• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 241
  • 74
  • 36
  • 35
  • 12
  • 10
  • 8
  • 6
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • Tagged with
  • 485
  • 485
  • 179
  • 116
  • 91
  • 90
  • 80
  • 63
  • 54
  • 50
  • 49
  • 46
  • 43
  • 42
  • 38
  • 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.
161

Mechanism and Therapeutic Potential of Statin-Mediated Inhibition of Tyrosine Kinase Receptors

Zhao, Tong Tong 27 October 2011 (has links)
Receptor tyrosine kinases (RTK) are key regulators of growth, differentiation and survival of epithelial cells and play a significant role in the development and progression of cancers derived from these tissues. In malignant cells, these receptors and their downstream signalling pathways are often deregulated, leading to cell hyper-proliferation, enhanced cell survival and increased metastatic potential. Furthermore, endothelial expressed RTKs regulate tumor angiogenesis allowing for tumor growth and maintenance by promoting their vascularization. Epithelial malignancies such as squamous cell carcinomas (SCC), non-small cell lung (NSCLC) and malignant mesotheliomas have very limited treatment options when presenting as metastatic disease. RTKs, particularly the epidermal growth factor (EGFR) and the vascular endothelial growth factor (VEGFR) receptors, have been shown to play significant roles in the pathogenesis of these tumor types. Statins are potent inhibitors of HMG-CoA reductase, the rate limiting enzyme of the mevalonate pathway, that are widely used as hypercholesterolemia treatments. The mevalonate pathway produces a variety of end products that are critical for many different cellular pathways, thus, targeting this pathway can affect multiple signalling pathways. Our laboratory has previously shown that lovastatin can induce tumor specific apoptosis especially in SCC and that 23% of recurrent SCC patients treated with lovastatin as a single agent showed disease stabilization in our Phase I clinical trial. Subsequently, our lab was able to demonstrate that lovastatin in combination with gefitinib, a potent inhibitor of the EGFR showed co-operative cytotoxicity when combined (Chapter 2). Furthermore, the pro-apoptotic and cytotoxic effects of these agents were found to be synergistic and to be manifested in several types of tumor cell lines including SCC, NSCLC and glioblastoma. I was able to expand upon these important findings and demonstrated that lovastatin, through its ability to disrupt the actin cytoskeleton, inhibited EGFR dimerization and activation (Chapter 3). This novel mechanism targeting this receptor has clinical implications as lovastatin treatment combined with gefitinib showed co-operative inhibitory effects on EGFR activation and downstream signalling. The RTK family of proteins share similar features with respect to activation, internalization and downstream signalling effectors. I further demonstrated that lovastatin can inhibit the VEGFR-2 in endothelial cells and mesotheliomas, where VEGF and its receptor are co-expressed driving their proliferation, and induces synergistic cytotoxicity in mesothelioma cells in combination with VEGFR-2 tyrosine kinase inhibitors (Chapter 4). These findings suggest that statins may augment the effects of a variety of RTK inhibitors in a similar fashion representing a novel combinational therapeutic approach in a wide repertoire of human cancers. More importantly, based on this work, we initiated a Phase I/II study evaluating high dose rosuvastatin and the EGFR inhibitor tarceva in SCC and NSCLC patients at our institute. This clinical evaluation will provide invaluable data that will play a role in developing this novel therapeutic strategy. Together, the work embodied in this thesis provides a model for the regulation of EGFR/VEGFR-2 activation and signalling by targeting the rho family of proteins that demonstrates a novel mechanism that can be exploited to refine current therapeutic paradigms.
162

Mechanism and Therapeutic Potential of Statin-Mediated Inhibition of Tyrosine Kinase Receptors

