Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 216
Filtrer
1.
Eur J Med Chem ; 230: 114114, 2022 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-35051746

RÉSUMÉ

C-mesenchymal-epithelia transition factor (c-Met) is highly expressed in various solid tumors such as gastric cancer, liver cancer, and lung cancer, playing a pivotal role in the growth, maintenance, and development of different tumor cells. In this study, three small-molecule fluorescent probes (5, 11, 16) targeting c-Met were developed, and their design strategies were also initially explored. In general, the fluorescence properties of the probes themselves could meet the imaging requirements, and they have shown sufficient inhibitory activities against c-Met, especially probe 16, reflecting the targeting and acceptance. Also, fluorescence polarization assays and flow cytometry analysis verified the binding between the probes and c-Met. Cell imaging confirmed that these probes could be used to label c-Met on living cells. It is of positive significance for the development of c-Met kinase inhibitors and tumor pathology research.


Sujet(s)
Colorants fluorescents , Protéines proto-oncogènes c-met/analyse , Lignée cellulaire tumorale , Humains , Tumeurs
2.
ACS Appl Mater Interfaces ; 13(46): 54817-54829, 2021 Nov 24.
Article de Anglais | MEDLINE | ID: mdl-34766499

RÉSUMÉ

Fast and selective recognition of molecules at the nanometer scale without labeling is a much desired but still challenging goal to achieve. Here, we show the use of high-speed atomic force microscopy (HS-AFM) for real-time and real-space recognition of unlabeled membrane receptors using tips conjugated with small synthetic macrocyclic peptides. The single-molecule recognition method is validated by experiments on the human hepatocyte growth factor receptor (hMET), which selectively binds to the macrocyclic peptide aMD4. By testing and comparing aMD4 synthesized with linkers of different lengths and rigidities, we maximize the interaction between the functionalized tip and hMET added to both a mica surface and supported lipid bilayers. Phase contrast imaging by HS-AFM enables us to discriminate nonlabeled hMET against the murine MET homologue, which does not bind to aMD4. Moreover, using ligands and linkers of small size, we achieve minimal deterioration of the spatial resolution in simultaneous topographic imaging. The versatility of macrocyclic peptides in detecting unlimited types of membrane receptors with high selectivity and the fast imaging by HS-AFM broaden the range of future applications of this method for molecular recognition without labeling.


Sujet(s)
Composés macrocycliques/composition chimique , Peptides/composition chimique , Protéines proto-oncogènes c-met/analyse , Silicates d'aluminium/composition chimique , Animaux , Humains , Ligands , Double couche lipidique/composition chimique , Composés macrocycliques/synthèse chimique , Souris , Microscopie à force atomique , Structure moléculaire , Nanotechnologie , Imagerie optique , Peptides/synthèse chimique , Propriétés de surface
3.
Article de Anglais | MEDLINE | ID: mdl-34476332

RÉSUMÉ

PURPOSE: MET exon 14 (METex14) skipping alterations are oncogenic drivers in non-small-cell lung cancer (NSCLC). We present a comprehensive overview of METex14 samples from 1,592 patients with NSCLC, associated clinicogenomic characteristics, potential mechanisms of acquired resistance, treatment patterns, and outcomes to MET inhibitors. METHODS: Hybrid capture-based comprehensive genomic profiling (CGP) was performed on samples from 69,219 patients with NSCLC. For treatment patterns and outcomes analysis, patients with advanced METex14-altered NSCLC were selected from the Flatiron Health-Foundation Medicine clinicogenomic database, a nationwide deidentified electronic health record-derived database linked to Foundation Medicine CGP for patients treated between January 2011 and March 2020. RESULTS: A total of 1,592 patients with NSCLC (2.3%) were identified with 1,599 METex14 alterations spanning multiple functional sites (1,458 of 60,244 tissue samples and 134 of 8,975 liquid samples). Low tumor mutational burden and high programmed death ligand 1 expression were enriched in METex14-altered samples. MDM2, CDK4, and MET coamplifications and TP53 mutations were present in 34%, 19%, 11%, and 42% of tissue samples, respectively. Comparing tissue and liquid cohorts, coalteration frequency and acquired resistance mechanisms, including multiple MET mutations, EGFR, ERBB2, KRAS, and PI3K pathway alterations, were generally similar. Positive percent agreement with the tissue was 100% for METex14 pairs collected within 1 year (n = 7). Treatment patterns showed increasing adoption of MET inhibitors in METex14-altered NSCLC after receipt of CGP results; the real-world response rate to MET inhibitors was 45%, and time to treatment discontinuation was 4.4 months. CONCLUSION: Diverse METex14 alterations were present in 2%-3% of NSCLC cases. Tissue and liquid comparisons showed high concordance and similar coalteration profiles. Characterizing common co-occurring alterations and immunotherapy biomarkers, including those present before or acquired after treatment, may be critical for predicting responses to MET inhibitors and informing rational combination strategies.


