• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 159
  • 52
  • 31
  • 12
  • 10
  • 8
  • 7
  • 7
  • 5
  • 3
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 337
  • 119
  • 54
  • 51
  • 42
  • 34
  • 32
  • 32
  • 30
  • 29
  • 28
  • 27
  • 26
  • 26
  • 24
  • 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.
181

Implication de la Galectine-3 dans le trafic intracellulaire de la mucine membranaire MUC1 et de son récepteur associé, l'EGFR , dans les cellules cancéreuses pancréatiques humaines

Merlin, Johann 14 December 2012 (has links) (PDF)
L'adénocarcinome pancréatique canalaire est un cancer de mauvais pronostic avec une survie à 5 ans inférieure à 5% et une médiane de survie d'environ 6 mois. Dès les stades précoces de la carcinogenèse pancréatique, la mucine membranaire MUC1, glycoprotéine de haute masse moléculaire, est surexprimée et présente des anomalies de distribution cellulaire, essentiellement une délocalisation membranaire vers le pôle basolatéral et une rétention à l'intérieur de la cellule. Sachant que MUC1 est capable d'interagir avec l'EGFR et de jouer un rôle sur la transduction des signaux, cette séquestration pourrait être à l'origine de signaux oncogéniques et est utilisée par les pathologistes comme indicateur de malignité après ponction sur des lésions pancréatiques. Cependant, les mécanismes permettant cette rétention cytoplasmique ne sont pas connus. Des études antérieures du laboratoire ont montré que le trafic de certaines glycoprotéines vers la membrane apicale des cellules épithéliales était dépendant de la Galectine 4. Dans ce travail, nous nous intéresserons à la Galectine 3, lectine endogène susceptible d'interagir avec les motifs glycanniques des mucines et aussi des récepteurs membranaires. Cette Galectine est surexprimée dans le cancer pancréatique et son expression est corrélée à une forte agressivité tumorale dans d'autres cancers. Le but de ce travail a été dans un premier temps de mettre au point des lignées cellulaires pancréatiques polarisées CAPAN-1 Knocked-down pour la Galectine-3. Dans un second temps d'étudier l'implication de la Galectine-3 : (i) dans le trafic intracellulaire de MUC1 et l'EGFR (ii) dans l'interaction entre MUC1 et l'EGFR (iii) sur les voies de signalisation de l'EGFR.Les résultats montrent que les cellules tumorales pancréatiques CAPAN-1 présentent les anomalies de distribution cellulaire de MUC1 observées dans les tumeurs pancréatiques humaines, notamment la rétention de MUC1 à l'intérieur des cellules. Le silencing de la Galectine-3 entraîne la disparition de cette anomalie de distribution cellulaire, MUC1 retrouvant la distribution membranaire normale. Nous avons montré que l'inhibition de l'expression de la Galectine-3 est associée (i) à une augmentation de la translocation nucléaire de l'EGFR (ii) à une inhibition de l'endoyctose de MUC1 et de l'EGFR en condition de sevrage. Ce phénomène s'accompagne d'une augmentation de l'interaction entre MUC1 et l'EGFR et d'une activation accrue de ce récepteur tyrosine kinase lorsqu'il est soumis à l'EGF : augmentation de la phosphorylation de l'EGFR et augmentation de la phosphorylation des MAPK Erk1 et 2. Mots clés : Galectine, Mucine, EGFR, cancer, endocytose, translocation nucléaire
182

