• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 34
  • 7
  • 6
  • 6
  • 4
  • 4
  • 2
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 76
  • 51
  • 44
  • 28
  • 23
  • 16
  • 9
  • 8
  • 8
  • 7
  • 7
  • 7
  • 6
  • 6
  • 6
  • 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

Avaliação da atividade do CHY-1, um novo análogo da miltefosina, como potencial inibidor da enzima CTP: fosfoetanolamina-citidilil-transferase, sobre o carcinoma de pulmão de não-pequenas células. / Evaluation of the activity of CHY-1, a novel miltefosine analogue, as a potential CTP: phosphoethanolamine cytidylyltransferase enzyme inhibitor against non-small cell lung cancer.

Sarah Fernandes Teixeira 18 August 2016 (has links)
O câncer de pulmão é um dos mais incidentes e letais, e, assim, a busca de novos fármacos é necessária. Atualmente o desenvolvimento de fármacos conta com abordagens computacionais que otimizam este processo. Dado que a fosfatidiletanolamina desempenha importantes papeis fisiológicos e uma das enzimas envolvidas na sua síntese, a CTP:fosfoetanolamina-citidilil-transferase (Pcyt2) é frequentemente superexpressa em células de câncer de pulmão, no presente trabalho, foram avaliados o potencial terapêutico de CHY-1, um análogo da miltefosina desenvolvido como inibidor da enzima Pcyt2, e os mecanismos inerentes à sua atividade antitumoral. O CHY-1 apresentou citotoxicidade superior ao seu protótipo e a outro inibidor da enzima Pcyt2, a meclizina. Além disso, as células malignas foram mais sensíveis ao CHY-1 do que as células não-tumorigênicas. Em conclusão, o presente trabalho evidencia o potencial do CHY-1 como um inibidor da enzima Pcyt2 e candidato a fármaco com atividade preferencial para câncer de pulmão. / Lung cancer is one of the most incident and lethal cancers, thus, the pursuit for new drugs is necessary. Nowadays, new drugs development has computational tools that improves this process. Once that phosphatidylethanolamine plays several important physiological roles and one of the enzymes of its production pathway, CTP:phosphoethanolamine cytidylyltransferase (Pcyt2), is usually overexpressed in lung cancer cells, therefore, this study aimed was to evaluate the antitumor effects of CHY-1, a miltefosine analogue developed as an inhibitor of Pcyt2 enzyme, and to investigate the mechanisms related to its antitumor action. CHY-1 was more cytotoxicity than its prototype, miltefosine, and was more cytotoxic than another inhibitor Pcyt2 enzyme, meclizine. Morevover, malignant cells were more sensitive to CHY-1 effects than non-tumorigenic cells. In conclusion, this work presents CHY-1 as an inhibitor of Pcyt2 enzyme and new candidate a drug with preferential activity on NSCLC cells.
62

Towards Novel Effective Combination Therapy for KRAS Mutant Non-Small Cell Lung Cancer

Kurim, Sara 12 April 2018 (has links)
Non-small-cell lung cancer (NSCLC) accounts for 80–85% of all lung cancers and is associated with significant mortality. As epidermal-growth-factor receptor (EGFR) is over-expressed in 80-90% of NSCLC, its inhibition via EGFR-Tyrosine Kinase inhibitors (EGFR-TKIs) is a main therapeutic strategy. However, patients with mutations in KRAS are resistant to EGFR-TKIs. A study in mutant KRAS-driven lung cancer in transgenic mice showed that tumor growth was dependent on the activity of focal adhesion kinase (FAK). Therefore, we hypothesized that KRAS-mutant NSCLC will be sensitive to FAK-TKIs and, given known FAK-EGFR cross-talk, FAK inhibition will sensitize KRAS-mutant NSCLC to EGFR-TKIs. We performed cell viability assays of WT versus mutant KRAS NSCLC cell lines following treatment with FAK-TKI alone or in combination with a clinically relevant EGFR-TKI. We found that KRAS-mutant cells were more sensitive to FAK-TKI than KRAS-WT NSCLC. In addition, we found that the combination treatment including FAK and EGFR TKIs resulted in reduced tumor cell viability as compared to treatment with either drug alone. This enhanced anti-tumor response could be due to FAK-TKI’s ability to down-regulate EGFR downstream targets. Our preliminary data suggests that in KRAS-mutant cells the drug combination appears to more effectively inhibit Akt activity than single drug treatment alone. This suggests an enhanced ability to impair cell survival following treatment with the drug combination. We also found that treatment with FAK TKI in KRAS mutant NSCLC cells resulted in increased activation of EGFR which was due in part to modulation of EGFR recycling and production of endogenous EGFR ligands. Thus, the combination of FAK- and EGFR-TKIs may be more effective in KRAS mutant NSCLC as treatment with EGFR-TKI overcomes the unexpected ‘side effect’ of treatment with FAK-TKI, namely activation of the EGFR pathway by this drug. The findings of our study are novel and have uncovered previously unrecognized outcomes of FAK inhibition on EGFR activity. Moreover, our data support the notion that the combination of FAK- and EGFR-TKIs could be an effective treatment for KRAS mutant NSCLC patients.
63