Sujet(s)
Carcinome pulmonaire non à petites cellules/génétique , Protéines proto-oncogènes c-met/génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Carcinome pulmonaire non à petites cellules/épidémiologie , Femelle , Humains , Mâle , Adulte d'âge moyen , Mutation/génétique , Protéines proto-oncogènes c-met/analyse
4.
Rev Esp Patol ; 54(3): 147-155, 2021.
Article de Espagnol | MEDLINE | ID: mdl-34175025

RÉSUMÉ

INTRODUCTION AND OBJECTIVES: Peritoneal relapse as an isolated form of recurrence in colon cancer occurs in 25% of cases during the first two years subsequent to a curative colectomy. Currently, the diagnostic limitations of imaging studies and the absence of predictive scales for peritoneal recurrence warrant "second look" surgery in high-risk patients. The aim of this study is to assess features of some epithelial-mesenchymal transition biomarkers (c-Met, IGF-1R and plexin ß1) in order to predict post-surgical peritoneal colonization and develop a mathematical model to predict carcinomatous relapse. METHODS: A retrospective study of the histopathological samples of 87 patients diagnosed with colon cancer who underwent radical resection was carried out, using immunohistochemical techniques for c-Met, IGF-1R and plexin ß1. The patients were divided into two groups; those who had presented peritoneal recurrence and those who only had risk factors for this kind of relapse. Every stained sample was assessed by the rate of stained cells and immunostaining intensity. A possible association between immunohistochemical findings and peritoneal relapse was evaluated. Statistical analysis of the biomarkers with higher prognostic value allowed a risk mathematical formula to be developed based on coefficients, providing a specific value to each biomarker and patient. RESULTS: c-Met expression in the primary tumour showed a high statistical trend (p: .074) while IGF-1 (p: .022) and plexin ß1 (p: .021) revealed a significative association with peritoneal relapse. However, the multivariate analysis selected c-Met y plexin ß1 as useful factors for a predictive mathematical model on peritoneal recurrence with a 75.8% sensitivity and 80.5% specificity in patients with a staining more than 50% for both biomarkers. CONCLUSION: c-Met and plexin B1 overexpression is related to an increased risk of peritoneal relapse in cases of colon cancer where a radical resection is feasible. The encouraging outcomes of the proposed mathematical model may prove useful clinically in the identification of candidates for carcinoprophylaxis.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Tumeurs du côlon/composition chimique , Tumeurs du côlon/anatomopathologie , Transition épithélio-mésenchymateuse , Tumeurs du péritoine/secondaire , Sujet âgé , Tumeurs du côlon/chirurgie , Femelle , Humains , Immunohistochimie , Mâle , Modèles théoriques , Protéines de tissu nerveux/analyse , Protéines proto-oncogènes c-met/analyse , Récepteur IGF de type 1/analyse , Récepteurs de surface cellulaire/analyse , Études rétrospectives , Facteurs de risque , Sensibilité et spécificité
5.
Int J Exp Pathol ; 102(3): 172-178, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33951261

RÉSUMÉ

The transmembrane protein, c-Met, is thought to be overexpressed and activated in colorectal cancer (CRC). This study explored its potential as a diagnostic tissue biomarker for CRC in a large human CRC tissue collection obtained from a randomized clinical trial. Tissue microarrays of matched normal colorectal epithelium and primary cancer were prepared from specimens obtained from 280 patients recruited to the MRC CLASICC trial (ISRCTN 74883561) and interrogated using immunohistochemistry for c-Met expression. The distribution and intensity of immunopositivity was graded using a validated, semi-quantifiable score, and differences in median scores analysed using the Wilcoxon signed-rank test. A receiver operating characteristic (ROC) curve was plotted to measure the diagnostic accuracy of c-Met as a biomarker in CRC. Epithelial cell membrane expression of c-Met differed significantly between CRC and normal colorectal tissue: median 12.00 (Interquartile range (IQR) 6-15) versus median 6.00 (IQR 2.70-12.00) respectively (P = <.0001). ROC-AUC analysis of c-Met expression yielded a CRC diagnostic probability of 0.66 (95% CI: 0.61 to 0.70; P < .0001). A score of ≥14.50 showed high specificity at 85.32% (95% CI 80.33%-89.45%) but sensitivity of only 30.92% (CI 25.37%-36.90%). Thus c-Met is consistently overexpressed in human CRC as compared to normal colorectal epithelium tissue. c-Met expression may have a role in diagnosis and prognostication if combined with other biomarkers.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Tumeurs colorectales/diagnostic , Protéines proto-oncogènes c-met/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/analyse , Tumeurs colorectales/métabolisme , Femelle , Humains , Mâle , Adulte d'âge moyen , Protéines proto-oncogènes c-met/analyse
6.
Sci Rep ; 11(1): 5468, 2021 03 09.
Article de Anglais | MEDLINE | ID: mdl-33750878

RÉSUMÉ

Keloids, tumor-like lesions that result from excessive scar formation, have no definitive treatment modality. Activation of c-mesenchymal-epithelial transition factor (c-Met) promotes cell proliferation and survival. Selective c-Met inhibitors, such as PHA-665752, may attenuate the activity of keloid fibroblasts and reduce keloid formation. Here, we aimed to evaluate the effect of PHA-665752, a second-generation selective small-molecule inhibitor of c-Met, on human keloid fibroblasts in vitro and in a mouse model. We performed in vitro cytotoxicity assays, scratch tests, western blotting, and immunofluorescence on human keloid fibroblasts. We also injected human fibroblasts into severe combined immunodeficient mice and measured the degree of nodule formation and skin histologic characteristics. We found that keloid fibroblast migration was inhibited by PHA-665752. Inhibitor treatment was also associated with lower expression of members of the hepatocyte growth factor/c-Met pathway, and lower fibroblast activity and collagen synthesis. In the in vivo experiments, PHA-665752-treated mice had lower nodule volumes and weights, accompanied by less inflammatory cell infiltration and collagen deposition, than those in control mice. These findings showed that although an in vivo model may not accurately represent the pathophysiology of human keloid development, PHA-665752 suppressed keloid fibroblast activity by inhibiting the c-Met-related tyrosine kinase pathway.


