Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Gastroenterology ; 161(5): 1601-1614.e23, 2021 11.
Article in English | MEDLINE | ID: mdl-34303658

ABSTRACT

BACKGROUND & AIMS: Promoted by pancreatitis, oncogenic KrasG12D triggers acinar cells' neoplastic transformation through acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia. Anterior gradient 2 (Agr2), a known inhibitor of p53, is detected at early stage of pancreatic ductal adenocarcinoma (PDAC) development. RNA polymerase II (RNAPII) is a key nuclear enzyme; regulation of its nuclear localization in mammalian cells represents a potential therapeutic target. METHODS: A mouse model of inflammation-accelerated KrasG12D-driven ADM and pancreatic intraepithelial neoplasia development was used. Pancreas-specific Agr2 ablation was performed to access its role in pancreatic carcinogenesis. Hydrophobic hexapeptides loaded in liposomes were developed to disrupt Agr2-RNAPII complex. RESULTS: We found that Agr2 is up-regulated in ADM-to-pancreatic intraepithelial neoplasia transition in inflammation and KrasG12D-driven early pancreatic carcinogenesis. Genetic ablation of Agr2 specifically blocks this metaplastic-to-neoplastic process. Mechanistically, Agr2 directs the nuclear import of RNAPII via its C-terminal nuclear localization signal, undermining the ATR-dependent p53 activation in ADM lesions. Because Agr2 binds to the largest subunit of RNAPII in a peptide motif-dependent manner, we developed a hexapeptide to interfere with the nuclear import of RNAPII by competitively disrupting the Agr2-RNAPII complex. This novel hexapeptide leads to dysfunction of RNAPII with concomitant activation of DNA damage response in early neoplastic lesions; hence, it dramatically compromises PDAC initiation in vivo. Moreover, the hexapeptide sensitizes PDAC cells and patient-derived organoids harboring wild-type p53 to RNAPII inhibitors and first-line chemotherapeutic agents in vivo. Of note, this therapeutic effect is efficient across various cancer types. CONCLUSIONS: Agr2 is identified as a novel adaptor protein for nuclear import of RNAPII in mammalian cells. Also, we provide genetic evidence defining Agr2-dependent nuclear import of RNAPII as a pharmaceutically accessible target for prevention and treatment in PDAC in the context of wild-type p53.


Subject(s)
Carcinoma in Situ/enzymology , Carcinoma, Pancreatic Ductal/enzymology , Mucoproteins/metabolism , Oncogene Proteins/metabolism , Pancreatic Neoplasms/enzymology , RNA Polymerase II/metabolism , Tumor Suppressor Protein p53/metabolism , Active Transport, Cell Nucleus , Animals , Antineoplastic Agents/pharmacology , Carcinoma in Situ/drug therapy , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Metaplasia , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mucoproteins/genetics , Mutation , Oligopeptides/pharmacology , Oncogene Proteins/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , RNA Polymerase II/genetics , Tumor Suppressor Protein p53/genetics
2.
J Biol Chem ; 295(8): 2348-2358, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31959629

ABSTRACT

The oncogenic receptor tyrosine kinase AXL is overexpressed in cancer and plays an important role in carcinomas of multiple organs. However, the mechanisms of AXL overexpression in cancer remain unclear. In this study, using HEK293T, Panc-1, and Panc-28 cells and samples of human pancreatic intraepithelial neoplasia (PanIN), along with several biochemical approaches and immunofluorescence microscopy analyses, we sought to investigate the mechanisms that regulate AXL over-expression in pancreatic ductal adenocarcinoma (PDAC). We found that AXL interacts with hematopoietic progenitor kinase 1 (HPK1) and demonstrate that HPK1 down-regulates AXL and decreases its half-life. The HPK1-mediated AXL degradation was inhibited by the endocytic pathway inhibitors leupeptin, bafilomycin A1, and monensin. HPK1 accelerated the movement of AXL from the plasma membrane to endosomes in pancreatic cancer cells treated with the AXL ligand growth arrest-specific 6 (GAS6). Moreover, HPK1 increased the binding of AXL to the Cbl proto-oncogene (c-Cbl); promoted AXL ubiquitination; decreased AXL-mediated signaling, including phospho-AKT and phospho-ERK signaling; and decreased the invasion capability of PDAC cells. Importantly, we show that AXL expression inversely correlates with HPK1 expression in human PanINs and that patients whose tumors have low HPK1 and high AXL expression levels have shorter survival than those with low AXL or high HPK1 expression (p < 0.001). Our results suggest that HPK1 is a tumor suppressor that targets AXL for degradation via the endocytic pathway. HPK1 loss of function may contribute to AXL overexpression and thereby enhance AXL-dependent downstream signaling and tumor invasion in PDAC.