Zhao, Tong Tong 27 October 2011 (has links)
Receptor tyrosine kinases (RTK) are key regulators of growth, differentiation and survival of epithelial cells and play a significant role in the development and progression of cancers derived from these tissues. In malignant cells, these receptors and their downstream signalling pathways are often deregulated, leading to cell hyper-proliferation, enhanced cell survival and increased metastatic potential. Furthermore, endothelial expressed RTKs regulate tumor angiogenesis allowing for tumor growth and maintenance by promoting their vascularization. Epithelial malignancies such as squamous cell carcinomas (SCC), non-small cell lung (NSCLC) and malignant mesotheliomas have very limited treatment options when presenting as metastatic disease. RTKs, particularly the epidermal growth factor (EGFR) and the vascular endothelial growth factor (VEGFR) receptors, have been shown to play significant roles in the pathogenesis of these tumor types. Statins are potent inhibitors of HMG-CoA reductase, the rate limiting enzyme of the mevalonate pathway, that are widely used as hypercholesterolemia treatments. The mevalonate pathway produces a variety of end products that are critical for many different cellular pathways, thus, targeting this pathway can affect multiple signalling pathways. Our laboratory has previously shown that lovastatin can induce tumor specific apoptosis especially in SCC and that 23% of recurrent SCC patients treated with lovastatin as a single agent showed disease stabilization in our Phase I clinical trial. Subsequently, our lab was able to demonstrate that lovastatin in combination with gefitinib, a potent inhibitor of the EGFR showed co-operative cytotoxicity when combined (Chapter 2). Furthermore, the pro-apoptotic and cytotoxic effects of these agents were found to be synergistic and to be manifested in several types of tumor cell lines including SCC, NSCLC and glioblastoma. I was able to expand upon these important findings and demonstrated that lovastatin, through its ability to disrupt the actin cytoskeleton, inhibited EGFR dimerization and activation (Chapter 3). This novel mechanism targeting this receptor has clinical implications as lovastatin treatment combined with gefitinib showed co-operative inhibitory effects on EGFR activation and downstream signalling. The RTK family of proteins share similar features with respect to activation, internalization and downstream signalling effectors. I further demonstrated that lovastatin can inhibit the VEGFR-2 in endothelial cells and mesotheliomas, where VEGF and its receptor are co-expressed driving their proliferation, and induces synergistic cytotoxicity in mesothelioma cells in combination with VEGFR-2 tyrosine kinase inhibitors (Chapter 4). These findings suggest that statins may augment the effects of a variety of RTK inhibitors in a similar fashion representing a novel combinational therapeutic approach in a wide repertoire of human cancers. More importantly, based on this work, we initiated a Phase I/II study evaluating high dose rosuvastatin and the EGFR inhibitor tarceva in SCC and NSCLC patients at our institute. This clinical evaluation will provide invaluable data that will play a role in developing this novel therapeutic strategy. Together, the work embodied in this thesis provides a model for the regulation of EGFR/VEGFR-2 activation and signalling by targeting the rho family of proteins that demonstrates a novel mechanism that can be exploited to refine current therapeutic paradigms.
163

A synthesis and biological screening of predicted inhibitors of Tyrosine Kinases, e.g. KDR, designed in silico / Synthèse et screening biologique d'inhibiteurs de tyrosine kinase, KDR, conçus in silico

Šramel, Peter 30 November 2017 (has links)
Les protéines kinases représentent le groupe d'enzymes qui servent d'intermédiaire pour la phosphorylation de protéines - le transfert d'un groupe phosphate de l'adénosine triphosphate(ATP) sur des chaînes latérales correspondantes de tyrosine, de serine ou de thréonine des acides aminées. La phosphorylation de protéines est un des outils les plus importants pour la régulation de l'activité cellulaire. La « signalisation » cellulaire par le récepteur de tyrosine kinase VEGFR2 (KDR) appartient aux réactions biochimiques clés influençant la croissance de tumeurs. L'inhibition thérapeutique de cette réaction à l'aide des composés de faible poids moléculaire spécifiques est devenue une stratégie utile dans le cadre des thérapies anticancéreuses. Ce travail a amené à la découverte de 16 substances biologiquement actives sur la base N,5-diaryloxazol-2-amine (IC50, VEGFR2 TK). D'excellents résultats ont été atteints notamment dans le cas des substances 189, 191, 211, 214, 220, 221, 223 et 4 qui montrent une activité inhibitrice inférieure à 500 nM. / Protein kinases represent a group of enzymes responsible for phosphorylation - transfer of aphosphate group from adenosine triphosphate (ATP) to tyrosine or serine/threonine residues. Protein phosphorylation is one of the most important tools regulating a cell activity. A cell "signalization" through an endothelial receptor tyrosine kinase VEGFR2 TK (KDR) is the important pathway influencing growth of a tumor. Small-molecule inhibitors of VEGFR2 TK (VEGFR2 TKls) have become an important tool for the treatment of various types of cancer. This dissertation thesis resulted in a discovery of 16 biologically active N,5-diaryloxazol-2-amines (IC50, VEGFR2 TK). Very good results were achieved especially with compounds 189, 191, 211, 214, 220, 221, 223 and 4 exhibiting the activity under 500 nM.
164

