Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Tumori ; 110(1): 49-59, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37574933

ABSTRACT

INTRODUCTION: The synthesis of the periprosthetic capsule during implant-based breast reconstruction is the result of a coordinate cascade of inflammatory events ending in a fibrous tissue deposition around the expander or implant. Although the development of small volumes of fluid is one of the complications of prosthetic-based breast reconstruction, the characterization of the periprosthetic effusions coupled with the micro-textured devices, that have been recently introduced after the recall of macro-textured ones, is still lacking. The investigation of these periprosthetic effusions and paired capsules in terms of immunological content were the primary and secondary aims of the present study, respectively. METHODS: For this, 68 women, 41 of whom had periprosthetic effusions at the time of expander replacement with implant, were recruited. For each case, capsule and healthy dermal tissues were taken and for women with periprosthetic effusion, peripheral blood was also collected. Periprosthetic effusions and peripheral blood were characterized by cytometry while capsules and dermal tissues by immunohistochemistry and Nanostring analysis. RESULTS: The results showed an increase of Th1, Th2 lymphocytes and a HLA-DR+bright CD16+ cells (likely representing monocytes-derived macrophages) in periprosthetic effusions in respect to peripheral blood. These pro-inflammatory cells were counterbalanced by the gain of suppressive CD4 Treg cells. In the corresponding capsules, immunohistochemistry revealed the absence of Th1 cells and the presence of tissutal FOXP3 Treg. No significant difference in expression of inflammatory-related genes between capsules and dermal tissues was present. CONCLUSIONS: These results suggest the presence of a Treg-controlled inflammation in both periprosthetic effusions and capsules.


Subject(s)
Breast Implants , Breast Neoplasms , Mammaplasty , Female , Humans , Mammaplasty/methods , Inflammation
2.
Cancer Biomark ; 38(3): 301-309, 2023.
Article in English | MEDLINE | ID: mdl-37545217

ABSTRACT

BACKGROUND: Pan-TRK inhibitors Entrectinib and Larotrectinib have been recently approved as tumor-agnostic therapies in NTRK1-2-3 rearranged patients and there is therefore an urgent need to identify reliable and accessible biomarkers for capturing NTRK fusions in the real-world practice. OBJECTIVE: We aim to assess the analytical validity of the recently released pan-TRK assay (Ventana), running a head-to-head comparison between immunohistochemistry and Archer FusionPlex Lung Panel (ArcherDX) that is designed to detect key fusions in 13 genes, also including NTRK1-3. METHODS: Pan-TRK IHC and NGS analysis were conducted on a retrospective/prospective cohort of 124 cancer patients (carcinomas, 93 cases; soft tissue sarcomas, 19; primary central nervous system tumours, 10; and neuroblastomas, 2). FISH data were available in most of the IHC/NGS discordant cases. RESULTS: A comparison between IHC and NGS results was carried out in 117 cases: among 30 pan-TRK positive cases, NTRK rearrangement by NGS was found in 11 (37%), while one of the 87 (1.1%) pan-TRK negative cases (a case of NSCLC) showed a TPM3-NRTK1 rearrangement by NGS. Accordingly, sensitivity and specificity of IHC in predicting NTRK status were 91.7% and 81.9%, respectively, while negative (NPV) and positive predictive value (PPV) were 98.8% and 36.7%, respectively. CONCLUSIONS: These data lead to suggest that IHC with VENTANA pan-TRK antibody can be a reliable screening tool for the identification of patients potentially bearing NTRK rearranged tumours.


