Academic literature on the topic 'MET/HGF'

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the lists of relevant articles, books, theses, conference reports, and other scholarly sources on the topic 'MET/HGF.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Journal articles on the topic "MET/HGF":

1

Toiyama, Y., K. Tanaka, H. Yasuda, S. Saigusa, H. Fujikawa, Y. Mohri, Y. Inoue, C. Miki, T. Tabata, and M. Kusunoki. "Use of co-expression of HGF and c-Met to predict peritoneal dissemination established by autocrine HGF/c-Met signaling in gastric cancer." Journal of Clinical Oncology 29, no. 4_suppl (February 1, 2011): 40. http://dx.doi.org/10.1200/jco.2011.29.4_suppl.40.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
40 Background: Epithelial mesencymal transition (EMT) promotes facilitates migration and invasion of epithelial tumour cells. EMT is induced by growth factors implicated in theses process such as hepatocyte growth factor (HGF). Our aim of this study is whether HGF/c-Met pathway is associated with metastasis of gastric cancer (GC), especially in peritoneal dissemination (PD). Methods: HGF and c-Met expression and EMT related molecules were evaluated using real-time PCR and immunohistochemistry in GC tissues. The role of HGF/c-Met pathway for EMT and anoikis was determined and c-Met TKI (SU11274) was tested for their ability to block HGF-induced biological effects in vitro and vivo. Results: In HGF(-)c-Met(+) GC cells,recombinant HGF promoted EMT phenotype characterized by morphology, impaired E-cadherin and induction of Vimentin. HGF promoted cell growth, invasiveness, migration ability and inhibition of anoikis. SU11274 blocked HGF-induced EMT and the biological effects in vitro. In contrast of HGF(+)c-Met(+) GC cells, HGF exposure was not affected biological outcome of EMT and anoikis but SU11274 blocked biological effect as same as in HGF(-)c-Met(+) GC cells. In vivo, HGF(+)c-Met(+) GC cell line only established PD and SU11274 intraperitoneally caused an inhibition of PD growth. Clinically, HGF expression was significantly positive correlated with c-Met expression in GC specimens. Increased HGF and c-Met demonstrated a significantly associated with poor prognosis and can predict PD, respectively. Furthermore, HGF was one of the independent factors for predicting PD. Immunohistochemical analysis showed HGF and c-Met were predominantly co-expressed in cancer cell of both primary GC and PD. Conclusions: We have demonstrated that HGF/c-Met pathway as an inducer of EMT and anoikis inhibition in GC cell. Co-expression of HGF and c-Met implicates its potential to promote PD in GC. Blocking the autocrine HGF/c-Met pathway may be clinically useful for the treatment of PD in GC. No significant financial relationships to disclose.
2

Zhang, Hongli, Qingqing Feng, Wei-Dong Chen, and Yan-Dong Wang. "HGF/c-MET: A Promising Therapeutic Target in the Digestive System Cancers." International Journal of Molecular Sciences 19, no. 11 (October 23, 2018): 3295. http://dx.doi.org/10.3390/ijms19113295.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
The HGF/c-MET pathway is active in the development of digestive system cancers, indicating that inhibition of HGF/c-MET signaling may have therapeutic potential. Various HGF/c-MET signaling inhibitors, mainly c-MET inhibitors, have been tested in clinical trials. The observed efficacy and adverse events of some c-MET inhibitors were not very suitable for treating digestive system cancers. The development of new HGF/c-MET inhibitors in preclinical studies may bring promising treatments and synergistic combination (traditional anticancer drugs and c-MET inhibitors) strategies provided anacceptable safety and tolerability. Insights into miRNA biology and miRNA therapeutics have made miRNAs attractive tools to inhibit HGF/c-MET signaling. Recent reports show that several microRNAs participate in inhibiting HGF/c-MET signaling networks through antagonizing c-MET or HGF in digestive system cancers, and the miRNAs-HGF/c-MET axis plays crucial and novel roles for cancer treatment. In the current review, we will discuss recent findings about inhibitors of HGF/c-MET signaling in treating digestive system cancers, and how miRNAs regulate digestive system cancers via mediating HGF/c-MET pathway.
3

Weimar, Iris S., Daphne de Jong, Egbert J. Muller, Toshikazu Nakamura, Joost M.H.H. van Gorp, Gijsbert C. de Gast, and Winald R. Gerritsen. "Hepatocyte Growth Factor/Scatter Factor Promotes Adhesion of Lymphoma Cells to Extracellular Matrix Molecules Via α4β1 and α5β1 Integrins." Blood 89, no. 3 (February 1, 1997): 990–1000. http://dx.doi.org/10.1182/blood.v89.3.990.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Hepatocyte growth factor (HGF )/scatter factor (SF ) is the ligand for a tyrosine kinase cell surface receptor encoded by the MET protooncogene (c-MET). HGF/SF can induce proliferation and motility in epithelial cells and promotes invasion of carcinoma cells and NIH3T3 fibroblasts transfected with both HGF/SF and c-MET genes. Our results show that HGF/SF and c-MET also play a role in adhesion and invasion of human lymphoma cells. c-MET mRNA is expressed in hemopoietic cells, such as hemopoietic progenitor cells (CD34+ cells) in bone marrow (BM) and mobilized peripheral blood, immature B cells in cord blood and BM, and germinal center B-centroblasts. In normal peripheral blood B cells, which are c-MET−, c-MET expression was induced by PMA, ConA, HGF/SF, and Epstein-Barr virus (EBV) infection. Using immunohistochemistry, we detected c-MET on the cell surface of large activated centroblasts in lymph nodes from patients with B-non–Hodgkin's lymphoma and Hodgkin's disease. In the latter group, c-MET expression correlated well with the presence of EBV. Because HGF/SF and c-MET promote metastasis of carcinoma cells, we studied the effects of c-MET stimulation by HGF/SF of B-lymphoma cells on properties relevant for metastasis, ie, adhesion, migration, and invasion. HGF/SF stimulated adhesion of the c-MET+ B-cell lines to the extracellular matrix molecules fibronectin (FN) and collagen (CN) in a dose dependent manner. However, adhesion to laminin was not affected by HGF/SF. Adhesion to FN was mediated by β1-integrins α4β1 (VLA4) and α5β1 (VLA5) since blocking antibodies against β1- (CD29), α4- (CD49d), or α5- (CD49e) integrin subunits, completely reversed the effect of HGF/SF. Furthermore, HGF/SF induced adhesion was abrogated by addition of genistein, which blocks protein tyrosine kinases, including c-MET. Addition of HGF/SF resulted in a sixfold increase in migration of c-MET B-lymphoma cells through Matrigel, compared to medium alone. In rat fibroblast cultures, HGF/SF doubled the number of c-MET+ B-lymphoma cells that invaded the fibroblast monolayer. In these adhesion, migration and invasion assays HGF/SF had no effect on c-MET− cell lines. In conclusion, c-MET is expressed or can be induced on immature, activated, and certain malignant B cells. HGF/SF increased adhesion of c-MET+ B-lymphoma cells to FN and CN, mediated via β1-integrins α4β1 and α5β1 , and furthermore promoted migration and invasion.
4