Genotype-phenotype studies in brain tumors

Ghasimi, Soma January 2013 (has links)
Meningioma and glioma are the most common primary brain tumors, but their etiologies are largely unknown. Although meningioma is usually benign, their intracranial location can lead to lethal consequences, and despite progress in surgery, radiotherapy, and chemotherapy the prognosis for patients with glioma remains poor. The only well-established environmental risk factor for meningioma and glioma is ionizing radiation. Evidence for inherited predisposition to meningioma and glioma is provided by a number of rare inherited syndromes where collectively these diseases account for only a small proportion of the twofold increased risk of brain tumors seen in first-degree relatives for meningioma and glioma patients. It is very possible that much of the excess familial risk is a consequence of co-inheritance of multiple low-risk genetic variations. With this in mind, the aims of the studies in this thesis were to discover genetic risk variants influencing the probability of acquiring the disease and to identify the association between risk variants on the tumor phenotype. To identify genetic variants influencing meningioma risk, a comprehensive tagging of the selected genes in a case-control study was performed. We identified nine risk variants in EGF, ERBB2, and LRIG2 genes. However, these findings could not be confirmed in another larger independent dataset. In addition, the study identified a correlation between LRIG2 protein expression and ER status when analyzed with different parameters. In a separate study with a larger sample of meningioma patients, the same correlation between LRIG2 and ER status was observed. To explore the potential association between reported germline risk variants and somatic genetic events, matched tumor and blood samples from glioma patients were analyzed by SNP array. The results identified correlations between EGFR gene variants and somatic aberrations within the EGFR locus and CDKN2A/B locus. To further study the relationship between germline risk variants and tumor phenotype, the same patient material was used and analyzed by three different techniques: SNP array, IHC, and FISH. The results revealed EGFR risk variants effecting copy number variation of the EGFR gene and the expression of the IDH1 and p53. Further comparison between different techniques such as SNP array and FISH analysis revealed the difficulty in achieving consistent results with different techniques. To summarize, the glioma studies show a link between genotype and phenotype where genetic risk variants in the EGFR gene were found to be associated with specific somatic aberrations. These associations are biologically interesting because EGFR is involved in multiple cellular processes. Additional studies of the direct functional role of these observations need to be conducted to elucidate the molecular mechanisms underlying the identified association between germline gene variants and somatic aberrations. For the meningioma studies, no significant risk variants influencing the disease were found but a correlation between LRIG2 and ER status was observed. This result suggests a potential role for the LRIG protein in the pathogenesis of meningioma, but more studies are needed to confirm this hypothesizes. / <p>Cancer research foundation in northern Sweden and Lions cancer research foundation at Umeå university</p>
183

111In-labeled Nimotuzumab Modified with Nuclear Localization Sequences (NLS): An Auger Electron-emitting Radiotherapeutic Agent for EGFR-overexpressing and Trastuzumab-resistant Breast Cancer

Fasih, Aisha 24 August 2011 (has links)
Objective: The cytotoxic property of anti-EGFR-1 monoclonal-antibody nimotuzumab modified with nuclear localization sequence and radiolabeled with 111In was evaluated in trastuzumab-resistant breast cancer cells. Methods: 111In-nimotuzumab-NLS was constructed and its immunoreactivity was determined. Cellular and nuclear uptake was evaluated by cell fractionation. Finally, the cytotoxicity of conjugates (111In-nimotuzumab/111In-nimotuzumab-NLS) was studied by clonogenic assays. Results: The immunoreactivity of 111In-nimotuzumab-NLS was conserved. 111In-nimotuzumab-NLS exhibited 2-fold higher nuclear translocation as compared to 111In-nimotuzumab in MDA-MB-468 cells. Nuclear importation of 111In-nimotuzumab-NLS in MDA-MB-468 cells was 4-fold and 6-fold higher than moderate and low EGFR expressing cell lines, respectively. Clonogenic survival (CS) for MDA-MB-468 cells showed 111In-nimotuzumab-NLS to be 10-folds and 60-folds more potent than 111In-nimotuzumab and nimotuzumab, respectively. Moderate killing for TrR1 and MDA-MB-231 was observed. 111In-hEGF showed significantly higher cytotoxicity and 2-fold higher γ-H2AX foci integrated density/nuclear-area as compared to 111In-nimotuzumab-NLS. Preserved selectivity of 111In-nimotuzumab-NLS makes it an excellent drug for treating cancers.
184

The combination of pan-ErbB tyrosine kinase inhibitor CI-1033 and lovastatin: A potential novel therapeutic approach in squamous cell carcinoma of the head and neck

Guimond, Tanya 28 September 2011 (has links)
The ErbB family of receptors are key regulators of growth, differentiation, migration and survival of epithelial cells. CI-1033 is an irreversible pan-ErbB tyrosine kinase inhibitor that has the ability to inhibit EGFR function but has shown limited therapeutic efficacy. Lovastatin targets the activity of HMG-CoA reductase, the rate-limiting step in the mevalonate pathway. In this study, the ability of lovastatin to potentiate the cytotoxic effects of CI-1033 was evaluated. The combination of lovastatin and CI-1033 exhibited some cooperative cytotoxic activity in a squamous cell carcinoma–derived cell line. This combination resulted in enhanced cell death by induction of a potent apoptotic response. Furthermore, this drug combination inhibited EGF-induced EGFR autophosphorylation and activation of the downstream signaling effectors, ERK and AKT. These findings suggest that combining lovastatin and tyrosine kinase inhibitors may represent a novel combinational therapeutic approach in squamous cell carcinoma of the head and neck.
185