Subject(s)
Lung Neoplasms , Humans , Immunohistochemistry , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Prospective Studies , Retrospective Studies , Workflow
3.
Biomedicines ; 11(7)2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37509688

ABSTRACT

In pseudomyxoma peritonei (PMP), KRAS and GNAS mutations are frequent. We hypothesized that these mutations may contribute to the suppression of antitumor immunity: KRAS may induce GMCSF expression, while GNAS may enhance the expression of cyclic adenosine monophosphate and A2AR signaling. This study aimed to explore possible mechanisms facilitated by KRAS and GNAS mutations for escaping immune surveillance. Additionally, we looked for new potential therapeutic and prognostic targets in this rare disease which is poorly characterized at the molecular level. GM-CSF, A2AR, CD73, CD39, and PD-L1 expression was investigated by immunohistochemistry in 40 PMPs characterized for GNAS and KRAS mutational status. Immune cell populations were studied by immunohistochemistry and nanostring nCounter®. Following the criteria of a prognostic nomogram reported for PMP, we stratified the patients into two different risk groups, with 28 "low-risk" and 12 "high-risk" patients. We observed the expression of GM-CSF (74%); CD39 (37%); CD73 (53%); A2AR (74%); and PD-L1 (16%) which was unrelated to GNAS or KRAS status. The tumor microenvironment showed the presence of CD4+ T cells (86%); CD8+ T cells (27%); CD20+ B (67%); CD15+ cells (86%); and CD163+ M2 macrophages (67%), while CD56+ NK cells were absent. CD163 expression (27%) in PMP tumor cells was associated with poor prognosis. GNAS mutation and A2AR expression were not associated with a specific immune transcriptional signature. However, the expression assay revealed 21 genes associated with prognosis. The "high-risk" patients exhibited worse progression-free survival (HR = 2.3, CI 95%: 1.1-5.1, p = 0.034) and significant downregulation of MET, IL8, PPARG, DTX4, HMGA1, ZIC2, WNT5B, and CCRL2. In conclusion, we documented the presence of immunosuppressive factors such as GM-CSF, A2AR, and PD-L1 in PMP. These factors were not associated with GNAS and KRAS status and could be explored as therapeutic molecular targets. Additionally, a set of potential prognostic biomarkers, including CD163 expression in tumor cells, deserve further investigation.

4.
Int J Mol Sci ; 24(4)2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36834892

ABSTRACT

Most oropharyngeal squamous cell carcinomas (OPSCCs) are human papillomavirus (HPV)-associated, high-risk (HR) cancers that show a better response to chemoradiotherapy and are associated with improved survival. Nucleophosmin (NPM, also called NPM1/B23) is a nucleolar phosphoprotein that plays different roles within the cell, such as ribosomal synthesis, cell cycle regulation, DNA damage repair and centrosome duplication. NPM is also known as an activator of inflammatory pathways. An increase in NPM expression has been observed in vitro in E6/E7 overexpressing cells and is involved in HPV assembly. In this retrospective study, we investigated the relationship between the immunohistochemical (IHC) expression of NPM and HR-HPV viral load, assayed by RNAScope in situ hybridization (ISH), in ten patients with histologically confirmed p16-positive OPSCC. Our findings show that there is a positive correlation between NPM expression and HR-HPV mRNA (Rs = 0.70, p = 0.03), and a linear regression (r2 = 0.55; p = 0.01). These data support the hypothesis that NPM IHC, together with HPV RNAScope, could be used as a predictor of transcriptionally active HPV presence and tumor progression, which is useful for therapy decisions. This study includes a small cohort of patients and, cannot report conclusive findings. Further studies with large series of patients are needed to support our hypothesis.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Oncogene Proteins, Viral , Oropharyngeal Neoplasms , Papillomavirus Infections , Humans , Carcinoma, Squamous Cell/pathology , Cyclin-Dependent Kinase Inhibitor p16 , DNA, Viral/genetics , Human Papillomavirus Viruses , Nucleophosmin , Oncogene Proteins, Viral/genetics , Oropharyngeal Neoplasms/pathology , Papillomaviridae/genetics , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck , Viral Load
5.
Oncologist ; 25(9): 780-786, 2020 09.
Article in English | MEDLINE | ID: mdl-32272500