BREAKING BARRIERS: BLOOD-BRAIN BARRIER PARADIGMS IN BRAIN METASTASES OF LUNG CANCER

Alexandra M Dieterly (9714149) 15 December 2020 (has links)
<p>A multitude of neurologic diseases are increasing in patients that both diminish quality and quantity of life. My dissertation research focused on unraveling the blood-brain barrier’s alterations (BBB), primarily in lung cancer brain metastases, the most common brain metastasis in patients. We optimized a reliable and reproducible mouse model for creating brain metastases using patient derived brain seeking cells of non-small lung cancer (NSCLC) using ultrasound-guided intracardiac injection. I then evaluated brain tissue with qualitative and quantitative immunofluorescence for individual components of the BBB. Using this experimental method, I was able to identify the specific shift of each BBB component over time in NSCLC brain metastases. I then used human brain metastases specimens to demonstrate the clinical relevance of my findings. These results show distinct alterations in the BBB, which have the potential for targeting therapeutic delivery to extend patient survival. I was also able to characterize a novel epithelial-mesenchymal (EMT) phenotype in vertebral metastases of NSCLC in our model, with features similar to those seen in human patients. Most recently, I analyzed patterns of paracellular permeability associated with each BBB component of NSCLC brain metastases which may provide direct passageways for therapeutic delivery. Altogether, this research offered foundational evidence for the future development of targeted novel therapeutics, including nanoparticles. Outside of the brain metastases field, we used an experimental framework to successfully characterize the BBB alterations in a traumatic brain injury model (bTBI). These findings provided the first description of this unique pathology and the framework for developing therapeutics in other neurologic diseases. Although my research work has focused on animal models of disease, future directions based on my research work include the developing a novel 3D BBB-on-chip device to enable high throughput novel therapeutic delivery through the BBB. Long-term, identifying targetable alterations in the restrictive BBB using <i>in vitro</i> and <i>in vivo</i> models provides a potential conduit for effective prevention and treatment of a myriad of neurologic diseases to prolong patient survival and quality of life.</p>
64

Regulation of ERK3 by KRAS signalling and its role in the growth of lung adenocarcinoma (LUAD) cells

Akunapuram, Shreya 09 August 2023 (has links)
No description available.
65

Développement d’un modèle de co-culture en trois dimensions de cellules de cancer du poumon et de fibroblastes

