic melanoma protein B (GPNMB) has previously been identified as a clinically relevant target in GBM and shown to be active in the tumor immune microenvironment. I found GPNMB to be upregulated in recurrent GBM and macrophage populations which can be exploited in a more comprehensive manner to treat GBM. Through a series of models, I elucidated how GPNMB influences GBM biology, its effectiveness as a target for Chimeric Antigen Receptor T-cells, and how it can be paired with CD133 therapies to provide better coverage of tumor cells. Together, these studies highlight how advances in pre-clinical models and technologies can be leveraged to develop new therapies in a rational manner. / Thesis / Doctor of Science (PhD) / Glioblastoma (GBM) remains an aggressive and incurable brain cancer despite decades of intense research. Treatment failure is due to the untargeted approaches currently undertaken in the clinic. The current work uses multiples methods to interrogate how GBM grows and develops over time. Using GBM samples from consenting patients, I investigated an important population of the tumor using a surface marker CD133 and CRISPR to study which genes influenced it. I then successfully validated SOX2 as a direct regulator of CD133 expression. Next, I combed multiple data sets for a target to kill GBM cells without harming healthy tissue in patients. I found Glycoprotein Non-Metastatic Melanoma Protein B (GPNMB) to be exploitable and used several experimental methods to investigate its role in GBM progression. Finally, we used a novel immunotherapy to eliminate cells which express GPNMB. Together, these findings could apply to the broader field of stem cell biology and be used for a more targeted method to eliminate the cancer entirely.

Page generated in 0.0311 seconds