ABSTRACT

BACKGROUND: Epstein-Barr virus (EBV)-positive gastric cancers (GCs) have been recently identified as a molecular subgroup showing excellent outcomes after surgery for early-stage disease and responsiveness to immune checkpoint inhibitors (ICIs) for metastatic stage. No data are available on the prevalence, clinical characteristics, and prognosis of this subgroup of GCs in the metastatic setting. MATERIALS AND METHODS: In this cohort study, we assessed the impact of EBV status in patients with metastatic GC treated with chemotherapy at two Italian institutions. RESULTS: Among the 175 cases analyzed, only 7 (4%) were EBV positive and all showed long-lasting and even complete responses to first-line chemotherapy with fluorouracil and platinum and a significantly better survival compared with EBV-negative patients (3-year overall survival: 80% vs. 20.1%; hazard ratio: 0.12). CONCLUSION: If confirmed in larger data sets, our results may give a strong rationale for investigating the addition of ICIs to chemotherapy, in order to maximize the chance of achieving durable and complete responses in this uncommon subtype of GC. IMPLICATIONS FOR PRACTICE: To date, no data are available on the prevalence and clinical characteristics of patients with Epstein-Barr virus (EBV)-positive metastatic gastric cancer (GC), a specific subtype of GC showing excellent outcomes after radical surgery in early-stage disease and responsiveness to immune checkpoint inhibitors (ICIs). This cohort study showed that patients with EBV-positive GC who did not receive ICIs had exceptional, long-lasting, and even complete responses to first-line chemotherapy with fluorouracil and platinum and a significantly better survival compared with EBV-negative patients. If confirmed in larger series, these results may give a strong rationale for investigating the combination of chemotherapy and ICIs to achieve durable and potentially complete response in this uncommon subtype of GC.


Subject(s)
Epstein-Barr Virus Infections , Stomach Neoplasms , Cohort Studies , Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human , Humans , Prognosis , Stomach Neoplasms/drug therapy
7.
Otolaryngol Head Neck Surg ; 161(4): 635-642, 2019 10.
Article in English | MEDLINE | ID: mdl-31286827

ABSTRACT

OBJECTIVE: There is no consensus on the follow-up modalities in patients with head and neck cancer. This study aims to describe the pattern and survival outcomes of recurrences/second primary cancers in patients undergoing an intensive radiologic and clinical follow-up. STUDY DESIGN: Retrospective analysis. SETTING: Single academic tertiary care center. SUBJECTS AND METHODS: All patients with stage III-IV head and neck cancer treated with chemoradiotherapy at our institution between 1998 and 2010 were retrospectively reviewed. Persistent/recurrent disease within 6 months since the curative treatment and second primary cancers outside the upper aerodigestive tract were excluded. Data were analyzed by descriptive statistics. Surveillance was planned every 3 months in the first year, then with increasing intervals till the fifth year. RESULTS: A total of 326 patients were included. Out of all detected cancer recurrences (n = 106, 32%), 38 (36%) were locoregional, 44 (41%) were distant, and 24 (23%) were second primary cancers. Approximately 70% of recurrences were clinically and/or radiologically discovered, while 30% were diagnosed due to the patients' symptoms. Of all clinically and/or radiologically discovered recurrences/second primary cancers (n = 74), 26 (35%) were curatively treated, with respect to 9 of the 32 (28%) diagnosed by symptoms. Median overall survival of recurrent curable cases did not significantly differ according to the detection modality (89 months by clinical/radiologic examination vs 85 by symptoms). CONCLUSIONS: Clinical and radiologic follow-up identified more recurrences/second primary cancers than the symptom-driven monitoring, but the curability of cancer recurrence was similar regardless of detection modality. Prospective trials are needed to define the most effective follow-up strategy in head and neck cancer.


Subject(s)
Aftercare , Head and Neck Neoplasms/therapy , Neoplasm Recurrence, Local/diagnostic imaging , Academic Medical Centers , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Female , Head and Neck Neoplasms/diagnostic imaging , Head and Neck Neoplasms/mortality , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Recurrence, Local/therapy , Neoplasms, Second Primary/therapy , Population Surveillance , Positron-Emission Tomography , Retrospective Studies , Salvage Therapy , Survival Analysis , Tomography, X-Ray Computed , Young Adult
8.
Sci Rep ; 9(1): 3545, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30837627