Interaktion einer Blockade des Rezeptors für den Epidermalen Wachstumsfaktor (EGFR) mit der Gabe von Keratinozyten-Wachstumsfaktor (KGF) bei der Strahlenreaktion der Mundschleimhaut – tierexperimentelle Untersuchungen an Mäusen

Fehrmann, Astrid 26 May 2010 (has links) (PDF)
Bei der Strahlentherapie fortgeschrittener Tumoren im Kopf-Hals-Bereich gilt die radiogene Mucositis enoralis als schwerwiegende und dosislimitierende frühe Nebenwirkung. Sehr häufig führt sie zu einer Unterbrechung der Behandlung, mit der Folge einer Reduktion der Tumorheilungschancen. Während einer fraktionierten Strahlenexposition kommt es in der Mundschleimhaut zu einer erhöhten Expression des Epidermalen Wachstumsfaktors (Epidermal Growth Factor, EGF) und dessen Rezeptors (EGFR). Durch eine Blockade des EGFR, als anerkannte Strategie zur Verbesserung der Tumorheilung, besteht deshalb die Gefahr, dass es zu einer Verschlimmerung der Schleimhaut-Nebenwirkungen kommt. Der Einsatz von Keratinozyten-Wachstumsfaktor (KGF) zeigt positive Ergebnisse bezüglich einer Reduktion der Schleimhautveränderungen. In dieser Arbeit wird deshalb im Tiermodell einerseits die Auswirkung einer Blockade des EGFR auf die Schleimhautreaktion, und andererseits eine mögliche Interaktion der Blockade mit der schleimhautschützenden Wirkung von KGF untersucht. Insgesamt kann keine signifikante Veränderung der Schleimhauttoleranz durch die EGFR-Inhibition mittels BIBX1382BF innerhalb der ersten beiden Wochen einer fraktionierten Bestrahlung festgestellt werden; lediglich das Auftreten ulzerativer Läsionen nach der zweiten Woche ist vorverlagert
186

Etude des relations entre les mutations EGFR/KRAS et les altérations de la voie p53/p14arf et caractérisation d'une nouvelle cible thérapeutique, le complexe neurotensine et son récepteur1, dans les cancers bronchiques non à petites cellules

Younes, Mohamad 26 September 2012 (has links) (PDF)
Nous avons étudié, le statut de la voie p53/p14arf dans l'adénocarcinome bronchique muté pour EGFR ou KRAS ainsi que l'impact du complexe NTS/NTSR1 sur la progression tumorale et la survie du cancer bronchique non à petites cellules. Dans une série de 96 patients, les mutations de TP53, KRAS et EGFR ont été détectées dans respectivement 45.2%, 15.8% et 34.4% des cas. La diminution de l'expression de p14arf a été observée dans 57.1% des cas. Le déficit de la voie p53/p14arf a été observé dans 83,3% et 76.9% des tumeurs mutées pour EGFR et KRAS respectivement, ce qui suggère que les altérations de la voie p53/p14arf sont des événements communs dans ces tumeurs mais pas systématiques. L'expression du NTSR1 a été mise en évidence par immunohistochimie dans 60.4% des adénocarcinomes de stades I (n= 139); 47% de stade II (n = 74) et 50% de stade III (n = 133). A l'analyse univariée et multivariée, l'expression du NTSR1, était associée significativement à un mauvais pronostic en termes de survie globale (p = 0,0081 pour les stade I, p = 0.0087 pour les stade II-III). Par contre, l'expression de NTSR1 n'était pas associée avec une différence significative en terme de survie globale pour le carcinome épidermoïde ou le carcinome à grandes cellules. Les études, in vivo et in vitro, ont montré que le complexe NTS/NTSR1 favorise la progression tumorale en augmentant l'expression HER2 et HER3 et la transactivation d' EGFR, HER2 et HER3 par l'activation de MMP1 et la libération de HB-EGF et de NRG1. Ces résultats suggèrent que le complexe NTS/NTSR1 contribue activement à l'agressivité et à la progression tumorale bronchique
187