Gallo, Simona, Valentina Sala, Stefano Gatti, and Tiziana Crepaldi. "Cellular and molecular mechanisms of HGF/Met in the cardiovascular system." Clinical Science 129, no. 12 (November 11, 2015): 1173–93. http://dx.doi.org/10.1042/cs20150502.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Met tyrosine kinase receptor, also known as c-Met, is the HGF (hepatocyte growth factor) receptor. The HGF/Met pathway has a prominent role in cardiovascular remodelling after tissue injury. The present review provides a synopsis of the cellular and molecular mechanisms underlying the effects of HGF/Met in the heart and blood vessels. In vivo, HGF/Met function is particularly important for the protection of the heart in response to both acute and chronic insults, including ischaemic injury and doxorubicin-induced cardiotoxicity. Accordingly, conditional deletion of Met in cardiomyocytes results in impaired organ defence against oxidative stress. After ischaemic injury, activation of Met provides strong anti-apoptotic stimuli for cardiomyocytes through PI3K (phosphoinositide 3-kinase)/Akt and MAPK (mitogen-activated protein kinase) cascades. Recently, we found that HGF/Met is also important for autophagy regulation in cardiomyocytes via the mTOR (mammalian target of rapamycin) pathway. HGF/Met induces proliferation and migration of endothelial cells through Rac1 (Ras-related C3 botulinum toxin substrate 1) activation. In fibroblasts, HGF/Met antagonizes the actions of TGFβ1 (transforming growth factor β1) and AngII (angiotensin II), thus preventing fibrosis. Moreover, HGF/Met influences the inflammatory response of macrophages and the immune response of dendritic cells, indicating its protective function against atherosclerotic and autoimmune diseases. The HGF/Met axis also plays an important role in regulating self-renewal and myocardial regeneration through the enhancement of cardiac progenitor cells. HGF/Met has beneficial effects against myocardial infarction and endothelial dysfunction: the cellular and molecular mechanisms underlying repair function in the heart and blood vessels are common and include pro-angiogenic, anti-inflammatory and anti-fibrotic actions. Thus administration of HGF or HGF mimetics may represent a promising therapeutic agent for the treatment of both coronary and peripheral artery disease.
5

De Silva, Dinuka M., Arpita Roy, Takashi Kato, Fabiola Cecchi, Young H. Lee, Kunio Matsumoto, and Donald P. Bottaro. "Targeting the hepatocyte growth factor/Met pathway in cancer." Biochemical Society Transactions 45, no. 4 (July 3, 2017): 855–70. http://dx.doi.org/10.1042/bst20160132.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Hepatocyte growth factor (HGF)-induced activation of its cell surface receptor, the Met tyrosine kinase, drives mitogenesis, motogenesis and morphogenesis in a wide spectrum of target cell types and embryologic, developmental and homeostatic contexts. Typical paracrine HGF/Met signaling is regulated by HGF activation at target cell surfaces, HGF binding-induced receptor activation, internalization and degradation. Despite these controls, HGF/Met signaling contributes to oncogenesis, tumor angiogenesis and invasiveness, and tumor metastasis in many types of cancer, leading to the rapid growth of pathway-targeted anticancer drug development programs. We review here HGF and Met structure and function, basic properties of HGF/Met pathway antagonists now in clinical development, and recent clinical trial results. Presently, the main challenges facing the effective use of HGF/Met-targeted antagonists for cancer treatment include optimal patient selection, diagnostic and pharmacodynamic biomarker development, and the identification and testing of effective therapy combinations. The wealth of basic information, analytical reagents and model systems available regarding normal and oncogenic HGF/Met signaling will continue to be invaluable in meeting these challenges and moving expeditiously toward more effective cancer treatment.
6

Kauma, Scott, Natalie Hayes, and Shannon Weatherford. "The Differential Expression of Hepatocyte Growth Factor and Met in Human Placenta*." Journal of Clinical Endocrinology & Metabolism 82, no. 3 (March 1, 1997): 949–54. http://dx.doi.org/10.1210/jcem.82.3.3806.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Abstract Met is the tyrosine kinase receptor for the ligand hepatocyte growth factor (HGF). Met/HGF plays an important role in epithelial cell proliferation, migration, and morphogenesis. HGF also plays a crucial role in placental development in the mouse. To determine whether HGF potentially has a similar role in human placental development, the production and localization of Met and HGF were determined in early second trimester and term placentas. Reverse transcription-PCR using specific primers demonstrated the expression of Met and HGF messenger ribonucleic acid in placental villi. HGF production was determined by enzyme-linked immunosorbent assay. HGF production over 48 h by second trimester placental villous explants in culture (810 pg/mg total protein·h) was 2.1-fold greater than that in term placental villous explants (380 pg/mg total protein·h; P < 0.01). Isolated trophoblast did not produce HGF, whereas isolated villous core tissues and villous core mesenchymal cells did produce HGF. Interleukin-1β treatment of placental villi or coculture of villous core mesenchymal cells with isolated trophoblast cells did not stimulate HGF production. Using immunohistochemistry, HGF localized to the villous core compartment with no localization to the trophoblast. In contrast, Met localized mainly to cytotrophoblast. These findings suggest that HGF produced by the villous core may act in a paracrine fashion to regulate trophoblast development or function through the HGF receptor, Met.
7