Subject(s)
Down-Regulation , Oncogenes , Pancreatic Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Cell Line, Tumor , Cytoplasm/metabolism , Endocytosis , Endosomes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Kaplan-Meier Estimate , MAP Kinase Signaling System , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Protein Binding , Protein Transport , Proteolysis , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Ubiquitination , Axl Receptor Tyrosine Kinase
3.
J Pathol ; 243(1): 65-77, 2017 09.
Article in English | MEDLINE | ID: mdl-28639695

ABSTRACT

Acinar-to-ductal metaplasia (ADM) is a reversible epithelial transdifferentiation process that occurs in the pancreas in response to acute inflammation. ADM can rapidly progress towards pre-malignant pancreatic intraepithelial neoplasia (PanIN) lesions in the presence of mutant KRas and ultimately pancreatic adenocarcinoma (PDAC). In the present work, we elucidate the role and related mechanism of glycogen synthase kinase-3beta (GSK-3ß) in ADM development using in vitro 3D cultures and genetically engineered mouse models. We show that GSK-3ß promotes TGF-α-induced ADM in 3D cultured primary acinar cells, whereas deletion of GSK-3ß attenuates caerulein-induced ADM formation and PanIN progression in KrasG12D transgenic mice. Furthermore, we demonstrate that GSK-3ß ablation influences ADM formation and PanIN progression by suppressing oncogenic KRas-driven cell proliferation. Mechanistically, we show that GSK-3ß regulates proliferation by increasing the activation of S6 kinase. Taken together, these results indicate that GSK-3ß participates in early pancreatitis-induced ADM and thus could be a target for the treatment of chronic pancreatitis and the prevention of PDAC progression. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Acinar Cells/enzymology , Carcinoma in Situ/prevention & control , Cell Transdifferentiation , Glycogen Synthase Kinase 3 beta/deficiency , Pancreas, Exocrine/enzymology , Pancreatic Ducts/enzymology , Pancreatic Neoplasms/prevention & control , Pancreatitis/enzymology , Acinar Cells/drug effects , Acinar Cells/pathology , Animals , Carcinoma in Situ/enzymology , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Cell Proliferation , Cell Transdifferentiation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Ceruletide , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Glycogen Synthase Kinase 3 beta/genetics , Homeodomain Proteins/genetics , Male , Metaplasia , Mice, Knockout , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/pathology , Pancreatic Ducts/drug effects , Pancreatic Ducts/pathology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology , Phenotype , Proto-Oncogene Proteins p21(ras)/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Time Factors , Trans-Activators/genetics , Tumor Necrosis Factor-alpha/pharmacology
4.
J Oral Pathol Med ; 45(10): 746-752, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26991907

ABSTRACT

BACKGROUND: PI3K-AKT-mTOR signaling pathway is associated with several cellular functions and is frequently changed in several malignancies. The aim of this study was to characterize the immunohistochemical expression pattern of components in PI3K-AKT-mTOR signaling pathway in oral epithelial dysplasia (OED), comparing to oral squamous cell carcinoma (OSCC) and non-dysplastic oral tissues (NDOT). METHODS: A total of 186 cases of NDOT, OED and OSCC were retrieved. Nuclear staining and cytoplasmic staining of the keratinocytes were considered positive, and the percentage of positive cells was calculated. RESULTS: Increased immunoreactivity from NDOT to OED and OSCC was seen for all proteins. In NDOT cases, positivity was found only for pS6 (52.9%) and p4EBP1 (13.5%). In OED, immunoreactivity was observed for pAKT (62.2%), pmTOR (28.6%), pS6 (70.8%), and p4EBP1 (42.9%). In OSCC cases, immunoreactivity was found for pAKT (83.3%), pmTOR (50%), pS6 (77.4%), and p4EBP1 (50%). The pAKT and pmTOR expression was higher in OED (<0.001, Fisher's exact test) and OSCC (<0.001, Fisher's exact test). CONCLUSION: Our study demonstrated higher pAKT and pmTOR expression during carcinogenesis of oral mucosa, differing considerably among OED and OSCC specimens when compared to NDOT. These proteins can be considered potential diagnostic markers for early detection of cancer.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Squamous Cell/enzymology , Head and Neck Neoplasms/enzymology , Mouth Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Biomarkers, Tumor/metabolism , Biopsy , Carcinogenesis/pathology , Carcinoma in Situ/enzymology , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cytoplasm/enzymology , Cytoplasm/metabolism , Cytoplasm/pathology , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/biosynthesis , Precancerous Conditions/enzymology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Protein Kinases/biosynthesis , Signal Transduction , Squamous Cell Carcinoma of Head and Neck , TOR Serine-Threonine Kinases/biosynthesis
5.
J Med Virol ; 86(9): 1534-41, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24898764