La surexpression et l'activation des récepteurs aux facteurs de croissance par des régulations autocrines ou paracrines à la neurotensine, conférant aux cellules une sensibilité aux inhibiteurs de tyrosine kinase / The overexpression and activation of growth factor receptor by neurotensin autocrine and paracrine regulation, confer on cells a sensitivity to tyrosine kinase inhibitors

Wu, Zherui 03 June 2015 (has links)
Les cancers hépatiques, bronchiques et mammaires sont responsables de 35 % de décès par le cancer en 2012. Les études sur des facteurs contribuant à la progression tumorale devraient approfondir nos connaissances sur la biologie de ces cancers et ouvrir de nouvelles voies pour le développement de stratégies thérapeutiques. Dans ce contexte, nous avons étudié l'impact de la neurotensine (NTS) et de son récepteur NTSR1 sur la progression tumorale et son rôle potentiel pour de futures applications cliniques. J’ai initié ce projet dans le carcinome hépatocellulaire (CHC) et participé aux projets dans le cancer du poumon et du sein qui avaient été initiés par les anciens doctorants de l’équipe. Dans le CHC, sur une série de 73 patients, la NTS et le NTSR1 ont été détectés dans respectivement 56 % et 64 % des cas. En utilisant deux modèles cellulaires nous avons montré que l’expression du NTSR1 est une cible de la voie wnt/β-caténine. Le couple NTS/NTSR1 augmente l’expression et l’activation de l’EGFR et favorise la croissance des tumeurs expérimentales et la capacité de migration et d’invasion des cellules. La régulation entre le complexe NTS/NTSR1 et les récepteurs des HERs a également été observée dans les cancers bronchiques et mammaires, la NTS induit l'expression et l'activation constitutive des récepteurs EGFR, HER2 et HER3, par l'intermédiaire de l'activation des métalloprotéinases qui libèrent les ligands “EGF-like” spécifiques d'EGFR et d’HER3. L’activation constitutive des HER par le couple NTS/NTSR1 mime les mutations activatrices des HERs, ainsi la réponse des tumeurs aux inhibiteurs de tyrosine kinase est potentialisée dans le cancer du poumon, du sein et du foie. / In 2012, Liver, lung and breast cancers represented 30% of new cancer cases, and 35% of cancer related deaths. Identification of factors contributing to tumor progression can strengthen our understanding of the cancer biology and suggest new therapeutic strategies. In this context, we studied the impact of neurotensin (NTS) and its receptor NTSR1 on tumor progression and its potential clinical application. I have initiated the project in hepatocellular carcinoma (HCC) and participated in the projects on lung and breast cancers initiated by former PhD students. In HCC, on a series of 73 patients, NTS and NTSR1 were detected in 56% and 64% of the cases, respectively. Meanwhile, I showed that NTSR1 expression is the target of the Wnt/¦Â-catenin pathway. The NTS / NTSR1 complex increases the expression and activation of EGFR and promotes the growth of experimental tumors and the ability of the cell for migration and invasion. The regulation between the NTS/NTSR1 complex and EGF receptors were also thoroughly studied in lung and mammary cancers. Indeed, NTS induced the expression and the constitutive activation of EGFR, HER2, and HER3, through the activation of metalloproteinases which released specific "EGF-like" ligands for EGFR and HER3. Constitutive activation of HERs by the NTS/NTSR1 complex mimics the activating mutations of HERs and therefore potentiates the tumor response to tyrosine kinase inhibitors treatment in liver, lung and breast cancers.
165

Mechanism and Therapeutic Potential of Statin-Mediated Inhibition of Tyrosine Kinase Receptors