Yamasaki, Koji, Shoichiro Mukai, Takahiro Nagai, Kozue Nakahara, Masato Fujii, Naoki Terada, Akinobu Ohno, et al. "Matriptase-Induced Phosphorylation of MET is Significantly Associated with Poor Prognosis in Invasive Bladder Cancer; an Immunohistochemical Analysis." International Journal of Molecular Sciences 19, no. 12 (November 22, 2018): 3708. http://dx.doi.org/10.3390/ijms19123708.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Hepatocyte growth factor (HGF) plays an important role in cancer progression via phosphorylation of MET (c-met proto-oncogene product, receptor of HGF). HGF-zymogen (pro-HGF) must be processed for activation by HGF activators including matriptase, which is a type II transmembrane serine protease and the most efficient activator. The enzymatic activity is tightly regulated by HGF activator inhibitors (HAIs). Dysregulated pro-HGF activation (with upregulated MET phosphorylation) is reported to promote cancer progression in various cancers. We retrospectively analyzed the expression of matriptase, phosphorylated-MET (phospho-MET) and HAI-1 in tumor specimens obtained from patients with invasive bladder cancer by immunohistochemistry. High expression of phospho-MET and increased expression of matriptase were significantly associated with poor prognosis, and high matriptase/low HAI-1 expression showed poorer prognosis. Furthermore, high expression of matriptase tended to correlate with phosphorylation of MET. Increased expression of matriptase may induce the ligand-dependent activation of MET, which leads to poor prognosis in patients with invasive bladder cancer.
8

Woolf, A. S., M. Kolatsi-Joannou, P. Hardman, E. Andermarcher, C. Moorby, L. G. Fine, P. S. Jat, M. D. Noble, and E. Gherardi. "Roles of hepatocyte growth factor/scatter factor and the met receptor in the early development of the metanephros." Journal of Cell Biology 128, no. 1 (January 1, 1995): 171–84. http://dx.doi.org/10.1083/jcb.128.1.171.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Several lines of evidence suggest that hepatocyte growth factor/scatter factor (HGF/SF), a soluble protein secreted by embryo fibroblasts and several fibroblast lines, may elicit morphogenesis in adjacent epithelial cells. We investigated the role of HGF/SF and its membrane receptor, the product of the c-met protooncogene, in the early development of the metanephric kidney. At the inception of the mouse metanephros at embryonic day 11, HGF/SF was expressed in the mesenchyme, while met was expressed in both the ureteric bud and the mesenchyme, as assessed by reverse transcription PCR, in situ hybridization, and immunohistochemistry. To further investigate the expression of met in renal mesenchyme, we isolated 13 conditionally immortal clonal cell lines from transgenic mice expressing a temperature-sensitive mutant of the SV-40 large T antigen. Five had the HGF/SF+/met+ phenotype and eight had the HGF/SF-/met+ phenotype. None had the HGF/SF+/met- nor the HGF/SF-/met- phenotypes. Thus the renal mesenchyme contains cells that express HGF/SF and met or met alone. When metanephric rudiments were grown in serum-free organ culture, anti-HGF/SF antibodies (a) inhibited the differentiation of metanephric mesenchymal cells into the epithelial precursors of the nephron; (b) increased cell death within the renal mesenchyme; and (c) perturbed branching morphogenesis of the ureteric bud. These data provide the first demonstration for coexpression of the HGF/SF and met genes in mesenchymal cells during embryonic development and also imply an autocrine and/or paracrine role for HGF/SF and met in the survival of the renal mesenchyme and in the mesenchymal-epithelial transition that occurs during nephrogenesis. They also confirm the postulated paracrine role of HGF/SF in the branching of the ureteric bud.
9

Tanaka, Ryota, Mizue Terai, Eric Londin, and Takami Sato. "The Role of HGF/MET Signaling in Metastatic Uveal Melanoma." Cancers 13, no. 21 (October 30, 2021): 5457. http://dx.doi.org/10.3390/cancers13215457.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (MET) signaling promotes tumorigenesis and tumor progression in various types of cancer, including uveal melanoma (UM). The roles of HGF/MET signaling have been studied in cell survival, proliferation, cell motility, and migration. Furthermore, HGF/MET signaling has emerged as a critical player not only in the tumor itself but also in the tumor microenvironment. Expression of MET is frequently observed in metastatic uveal melanoma and is associated with poor prognosis. It has been reported that HGF/MET signaling pathway activation is the major mechanism of treatment resistance in metastatic UM (MUM). To achieve maximal therapeutic benefit in MUM patients, it is important to understand how MET signaling drives cellular functions in uveal melanoma cells. Here, we review the HGF/MET signaling biology and the role of HGF/MET blockades in uveal melanoma.
10

Czyz, Malgorzata. "HGF/c-MET Signaling in Melanocytes and Melanoma." International Journal of Molecular Sciences 19, no. 12 (December 3, 2018): 3844. http://dx.doi.org/10.3390/ijms19123844.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Hepatocyte growth factor (HGF)/ mesenchymal-epithelial transition factor (c-MET) signaling is involved in complex cellular programs that are important for embryonic development and tissue regeneration, but its activity is also utilized by cancer cells during tumor progression. HGF and c-MET usually mediate heterotypic cell–cell interactions, such as epithelial–mesenchymal, including tumor–stroma interactions. In the skin, dermal fibroblasts are the main source of HGF. The presence of c-MET on keratinocytes is crucial for wound healing in the skin. HGF is not released by normal melanocytes, but as melanocytes express c-MET, they are receptive to HGF, which protects them from apoptosis and stimulates their proliferation and motility. Dissimilar to melanocytes, melanoma cells not only express c-MET, but also release HGF, thus activating c-MET in an autocrine manner. Stimulation of the HGF/c-MET pathways contributes to several processes that are crucial for melanoma development, such as proliferation, survival, motility, and invasiveness, including distant metastatic niche formation. HGF might be a factor in the innate and acquired resistance of melanoma to oncoprotein-targeted drugs. It is not entirely clear whether elevated serum HGF level is associated with low progression-free survival and overall survival after treatment with targeted therapies. This review focuses on the role of HGF/c-MET signaling in melanoma with some introductory information on its function in skin and melanocytes.