Sujet(s)
Prolifération cellulaire/effets des médicaments et des substances chimiques , Fibroblastes/effets des médicaments et des substances chimiques , Chéloïde/traitement médicamenteux , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Bibliothèques de petites molécules/pharmacologie , Animaux , Lignée cellulaire , Modèles animaux de maladie humaine , Fibroblastes/anatomopathologie , Humains , Chéloïde/anatomopathologie , Mâle , Souris , Souris SCID , Protéines proto-oncogènes c-met/analyse , Bibliothèques de petites molécules/usage thérapeutique
7.
Clin Cancer Res ; 27(7): 2100-2110, 2021 04 01.
Article de Anglais | MEDLINE | ID: mdl-33451980

RÉSUMÉ

PURPOSE: Pancreatic cancer is an aggressive disease associated with a poor 5-year overall survival. Most patients are ineligible for surgery due to late diagnosis and are treated primarily with chemotherapy with very limited success. Pancreatic cancer is relatively insensitive to chemotherapy due to multiple factors, including reduced bioavailability of drugs to tumor cells. One strategy to improve drug efficacy with reduced toxicity is the development of antibody-drug conjugates (ADC), which have now been used successfully to treat both solid and liquid tumors. Here, we evaluate the efficacy of TR1801-ADC, a newly developed ADC composed of a MET antibody conjugated to the highly potent pyrrolobenzodiazepine toxin-linker, tesirine. EXPERIMENTAL DESIGN: We first evaluated MET expression and subcellular localization in pancreatic cancer cell lines, human tumors, and patient-derived xenografts (PDX). We then tested TR1801-ADC efficacy in vitro in pancreatic cancer cell lines. Preclinical evaluation of TR1801-ADC efficacy was conducted on PDXs selected on the basis of their MET expression level. RESULTS: We show that MET is highly expressed and located at the plasma membrane of pancreatic cancer cells. We found that TR1801-ADC induces a specific cytotoxicity in pancreatic cancer cell lines and a profound tumor growth inhibition, even in a gemcitabine-resistant tumor. We also noted synergism between TR1801-ADC and gemcitabine in vitro and an improved response to the combination in vivo. CONCLUSIONS: Together, these results suggest the promise of agents such as TR1801-ADC as a novel approach to the treatment of pancreatic cancer.


Sujet(s)
Carcinome du canal pancréatique/traitement médicamenteux , Désoxycytidine/analogues et dérivés , Immunoconjugués/usage thérapeutique , Tumeurs du pancréas/traitement médicamenteux , Protéines proto-oncogènes c-met/immunologie , Animaux , Carcinome du canal pancréatique/mortalité , Lignée cellulaire tumorale , Désoxycytidine/usage thérapeutique , Résistance aux médicaments antinéoplasiques , Humains , Mâle , Souris , Tumeurs du pancréas/mortalité , Protéines proto-oncogènes c-met/analyse , Tests d'activité antitumorale sur modèle de xénogreffe ,
8.
Int J Oncol ; 57(5): 1145-1156, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-33300049

RÉSUMÉ

Aberrant DNA methylation is widely observed in various types of cancer, and expression of microRNAs (miRNAs/miRs) is suppressed by DNA methylation. The present study explored tumor suppressor miRNAs downregulated by DNA methylation in endometrial cancer cells, as the basis of a novel therapeutic approach for endometrial cancer. Among 821 candidate miRNAs, miR­34b was identified as an upregulated miRNA after demethylation treatment in all four endometrial cancer cell lines (HEC­108, SNG­II, Ishikawa and HHUA) examined. miR­34b expression with or without demethylation treatment in cancer cells was confirmed by TaqMan quantitative PCR. MYC and MET, the predicted target genes of miR­34b, were downregulated at both the RNA and protein levels following miR­34b overexpression. Following miR­34b treatment, inhibition of cell growth and invasion, and cell cycle arrest were observed in HEC­108 cells. Sensitivity to paclitaxel was increased in cancer cells with miR­34b overexpression, compared with untreated cancer cells, but this difference was not identified for cisplatin or doxorubicin. In vivo, combination treatment with miR­34b and paclitaxel markedly reduced tumor growth compared with treatment with negative control miRNA and paclitaxel. These data suggest that miR­34b enhances paclitaxel sensitivity in endometrial cancer cells, and that miR­34b and MET are key targets for treatment of endometrial cancer. The present results may contribute to the development of combination treatment with a demethylation agent, miR­34b mimic or MET inhibitor and an anticancer drug.


Sujet(s)
Tumeurs de l'endomètre/traitement médicamenteux , microARN/physiologie , Paclitaxel/pharmacologie , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Méthylation de l'ADN , Résistance aux médicaments antinéoplasiques , Tumeurs de l'endomètre/génétique , Tumeurs de l'endomètre/anatomopathologie , Femelle , Humains , Souris , Souris de lignée BALB C , Protéines proto-oncogènes c-met/analyse , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/physiologie , Protéines proto-oncogènes c-myc/analyse , Protéines proto-oncogènes c-myc/physiologie
9.
Hum Pathol ; 105: 31-36, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32916162