Zhao, Tong Tong January 2011 (has links)
Receptor tyrosine kinases (RTK) are key regulators of growth, differentiation and survival of epithelial cells and play a significant role in the development and progression of cancers derived from these tissues. In malignant cells, these receptors and their downstream signalling pathways are often deregulated, leading to cell hyper-proliferation, enhanced cell survival and increased metastatic potential. Furthermore, endothelial expressed RTKs regulate tumor angiogenesis allowing for tumor growth and maintenance by promoting their vascularization. Epithelial malignancies such as squamous cell carcinomas (SCC), non-small cell lung (NSCLC) and malignant mesotheliomas have very limited treatment options when presenting as metastatic disease. RTKs, particularly the epidermal growth factor (EGFR) and the vascular endothelial growth factor (VEGFR) receptors, have been shown to play significant roles in the pathogenesis of these tumor types. Statins are potent inhibitors of HMG-CoA reductase, the rate limiting enzyme of the mevalonate pathway, that are widely used as hypercholesterolemia treatments. The mevalonate pathway produces a variety of end products that are critical for many different cellular pathways, thus, targeting this pathway can affect multiple signalling pathways. Our laboratory has previously shown that lovastatin can induce tumor specific apoptosis especially in SCC and that 23% of recurrent SCC patients treated with lovastatin as a single agent showed disease stabilization in our Phase I clinical trial. Subsequently, our lab was able to demonstrate that lovastatin in combination with gefitinib, a potent inhibitor of the EGFR showed co-operative cytotoxicity when combined (Chapter 2). Furthermore, the pro-apoptotic and cytotoxic effects of these agents were found to be synergistic and to be manifested in several types of tumor cell lines including SCC, NSCLC and glioblastoma. I was able to expand upon these important findings and demonstrated that lovastatin, through its ability to disrupt the actin cytoskeleton, inhibited EGFR dimerization and activation (Chapter 3). This novel mechanism targeting this receptor has clinical implications as lovastatin treatment combined with gefitinib showed co-operative inhibitory effects on EGFR activation and downstream signalling. The RTK family of proteins share similar features with respect to activation, internalization and downstream signalling effectors. I further demonstrated that lovastatin can inhibit the VEGFR-2 in endothelial cells and mesotheliomas, where VEGF and its receptor are co-expressed driving their proliferation, and induces synergistic cytotoxicity in mesothelioma cells in combination with VEGFR-2 tyrosine kinase inhibitors (Chapter 4). These findings suggest that statins may augment the effects of a variety of RTK inhibitors in a similar fashion representing a novel combinational therapeutic approach in a wide repertoire of human cancers. More importantly, based on this work, we initiated a Phase I/II study evaluating high dose rosuvastatin and the EGFR inhibitor tarceva in SCC and NSCLC patients at our institute. This clinical evaluation will provide invaluable data that will play a role in developing this novel therapeutic strategy. Together, the work embodied in this thesis provides a model for the regulation of EGFR/VEGFR-2 activation and signalling by targeting the rho family of proteins that demonstrates a novel mechanism that can be exploited to refine current therapeutic paradigms.
166

Identification de voies de résistance aux inhibiteurs de tyrosine kinase dans la leucémie myéloïde chronique par criblage CRISPR-Cas9. / Genome-wide CRISPR-Cas9 Screening to Identify Pathways Involved in Tyrosine Kinase Inhibitor Resistance in Chronic Myeloid Leukemia

Lewis, Matthieu 12 April 2019 (has links)
La caractérisation des tumeurs malignes et la compréhension des mécanismes de résistance aux traitements anticancéreux sont essentielles pour la découverte de nouvelles cibles thérapeutiques. Les criblages génétiques, devenus encore plus puissants avec la technologie d’édition du génome CRISPR-Cas9, le séquençage nouvelle génération et la bioinformatique, sont des outils formidables pour décrypter de nouveaux mécanismes cellulaires, dont la résistance au traitement. La leucémie myéloïde chronique (LMC) est un syndrome myéloprolifératif qui est caractérisé par l’anomalie génétique t(9;22). Cette aberration chromosomique est à l’origine du gène de fusion BCR-ABL1 qui code l’oncogène du même nom responsable de la prolifération anarchique des cellules. L’imatinib mesylate, un inhibiteur de tyrosine kinase, élimine de manière spécifique les cellules leucémiques en ciblant et en bloquant l’activité kinase de cette protéine. Malheureusement, comme pour tout type de thérapie ciblée, une résistance au traitement survient chez certains patients. Afin de repérer des nouvelles voies de résistance à cet inhibiteur de tyrosine kinase, nous avons effectué un criblage génétique avec la librairie « genome-scale CRISPR knock-out » (GeCKO v2) in vitro dans la lignée cellulaire K562. Nous avons découvert plusieurs gènes qui semblent être essentiels pour la réponse au traitement par imatinib, tels que les facteurs pro-apoptotiques BIM et BAX, ou le répresseur de la voie des MAPK, SPRED2. Le rétablissement spécifique de l’apoptose dans les cellules BIM knock-out (KO) par des BH3-mimétiques, ou l’inhibition ciblée de la voie MAPK dans la lignée SPRED2 KO sensibilise de nouveau les lignées résistantes. Dans ce travail, nous avons découvert des mécanismes de résistance déjà connus (l’apoptose, la voie MAPK…) mais nous avons également démontré l’implication de voies peu connues telles que le complexe Mediator, la maturation de ARNm et l’ubiquitinylation de protéines. Spécifiquement cibler ces lésions génétiques avec des thérapies ciblées combinées peut permettre de surmonter les phénotypes de résistance et ouvre la porte à l’utilisation de l’oncologie de précision. / The characterization of malignant tumour growth and the understanding of resistance mechanisms to treatment in cancer is of utmost importance for the discovery of novel “druggable” targets. Efficient genetic screening, now even more possible with the convergence of CRISPR-Cas9 gene editing technology, next-generation sequencing and bioinformatics, is an important tool for deciphering novel cellular processes, such as resistance to treatment in cancer. Chronic myeloid leukemia (CML) is a myeloproliferative disorder characterised by the t(9;22) genetic abnormality, which encodes the driver of CML, the BCR-ABL1 fusion protein. Imatinib mesylate, a tyrosine kinase inhibitor, specifically eliminates CML cells by targeting and blocking the kinase activity of this protein, yet, as for all targeted therapies in cancer, resistance to treatment exists. In order to discover alternative BCR-ABL1 independent mechanisms of imatinib resistance, we utilized the genome-scale CRISPR knock-out library GeCKO v2 to screen for imatinib sensitising genes in vitro on K562 cells. We revealed genes that seem essential for imatinib induced cell death, such as pro-apoptotic genes (BIM, BAX) or MAPK inhibitor SPRED2. Specifically re-establishing apoptotic capabilities in BIM knock-out (KO) cells with BH3-mimetics, or inhibiting MAP-kinase signalling in SPRED2 KO cells with MEK inhibitors restores sensitivity to imatinib, overcoming resistance phenotypes. In this work, we discovered previously identified pathways (apoptosis, MAP-kinase signalling) and novel pathways that modulate response to imatinib in CML cell lines, such as the implication of the Mediator complex, mRNA processing and protein ubiquitinylation. Targeting these specific genetic lesions with combinational therapy can overcome resistance phenotypes and paves the road for the use of precision oncology.
167