Dissertations / Theses on the topic "MET/HGF":

1

Ding, Shunli. "Rôle du couple HGF/C-MET dans l'angiogenèse." Paris 7, 2004. http://www.theses.fr/2004PA077188.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Moore, Amy Elizabeth. "The role of HGF/Met signalling in colorectal tumorigenesis." Thesis, University of Bristol, 2012. http://ethos.bl.uk/OrderDetails.do?uin=uk.bl.ethos.544331.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Besret, Soizic. "Ligations chimiques : synthèse d'inhibiteurs extracellulaires de la signalisation HGF/SF-MET." Phd thesis, Université du Droit et de la Santé - Lille II, 2011. http://tel.archives-ouvertes.fr/tel-00630962.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Les peptides constituent une famille de biomolécules dont l'utilisation dans différents domaines thérapeutiques (cancer, diabète, sida) s'est fortement développée ces dernières années. Le défi pour les chimistes consiste à y accéder grâce à de nouvelles méthodes fiables et efficaces. La première partie de notre travail a d'abord été orientée vers le développement deux méthodes de ligations non natives efficaces et complémentaires de celles existant. La première méthode, appelée ligation thiocarbamate, permet d'obtenir des peptides alkylthiocarbamate avec de très bons rendements, alors que la seconde, appelée ligation azaGly, aboutit à la formation d'un azaGlypeptide. La seconde partie de cette thèse traite de la conception et synthèse de nouveaux peptides susceptibles d'inhiber la signalisation HGF/SF-MET. Le récepteur à activité tyrosine kinase MET et son ligand, l'HGF/SF (Hepatocyte Growth Factor/Scattor Factor), sont des cibles de choix pour une thérapie anti-cancéreuse. La ligation thiocarbamate, précédemment décrite, et la ligation thioéther plus classique ont été utilisées pour préparer une chimiothèque de peptides sulfonatés d'inhiber cette signalisation de façon extracellulaire. La capacité de liaison des composés de la chimiothèque avec le domaine extracellulaire de MET a été évaluée grâce à la technologie biopuces. L'activité biologique (tests MTT, d'activité kinase) des meilleurs produits a été ensuite évaluée.
4

Murat, Cahue de Bernardis. "Estudo molecular dos componentes da via de sinalização HGF/MET em insulinomas." Universidade de São Paulo, 2013. http://www.teses.usp.br/teses/disponiveis/5/5135/tde-03102013-110107/.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Os insulinomas são os tumores neuroendócrinos pancreáticos funcionantes mais frequentes, entretanto, os aspectos moleculares envolvidos em sua tumorigênese precisam ser melhor esclarecidos. As características morfológicas e histoquímicas dos insulinomas não conseguem predizer completamente seu comportamento biológico, apenas o fenótipo invasivo local e a presença de metástase são as formas confiáveis do diagnóstico maligno. A presente investigação teve por objetivos analisar a expressão gênica por reação em cadeia da polimerase (PCR) em tempo real e a expressão protéica por imuno-histoquímica dos componentes da via de sinalização do fator de crescimento hepatocítico (HGF) e seu receptor (c-MET) em 27 amostras de insulinomas, sendo: 16 tumores benignos grau 1 (G1), seis tumores benignos grau 2 (G2), dois insulinomas malignos grau 3 (G3) e três metástases hepáticas. Além disso, realizou-se a pesquisa de mutações somáticas no gene MET. Observou-se (1) o aumento da expressão dos genes HGF, MET e ST14 (codificante para a matriptase) e a baixa expressão do gene HAI-1 (codificante para a protease inibidora tipo-kunitz do tipo 1) nos insulinomas malignos e metástases quando comparados aos insulinomas benignos G1; (2) uma correlação positiva entre a expressão do mRNA do gene MET e o gene ST14 e índice proliferativo Ki-67, bem como uma correlação inversa entre a expressão do mRNA do gene HAI-1 e os genes MET, HGF, ST14 e o índice mitótico; (3) uma correlação positiva entre a expressão do gene ST14 e a expressão do mRNA do gene HGF; (4) maior expressão protéica de c-MET nos insulinomas malignos G3 em relação aos insulinomas G1/G2 e (5) ausência de mutações nos éxons 2, 10, 14, 16, 17 e 19 do gene MET. Concluiu-se que os genes HGF, MET, ST14 e HAI-1 estão diferencialmente expressos entre insulinomas malignos e benignos, o que pode ter implicações diagnósticas e terapêuticas
In an attempt to better understand the molecular processes involved in the tumourigenesis of islet beta-cells, the present study evaluated the expression of genes belonging to the hepatocyte growth factor and its receptor (HGF/MET) system, namely, MET, HGF; HGFAC and ST14 (encode HGF activator and matriptase, respectively, two serine proteases that catalyze conversion of pro-HGF to active HGF); and SPINT1 and SPINT2 (encode serine peptidase inhibitors Kunitz type 1 and type 2, respectively, two potent inhibitors of HGF activator and of matriptase) in 27 sporadic insulinomas; 16 grade 1 (G1), six grade 2 (G2), two grade 3 (G3) and three hepatic metastases. Quantitative reverse-transcriptase polymerase chain reaction was employed to assess RNA expression of the target genes and immunohistochemical analysis was used to evaluate the expression of MET and SPINT1. Somatic mutations of MET gene were searched by direct sequencing of exons 2, 10, 14, 16, 17 and 19. Overexpression of MET was observed in grouped G3 insulinomas and metastases concomitantly with upregulation of the genes encoding HGF and matriptase and downregulation of SPINT1. Positive correlations were observed between MET RNA expression and Ki-67 proliferation index while a negative correlation was detected between SPINT1 expression and the mitotic index. No somatic mutations were found in MET gene. The final effect of the increased expression of HGF, its activator (matriptase) and its specific receptor (MET) together with a decreased expression of one potent inhibitor of matriptase (SPINT1) is probably a contribution to tumoural progression and malignancy in insulinomas
5