ABSTRACT

Vulval intraepithelial neoplasia is a precursor of vulval cancer and is commonly caused by infection with Human Papillomavirus (HPV). Development of topical treatments for vulval intraepithelial neoplasia requires appropriate in vitro models. This study evaluated the feasibility of primary culture of vulval intraepithelial neoplasia biopsy tissue to produce cell lines for use as in vitro models. A potentially immortal cell line was produced which gave rise to three monoclonal lines. These lines were characterized for HPV genomic integration and for viral gene expression using ligation-mediated PCR and quantitative PCR. Distinct patterns of viral integration and gene expression were observed among the three lines. Integration and expression data were validated using deep sequencing of mRNA. Gene ontology analyses of these data also demonstrated that expression of the HPV16 E4 and E5 proteins resulted in substantial changes in the composition of the cell membrane and extracellular space, associated with alterations in cell adhesion and differentiation. These data illustrate the diverse patterns of HPV gene expression potentially present within a single lesion. The derived cell lines provide useful models to investigate the biology of vulval intraepithelial neoplasia and the interactions between different HPV gene products and potential therapeutic agents.


Subject(s)
Carcinoma in Situ/virology , Human papillomavirus 16/genetics , Oncogene Proteins, Viral/genetics , Vulvar Neoplasms/virology , Carcinoma in Situ/enzymology , Cell Line, Tumor , Female , Gene Expression , Gene Ontology , Human papillomavirus 16/enzymology , Humans , Middle Aged , Oncogene Proteins, Viral/biosynthesis , RNA, Messenger , Sequence Analysis, RNA , Tumor Cells, Cultured , Vulvar Neoplasms/enzymology
6.
Br J Cancer ; 109(3): 694-703, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23839495

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Improvements in the understanding of its molecular mechanism and the characterisation of CRC-specific biomarkers facilitating early detection are considered to increase overall survival. METHODS: A meta-analysis of microarray and Serial Analysis of Gene Expression (SAGE) has been performed to identify differentially regulated genes in CRC. Dipeptidase 1 (DPEP1/MDP/RDP) and Syntenin-2 (SDCBP2/SITAC18) were found to be differentially expressed in tumour tissue compared with normal mucosa. Expression of DPEP1 was assessed in a validation set of 87 normal mucosa samples, 20 hyperplastic polyps, 46 CR adenomas with low- and high-grade intraepithelial neoplasia (IEN) and 217 well-documented CRCs by immunohistochemistry and partially by immunoblotting and real-time PCR. RESULTS: Expression of DPEP1 was specifically increased in human CRC tissue samples compared with normal mucosa (P<0.0001, Mann-Whitney U-test), showing a striking upregulation in high-grade compared with low-grade IEN. Furthermore, high DPEP1 expression was found to strongly correlate with histological stage (P<0.0001, chi-square test) as well as localisation (P<0.0001, chi-square test) and has been recognised as an independent adverse prognostic factor, showing significant prognostic values with an ROC (receiver operating characteristic)-AUC of 0.9230. CONCLUSION: Dipeptidase 1 has been identified as an excellent marker of high-grade IEN and CRC, and may thus be applied for screening of early neoplastic lesions and for prognostic stratification.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Dipeptidases/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma in Situ/genetics , Colorectal Neoplasms/genetics , Dipeptidases/genetics , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Humans , Neoplasm Grading , Prognosis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
7.
Gynecol Oncol ; 131(2): 445-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23927961