Identification des évènements de signalisation associés à la prolifération autonome induite par le récepteur mutant FLT3-ITD dans les cellules myéloïdes / Identification of the signaling events associated with FLT3-ITD mutant receptor-induced constitutive proliferation in myeloid cells

Habif, Guillaume 17 December 2009 (has links)
Le récepteur tyrosine kinase FLT3 est impliqué dans le maintien et le renouvellement des cellules souches et des progéniteurs hématopoïétiques. Dans environ 30% des cas de leucémies aiguës myéloïdes, il est activé constitutivement par des mutations, dont les plus fréquentes impliquent une duplication de séquence dans le domaine juxtamembranaire (Internal Tandem Duplication, ou ITD). Des inhibiteurs chimiques de FLT3 ont été développés dans le cadre de thérapies anti-cancéreuses, mais leurs essais cliniques se sont révélés assez décevants avec des effets essentiellement transitoires. Par ailleurs, certaines études ont mis en évidence une signalisation intracellulaire spécifique au mutant FLT3-ITD, comme l’activation spécifique de STAT5a. Ces données soulignent la nécessité d’étudier exhaustivement et en détail la signalisation intracellulaire induite par le récepteur FLT3 et ses mutants oncogéniques dans des modèles pertinents, avec l’espoir d’identifier de nouvelles cibles thérapeutiques. Nous avons surexprimé FLT3 et sa forme mutante ITD dans la lignée murine de progéniteurs myéloïdes FDCP-1/Fms. Ce modèle s’est avéré représentatif des processus observés in vivo en termes de survie, de prolifération et de différenciation monocytaire. Nous l’avons alors utilisé dans une approche protéomique pour identifier des protéines différemment phosphorylées et/ou exprimées entre les deux cas. L’utilisation d’ARN interférant et la surexpression des protéines candidates ou de leurs mutants a permis de révéler l’implication fonctionnelle de plusieurs d’entre elles dans la signalisation FLT3, telles que Hcls1, Ezrin, et PAK1 qui sont toutes des régulateurs du cytosquelette / The FLT3 receptor is involved in stem cells and myeloid progenitors self renewal processes. In about 30% of the acute myeloid leukemia cases, this receptor is mutated and constitutively active, the most common mutation being duplication of sequences in the juxtamembrane domain (Internal Tandem Duplication, ITD). Many chemical inhibitors of FLT3 have been developed for anti-cancer therapies but the clinical trials were a bit disappointing, showing mainly transient effect on blast reduction. Several studies have shown that FLT3-ITD triggers a different signaling from the wild-type receptor, like the specific activation of STAT5a. These data show the necessity of the exhaustive and detailed study of the intracellular signaling induced by FLT3 and its oncogenic mutants, to identify new therapeutic targets. We have overexpressed wild-type and ITD mutant forms of FLT3 in the murine myeloid progenitors cell line FDCP-1/Fms. This model proved it-self representative of the in vivo processes described in the literature in terms of survival, proliferation and monocytic differentiation. Consequently, we have used it for a proteomic approach to identify differentially expressed and/or phosphorylated proteins depending on FLT3 status. Using RAN interference and overexpression of these identified candidate proteins, we have demonstrated the functional involvement of several of them in FLT3 signaling, including Hcls1, Ezrin, and PAK1, which all regulate the cytoskeleton
168