Mekki, Meriem Sarah. "Conséquences de l'hypoxie sur la régulation de la signalisation HGF/SF-MET." Thesis, Lille 2, 2015. http://www.theses.fr/2015LIL2S047/document.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Le récepteur à activité tyrosine kinase MET et son ligand le facteur de croissance des hepatocytes (Hepatocyte Growth Factor/Scattor Factor (HGF/SF)) sont essentiels pour la migration, la morphogenèse et la survie des cellules épithéliales. En plus de son implication et son importance physiologiques, la dérégulation de la signalisation de MET favorise la progression et l’invasion tumorales dans plusieurs types de cancers. Au sein des tumeurs, l’hypoxie est également un phénomène crucial qui induit une réponse adaptative menant à l’invasion, la métastase cancéreuses et la résistance aux traitements.Nous avons démontré que dans des conditions hypoxiques, la phosphorylation de MET induite par sa liaison au ligand, des mutations activatrices ou sa surexpression, est diminuée de manière importante in vitro et in vivo dans des modèles de tumeurs expérimentales chez la souris. Cette baisse de phosphorylation est très rapide et est réversible quand les cellules sont replacées en normoxie. Alors que la phosphorylation de GAB1, principal partenaire de MET, est également diminuée en hypoxie, l’activation des voies de signalisation en aval AKT et ERK n’est pas affectée et reste bien dépendante de l’activité du récepteur et du recrutement de GAB1. De la même façon, l’HGF/SF induit des réponses de motilité, de dispersion, de morphogenèse et de survie similaires en normoxie et en hypoxie. De manière intéressante, le traitement par deux inhibiteurs de tyrosine kinase (ITK) ciblant MET (PHA-665752 et SU11274) est moins efficace en hypoxie pour inhiber les voies de signalisation AKT et ERK ainsi les réponses cellulaires induites par MET. Comme pour la phosphorylation de MET, la résistance à ces ITK est un phénomène réversible. Ainsi, alors que l’hypoxie n’affecte pas les voies de signalisation en aval ni les effets biologiques, elle diminue la sensibilité de MET aux ITK induisant donc une résistance immédiate. L’ensemble de ces données pourrait fournir de nouvelles perspectives dans l’utilisation des thérapies ciblant MET dans les tumeurs solides
The receptor tyrosine kinase MET and its ligand the Hepatocyte Growth Factor/Scattor Factor (HGF/SF) are essential for migration, morphogenesis and survival of epithelial cells. Beside its physiological involvement, deregulation of MET signaling has been shown to promote tumor progression and invasion in many cancers. Inside the tumors, hypoxia is also a crucial phenomenon promoting an adaptive response able to induce invasion, metastasis and resistance to treatment.We show that under hypoxia, MET phosphorylation induced by ligand-stimulation, activating mutation or overexpression, is drastically decreased both in cell culture and in experimental tumors. This decrease in MET phosphorylation occurs within minutes and is reversible when cells are returned to normoxia. While phosphorylation of the proximal signaling adaptor GAB1 is also decreased in hypoxia, activation of the downstream kinases ERK and AKT is not affected, but is still dependent on MET receptor activity. Consistently, several cellular responses induced by HGF/SF, including motility, morphogenesis or survival, are still efficiently induced. Interestingly, treatment with two tyrosine kinase inhibitors targeting MET (PHA-665752 and SU11274) are less efficient to inhibit the downstream kinases ERK and AKT and cellular responses induced by MET in hypoxia compared to normoxia. Similarly to MET phosphorylation, this resistance to TKI is a reversible phenomenon. Therefore, while hypoxia does not affect downstream signaling and cellular responses, it decreases MET sensitivity to TKIs targeting the receptor thus providing an immediate resistance. This may provide new insights in the use of MET targeted therapies in solid tumors
6

Tjin, Esther Pit Mien. "The HGF/MET and WNT signaling pathways in B cell differentiation and neoplasia." [S.l. : Amsterdam : s.n.] ; Universiteit van Amsterdam [Host}, 2005. http://dare.uva.nl/document/17835.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Stefan, Monica. "Identifizierung eines neuen Interaktionspartners des HGF-Rezeptors c-Met SHIP-1 spielt eine Schlüsselrolle bei der Vermittlung der HGF-induzierten Tubulogenese von Epithelzellen /." [S.l.] : [s.n.], 2001. http://deposit.ddb.de/cgi-bin/dokserv?idn=961707232.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Arlt, Franziska. "Das neu identifizierte Gen MACC1 ist ein Regulator des HGF/Met-Signalweges und ist prognostisch für die Metastasierung des Kolonkarzinoms." Doctoral thesis, Humboldt-Universität zu Berlin, Mathematisch-Naturwissenschaftliche Fakultät I, 2009. http://dx.doi.org/10.18452/15974.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Das Kolonkarzinom ist eine der häufigsten Tumorerkrankungen weltweit. Etwa 50 % der Patienten entwickeln Fernmetastasen. Diese haben eine sehr schlechte Überlebensprognose. Deshalb fokussiert die Forschung auf die Identifizierung neuer, molekularer Marker für eine verbesserte Metastasierungsvorhersage. Identifizierte Hochrisiko-Patienten könnten somit rechtzeitig eine individualisierte, intensivere Therapie erhalten. MACC1 (Metastasis-associated in colon cancer 1) ist ein neu identifiziertes Gen, das in Kolonkarzinomen und deren Fernmetastasen überexprimiert wird. Die Domänenstruktur von MACC1 ist kennzeichnend für Proteine der Rezeptor-Tyrosinkinase-Signalwege. Ziel dieser Arbeit war die Aufklärung der zellulären Funktion von MACC1 und seiner Rolle in der Tumorprogression sowie die Evaluierung von MACC1 als molekularer Metastasierungsmarker. MACC1-überexprimierende Tumorzellen zeigten in Abhängigkeit von der Domänenstruktur in in vitro Assays ein erhöhtes migratorisches, invasives und proliferatives Potential. Der Einfluss von MACC1 auf die Metastasierungskapazität von Tumorzellen konnte auch im Tiermodell belegt werden. Der Hepatocyte-growth-factor (HGF) induziert die epitheliale-mesenchymale Transition MACC1-exprimierender Zellen und die nukleäre Translokation von MACC1. Die Expression des HGF-Rezeptors Met war in diesen Zellen stark erhöht. Reportergen-Studien bestätigten die transkriptionelle Regulation von Met durch MACC1. Die Analyse humaner Kolonkarzinome ergab eine signifikant höhere MACC1 Expression in Primärtumoren mit metachroner Fernmetastasierung. MACC1 ist ein neu identifizierter Regulator des HGF/Met-Signalweges und trägt somit entscheidend zur Determinierung des metastatischen Potentials von Tumorzellen bei. MACC1 hat großes Potential als neuer, prognostischer Marker für die Metastasierung des Kolonkarzinoms und ist ein Kandidatengen als Ziel effektiver, molekularer Interventionsstrategien zur Metastasierungs-Prävention.
Colon cancer is one of the most frequent malignant diseases worldwide. About 50% of the patients develop distant metastasis. These patients have only few therapy options and very poor survival rates. Therefore cancer research focuses on the identification of novel molecular markers to provide a better prognosis of the metastatic risk. Identified high-risk patients would get access to an early, individualized therapy. MACC1 (metastasis associated in colon cancer 1) is a newly identified gene that is overexpressed in colon carcinomas and their distant metastases. The MACC1 domain structure is characteristic for proteins of the receptor tyrosine kinase signalling pathways. Aim of this study was the analysis of the cellular function of MACC1, its role in tumor progression and its evaluation as a molecular, prognostic marker for metastasis. MACC1 overexpressing tumor cells revealed higher migratory, invasive, and proliferative potential in in vitro assays. The impact of MACC1 on the metastatic potential of tumors was also shown in mouse models. The hepatocyte growth factor (HGF) induced epithelial-mesenchymal-transition in MACC1 positive cells and MACC1´s nuclear translocation. Expression of the HGF receptor Met was strongly elevated in these cells. Reporter gene experiments confirmed the transcriptional regulation of Met by MACC1. Analyses in human colon carcinomas showed a significantly higher MACC1 expression in tumors that developed distant metastases. MACC1 is a newly identified regulator of the HGF/Met signalling pathway. It contributes decisively to the metastatic capacity of tumor cells. MACC1 has great potential as new prognostic marker for colon cancer metastasis and is a promising candidate as target for effective, molecular intervention strategies for metastasis prevention.
9