RÉSUMÉ

The separation of benign from malignant mesothelial proliferations can be a difficult problem for the surgical pathologist. c-MET is a receptor tyrosine kinase that is overexpressed and detectable by immunohistochemistry in many malignancies, including malignant mesothelioma. Whether c-MET is also expressed in benign mesothelial reactions is unclear from the literature. To determine whether c-MET immunohistochemistry can separate benign from malignant mesothelial processes, we stained 2 tissue microarrays containing 33 reactive epithelioid mesothelial proliferations (E-RMPs), 23 reactive spindle cell mesothelial proliferations, 45 epithelioid malignant mesotheliomas (EMMs), and 26 sarcomatoid/desmoplastic mesotheliomas (SMMs) for c-MET and compared the results with immunohistochemistry for two established markers, BAP1 and methylthioadenosine phosphorylase (MTAP). Membrane staining for c-MET was evaluated using a 12-point H-score classified as negative (score = 0), trace (score = 1-3), moderate (score = 4-6), and strong (score = 8-12). Staining was seen in only 3 of 33 (all trace) E-RMPs compared with 36 of 45 (80%) EMMs (chi-square comparing reactive and malignant = 39.80, p = 1.2 × 10-8). The H-score was >3 (moderate or strong) in 24 of 45 (53%) EMMs. Addition of BAP1 staining to the c-MET-negative/trace EMM increased sensitivity to 75% (32/42), whereas similar addition of MTAP staining increased sensitivity to 77% (33/43). No benign spindle cell proliferations showed staining compared with 10 of 26 (38%) positive SMMs, but only 4 (15%) SMMs were classified as moderate or strong. We conclude that moderate/strong c-MET staining can be used to support a diagnosis of EMM vs an epithelial reactive proliferation. c-MET is too insensitive to use for detecting SMM.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Prolifération cellulaire , Épithélium/enzymologie , Immunohistochimie , Mésothéliome malin/enzymologie , Protéines proto-oncogènes c-met/analyse , Diagnostic différentiel , Épithélium/anatomopathologie , Humains , Mésothéliome malin/anatomopathologie , Protéines associées aux microtubules/analyse , Valeur prédictive des tests , Récepteur ErbB-3/analyse , Analyse sur puce à tissus , Protéines suppresseurs de tumeurs/analyse , Ubiquitin thiolesterase/analyse
10.
Mol Oncol ; 14(11): 2713-2726, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32946618

RÉSUMÉ

Experimental studies suggest that hepatocyte growth factor (HGF) and its transmembrane tyrosine kinase receptor, Met, in part also relying on Akt kinase activity, mediate radioresistance. We investigated the importance of these biomarkers for the risk of ipsilateral breast tumour recurrence (IBTR) after adjuvant radiotherapy (RT) in primary breast cancer. HGF, phosphorylated Met (pMet) and phosphorylated Akt (pAkt) were evaluated immunohistochemically on tissue microarrays from 1004 patients in the SweBCG91-RT trial, which randomly assigned patients to breast-conserving therapy, with or without adjuvant RT. HGF was evaluated in the stroma (HGFstr ); pMet in the membrane (pMetmem ); HGF, pMet and pAkt in the cytoplasm (HGFcyt , pMetcyt , pAktcyt ); and pAkt in the nucleus (pAktnuc ). The prognostic and treatment predictive effects were evaluated to primary endpoint IBTR as first event during the first 5 years. Patients with tumours expressing low levels of HGFcyt and pMetcyt and high levels of pAktnuc derived a larger benefit from RT [hazard ratio (HR): 0.11 (0.037-0.30), 0.066 (0.016-0.28) and 0.094 (0.028-0.31), respectively] compared to patients with high expression of HGFcyt and pMetcyt , and low pAktnuc [HR: 0.36 (0.19-0.67), 0.35 (0.20-0.64) and 0.47 (0.32-0.71), respectively; interaction analyses: P = 0.052, 0.035 and 0.013, respectively]. These differences remained in multivariable analysis when adjusting for patient age, tumour size, histological grade, St Gallen subtype and systemic treatment (interaction analysis, P-values: 0.085, 0.027, and 0.023, respectively). This study suggests that patients with immunohistochemically low HGFcyt , low pMetcyt and high pAktnuc may derive an increased benefit from RT after breast-conserving surgery concerning the risk of developing IBTR.


Sujet(s)
Tumeurs du sein/diagnostic , Tumeurs du sein/radiothérapie , Facteur de croissance des hépatocytes/analyse , Protéines proto-oncogènes c-akt/analyse , Protéines proto-oncogènes c-met/analyse , Tumeurs du sein/anatomopathologie , Tumeurs du sein/chirurgie , Femelle , Études de suivi , Humains , Mastectomie partielle , Phosphorylation , Pronostic , Résultat thérapeutique
11.
Hum Pathol ; 104: 42-53, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32702402