The Role of c-Src in E-Cadherin Activity

Robert Mclachlan Unknown Date (has links)
Cadherin-based cell-cell contacts are prominent sites for phosphotyrosine signalling, being enriched in tyrosine-phosphorylated proteins, tyrosine kinases and phosphatases. The functional interplay between cadherin adhesion and tyrosine kinase signalling, however, is complex and incompletely understood. In my thesis I have tested the hypothesis that c-Src contributes positively to cadherin biology by functioning as part of an adhesion activated cell-signalling pathway. I found that c-Src is active at both established and reforming cell-cell contacts, and c-Src can be activated by homophilic ligation of the adhesion receptor. However, c-Src has a biphasic impact on cadherin function, exerting a positive supportive role at lower signal strengths, but inhibiting function at high signal strengths. Inhibiting c-Src under circumstances when it is activated by cadherin adhesion decreased several measures of cadherin function. This suggests that the cadherin-activated c-Src signalling pathway serves positively to support cadherin function, while quantitative changes in signal strength may result in qualitative differences in functional outcome. Finally, my data implicated PI3-kinase signalling and cortactin as potential targets for cadherin-activated c-Src signalling. By inhibiting protein tyrosine phosphatases with pervanadate, I found that tyrosine phosphatase activity and not just protein binding was required to stimulate Src activity in response to cadherin ligation. I identified the tyrosine phosphatase RPTPα as a possible regulator of cadherin-activated Src signalling. RPTPα localises to cell-cell adhesions and it is found in a complex with E-cadherin and c-Src. Furthermore, knockdown of RPTPα disrupted the integrity of cadherin-based contacts and the activity of Src at these cell-cell contacts. This suggests that in response to cadherin-homophilic ligation PTP activity is required to stimulate Src signalling. Finally, I identified a novel pathway by which aberrant growth factor signalling could be downregulating cadherin function and promoting the invasion of epithelial cells. Stimulating cells with high levels of EGF revealed that aberrant epidermal growth factor signalling could disrupt cadherin-activated cell signalling. The integrity of cadherin-based contacts and the activity of Src at the cell-cell contacts were both disrupted in the presence of high levels of EGF. Analysis of E-cadherin and RPTPα immunoprecipitates suggested that activation of cadherin-bound EGFR might disrupt Src activation by displacing E-cadherin-RPTPα binding. Finally, analysing the subcellular distribution of these proteins revealed that, in response to high levels of EGF, E-cadherin, β-catenin, EGFR and pEGFR are internalised together in phospho-cortactin-rich endosomal-like structures. Therefore I propose that E-cadherin adhesion activates a cell-signalling pathway involving c-Src that functions to dynamically regulate the actin cytoskeleton and to maintain the adhesive strength of cell-cell adhesions. Perturbation of cadherin-activated Src signalling downregulates cadherin function and promotes the disassembly of cell-cell adhesive contacts. The concept of a cadherin-activated Src signalling pathway provides a new way to think about cadherin biology. Instead of merely functioning as passive glue holding two cells together, E-cadherin functions as an adhesion-activated signalling receptor. Dysregulation of E-cadherin-activated Src signalling and downregulation of cell-cell adhesions could be a mechanism promoting the invasion and metastasis of epithelial tumours.
188