Etude des mécanismes de résistances primaire et acquise aux inhibiteurs du récepteur de l’Epidermal Growth Factor dans le cancer bronchique non à petites cellules / Mechanisms of primary and acquired resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer

Cortot, Alexis 21 December 2010 (has links)
Les inhibiteurs de tyrosine kinase (ITK) du récepteur à l’Epidermal Growth Factor (EGFR) constituent un nouveau traitement du cancer bronchique non à petites cellules (CBNPC), particulièrement efficace chez les patients porteurs d’une mutation d’EGFR (EGFR M+). Néanmoins, certains de ces patients peuvent avoir une résistance primaire à ces traitements, et les autres développent inéluctablement une rechute correspondant au phénomène de résistance acquise. L’objectif de ce travail était d’étudier les mécanismes de résistances primaire et acquise chez les patients avec CBNPC EGFR M+.Nous avons montré que le statut mutationnel de KRAS était le même dans la tumeur primitive et les métastases dans la majorité des cas de CBNPC. Dans quelques cas cependant, le statut différait entre tumeur primitive et métastase, ce qui soulève la question d’une dissociation de la réponse aux ITK, les mutations de KRAS étant associées à une résistance primaire au traitement.Nous avons par ailleurs mis en évidence la fréquente inactivation de la voie p53, soit par mutation de TP53 soit par diminution d’expression de p14arf, dans les tumeurs EGFR M+, qui pourrait être une des causes des variations de réponse aux ITK chez les patients EGFR M+. Enfin, nous avons montré que la résistance à deux ITK de nouvelle génération, PF299804 et WZ4002, passait par un phénomène en deux étapes impliquant d’une part l’activation de la voie IGF1R, via la diminution de l’expression d’IGFBP3, et d’autre part celle de la voie des MAP kinases. Ces travaux pourraient déboucher sur des stratégies thérapeutiques innovantes chez les patients présentant une résistance acquise aux ITK de nouvelle génération. / Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitors (TKI) provide clinical efficacy in Non Small Cell Lung Cancer (NSCLC) patients, especially in the presence of an EGFR mutation. However, some EGFR mutated patients display primary resistance to EGFR TKI, and the others will ultimately develop acquired resistance. We focused our work on mechanisms of primary and acquired resistance in EGFR mutated NSCLC.We first showed that KRAS mutational status is the same in primary NSCLC and matched metastases in most of the cases. However, in some patients, we found a discordant KRAS status between primary tumors and metastases, which could translate into a discordant response to EGFR TKI, since KRAS mutations are associated with resistance to EGFR TKI.We also showed that EGFR mutated tumors are associated in most of the cases with an inactivation of the p53 pathway, either through a TP53 mutation or through loss of expression of p14arf, which could account for some of the variations observed in the response to TKI in EGFR mutated patients.Last, we showed that acquired resistance to two new generation EGFR TKI, PF299804 and WZ4002, occurred through a multistep process involving activation of the IGF1R pathway through downregulation of IGFBP3 and activation of the MAP kinase pathway. These results provide new insights into the treatment of NSCLC in EGFR mutated patients with acquired resistance to new generation TKI.
169