Sy, Emmanuel 08 1900 (has links)
Le cancer du poumon non-à-petites-cellules (CPNPC) représente 85% des cas de cancer du poumon. Cependant, les modèles utilisés de culture cellulaire en deux dimensions (2D) représentent partiellement les caractéristiques physiopathologiques du CPNPC. Notre objectif est de réaliser un modèle in vitro plus représentatif des caractéristiques de ce type de cancer. La culture tridimensionnelle (3D) dans laquelle les cellules forment un sphéroïde, est considérée comme un modèle plus fidèle aux tumeurs retrouvées chez les patients grâce à la structure et aux interactions intercellulaires du modèle. Pour mieux représenter le microenvironnement tumoral, nous intégrons une lignée cellulaire de fibroblastes dans le sphéroïde de CPNPC, afin d’imiter l’interaction entre cellules cancéreuses et cellules stromales. En effet, bien que les fibroblastes représentent un faible pourcentage des cellules au sein des tumeurs de CPNPC, elles jouent un rôle prépondérant dans la biologie tumorale et la réponse aux médicaments. Nous avons testé différentes lignées de fibroblastes et différents ratios de co-culture afin de déterminer les conditions optimales de notre modèle de co-culture 3D. Après 7 jours de co-culture, les cellules cancéreuses démontrent un potentiel migratoire plus élevé lorsque mesuré dans des chambres de Boydens. Cet effet n’est dépendant ni de la prolifération, ni d’un changement de phase dans le cycle cellulaire. Nous avons caractérisé la localisation des fibroblastes au sein du sphéroïde par des expériences de microscopie à fluorescence. L’expression de la protéine α-SMA du cytosquelette a aussi été déterminée par immunofluorescence. Nous avons par la suite établi un modèle de co-culture sur 24 jours afin de maximiser la communication entre les cellules cancéreuses et les fibroblastes. Ce modèle de co-culture long terme a par la suite été analysé selon son contenu en cytokines, chimiokines et métabolites. Enfin, nous avons réalisé un criblage de médicaments au jour 24 de notre co-culture long terme afin d’évaluer une réponse thérapeutique des cellules cancéreuses, dans des conditions plus semblables au microenvironnement tumoral. / Non-small cell lung cancer (NSCLC) accounts for 85% of all lung cancer cases. However, the two-dimensional (2D) cell culture models used represent partially the pathophysiological characteristics of NSCLC. Our goal is to develop an in vitro model that is more representative of the characteristics of this type of cancer. The three-dimensional (3D) culture, in which the cells form a spheroid, is considered to be a model more faithful to the tumors found in patients due to the structure and intercellular interactions of the model. In order to better represent the tumor microenvironment, we are integrating a fibroblast cell line into the spheroid of NSCLC to mimic the interaction between cancer cells and stromal cells. Although fibroblasts represent a small percentage of cells in NSCLC tumors, they play a major role in tumor biology and drug response. We tested different fibroblast cell lines and co-culture ratios to determine the optimal conditions of our 3D co-culture model. After 7 days of co-culture, the cancer cells show a higher migratory potential when measured in Boydens chambers. This effect is not dependent on proliferation or change of phase in the cell cycle. We characterized the localization of fibroblasts within the spheroid by fluorescence microscopy experiments. The expression and localization of the cytoskeletal protein α-SMA was also determined by immunofluorescence. We then established a 24-day co-culture model to maximize communication between cancer cells and fibroblasts. This long-term co-culture model was subsequently analyzed for cytokine, chemokine and metabolite content. Finally, we performed drug screening on day 24 of our long-term co-culture to evaluate a therapeutic response of cancer cells under conditions more similar to the tumor microenvironment.
66

LKB1 Loss in Lung Adenocarcinoma

Koenig, Michael J. 28 August 2019 (has links)
No description available.
67

The role of MMP10 in non-small cell Lung cancer, and pharmacological evaluation of its potential as a target for therapeutic intervention. Investigation of the role of MMP10 in the tumour microenvironment of non-small cell lung cancer using gene, protein and mass spectrometry approaches to determine MMP10’s potential in drug development strategies

Bin Saeedan, Abdulaziz S.A. January 2014 (has links)
Non-Small Cell Lung Cancer (NSCLC), which accounts for 80% of all lung cancer cases, is associated with resistance to chemotherapy and poor prognosis. Exploitation of NSCLC-upregulated pathways that can either be targeted by novel therapeutics or used to improve the tumour-delivery of current chemotherapeutics are required. Among the matrix metalloproteinases (MMPs) that are essential for tumour development, MMP10 is a potential candidate as a therapeutic target based on its expression and contribution to NSCLC development. This research aims to explore the expression and functions of MMP10 in the tumour microenvironment of NSCLC and evaluate the potential of MMP10 as a target for therapeutic intervention. Herein, MMP10 expression at gene and protein levels were analysed in a panel of NSCLC cell lines using RT-PCR and Western blotting analysis. To determine MMP10 functional relevance, an in vitro angiogenesis assay using cell conditioned media was carried out. To identify specific peptide sequences for the design of prodrugs rationalised to be MMP10 activated, in vitro substrate cleavage studies were performed using a mass spectrometry approach to differentiate between MMP10 and the structurally similar MMP3. This study demonstrates that MMP10 is highly expressed in NSCLC and that high levels of MMP10 are associated with induction of angiogenesis, a crucial process supporting tumour growth. In addition to the achievement of having been able to differentiate between closely similar MMP3 and MMP10 through carefully monitoring the hydrolysis rate of compound 444259 (a known MMP substrate), data generated herein provides the basis for further studies to exploit MMP10 as a prodrug-activator. / Full text was made available at the end of the embargo period, 12th Dec 2019
68