FRMD8 is a novel regulator of iRhom-dependent ADAM17 activity

Künzel, Ulrike January 2017 (has links)
A disintegrin and metalloprotease (ADAM) 17 cleaves and releases membrane-tethered pro-forms of several signalling molecules from the plasma membrane, including the inflammatory cytokine tumour necrosis factor alpha (TNFα) and ligands of the epidermal growth factor receptor (EGFR). Due to the important functions of its substrates, ADAM17 activity has to be tightly controlled, and its misregulation has implications for inflammation and cancer. The multi-pass membrane proteins iRhom1 and iRhom2 are members of the conserved rhomboid-like superfamily and control ADAM17 activity by several mechanisms throughout the secretory pathway. First, iRhoms facilitate trafficking of the catalytically inactive proenzyme form of ADAM17 from the endoplasmic reticulum (ER) to the Golgi apparatus, where the inhibitory pro-domain of ADAM17 is removed. Subsequently, iRhoms exert a different form of control of ADAM17 at the plasma membrane, this time on stimulus-induced ADAM17 activity, its substrate specificity, and its stability. iRhoms ultimately regulate the release of ADAM17 substrates, and are consequently key players in TNF&alpha; and EGFR signalling. However, it remains unclear how iRhom function itself is regulated posttranslationally, and whether iRhoms require co-factors to exert their roles as ADAM17 regulators. The goal of my project was to shed light into these questions by identifying new iRhom interaction partners. I developed a mass spectrometry-based screen to identify new binding partners of human iRhoms using co-immunoprecipitation. The top hit of the screen was the poorly characterised FERM domain-containing protein 8 (FRMD8), which binds to both iRhom1 and iRhom2. FRMD8 was found to play a crucial role in the iRhom/ADAM17 pathway because FRMD8 knockdown and knockout in HEK293T cells significantly reduced the levels of mature ADAM17 and the release of ADAM17 substrates. The closely related metalloprotease ADAM10 was not affected by the loss of FRMD8, implying that FRMD8 is not a general regulator of ADAM metalloproteases. Interaction studies revealed that FRMD8 binds to the cytosolic N-terminus of iRhom2 throughout the entire secretory pathway. FRMD8 loss does not affect the ER-to-Golgi trafficking of iRhom2 but plays a role in stabilising iRhom2 at the plasma membrane by preventing the lysosomal degradation of both iRhom2 and mature ADAM17. Using human induced pluripotent stem cell (hiPSC)-derived macrophages, I showed that FRMD8 regulates mature ADAM17 levels and the ADAM17-dependent release of TNF&alpha; in human macrophages. Studies in FRMD8 knockout (KO) mice confirmed the reduced mature ADAM17 levels in all mouse tissues tested, further supporting the conclusion that FRMD8 is a novel regulator of the iRhom/ADAM17 pathway with physiological relevance in mammals. Finally, I showed that the interaction of FRMD8 and iRhom, which are both conserved from Drosophila to human, is also conserved. Furthermore, loss of the FRMD8 orthologue in flies, Bili, leads to motility defects and shows similarity to the loss of iRhom in flies. These results suggest that FRMD8 is a novel regulator of iRhom function in mammals and Drosophila.
189

A novel mechanism for the anti-cancer activity of aspirin and its analogues

Bashir, Asma'u Ismail Junaidu January 2017 (has links)
Colorectal cancer (CRC), which includes cancer of the large bowel and rectum is the third most common cancer in men and the second in women and there is a poorer survival rate in less developed regions of the world such as West Africa mainly due to the ‘out of reach’ costs of chemotherapy. Evidence suggests that aspirin, a non-steroidal anti-inflammatory drug (NSAID) has the potential to decrease incidence of, or mortality from, a number of cancers including CRC through several mechanisms of action. However, this evidence is dampened by aspirin’s gastrointestinal (GI) toxicity, which have been found to be mostly age-dependent. The search for potential aspirin-related compounds with the same or better cytotoxic effects against cancer cells accompanied by a safer toxicity profile has been ongoing over the years and led to us to synthesise a number of novel aspirin analogues. One of the mechanisms of action suggested for the anticancer property of aspirin is the COX-dependent pathway. In this thesis SW480 cell line, a CRC cell line that is COX-2 negative and mismatch repair (MMR) proficient was used to study the possible COX-independent mechanism of action for aspirin, its analogues and diflunisal at 0.5 mM. Diflunisal was included in this study because it is also a salicylate with reports of having cytotoxic effects. OE33 and FLO1 oesophageal cancer cells were also employed in the epidermal growth factor receptor (EGFR) and synergy experiments to show effects were not just specific to SW480 cells alone. These aspirin analogues were synthesised, identified using nuclear magnetic resonance (NMR) and infra-red (IR) spectroscopy, and tested for purity using thin layer chromatography (TLC) and melting point. The findings of this study suggest that these compounds breakdown into salicylates and perturb epidermal growth factor (EGF) internalization with PN517 (fumaryldiaspirin) and PN590 (ortho-thioaspirin) also driving EGF co-localization with early-endosome antigen-1 (EEA1). The perturbation of the internalization of EGF by aspirin and PN517 was also observed by a time-lapse assay using live confocal imaging. These compounds also had specific effects on different tyrosine phosphorylation sites of the EGFR, with none but PN590 inhibiting 4 phosphorylation at Y1068, and all but PN502 (ortho-aspirin), PN548 (meta-aspirin) and PN549 (para-aspirin) inhibiting phosphorylation at Y1045 and Y1173. Given that the EGF internalization assay involved the cells being treated with compounds for 2 h, cells were also treated for this same time period and probed with pEGFR 1045, which resulted in the compounds having no significant effect on phosphorylation at that site which is responsible for the ubiquitination of the EGFR. Most of these compounds were apoptotic with some showing a combination of apoptosis and necrosis. Aspirin and its isomers drove apoptotic cell death in SW480 cells via the BCL2-BAX pathway while the thioaspirins appear to follow the p21 pathway by decreasing the expression of the protein. In addition, it was shown that PN502 (aspirin), PN517 and PN590 had synergistic effects when used in combination with oxaliplatin at ED50, ED75 and ED90 in SW480 CRC cells. The cytotoxicity of these compounds individually or in combination was determined using MTT assay followed by the use of the CompuSyn and CalcuSyn software to calculate combination index (CI), which indicated whether a drug combination was synergistic, antagonistic or additive. PN517 and PN524 were synergistic when used in combination with cisplatin in OE33 oesophageal cancer cells. Effect of these compounds on the EGFR indicates a delay or disruption of the signalling pathway involved in the proliferation of cancer cells, thus, translating into protection against tumour formation or progression while the synergistic effects of these compounds when used in combination with platinum compounds can provide patients with less toxic chemotherapeutic regimen especially in patients with CRC tumours that harbour mutant TP53 gene and normally resistant to oxaliplatin. It is therefore proposed that the perturbation of EGF internalization is a novel mechanism of action for aspirin and its analogues in cancer therapy. These positive findings shed light on the understanding of the possible mechanism of action for aspirins and gives hope for a more affordable, less toxic therapy for the prevention, treatment and management of cancer.
190