ABSTRACT

The HER2 splice variant characterized by the deletion of exon 16 and denominated as d16HER2, is associated with HER2-positive breast cancer (BC) aggressiveness, stemness, and trastuzumab susceptibility and is considered to be a "flag" of HER2 dependence. However, with the exception of quantitative real-time PCR analysis, easily reproducible assays are still lacking to clinically detect and quantify the d16HER2 expression. Further, no data on d16HER2 expression and its potential role are available in HER2-positive gastrointestinal malignancies. Here, we used a novel RNA in situ hybridization technique (BaseScope) to discriminate d16HER2 variant expression from the wild type isoform (WTHER2) and to assess their levels across different HER2-positive histological samples. Our results demonstrate the existence of outliers, with d16HER2 mRNA high scores restricted to HER2-positive gastric cancer (GC) and colorectal cancer (CRC) coupled with increased d16HER2 expression compared with BC. Consistent with previously reported data on BC, experiments performed in HER2-positive GC patient-derived xenografts suggest that increased d16HER2 expression is associated with a clinical benefit/response to single-agent trastuzumab. Therefore, d16HER2 may be considered as a "flag" of HER2 dependence in GC and can be clinically investigated as a marker of trastuzumab susceptibility in several other HER2-driven cancers, including CRC. As a clinical proof-of-concept, we indicate that high d16HER2 mRNA scores are exclusively found in patients with a long-term benefit from trastuzumab exceeding 12 months (clinical "outliers"), and that d16HER2 expression is also increased in circulating tumor-released exosomes obtained from baseline plasma samples of long-term responders.


Subject(s)
Breast Neoplasms/pathology , Exons/genetics , Gene Expression Regulation, Neoplastic , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic , Humans , MCF-7 Cells , Mice , Protein Isoforms/genetics , Protein Isoforms/metabolism
9.
Clin Cancer Res ; 24(5): 1082-1089, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29208673

ABSTRACT

Purpose: Refining the selection of HER2-positive metastatic gastric cancer patient candidates for trastuzumab is a challenge of precision oncology. Preclinical studies have suggested several genomic mechanisms of primary resistance, leading to activation of tyrosine kinase receptors other than HER2 or downstream signaling pathways.Experimental Design: We carried out this multicenter, prospective, case-control study to demonstrate the negative predictive impact of a panel of candidate genomic alterations (AMNESIA panel), including EGFR/MET/KRAS/PI3K/PTEN mutations and EGFR/MET/KRAS amplifications. Hypothesizing a prevalence of candidate alterations of 30% and 0% in resistant and sensitive HER2-positive patients, respectively, 20 patients per group were needed.Results: AMNESIA panel alterations were significantly more frequent in resistant (11 of 20, 55%) as compared with sensitive (0% of 17) patients (P < 0.001), and in HER2 IHC 2+ (7 of 13, 53.8%) than 3+ (4 of 24, 16.7%) tumors (P = 0.028). Patients with tumors bearing no candidate alterations had a significantly longer median progression-free [5.2 vs. 2.6 months; HR, 0.34; 95% confidence interval (CI), 0.07-0.48; P = 0.001] and overall survival (16.1 vs. 7.6 months; HR, 0.38; 95% CI, 0.09-0.75; P = 0.015). The predictive accuracy of the AMNESIA panel and HER2 IHC was 76% and 65%, respectively. The predictive accuracy of the combined evaluation of the AMNESIA panel and HER2 IHC was 84%.Conclusions: Our panel of candidate genomic alterations may be clinically useful to predict primary resistance to trastuzumab in patients with HER2-positive metastatic gastric cancer and should be further validated with the aim of molecularly stratifying HER2-addicted cancers for the development of novel treatment strategies. Clin Cancer Res; 24(5); 1082-9. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor/genetics , Drug Resistance, Neoplasm/genetics , Stomach Neoplasms/drug therapy , Trastuzumab/pharmacokinetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/therapeutic use , Case-Control Studies , Female , Humans , Male , Middle Aged , Mutation , Patient Selection , Precision Medicine/methods , Progression-Free Survival , Prospective Studies , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Response Evaluation Criteria in Solid Tumors , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Survival Analysis , Trastuzumab/pharmacology , Trastuzumab/therapeutic use
10.
Oncotarget ; 8(29): 47780-47789, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28562354