PRONTOX – proton therapy to reduce acute normal tissue toxicity in locally advanced non-small-cell lung carcinomas (NSCLC): study protocol for a randomised controlled trial

Zschaeck, Sebastian, Simon, Monique, Löck, Steffen, Troost, Esther G. C., Stützer, Kristin, Wohlfahrt, Patrick, Appold, Steffen, Makocki, Sebastian, Bütof, Rebecca, Richter, Christian, Baumann, Michael, Krause, Mechthild 17 March 2017 (has links) (PDF)
Background Primary radiochemotherapy with photons is the standard treatment for locally advanced-stage non-small cell lung cancer (NSCLC) patients. Acute radiation-induced side effects such as oesophagitis and radiation pneumonitis limit patients’ quality of life, and the latter can be potentially life-threatening. Due to its distinct physical characteristics, proton therapy enables better sparing of normal tissues, which is supposed to translate into a reduction of radiation-induced side effects. Methods/design This is a single-centre, prospective, randomised controlled, phase II clinical trial to compare photon to proton radiotherapy up to 66 Gy (RBE) with concomitant standard chemotherapy in patients with locally advanced-stage NSCLC. Patients will be allocated in a 1:1 ratio to photon or proton therapy, and treatment will be delivered slightly accelerated with six fractions of 2 Gy (RBE) per week. Discussion The overall aim of the study is to show a decrease of early and intermediate radiation-induced toxicity using proton therapy. For the primary endpoint of the study we postulate a decrease of radiation-induced side effects (oesophagitis and pneumonitis grade II or higher) from 39 to 12%. Secondary endpoints are locoregional and distant failure, overall survival and late side effects. Trial registration Registered at ClinicalTrials.gov with Identifier NCT02731001 on 1 April 2016.
69

Retrospektive Analyse von Diagnostik, Klinik und Verlauf bei Patienten mit Vena-cava-superior-Syndrom (obere Einflussstauung) / A retrospective analysis of the diagnosis, treatment and course in patients with superior vena cava syndrome

Bertram, Nick 04 March 2015 (has links)
No description available.
70

PRONTOX – proton therapy to reduce acute normal tissue toxicity in locally advanced non-small-cell lung carcinomas (NSCLC): study protocol for a randomised controlled trial

Zschaeck, Sebastian, Simon, Monique, Löck, Steffen, Troost, Esther G. C., Stützer, Kristin, Wohlfahrt, Patrick, Appold, Steffen, Makocki, Sebastian, Bütof, Rebecca, Richter, Christian, Baumann, Michael, Krause, Mechthild 17 March 2017 (has links)
Background Primary radiochemotherapy with photons is the standard treatment for locally advanced-stage non-small cell lung cancer (NSCLC) patients. Acute radiation-induced side effects such as oesophagitis and radiation pneumonitis limit patients’ quality of life, and the latter can be potentially life-threatening. Due to its distinct physical characteristics, proton therapy enables better sparing of normal tissues, which is supposed to translate into a reduction of radiation-induced side effects. Methods/design This is a single-centre, prospective, randomised controlled, phase II clinical trial to compare photon to proton radiotherapy up to 66 Gy (RBE) with concomitant standard chemotherapy in patients with locally advanced-stage NSCLC. Patients will be allocated in a 1:1 ratio to photon or proton therapy, and treatment will be delivered slightly accelerated with six fractions of 2 Gy (RBE) per week. Discussion The overall aim of the study is to show a decrease of early and intermediate radiation-induced toxicity using proton therapy. For the primary endpoint of the study we postulate a decrease of radiation-induced side effects (oesophagitis and pneumonitis grade II or higher) from 39 to 12%. Secondary endpoints are locoregional and distant failure, overall survival and late side effects. Trial registration Registered at ClinicalTrials.gov with Identifier NCT02731001 on 1 April 2016.

Page generated in 0.0386 seconds