RÉSUMÉ

Using tissue microarrays, it was shown that membranous C-terminal MET immunoreactivity and ectodomain (ECD) shedding are associated with poor prognosis in oral cancer. Seen the potential diagnostic value, extrapolation of these results to whole-tissue sections was investigated. Because MET orchestrates epithelial-to-mesenchymal transition (EMT), the results were benchmarked to loss of E-cadherin, a readout for EMT known to be associated with poor prognosis. C-terminal MET, N-terminal MET, and E-cadherin immunoreactivities were examined on formalin-fixed paraffin-embedded parallel sections of 203 oral cancers using antibody clones D1C2, A2H2-3, and NCH-38. Interantibody and intra-antibody relations were examined using a novel scoring system, nonparametric distribution, and median tests. Survival analyses were used to examine the prognostic value of the observed immunoreactivities. Assessment of the three clones revealed MET protein status (no, decoy, transmembranous C-terminal positive), ECD shedding, and EMT. For C-terminal MET-positive cancers, D1C2 immunoreactivity is independently associated with poor overall survival (hazard ratio [HR] = 2.40; 95% confidence interval [CI] = 1.25 to 4.61; and P = 0.008) and disease-free survival (HR = 1.83; 95% CI = 1.07-3.14; P = 0.027). For both survival measures, this is also the case for ECD shedding (43.4%, with HR = 2.30; 95% CI = 1.38 to 3.83; and P = 0.001 versus HR = 1.87; 95% CI = 1.19-2.92; P = 0.006) and loss of E-cadherin (55.3%, with HR = 2.21; 95% CI = 1.30 to 3.77; and P = 0.004 versus HR = 1.90; 95% CI = 1.20-3.01; P = 0.007). The developed scoring system accounts for MET protein status, ECD shedding, and EMT and is prognostically informative. These findings may contribute to development of companion diagnostics for MET-based targeted therapy.


Sujet(s)
Antigènes CD/analyse , Marqueurs biologiques tumoraux/analyse , Cadhérines/analyse , Immunohistochimie , Tumeurs de la bouche/composition chimique , Protéines proto-oncogènes c-met/analyse , Carcinome épidermoïde de la tête et du cou/composition chimique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Transition épithélio-mésenchymateuse , Femelle , Humains , Mâle , Adulte d'âge moyen , Tumeurs de la bouche/mortalité , Tumeurs de la bouche/anatomopathologie , Valeur prédictive des tests , Pronostic , Domaines protéiques , Protéolyse , Carcinome épidermoïde de la tête et du cou/mortalité , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Analyse sur puce à tissus
12.
Biochim Biophys Acta Gen Subj ; 1864(10): 129650, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32522525

RÉSUMÉ

BACKGROUND: The c-MET oncoprotein drives cancer progression in a variety of tumors through its signaling transduction pathways. This oncoprotein is also degraded by multiple mechanisms involving the lysosome, proteasome and cleavage by proteases. Targeting c-MET degradation pathways may result in effective therapeutic strategies. SCOPE OF REVIEW: Since the discovery of oncogenic functions of c-MET, there has been a great deal of effort to develop anti-cancer drugs targeting this oncoprotein. Unexpectedly, novel di-2-pyridylketone thiosemicarbazones that demonstrate marked anti-tumor activity, down-regulate c-MET through their ability to bind intracellular iron and via mechanisms including, down-regulation of MET mRNA, enhanced lysosomal processing and increased metalloprotease-mediated cleavage. MAJOR CONCLUSIONS: The c-MET oncoprotein regulation and degradation pathways are complex. However, with increasing understanding of its degradation mechanisms, there is also greater opportunities to therapeutically target these pathways. GENERAL SIGNIFICANCE: Understanding the mechanisms of degradation of c-MET protein and its regulation could lead to novel therapeutics.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Protéolyse/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-met/métabolisme , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques immunologiques/composition chimique , Antinéoplasiques immunologiques/pharmacologie , Évolution de la maladie , Découverte de médicament , Humains , Thérapie moléculaire ciblée , Tumeurs/génétique , Protéines proto-oncogènes c-met/analyse , Protéines proto-oncogènes c-met/génétique , Thiosemicarbazones/composition chimique , Thiosemicarbazones/pharmacologie
13.
Biochem Biophys Res Commun ; 524(2): 378-384, 2020 04 02.
Article de Anglais | MEDLINE | ID: mdl-32005519

RÉSUMÉ

Signaling adaptor protein Crk has been shown to play an important role in various human cancers. Crk links tyrosine kinases and guanine nucleotide exchange factors (GEFs) such as C3G and Dock180 to activate small G-proteins Rap and Rac, respectively. In pancreatic cancer, various molecular targeted therapies have provided no significant therapeutic benefit for the patients so far due to constitutive activation of KRAS by frequent KRAS mutation. Therefore, the establishment of novel molecular targeted therapy in KRAS-independent manner is required. Here, we investigated a potential of Crk as a therapeutic target in pancreatic cancer. Immunohistochemistry on human pancreatic cancer specimens revealed that the patients with high expression of Crk had a worse prognosis than those with low expression. We established Crk-knockdown pancreatic cancer cells by siRNA using PANC-1, AsPC-1, and MIA PaCa-2 cells, which showed decreased cell proliferation, invasion, and adhesion. In Crk-knockdown pancreatic cancer cells, the decrease of c-Met phosphorylation was observed. In the orthotopic xenograft model, Crk depletion prolonged survival of mice significantly. Thus, signaling adaptor protein Crk is involved in malignant potential of pancreatic cancer associated with decrease of c-Met phosphorylation, and Crk can be considered to be a potential therapeutic molecular target.