ABSTRACT

The prognostic value of pre-treatment Epstein-Barr Virus (EBV) DNA viral load for non-endemic, locally-advanced, EBV-related nasopharyngeal cancer (NPC) patients is yet to be defined. All patients with EBV encoded RNA (EBER)-positive NPC treated at our Institution from 2005 to 2014 with chemotherapy (CT) concurrent with radiation (RT) +/- induction chemotherapy (ICT) were retrospectively reviewed. Pre-treatment baseline plasma EBV DNA (b-EBV DNA) viral load was detected and quantified by PCR. Median b-EBV DNA value was correlated to potential influencing factors by univariate analysis. Significant variables were then extrapolated and included in a multivariate linear regression model. The same variables, including b-EBV DNA, were correlated with Disease Free Survival (DFS) and Overall Survival (OS) by univariate and multivariate analysis.A total of 130 locally-advanced EBER positive NPC patients were evaluated. Overall, b-EBV DNA was detected in 103 patients (79.2%). Median viral load was 554 copies/mL (range 50-151075), and was positively correlated with T stage (p=0.002), N3a-b vs N0-1-2 stage (p=0.048), type of treatment (ICT followed by CTRT, p=0.006) and locoregional and/or distant disease recurrence (p=0.034). In the overall population, DFS and OS were significantly longer in patients with pre-treatment negative EBV DNA than in positive subjects at the multivariate analysis.Negative b-EBV DNA can be considered as prognostic biomarker of longer DFS and OS in NPC in non-endemic areas. This finding needs confirmation in larger prospective series, with standardized and inter-laboratory harmonized method of plasma EBV DNA quantification.


Subject(s)
DNA, Viral , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/etiology , Viral Load , Adolescent , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/therapy , Neoplasm Staging , Prognosis , ROC Curve , Young Adult
11.
Br J Cancer ; 117(3): 347-352, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28654634

ABSTRACT

BACKGROUND: Combined MET and BRAF inhibition showed clinical benefit in a patient with rectal cancer carrying BRAFV600E and MET amplification. However after 4 months, acquired resistance emerged and the patient deceased shortly after disease progression. The mechanism of resistance to this drug combination is unknown. METHODS: We analysed plasma circulating tumour DNA obtained at progression by exome sequencing and digital PCR. MET gene and mRNA in situ hybridisation analyses in two bioptic specimens obtained at progression were used to confirm the plasma data. RESULTS: We identified in plasma MET gene hyper-amplification as a potential mechanism underlying therapy resistance. Increased MET gene copy and transcript levels were detected in liver and lymph node metastatic biopsies. Finally, transduction of MET in BRAF mutant colorectal cancer cells conferred refractoriness to BRAF and MET inhibition. CONCLUSIONS: We identified in a rectal cancer patient MET gene hyper-amplification as mechanism of resistance to dual BRAF and MET inhibition.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA, Neoplasm/blood , Drug Resistance, Neoplasm/genetics , Proto-Oncogene Proteins c-met/genetics , Rectal Neoplasms/drug therapy , Rectal Neoplasms/genetics , Cell Line , Crizotinib , Disease Progression , Fatal Outcome , Gene Amplification , Humans , Indoles/administration & dosage , Middle Aged , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Rectal Neoplasms/pathology , Sulfonamides/administration & dosage , Vemurafenib
12.
Clin Cancer Res ; 23(10): 2414-2422, 2017 May 15.
Article in English | MEDLINE | ID: mdl-27780856