Rocha, Angélica Gomes 1989. "Estudo da expressão proteica da via HGF/c-Met/STAT3 no carcinoma diferenciado da tiroide." [s.n.], 2014. http://repositorio.unicamp.br/jspui/handle/REPOSIP/310289.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Orientadores: Laura Sterian Ward, Antônio Hugo José Fróes Marques Campos
Dissertação (mestrado) - Universidade Estadual de Campinas, Faculdade de Ciências Médicas
Made available in DSpace on 2018-08-26T01:02:11Z (GMT). No. of bitstreams: 1 Rocha_AngelicaGomes_M.pdf: 1644333 bytes, checksum: e3503fb6e2c8209f7a6696df111d6a78 (MD5) Previous issue date: 2014
Resumo: Marcadores de malignidade, especialmente capazes de distinguir lesões de padrão folicular, que sejam de fácil implantação na rotina do diagnóstico de nódulos tireoidianos, continuam sendo extremamente necessários, dado o crescente aumento de nódulos tireoidianos diagnosticados nos últimos anos. A via HGF/c-Met/STAT3 está relacionada com desenvolvimento e progressão tumoral, sendo que a expressão de c-Met, HGF e de STAT3 foram descritas em grande parte dos carcinomas papilíferos de tireoide (CPT), mas não em tecido tireoidiano normal, sugerindo sua relação com o desenvolvimento e progressão do CPT. Para avaliar a utilidade da expressão proteica de c-Met, HGF, STAT3, e de sua proteína fosforilada (pSTAT3) no diagnóstico e no prognóstico de pacientes com nódulos tireoidianos, analisamos 356 tecidos tireoidianos, sendo 153 carcinomas papilíferos (CPT), dos quais 95 eram clássicos (CPC), 47 carcinomas papilíferos variante folicular (CPVF), e 11 carcinomas papilíferos de células altas (CPCA); 34 carcinomas foliculares (CFT), 34 adenomas foliculares (AF), 124 bócios e 11 tecidos normais. Todos os pacientes foram tratados e acompanhados de acordo com um mesmo protocolo padrão por 1-10 anos (Mo=5 anos). Áreas representativas do tecido foram selecionadas para a construção de uma lâmina de tissue microarray (TMA) que foi submetida à técnica de imunoistoquímica e analisada pelo score de Allred. A expressão citoplasmática de c-Met foi capaz de diferenciar nódulos malignos de benignos (p<0,0001, sensibilidade 86%, especificidade 76%, VPP 77%, VPN 86%); CPT de CF (p=0,0003, sensibilidade 96%, especificidade 31%, VPP 87%, VPN 63%); variante folicular de CPT de CF (p=0,0232 sensibilidade 93%, especificidade 31%, VPP 66%, VPN 77%); assim como variante folicular de CPT de AF (p=0,0003, sensibilidade 93%, especificidade 50%, VPP 68%, VPN 87%). Além disso, a expressão de c-Met se correlacionou com tireoidite (p<0,0001) e multifocalidade (p=0,0028), mas não com presença de cápsula, invasão, tamanho do tumor, estadiamento TNM, e presença de metástase no diagnóstico e na evolução. A expressão nuclear de STAT3 diferenciou os nódulos benignos dos malignos (p<0,0001, sensibilidade 83%, especificidade 74%, VPP 75%, VPN 83%); CF de AF (p=0,0457, sensibilidade 80%, especificidade 52%, VPP 65%, VPN 71%); bócios de CPT variante folicular (p<0,001, sensibilidade 89%, especificidade 65%, VPP 91%, VPN 60%); bócio de CF (p<0,0001, sensibilidade 89%, especificidade 80%, VPP 95%, VPN 60%); bócio de AF (p=0,0005, sensibilidade 89%, especificidade 80%, VPP 95%, VPN 60%). Além disso, a expressão de STAT3 se correlacionou com tireoidite (p=0,0095) e multifocalidade (p<0,0001), mas não com presença de cápsula, invasão, tamanho do tumor, estadiamento TNM, e presença de metástase no diagnóstico e na evolução. A expressão de pSTAT3 e HGF não auxiliou no diagnóstico dos nódulos, e tampouco se correlacionou com características de agressividade dos tumores. Conclui-se que as proteínas c-Met e STAT3 podem ser consideradas marcadores clínicos úteis na rotina de laboratórios, uma vez que foram capazes de diferenciar os nódulos malignos dos benignos, alguns tipos histológicos dos nódulos, além de se correlacionarem com fatores de agressividade dos tumores
Abstract: Malignancy markers, especially the ones that are capable of distinguishing follicular lesions and with easy deployment in the routine diagnosis of thyroid nodules are much needed, given the increasing number of thyroid nodules in recent years. The HGF/c-Met/STAT3 pathway is related to the development and progression of many types of cancers, and c-Met, HGF and STAT3 expression were described in most papillary thyroid carcinomas (PTC), but not in normal thyroid tissue, suggesting it is related with the development and progression of PTC. To evaluate the usefulness of c-Met, HGF, STAT3, and its phosphorylated form (pSTAT3) protein expression in the diagnosis and prognosis of patients with thyroid nodules, we analyzed 356 thyroid tissues, including 153 papillary carcinomas (PTC), 95 classical type, 47 follicular variants of papillary carcinoma, and 11 tall cells carcinomas; 34 follicular carcinomas (FC), 34 follicular adenomas (FA), 124 goiters and 11 normal tissues. All patients were treated and monitored according to the same standard protocol for 1-10 years (Mo = 5 years). Representative tissue areas were selected for the construction of a tissue microarray (TMA) which was subjected to immunohistochemistry and analyzed by the Allred score. The cytoplasmic expression of c-Met was able to differentiate malignant from benign nodules (p <0.0001, sensitivity 86%, specificity 76%, PPV 77%, 86% NPV); PTC from FCT (p = 0.0003, sensitivity 96%, specificity 31%, PPV 87%, 63% NPV); follicular variant of PTC from FCT (p = 0.0232 sensitivity 93%, specificity 31%, PPV 66%, NPV 77%); as well as follicular variant of CPT from FA (p = 0.0003, sensitivity 93%, specificity 50%, PPV 68%, 87% NPV). Furthermore, c-Met expression was correlated to the presence of thyroiditis (p<0.0001) and multifocality (p=0.0028), but not with the presence of capsule, invasion, tumour size, TNM staging, and metastasis at diagnosis or evolution of the disease. The nuclear expression of STAT3 differentiated benign from malignant nodules (p <0.0001, sensitivity 83%, specificity 74%, PPV 75%, NPV 83%); FCT from FA (p = 0.0457, sensitivity 80%, specificity 52%, PPV 65%, NPV 71%); goiter follicular variant of PTC (p <0.001, sensitivity 89%, specificity 65%, PPV 91%, 60% NPV); goiter from FCT (p <0.0001, sensitivity 89%, specificity 80%, PPV 95%, NPV 60%); goiter from FA (p = 0.0005, sensitivity 89%, specificity 80%, PPV 95%, NPV 60%). In addition, STAT3 expression was associated with thyroiditis (p=0.0095) and multifocality (p<0.0001), but not with the presence of capsule, invasion, tumour size, TNM staging, and metastasis at diagnosis or evolution of the disease. The expression of pSTAT3 and HGF did not help the diagnostic of nodules and was not correlated with any tumour characteristic of aggressiveness. We concluded that c-Met and STAT3 could be useful as molecular markers in laboratory routine, helping to differentiate malignant from benign nodules, and some histological types of nodules, and was also associated with some tumour characteristics of aggressiveness
Mestrado
Ensino em Saúde
Mestra em Clínica Médica
10