Sujet(s)
Tumeurs du pancréas/anatomopathologie , Protéines proto-oncogènes c-crk/métabolisme , Protéines proto-oncogènes c-met/métabolisme , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Humains , Souris , Tumeurs du pancréas/diagnostic , Tumeurs du pancréas/métabolisme , Phosphorylation , Pronostic , Protéines proto-oncogènes c-crk/analyse , Protéines proto-oncogènes c-met/analyse
14.
Oncology ; 98(3): 186-194, 2020.
Article de Anglais | MEDLINE | ID: mdl-31846974

RÉSUMÉ

BACKGROUND: The clinical course of hepatocellular carcinoma (HCC) is complicated, because it often recurs and shows multiple lesions, some of which progress to a more malignant form, shortening the life of the patient. The hepatocyte growth factor receptor c-Met has been shown to play an important role in the pathogenesis of HCC, but the influence of c-Met expression on the clinical course of HCC remains to be fully elucidated. METHODS: We randomly selected and included 600 tumor specimens obtained from the primary and recurrent lesions of 319 HCC cases between 1995 and 2007. The expression of c-Met was determined by immunohistochemistry using archived formalin-fixed paraffin-embedded samples. We analyzed the correlation between c-Met expression and clinical parameters, including survival. In addition, we examined c-Met expression in the malignant transition of HCC in all cases including recurrent lesions. RESULTS: Survival analysis using the multivariate Cox proportional-regression model revealed that the prognosis was significantly better in the primary cases with high c-Met expression than in those with low c-Met expression (hazard ratio 0.159, 95% confidence interval 0.065-0.391; p < 0.001). During the course of recurrence, some cases with high c-Met expression returned to low c-Met expression. Among 40 cases with high c-Met expression, 29 survived more than 2 years after detecting the high c-Met expression. CONCLUSION: High expression of c-Met may be a prognostic factor for a good, rather than a poor, HCC prognosis. The involvement of c-Met expression in the malignant transition of recurrent HCC is obscure.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Carcinome hépatocellulaire/enzymologie , Tumeurs du foie/enzymologie , Récidive tumorale locale , Protéines proto-oncogènes c-met/analyse , Sujet âgé , Carcinome hépatocellulaire/mortalité , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/thérapie , Évolution de la maladie , Femelle , Humains , Tumeurs du foie/mortalité , Tumeurs du foie/anatomopathologie , Tumeurs du foie/thérapie , Mâle , Adulte d'âge moyen , Facteurs de risque , Facteurs temps , Résultat thérapeutique , Régulation positive
15.
Immunology ; 159(1): 96-108, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31596953

RÉSUMÉ

Among various solid tumours, gastric cancer (GC) is one of the leading causes of cancer-related deaths worldwide. Expansion into the peritoneal cavity, which results from dissemination of diffuse cancer cells, is the main cause of mortality in gastric adenocarcinoma patients. Therefore, investigation of putative biomarkers involved in metastasis is prerequisite for GC management. In an effort to discover potential tumour markers associated with peritoneal metastasis of GC, a semi-synthetic human scFv library (Tomlinson I) was used to isolate novel antibody fragments recognizing MKN-45, a poorly differentiated diffuse gastric adenocarcinoma cell line. Four rounds of subtractive selection each consisting of extensive pre-absorption of phage library with NIH-3T3 murine embryonic fibroblasts and AGS (a well-differentiated intestinal gastric adenocarcinoma) cell line were carried out prior to positive selection on MKN-45 target cells. ELISA-based screening of 192 phage-displayed scFv clones indicated 21 high-affinity binders with specific staining of MKN-45 compared with AGS cells. Diversity analysis of the selected phage-scFvs resulted in five distinct sequences with multiple frequency. Further analysis by ELISA and flow cytometry verified three clones that specifically recognized MKN-45 cells. Liquid chromatography-mass spectrometry analysis of the scFv-immunoprecipitated proteins has led to identification of c-Met, HSP90 α and HSP90 ß as candidate biomarkers associated with diffuse GC. Immunohistochemistry revealed the capability of purified scFvs to differentiate diffuse and intestinal gastric adenocarcinoma. Taken together, the isolated MKN-45-specific scFv fragments and their cognate antigens would be beneficial in screening and management as well as targeting and therapy of the diffuse gastric adenocarcinoma.


Sujet(s)
Adénocarcinome/immunologie , Marqueurs biologiques tumoraux/analyse , Bioprospection/méthodes , Techniques d'exposition à la surface cellulaire , Protéines du choc thermique HSP90/analyse , Protéines proto-oncogènes c-met/analyse , Anticorps à chaîne unique/immunologie , Tumeurs de l'estomac/immunologie , Adénocarcinome/anatomopathologie , Animaux , Lignée cellulaire tumorale , Chromatographie en phase liquide à haute performance , Diagnostic différentiel , Humains , Immunohistochimie , Souris , Cellules NIH 3T3 , Valeur prédictive des tests , Anticorps à chaîne unique/génétique , Tumeurs de l'estomac/anatomopathologie , Spectrométrie de masse en tandem
16.
World Neurosurg ; 126: e1042-e1049, 2019 Jun.
Article de Anglais | MEDLINE | ID: mdl-30878754

RÉSUMÉ

OBJECTIVE: c-Met has been shown to be associated with tumor growth in several human cancers. This study aims to evaluate the correlation between the c-Met expression and histopathologic/clinical characteristics. METHODS: A total of 153 patients with histologically defined World Health Organization grade II-IV diffuse astrocytic and oligodendroglial tumors were analyzed. RESULTS: For each histopathologic diagnosis, the number of cases and positive rate of c-Met expression are as follows: oligodendroglioma, IDH-mutant, and 1p19q codeletion (OD): 16 cases, 6.3%; anaplastic oligodendroglioma, IDH-mutant, and 1p19q codeletion (AO): 11 cases, 36.4%; diffuse astrocytoma (DA), IDH-mutant: 21 cases, 28.6%; anaplastic astrocytoma (AA), IDH- mutant: 15 cases, 20%; glioblastoma, IDH-mutant: 2, 100%, DA, IDH-wildtype: 9 cases, 33.3%; AA, IDH-wildtype: 20 cases, 30.0%; and glioblastoma, IDH-wildtype: 59 cases, 52.5%. c-Met expression was correlated with progression-free survival in oligodendroglial tumors and glioblastoma, IDH-wildtype. Furthermore, it was correlated with overall survival in AO, oligodendroglial tumors, DA, IDH-mutant, DA, IDH-wildtype, and glioblastoma, IDH-wildtype, and tend to be correlated with overall survival in IDH-mutant lower-grade astrocytic tumors. CONCLUSIONS: c-Met expression was revealed to be a useful marker for prognosis prediction in IDH-mutant lower-grade gliomas and glioblastoma, IDH-wildtype, representing a new independent prognostic marker that can be easily measured.