ABSTRACT

Purpose: Even if RAS-BRAF wild-type and HER2/MET-negative metastatic colorectal cancer (mCRC) patients frequently respond to anti-EGFR mAbs, acquired resistance almost invariably occurs. Mechanisms of resistance to EGFR blockade include the emergence of KRAS, NRAS, and EGFR extracellular domain mutations as well as HER2/MET alterations. However, these findings derive from retrospective studies that analyzed one single resistance mechanism at a time; moreover, it is still unclear how molecular heterogeneity affects clonal evolution in patients. In this work, we aimed at extensively characterizing and correlating the molecular characteristics of tissue- and blood-based data in a prospective cohort of patients with mCRC who received anti-EGFR antibodies.Experimental design: Twenty-two RAS-BRAF wild-type, HER2/MET-negative mCRC patients progressing on anti-EGFR therapy after initial response underwent rebiopsy. Next-generation sequencing and silver in situ hybridization (SISH)/IHC analyses were performed both on archival tumors and postprogression samples. Circulating tumor (ctDNA) molecular profiles were obtained in matched tissue-plasma samples.Results:RAS mutations and HER2/MET amplification were the most frequently detected resistance mechanisms in both tissue and blood sample analysis. On the other hand, BRAF and EGFR ectodomain mutations were much rarer. Patients with acquired MET amplification showed worse PFS on anti-EGFRs. We detected both intralesion heterogeneity, as suggested by co-occurrence of different resistance mechanisms in the same sample, and interlesion heterogeneity. The combined analysis of tissue and blood (ctDNA) results highlights the complexity of clonal evolution triggered by EGFR blockade.Conclusions: Our results indicate that it may be extremely challenging to target the complex landscape of molecular heterogeneity associated with emergence of resistance to targeted therapies in patients with mCRC. Clin Cancer Res; 23(10); 2414-22. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Colorectal Neoplasms/drug therapy , ErbB Receptors/genetics , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/genetics , Adult , Aged , Biomarkers, Tumor/blood , Circulating Tumor DNA/blood , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Female , GTP Phosphohydrolases/genetics , Humans , Male , Membrane Proteins/genetics , Middle Aged , Mutation , Neoplasm Metastasis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics
13.
J Clin Pathol ; 68(8): 661-4, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25934842

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that modulate gene expression by binding to complementary sequences on target messenger RNA transcripts. In situ hybridisation (ISH) methods have been applied to the study of miRNA in tissue samples in order to understand which is the source of the miRNA of interest. In this paper, the authors describe a novel semi-automated bright field ISH method to visualise miRNAs in formalin fixed paraffin embedded tissue sections. The relevance of this work resides in the use of 3,3'-diaminobenzidine and peroxidase as the detection method, which provides a good defined deposition within tissues This method, which reveals the cells of origin of specific miRNAs, will enable investigators to further explore the biological role of miRNAs.


Subject(s)
In Situ Hybridization/methods , MicroRNAs/genetics , Neoplasms/genetics , Paraffin Embedding , Automation, Laboratory , Benzene Derivatives , Biopsy , Cell Line, Tumor , Digoxigenin , Fixatives , Formaldehyde , Humans , Neoplasms/pathology , Peroxidase , Predictive Value of Tests , Tissue Fixation , Workflow
14.
Int J Mol Sci ; 14(9): 18056-77, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-24005867

ABSTRACT

Mesenchymal-epithelial transition (MET) is a member of a distinct subfamily of heterodimeric receptor tyrosine kinase receptors that specifically binds the hepatocyte growth factor (HGF). Binding to HGF leads to receptor dimerization/multimerization and phosphorylation, resulting in its catalytic activation. MET activation drives the malignant progression of several tumor types, including colorectal cancer (CRC), by promoting signaling cascades that mainly result in alterations of cell motility, survival, and proliferation. MET is aberrantly activated in many human cancers through various mechanisms, including point mutations, gene amplification, transcriptional up-regulation, or ligand autocrine loops. MET promotes cell scattering, invasion, and protection from apoptosis, thereby acting as an adjuvant pro-metastatic gene for many tumor types. In CRC, MET expression confers more aggressiveness and worse clinical prognosis. With all of this rationale, inhibitors that target the HGF/MET axis with different types of response have been developed. HGF and MET are new promising targets to understand the pathogenesis of CRC and for the development of new, targeted therapies.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Hepatocyte Growth Factor/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...