Brk tyrosine kinase signaling in the gastrointestinal tract

Hägebarth, Andrea 07 December 2005 (has links)
Die Tyrosin Kinase Brk stellt den Prototypen nicht N-terminal myristoylierter, Nicht-Rezeptor Tyrosin Kinasen dar. Die Expression dieser Kinase ist auf epitheliale Gewebe beschränkt und wird während der Entwicklung differentiell reguliert. In normalen Geweben ist die Brk Expression auf nichtproliferierende, terminal differenzierte Zellen beschränkt. Um die regulatorische Funktion von Brk im murinen Darmepithel zu untersuchen, wurde das brk Gen in der Maus inaktiviert. Brk knockout Mäuse zeigten keine offensichtlichen Defekte in ihrer Entwicklung jedoch eine erweiterte Proliferationszone in den Krypten des Darmepithels und verlängerte Villi. Die Inaktivierung von Brk führte zu einer erhöhten Akkummulation von nukleärem (-catenin sowie einer Hochregulierung des (-catenin Zielgens c-myc in den Krypten der knockout Mäuse. Zusätzlich zeigten Brk knockout Mäuse eine Aktivierung des Akt-Signaltransduktionswegs in ihrem Darmepithel. Im Gegensatz zu Wildtyp Mäusen waren Brk knockout Mäuse resistent gegenüber (-Strahlung, was die Anhäufung onkogener Mutationen und damit die Entwicklung von Krebs fördert. Eine Induktion der Expression des Brk-Proteins im Darmepithel behandelter Wildtyp Mäuse wurde festgestellt. Weiterhin traten bei Brk knockout Mäusen chronische Entzündungen des Darmepithels sowie eine erhöhte Sensibilität gegenüber dem Reizmittel DSS auf. Im Gegensatz dazu, zeigten Wildtyp Mäuse eine mit der Literatur Übereinstimmende Reaktion zu DSS verbunden mit einer Induktion der Brk Expression im Darmepithel. Zusammenfassend kann gesagt werden, dass die Brk Tyrosin Kinase eine entscheidende Rolle in der Aufrechterhaltung der Homöstase und Integrität des Darmepithels spielt. Insbesondere scheint Brk als wichtiger Faktor zur Bestimmung der Sensitivität epithelialer Zellen zu genotoxischem Stress zu fungieren. Entgegen der bisher vermuteten onkogenen Funktion in epithelialen Tumoren scheint Brk im normalen Darmepithel "Tumor Suppressor" Ähnliche Funtionen innezuhaben. / The Breast tumor kinase Brk is a prototypical non-myristoylated, non-receptor tyrosine kinase. Brk expression is epithelial-specific and ,in normal tissues, restricted to cells exiting the cell cycle and undergoing terminal differentiation. To determine the biological role of Brk in the gastrointestinal tract, we disrupted mouse brk by homologous recombination. Loss of Brk in the mouse resulted in increased intestinal epithelial cell turnover and the appearance of longer small intestinal villi. Brk deficient mice displayed enhanced accumulation of nuclear (-catenin and upregulation of the (-catenin target gene c-myc in the crypt compartment of small and large intestine. In addition, Brk deficient mice exhibited increased Akt kinase activity. Even though, there was no corresponding difference in base-line apoptosis in untreated wild-type and knockout animals. However, subjected to (-irradiation, Brk deficient animals were significantly impaired in the apoptotic response. Wild-type mice, however, exhibited normal levels of apoptosis following (-irradiation accompanied by a rapid induction of Brk expression in crypt cells. Furthermore, chronic inflammation was observed in Brk deficient mice, and they showed increased susceptibility to a colon injury model utilizing DSS. Interestingly, wild-type mice exhibited a significant upregulation of nuclear Brk protein throughout the intestinal epithelium in response to DSS. These recent findings suggest that Brk plays a crucial role in the maintenance of intestinal tissue homeostasis and integrity. In addition, Brk may function to protect the intestinal epithelium against DNA-replication-induced errors and hence the development of cancer. Contrary to reported oncogenic properties of Brk in other epithelial tissues, Brk appears to have tumor suppressor-like functions in the mouse gastrointestinal epithelium.
170

Comparação da eficácia do mesilato de imatinibe com a vimblastina associada a prednisona no tratamento do mastocitoma canino: estudo clínico, histopatológico, imunohistoquímico e molecular / Comparison of the efficacy of imatinib mesylate with vinblastine and prednisone in the treatment of canine mast cell tumor: clinical, histological, immunohistochemical and molecular study