ABSTRACT

OBJECTIVES: Inherited genetic variability contributes to susceptibility to cervical cancer. We investigated the association of single nucleotide polymorphisms (SNPs) in the human epidermal growth factor receptor (ERBB) family with cervical cancer. METHODS: We used the transmission disequilibrium test (TDT) to look for excessive transmission of tag single nucleotide polymorphisms (tSNPs) in ERBB family members EGFR, ERBB2, ERBB3, and ERBB4 in a large sample of women with invasive and in situ cervical cancer and their biological parents (628 trios). The study used a discovery set of trios (244) analyzed by Illumina GoldenGate in which SNPs reaching a P<.05 were re-tested by TaqMan in the combined set of 628. We also explored collaborative effects of different ERBB alleles. RESULTS: Based on single SNP TDT tests we identified 16 significant SNPs in the discover stage and six of 14 SNPs that could be assayed by TaqMan were significantly overtransmitted in women with cervical cancer in the combined replication set. Four SNPs were located in intron 1 of EGFR and two SNPs in intron 24 of ERBB4. The EGFR variants are located near multiple enhancers, silencers, and the previously identified functional common polymorphisms in intron 1. CONCLUSIONS: Our data provide evidence for the involvement of intron 1 EGFR variants and intron 24 ERBB4 variants in modulating risk for the development of in situ and invasive cervical cancer. These variants should be examined in additional populations and functional studies would be needed to confirm this hypothesis.


Subject(s)
ErbB Receptors/genetics , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/genetics , Adult , Carcinoma in Situ/enzymology , Carcinoma in Situ/genetics , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Female , Genotype , Humans , Introns , Linkage Disequilibrium , Polymorphism, Single Nucleotide , Receptor, ErbB-4
8.
Gut ; 61(9): 1315-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22068166

ABSTRACT

BACKGROUND AND AIMS: Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death in the USA. Surgical resection is the only effective treatment; however, only 20% of patients are candidates for surgery. The ability to detect early PDAC would increase the availability of surgery and improve patient survival. This study assessed the feasibility of using the enzymatic activity of cathepsin E (Cath E), a protease highly and specifically expressed in PDAC, as a novel biomarker for the detection of pancreas-bearing pancreatic intraepithelial neoplasia (PanIN) lesions and PDAC. METHODS: Pancreas from normal, chronic pancreatitis and PDAC patients was assessed for Cath E expression by quantitative real-time PCR and immunohistochemistry. Human PDAC xenografts and genetically engineered mouse models (GEMM) of PDAC were injected with a Cath E activity selective fluorescent probe and imaged using an optical imaging system. RESULTS: The specificity of Cath E expression in PDAC patients and GEMM of pancreatic cancer was confirmed by quantitative real-time PCR and immunohistochemistry. The novel probe for Cath E activity specifically detected PDAC in both human xenografts and GEMM in vivo. The Cath E sensitive probe was also able to detect pancreas with PanIN lesions in GEMM before tumour formation. CONCLUSIONS: The elevated Cath E expression in PanIN and pancreatic tumours allowed in-vivo detection of human PDAC xenografts and imaging of pancreas with PanIN and PDAC tumours in GEMM. Our results support the usefulness of Cath E activity as a potential molecular target for PDAC and early detection imaging.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma in Situ/diagnosis , Carcinoma, Pancreatic Ductal/diagnosis , Cathepsin E/metabolism , Diagnostic Imaging/methods , Pancreatic Neoplasms/diagnosis , Precancerous Conditions/diagnosis , Animals , Biomarkers, Tumor/genetics , Carcinoma in Situ/enzymology , Carcinoma, Pancreatic Ductal/enzymology , Cathepsin E/genetics , Cell Line, Tumor , DNA Primers/chemistry , Disease Models, Animal , Early Diagnosis , Feasibility Studies , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Oligonucleotide Probes/chemistry , Pancreatic Neoplasms/enzymology , Precancerous Conditions/enzymology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
9.
Mod Pathol ; 25(5): 758-66, 2012 May.
Article in English | MEDLINE | ID: mdl-22222635