Expressão do receptor do fator de crescimento epidérmico em adenocarcinoma esofágico : relação com estágio tumoral e sobrevida após esofagectomia / Epidermal growth factor receptor expression in esophageal adenocarcinoma : relationship with tumor stage and survival after esophagectomy

Navarini, Daniel January 2011 (has links)
Introdução e objetivos: O adenocarcinoma de esôfago (AE) é um tumor agressivo, com o aumento da incidência em países ocidentais. Vários marcadores prognósticos têm sido propostos, incluindo o Receptor do Fator de Crescimento Epitelial (EGFR). O objetivo deste estudo foi avaliar se a expressão do EGFR está relacionada com o estadiamento tumoral e com a sobrevida. Métodos: Trata-se de uma coorte histórica na qual 70 pacientes consecutivos com AE atendidos entre 2000 a 2009 foram considerados elegíveis para o estudo. As peças cirúrgicas dos pacientes submetidos a esofagectomia transhiatal foram avaliadas para estabelecer a expressão do EGFR e analisadas em relação às variáveis dos pacientes. A sobrevida foi determinada de acordo com os registros nos prontuários médicos dos pacientes ou por contato telefônico com familiares. Resultados: Dos 70 pacientes, 37 (53%) preencheram os critérios para inclusão no estudo. A expressão do EGFR foi positiva em 16 pacientes (43%) e foi mais freqüente nos estágios tumorais mais avançados, TNM (I e II = 0% vs III = 47% vs IV = 100%, P <0,001). A sobrevida média em meses, foi significativamente menor no grupo de pacientes com expressão do EGFR (10,5 vs 21,7, P = 0,001) em comparação com pacientes sem expressão do EGFR. Houve maior expressão do EGFR em neoplasias com pobre diferenciação tumoral em relação aos bem diferenciados e moderadamente diferenciados. Conclusão: Em pacientes com adenocarcinoma de esôfago tratados com esofagectomia, a expressão de EGFR está relacionada com maior estádio TNM e menor sobrevida. A expressão do EGFR pode ser assumida como um marcador de prognóstico para o adenocarcinoma de esôfago. / Background and aims: Esophageal adenocarcinoma (EA) is an aggressive tumor with increasing incidence in occidental countries. Several prognostic biomarkers have been proposed, including epidermal growth factor receptor (EGFR). The aim of this study was to assess whether EGFR expression predicts tumor staging and survival. Methods: In this historical cohort, 70 consecutive patients with EA managed between 2000 and 2009 were considered eligible for the study. Surgical specimens from those treated with esophagectomy were evaluated to establish EGFR expression and tumor differentiation. Survival was determined according to medical register or patient contact. Results: Among 70 patients, 37 (53%) underwent esophagectomy without pre-surgical chemotherapy and composed the study population. Of these, EGFR expression was found in 16 patients (43%). EGFR expression was more frequent as higher was TNM staging (I and II = 0% vs. III = 47% vs. IV = 100%; P < 0.001). Average survival in months was significantly shorter in the group of patients with EGFR expression (10.5 vs. 21.7; P = 0.001). Conclusions: In patients with esophageal adenocarcinoma treated with esophagectomy, EGFR expression was related with higher TNM staging and shorter survival. EGFR expression can be assumed as a prognostic marker for esophageal adenocarcinoma.

Page generated in 0.0274 seconds