Chmielowiec, Jolanta. "The role of the Met tyrosine kinase receptor in skin maintenance and regeneration." Doctoral thesis, Humboldt-Universität zu Berlin, Mathematisch-Naturwissenschaftliche Fakultät I, 2007. http://dx.doi.org/10.18452/15690.

Full text
APA, Harvard, Vancouver, ISO, and other styles
Abstract:
Met und der korrespondierende Ligand HGF/SF sind im hyperproliferativen Epithelium von Hautwunden exprimiert. Aus diesem Grund ist es wahrscheinlich, dass der Rezeptor und sein Ligand in autokriner Weise wechselwirken und wichtige Funktionen für den Heilungsprozess der Haut besitzen. Unter Verwendung der Keratin 14 Cre-Rekombinase konnte ein „Knockout“ des Met-Rezeptors spezifisch in der Epidermis erzielt werden. In der Tat zeigten die Ergebnisse, dass Met für die Re-epithelisierung in Wundschlussprozessen essentiell ist, da in den an der Wundheilung beteiligten Keratinozyten keine Rekombination des Met-Gens stattgefunden hat. In Met-Mausmutanten war der Wundschlussprozess verlangsamt, denn er erfolgte ausschließlich durch wenige Keratinozyten, in denen die Cre-Rekombinase keine Rekombination bewirkte. Met konnte als erstes Gen identifiziert werden, das absolut erforderlich für Re epithelisierungsprozesse von Wunden ist. Diese Arbeit trägt daher wesentlich zum Verständnis der Regulation von Wundheilungsprozessen bei.
Met and its ligand, HGF/SF are expressed in the hyperproliferative epithelium of the wound. This suggests that receptor and ligand may act in an autocrine manner to promote wound healing in the skin. Using Keratin 14 cre recombinase, Met receptor was specifically knockout in the epidermis. In this way, it was demonstrated that Met receptor is essential for wound healing process and that keratinocytes, which contributed to the wound closure were Met-postitive. In the Met mutant mice, wound closure was slightly attenuated, but occurred exclusively by a few keratinocytes that had escaped recombination. Met is therefore the fist gene, which is absolutely required for re-epithelialization of the wound. This finding is fundamental for understanding the regulation of wound healing process.

Books on the topic "MET/HGF":

1

1948-, Goldberg I. D., and Rosen E. M, eds. Hepatocyte growth factor-scatter factor (HGF-SF) and the C-met receptor. Basel: Birkhäuser, 1992.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
2

Somerset, David Alan. Hepatocyte growth factor (HGF) and its receptor c-met, in healthy and pathological human placentae. Birmingham: University of Birmingham, 2001.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
3

Alami, Jennifer. The role of HGF and met in Wilms tumour. 2004.

Find full text
APA, Harvard, Vancouver, ISO, and other styles
4

Long, Isolde Marie Seiden. Functional relationships between hepatocyte growth factor receptor (HGF)/Met signaling and Ki-ras oncogenic mutation in colon carcinoma cells. 2005.

Find full text
APA, Harvard, Vancouver, ISO, and other styles

Book chapters on the topic "MET/HGF":