ABSTRACT

Aldo-keto reductase family 1B10 (AKR1B10) exhibits more restricted lipid substrate specificity (including farnesal, geranylgeranial, retinal and carbonyls), and metabolizing these lipid substrates has a crucial role in promoting carcinogenesis. Overexpression of AKR1B10 has been identified in smoking-related carcinomas such as lung cancer. As development of pancreatic cancer is firmly linked to smoking, the aim of the present study was to examine the expression and oncogenic role of AKR1B10 in pancreatic adenocarcinoma. AKR1B10 expression was analyzed in 50 paraffin-embedded clinical pancreatic cancer samples using immunohistochemistry. Oncogenic function of AKR1B10 was examined in pancreatic carcinoma cells in vitro using western blotting and siRNA approaches, mainly on cell apoptosis and protein prenylation including KRAS protein and its downstream signals. Immunohistochemistry analysis revealed that AKR1B10 overexpressed in 70% (35/50) of pancreatic adenocarcinomas and majority of pancreatic intraepithelial neoplasia, but not in adjacent morphologically normal pancreatic tissue. Compared with a normal pancreatic ductal epithelial cell (HPDE6E7), all of the six cultured pancreatic adenocarcinoma cell lines had an overexpression of AKR1B10 using immunoblotting, which correlated with increase of enzyme activity. siRNA-mediated silencing of AKR1B10 expression in pancreatic cancer cells resulted in (1) increased cell apoptosis, (2) increased non-farnesyled HDJ2 protein and (3) decreased membrane-bound prenylated KRAS protein and its downstream signaling molecules including phosphorylated ERK and MEK and membrane-bound E-cadherin. Our findings provide first time evidence that AKR1B10 is a unique enzyme involved in pancreatic carcinogenesis possibly via modulation of cell apoptosis and protein prenylation.


Subject(s)
Adenocarcinoma/pathology , Aldehyde Reductase/metabolism , Carcinoma in Situ/pathology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/pathology , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Aldehyde Reductase/genetics , Aldo-Keto Reductases , Apoptosis , Biomarkers, Tumor/metabolism , Carcinoma in Situ/enzymology , Carcinoma in Situ/genetics , Cell Line, Tumor , Gene Knockdown Techniques , Gene Silencing , HSP40 Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry/methods , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Protein Prenylation
10.
J Gastroenterol Hepatol ; 27(11): 1752-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22742976

ABSTRACT

BACKGROUND AND AIM: Abnormal expression of Fragile Histidine Triad (Fhit), E-cadherin and p53 is observed in esophageal squamous cell carcinoma. It has recently been reported that aberrant expression of activation-induced cytidine deaminase (AID) in gastric epithelium leads to the accumulation of nucleotide alterations in the p53 gene. However, little is known about the association between these molecular events and the clinicopathological characteristics of early stage esophageal squamous neoplasia, especially in endoscopically resected tumors. METHODS: Esophageal squamous neoplasias (n = 49) comprising nine cases of low-grade intraepithelial neoplasia (LGIN), 22 of high-grade intraepithelial neoplasia/carcinoma in situ (HGIN/CIS) and 18 of invasive cancers, were endoscopically resected. Their expression of the tumor-related proteins: Fhit, E-cadherin, p53 and AID was assessed using immunohistochemical methods, and the relationship between protein expression and clinicopathological data was examined. RESULTS: Reduced or absent Fhit and E-cadherin expression was detected in 22% and 0% of LGIN cases, 73% and 14% of HGIN/CIS cases, and 94% and 61% of invasive cancer cases, respectively, showing progressive increases during neoplastic progression (Fhit: P < 0.01, E-cadherin: P < 0.01). Although p53 and AID were overexpressed in these samples, no change in their expression occurred during neoplastic progression. Moreover, p53 expression was not significantly associated with AID expression. CONCLUSIONS: These results indicate that a decrease in Fhit and E-cadherin expression could be related to the development and progression of esophageal squamous neoplasia, and that the expression of p53 was independent of aberrant AID expression in the early stage of esophageal carcinogenesis.


Subject(s)
Carcinoma in Situ/metabolism , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Acid Anhydride Hydrolases/metabolism , Aged , Aged, 80 and over , Alcohol Drinking , Cadherins/metabolism , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Chi-Square Distribution , Cytidine Deaminase/metabolism , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Proteins/metabolism , Smoking , Statistics, Nonparametric , Tumor Suppressor Protein p53/metabolism
11.
Am J Dermatopathol ; 34(7): 723-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22534634