Macêdo, Thais Rodrigues 22 August 2014 (has links)
O objetivo deste trabalho foi avaliar a eficácia do mesilato de imatinibe, em comparação com a quimioterapia usual com vimblastina e prednisona, no tratamento do mastocitoma canino e descrever os efeitos colaterais apresentados pelas medicações. Bem como analisar a expressão do VEGF (fator de crescimento endotelial), a relação da expressão do gene c-kit por RT-PCR e marcação imunoistoquímica do KIT com a presença de mutações na justamembrana e a relação desta mutação com a resposta à terapia. Para tanto foram incluídos 29 animais com diagnóstico citológico de mastocitoma, estes animais foram submetidos a tomografia computadorizada para determinação das medidas das formações cutâneas e em seguida divididos em 2 grupos. O grupo 1 foi tratado com o protocolo quimioterápico de vimblastina e prednisona por 12 semanas e o grupo 2 com o mesilato de imatinibe na dose de 10 mg/Kg a cada 24 horas por 8 semanas. A avaliação da resposta ao tratamento foi realizada com mensurações periódicas das formações com paquímetro e nova tomografia ao final do tratamento para mensuração do maior diâmetro e volume tumoral. Um fragmento das formações cutâneas foi coletado antes do início do tratamento para graduação histológica da neoplasia, determinação do índice mitótico e imunomarcação para KIT, VEGF e Ki- 67. Parte do material coletado teve o DNA e RNA extraídos e posterior sequenciamento dos exons 11 do gene c-kit e determinação da expressão deste e do seu ligante por RT-PCR. A toxicidade a medicação foi avaliada segundo as normas do VCOG 1.1.A taxa de resposta do grupo VP foi de 7,7 % e no grupo MI de 28,6%, embora os pacientes tratados com mesilato de imatinibe tenham apresentado maior chance de resposta a terapia, não foi observado diferença entre os dois grupos. Os dois protocolos foram bem tolerados, os pacientes do grupo MI d menor número de efeitos colaterais. O grau histológico, Indice mitótico, padrão imunohistoquimico do KIT, além da quantificação do ki-67 foram homogêneos nos dois grupos e não influenciaram na resposta ao tratamento. A quantificação do VEGF foi mais intensa nos pacientes com remissão parcial e total. Não foi observado relação entre a quantificação do KIT e a expressão do gene c-kit, que foi maior nos pacientes que responderam ao tratamento, porém a associação desta com a resposta a terapia não pode ser determinada. Mutações ativantes no exon11 do gene c-kit não foram identificadas. O tratamento com o mesilato de imatinibe é bem tolerado pelos animais, no entanto este não se mostrou superior ao protocolo padrão de quimioterapia para o tratamento do mastocitoma; este resultado pode ter sido influenciado pelo número de animais incluídos no estudo. Mutações em outros domínios do receptor KIT e a ação do ITK em receptores como do PDGF e o VEGF podem estar relacionados a resposta a esta classe de fármacos observada neste estudo, a despeito da ausência de mutações ativantes no exon 11 do gene c-kit. / The objective of this study was to evaluate the efficacy of imatinib mesylate, compared with the usual chemotherapy with vinblastine and prednisone in the treatment of canine mast cell tumor and describe the side effects submitted by medications. Well as analyzing the expression of VEGF (vascular endothelial growth factor), the relationship between the expression of c-kit gene by RT-PCR and immunohistochemical staining of KIT with the presence of mutations in the juxtamembrane and the relationship of this mutation with response to therapy. For both 29 animals with cytological diagnosis of mast cell tumor were included, these animals underwent computed tomography to determine the measured skin formations and then divided into 2 groups. Group 1 was treated with the chemotherapeutic protocol vinblastine and prednisone for 12 weeks and the second group with the imatinib mesylate in a dose of 10 mg / kg every 24 hours for 8 weeks. The assessment of response to treatment was performed with periodic measurements of the formations Caliper and a new computed tomography in the end of treatment to measure the largest tumor diameter and volume. A fragment of skin formations was collected before the initiation of treatment for histological grading, determination of mitotic index, KIT and VEGF staining patterns and the proliferation marker Ki67. Part of the collected material was extracted RNA and DNA and subsequent sequencing of 11 exons of the c-kit gene and determination and expression of its ligand by RT-PCR. The medication toxicity was evaluated according to the standards of VCOG 1.1.A response rate of the VP group was 7.7% and 28.6% MI group, although patients treated with imatinib had a higher chance of response therapy, no difference in response between the two groups was observed. The two protocols were well tolerated, patients in the MI group had a smaller number of side effects. The histological grade, mitotic index, staining patterns KIT, beyond the quantification of Ki-67 were homogeneous in both groups and did not influence the response to treatment. Quantification of VEGF was intensely in patients with partial and total remission. It was no relationship between KIT and quantification of the expression of c-kit gene, which was higher in patients who responded to treatment, but its association with response to therapy cant be determined. Exon11 activating mutations in the c-kit gene were not identified. Treatment with imatinib mesylate is well tolerated by the animals, however this was not superior to standard chemotherapy protocol for the treatment of mast cell tumors; this result may have been influenced by the number of animals included in the study. Mutations in other domains of the KIT receptor and action in ITK receptors as PDGF and VEGF may be related to response to this class of drugs in this study, despite the absence of activating mutations in exon 11 of c-kit gene.

Page generated in 0.0851 seconds