Sujet(s)
Marqueurs biologiques tumoraux/analyse , Tumeurs du cerveau/anatomopathologie , Gliome/anatomopathologie , Protéines proto-oncogènes c-met/biosynthèse , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/mortalité , Femelle , Gliome/métabolisme , Gliome/mortalité , Humains , Isocitrate dehydrogenases/génétique , Mâle , Adulte d'âge moyen , Mutation , Pronostic , Protéines proto-oncogènes c-met/analyse , Études rétrospectives , Jeune adulte
17.
Gene Ther ; 26(3-4): 93-108, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30683895

RÉSUMÉ

Tumor heterogeneity, within and between tumors, may have severe implications for tumor therapy, especially for targeted gene therapy, where single-targeted approaches often result in limited efficacy and therapy resistance. Polymer-formulated nonviral vectors provide a potent delivery platform for cancer therapy. To improve applicability for future clinical use in a broad range of patients and cancer types, a dual-targeting approach was performed. Synthetic LPEI-PEG2kDa-based polymer backbones were coupled to two tumor-specific peptide ligands GE11 (EGFR-targeting) and cMBP (cMET-targeting). The dual-targeting approach was used to deliver the theranostic sodium iodide symporter (NIS) gene to hepatocellular cancer. NIS as auspicious theranostic gene allows noninvasive imaging of functional NIS gene expression and effective anticancer radioiodide therapy. Enhanced tumor-specific transduction efficiency of dual-targeted polyplexes compared to single-targeted polyplexes was demonstrated in vitro using tumor cell lines with different EGFR and cMET expression and in vivo by 124I-PET-imaging. Therapeutic efficacy of the bispecific concept was mirrored by significantly reduced tumor growth and perfusion, which was associated with prolonged animal survival. In conclusion, the dual-targeting approach highlights the benefits of a bifunctional strategy for a future clinical translation of the bioimaging-based NIS-mediated radiotherapy allowing efficient targeting of heterogeneic tumors with variable receptor expression levels.


Sujet(s)
Carcinome hépatocellulaire/génétique , Thérapie génétique/méthodes , Nanomédecine théranostique/méthodes , Animaux , Carcinome hépatocellulaire/thérapie , Lignée cellulaire tumorale , Systèmes de délivrance de médicaments/méthodes , Récepteurs ErbB/analyse , Récepteurs ErbB/génétique , Femelle , Expression des gènes/génétique , Techniques de transfert de gènes , Hétérogénéité génétique , Hétérogreffes , Humains , Ligands , Tumeurs du foie/génétique , Souris , Souris nude , Peptides/synthèse chimique , Peptides/génétique , Polymères , Protéines proto-oncogènes c-met/analyse , Protéines proto-oncogènes c-met/génétique
18.
Gastric Cancer ; 22(2): 335-343, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-29951752

RÉSUMÉ

BACKGROUND: Receptor tyrosine kinases (RTKs) play critical roles in gastric cancer (GC) progression and are potential targets for novel molecular-targeted agents or photo-immunotherapies. During patient selection, targeted biopsy is the first step. However, heterogeneous expression of RTKs based on the macroscopic appearance in GC has not been extensively addressed. Accordingly, in this study, we evaluated differences in RTK expression associated with macroscopic appearance in GC. METHODS: In total, 375 consecutive patients who had undergone gastrectomy at the National Cancer Center Hospital East and who had histologically proven adenocarcinoma, available archived tumor sample, and no history of chemotherapy were enrolled in this study. For these cases, tissue microarray (TMA) samples were examined using immunohistochemistry (IHC). Based on the results of IHC, cases were selected for detailed examination. We re-evaluated IHC scores in more than three tumor blocks per case and comparatively evaluated differences in IHC expression in RTKs between the mucosal portion (MuP) and invasive portion (InP). RESULTS: Human epidermal growth factor receptor 2 (HER2)-, epidermal growth factor receptor (EGFR)-, and mesenchymal epithelial transition factor (c-MET)-positive rates were 6, 9, and 20%, respectively. Twenty-two cases were then analyzed to assess differences in IHC expression levels in the same lesion. Concordance rates of positive staining of HER2, EGFR, and MET between MuP and whole tumor were 100, 40, and 56% and those with InP were 46, 100, and 56%. CONCLUSIONS: To avoid underestimating expression status, biopsies must be taken from MuP for HER2, InP for EGFR, and both proportions for c-MET.