1

Ray, Mandira, J. G. Garcia, and Ravi Salgia. "HGF/c-MET Signaling in Advanced Cancers." In Cancer Genome and Tumor Microenvironment, 273–92. New York, NY: Springer New York, 2009. http://dx.doi.org/10.1007/978-1-4419-0711-0_12.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Vande Woude, G. F., M. Jeffers, J. Cortner, G. Alvord, I. Tsarfaty, and J. Resau. "Met-HGF/SF: Tumorigenesis, Invasion and Metastasis." In Ciba Foundation Symposium 212 - Plasminogen-Related Growth Factors, 119–32. Chichester, UK: John Wiley & Sons, Ltd., 2007. http://dx.doi.org/10.1002/9780470515457.ch8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Thewke, Douglas P., Jianqun Kou, Makenzie L. Fulmer, and Qian Xie. "The HGF/MET Signaling and Therapeutics in Cancer." In Current Human Cell Research and Applications, 155–81. Singapore: Springer Singapore, 2017. http://dx.doi.org/10.1007/978-981-10-7296-3_8.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Zambelli, Alberto, Giuseppe Biamonti, and Angela Amato. "HGF/c-Met Signalling in the Tumor Microenvironment." In Advances in Experimental Medicine and Biology, 31–44. Cham: Springer International Publishing, 2020. http://dx.doi.org/10.1007/978-3-030-47189-7_2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Karsten, Ruud, and Joost Roeters. "Behandeling Van Een PatiëNt Met Erfelijke Of Idiopathische Gingivafibromatose (HGF)." In Tandheelkundige casuïstiek, 481–92. Houten: Bohn Stafleu van Loghum, 2008. http://dx.doi.org/10.1007/978-90-313-8811-0_92.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Abounader, Roger, and John Laterra. "HGF/c-Met Signaling and Targeted Therapeutics in Brain Tumors." In CNS Cancer, 933–52. Totowa, NJ: Humana Press, 2009. http://dx.doi.org/10.1007/978-1-60327-553-8_39.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Chandel, Vaishali, Sibi Raj, Ramesh Choudhari, and Dhruv Kumar. "Role of c-Met/HGF Axis in Altered Cancer Metabolism." In Cancer Cell Metabolism: A Potential Target for Cancer Therapy, 89–102. Singapore: Springer Singapore, 2020. http://dx.doi.org/10.1007/978-981-15-1991-8_7.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Corso, Simona, and Silvia Giordano. "Mechanisms of Resistance to Molecular Therapies Targeting the HGF/MET Axis." In Resistance to Targeted Anti-Cancer Therapeutics, 67–87. Cham: Springer International Publishing, 2018. http://dx.doi.org/10.1007/978-3-319-67932-7_4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Cortner, J., G. F. Vande Woude, and S. Rong. "The Met-HGF/SF autocrine signaling mechanism is involved in sarcomagenesis." In Experientia Supplementum, 89–121. Basel: Birkhäuser Basel, 1995. http://dx.doi.org/10.1007/978-3-0348-9070-0_6.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Joffre, Carine, Rachel Barrow, Ludovic Ménard, and Stéphanie Kermorgant. "RTKs as Models for Trafficking Regulation: c-Met/HGF Receptor-c-Met Signalling in Cancer—Location Counts." In Vesicle Trafficking in Cancer, 261–77. New York, NY: Springer New York, 2013. http://dx.doi.org/10.1007/978-1-4614-6528-7_13.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Conference papers on the topic "MET/HGF":

1

Chen, Dawei, Ziyong Sun, Likun Zhang, Jie Cai, Davy X. Ouyang, Jean-Pierre Wery, and Henry Li. "Abstract 747: Paracrine c-MET/HGF HCC PDX: evaluation of a biologics targeting c-MET." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-747.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Wortinger, Mark A., Wei Zeng, Wei Jennifer Yang, Victoria Peek, Lei Yan, Jirong Lu, Chi-Kin Chow, et al. "Abstract 695: LA480, a c-Met antibody with neutralization and internalization properties, inhibits HGF-dependent and HGF-independent c-Met pathway activation and tumor growth." In Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC. American Association for Cancer Research, 2010. http://dx.doi.org/10.1158/1538-7445.am10-695.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Gordon, Marcia, Alexandra Croft, Chi-Ming Li, Sean Taylor, Sean R. Caenepeel, and Kim Quon. "Abstract LB-241: HGF promotes resistance to MET inhibitor AMG337 in MET-amplified gastric cancer cells." In Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA. American Association for Cancer Research, 2015. http://dx.doi.org/10.1158/1538-7445.am2015-lb-241.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Owusu, Benjamin Y., Phanindra K. Venukadasula, Robert A. Galemmo, and Lidija Klampfer. "Abstract 1583: SRI31215, a novel inhibitor of oncogenic HGF/MET signaling." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-1583.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Yu, Yanlin, Liping Huang, and Glenn Merlino. "Abstract 4114: Blocking Ezrin can inhibit HGF/Met signaling-induced metastasis." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-4114.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Hughes, Veronica S., and Dietmar W. Siemann. "Abstract 4102: Effect of HGF concentrations on c-Met inhibition investigation." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-4102.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Vierkant, Robert A., Kimberly R. Kalli, Kristin L. White, Melissa C. Larson, Brooke L. Fridley, Gary L. Keeney, Trynda N. Oberg, et al. "Abstract 893: Associations between HGF, MET, and phospho-MET expression, overall survival, andHGFgenotypes in epithelial ovarian cancer." In Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL. American Association for Cancer Research, 2011. http://dx.doi.org/10.1158/1538-7445.am2011-893.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Chen, Dawei, Xiaoming Song, Jie Cai, Taiping Chen, Yiyou Chen, and Henry Q. X. Li. "Abstract 1322: Autocrine c-met/HGF HCC patient derived xenograft (PDX) models for evaluating c-met inhibitors." In Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL. American Association for Cancer Research, 2012. http://dx.doi.org/10.1158/1538-7445.am2012-1322.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Taniguchi, Hirokazu, Shinji Takeuchi, Koji Fukuda, Tadaaki Yamada, Shuichi Sakamoto, Manabu Kawada, and Seiji Yano. "Abstract 4763: Targeted therapy by MET inhibitors against small-cell lung cancer with aberrant activation of HGF/MET pathway." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-4763.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Hultberg, Anna, Cristina Basilico, Cristophe Blanchetot, Natalie D. Jonge, Valérie Hanssens, Gitte D. Boeck, Alessia Mira, et al. "Abstract LB-330: Four individually druggable Met hotspots mediate HGF-driven tumor progression." In Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA. American Association for Cancer Research, 2014. http://dx.doi.org/10.1158/1538-7445.am2014-lb-330.

Full text
APA, Harvard, Vancouver, ISO, and other styles

Reports on the topic "MET/HGF":

1

Bordeaux, Jennifer. Met (HGF Receptor) in Breast Cancer. Fort Belvoir, VA: Defense Technical Information Center, September 2010. http://dx.doi.org/10.21236/ada535659.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Bordeaux, Jennifer. Met (HGF Receptor) in Breast Cancer. Fort Belvoir, VA: Defense Technical Information Center, September 2009. http://dx.doi.org/10.21236/ada515399.

Full text
APA, Harvard, Vancouver, ISO, and other styles

To the bibliography