Sujet(s)
Adénocarcinome/anatomopathologie , Protéines proto-oncogènes c-met/biosynthèse , Récepteur ErbB-2/biosynthèse , Tumeurs de l'estomac/anatomopathologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/analyse , Récepteurs ErbB/analyse , Récepteurs ErbB/biosynthèse , Femelle , Humains , Mâle , Adulte d'âge moyen , Protéines proto-oncogènes c-met/analyse , Récepteur ErbB-2/analyse
19.
Cell Physiol Biochem ; 51(5): 2052-2064, 2018.
Article de Anglais | MEDLINE | ID: mdl-30522102

RÉSUMÉ

BACKGROUND/AIMS: Alpha-fetoprotein-producing colorectal cancer (AFPP-CRC) is quite rarely seen. This study aimed to elucidate the clinicopathologic characteristics and prognostic factors of AFPP-CRC. METHODS: Among 5,051 colorectal cancer patients receiving surgery in the Fudan University Shanghai Cancer Center from 2006 to 2016, we identified 78 patients with elevated serum level of AFP (> 10 µg/L) preoperatively. A propensity score matching (PSM) analysis was performed which matched 75 AFPP-CRC patients to the same number of AFP-negative colorectal cancer (AFPN-CRC) patients. Kaplan-Meier curves were compared using the log-rank test and multivariable analysis was performed to evaluate the effect of AFP-positivity while adjusting confounding factors. 27 patients were available for immunohistochemical analysis. We conducted functional experiments to characterize the tumorigenicity of AFP. RESULTS: Patients with AFPP-CRC had a significantly higher incidence of advanced TNM stage and liver metastasis. Overall survival was significantly different between two groups before and after PSM, and AFP-positivity was one of the strongest predictors of overall survival in the multivariable model (HR 4.11, CI 95%: 1.43-11.76, p = 0.009) after PSM. We further investigated prognostic factors affecting prognosis in AFPP-CRC and found that the presence of liver metastasis was the only independent prognostic factor (HR 4.95, CI 95%: 1.48-16.48, p = 0.009). AFP expression was significantly positively correlated with HGF and c-Met expression. Transwell invasion assay revealed significantly increased cell motility with AFP overexpression. CONCLUSION: AFP-positivity is a significant negative predictor of overall survival in patients with colorectal cancer, which may be mediated by HGF/c-Met signaling pathway.


Sujet(s)
Côlon/anatomopathologie , Tumeurs colorectales/sang , Tumeurs colorectales/anatomopathologie , Rectum/anatomopathologie , Alphafoetoprotéines/analyse , Lignée cellulaire tumorale , Mouvement cellulaire , Tumeurs colorectales/diagnostic , Femelle , Facteur de croissance des hépatocytes/analyse , Humains , Mâle , Adulte d'âge moyen , Pronostic , Protéines proto-oncogènes c-met/analyse , Analyse de survie
20.
Eur J Cancer ; 105: 41-49, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30391779

RÉSUMÉ

BACKGROUND: Anti-HER2 therapy has not demonstrated a survival advantage in the second-line setting of patients with HER2-positive gastric cancer. We conducted this study to assess changes in HER2 status and to identify possible biomarkers for acquired resistance after the use of trastuzumab as the first-line therapy. PATIENTS AND METHODS: Patients with advanced or recurrent HER2-positive gastric adenocarcinoma who were diagnosed with progressive disease after the first-line trastuzumab-based therapy and developed pathologically confirmed adenocarcinoma within 3 months after completion of trastuzumab-based therapy were enrolled in this study. We collected re-biopsied samples from the HER2-positive patients who had developed resistance to trastuzumab and re-evaluated their HER2 status. Amplification of EGFR and c-met, as well as PIK3CA mutation, were comparatively analysed when samples were available. RESULTS: Among 33 eligible patients, loss of HER2 was identified in 20 patients (60.6%) with refractory disease. Immunohistochemistry showed that the rate of HER2 overexpression was greatly reduced after therapy (pre-HER2 3+: 24 [72.7%] vs. post-HER2 3+: 13 [39.4%]). We found that the use of fixatives other than 10% neutral buffered formalin significantly reduced the HER2-positive rate. EGFR amplification, c-met amplification and PIK3CA mutation before and after trastuzumab-based therapy were observed in 10.3% and 3.8%, 17.9% and 4.2% and 4.0% and 4.2% of cases, respectively. CONCLUSION: Re-evaluation of HER2 status is needed to determine the appropriate use of anti-HER2-targeted therapy after disease progression. Our results also highlight the importance of formalin fixation conditions for HER2 testing.


Sujet(s)
Adénocarcinome/composition chimique , Antinéoplasiques immunologiques/pharmacologie , Résistance aux médicaments antinéoplasiques , Gènes erbB-2 , Protéines tumorales/analyse , Récepteur ErbB-2/analyse , Tumeurs de l'estomac/composition chimique , Trastuzumab/pharmacologie , Adénocarcinome/traitement médicamenteux , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Antinéoplasiques immunologiques/usage thérapeutique , Biopsie/méthodes , Phosphatidylinositol 3-kinases de classe I/analyse , Phosphatidylinositol 3-kinases de classe I/génétique , ADN tumoral/analyse , Évolution de la maladie , Résistance aux médicaments antinéoplasiques/génétique , Récepteurs ErbB/analyse , Récepteurs ErbB/génétique , Faux négatifs , Femelle , Fixateurs/pharmacologie , Formaldéhyde/pharmacologie , Amplification de gène , Gènes erbB-1 , Humains , Immunohistochimie , Mâle , Protéines tumorales/génétique , Études prospectives , Protéines proto-oncogènes c-met/analyse , Protéines proto-oncogènes c-met/génétique , Récepteur ErbB-2/génétique , Récepteur ErbB-2/immunologie , Récidive , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Fixation tissulaire , Trastuzumab/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...