ABSTRACT

Given the established role of matrix metalloproteinases (MMPs) in physiological processes in the skin, we investigated the expression of MMP-2, MMP-9, and MMP-14 to evaluate their role in the grading and development of atypical epithelial lesions. Immunohistochemistry was performed using antibodies against these MMPs in actinic keratosis (AK; n = 24), squamous cell carcinoma (SCC) in situ (SCCIS; n = 27), SCC well differentiated (SCCWD; n = 28), and SCC moderately to poorly differentiated (SCCMPD; n = 20). Tumoral and stromal expression was assessed by intensity (SI) and percentage positivity (PC). The mean of the total score, calculated by adding intensity and percentage positivity, was used for statistical analyses. In AK, SCCIS, SCCWD, and SCCMPD, mean tumoral MMP-2 expression was 3.33, 4.07, 4.46, and 3.40, respectively (P = NS for all) and stromal expression was 1.42, 3.26, 3.07, and 1.55 respectively (P < 0.05 for AK vs. SCCIS/SCCWD and SCCMPD vs. SCCIS/SCCWD); mean tumoral MMP-9 expression was 4.33, 4.11, 4.46, and 3.35, respectively, and stromal expression was 4.29, 4.41, 4.75, and 4.60, respectively (P = NS for all) and, mean tumoral MMP-14 expression was 1.58, 2.41, 0.32, and 0.35, respectively (P < 0.05 AK vs. SCCWD and SCCIS vs. SCCWD/SCCMPD) and stromal expression was 3.04, 3.52, 0.46, and 0.60, respectively (P < 0.05 for AK vs. SCCWD/SCCMPD). Only MMP-14 showed a statistically significant linear trend with decreasing values for tumoral and stromal expression with invasion suggesting that it might be of use as a prognosticator. Enhanced stromal MMP-2 expression in SCCIS and SCCWD relative to AK suggests that it may be of relevance to disease progression.


Subject(s)
Carcinoma in Situ/enzymology , Carcinoma, Squamous Cell/enzymology , Keratosis, Actinic/enzymology , Matrix Metalloproteinase 14/analysis , Matrix Metalloproteinase 2/analysis , Matrix Metalloproteinase 9/analysis , Skin Neoplasms/enzymology , Analysis of Variance , Biopsy , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Disease Progression , Enzyme Activation , Humans , Immunohistochemistry , Keratosis, Actinic/pathology , Linear Models , Neoplasm Invasiveness , Skin Neoplasms/pathology , Stromal Cells/enzymology , Stromal Cells/pathology
12.
Ann Diagn Pathol ; 16(5): 354-61, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22575501

ABSTRACT

Heparanase and cyclooxygenase-2 (COX-2) are 2 key enzymes that modulate diverse physiological processes during embryonic development and in adult life. Their deregulations have been implicated in the growth and progression of many cancer types. To date, comparatively little is known about the roles of these molecules during oral carcinogenesis. The aim of this study was to investigate the expression patterns of heparanase and COX-2 during progression of oral epithelial dysplasia (OED) to carcinoma. In situ hybridization and immunohistochemistry were performed on 5 cases of normal mucosa, 15 cases of OED, 5 cases of carcinoma in situ and/or microinvasive carcinoma, and 40 cases of oral squamous cell carcinoma (OSCC). Results demonstrated that heparanase and COX-2 messenger RNA and protein were absent in normal oral mucosa but were coexpressed in increasing intensity as OED progressed to OSCC. Concomitant heparanase- and COX-2-positive staining in the stromal cells suggests that OED/OSCC progression may be modulated by stromal-cancer cell interactions. Diffuse intense staining of poorly differentiated OSCC compared with staining localized to tumor nest periphery in well- and moderately differentiated OSCC suggests that heparanase and COX-2 overexpressions correlated with tumor grade. Strong expression of these enzymes in tumor cells at the advancing front suggests a role in local tumor spread. These results, taken together, suggest that heparanase and COX-2 might play complementary roles in the stepwise progression of OED to carcinoma.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma in Situ/genetics , Carcinoma, Squamous Cell/genetics , Cyclooxygenase 2/genetics , Gene Expression Regulation, Enzymologic/physiology , Glucuronidase/genetics , Mouth Neoplasms/genetics , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/enzymology , Cyclooxygenase 2/metabolism , Disease Progression , Epithelial Cells/enzymology , Epithelial Cells/pathology , Glucuronidase/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Mouth Mucosa/pathology , Mouth Neoplasms/diagnosis , Mouth Neoplasms/enzymology , Neoplasm Invasiveness , RNA, Messenger/metabolism
13.
BMC Cancer ; 11: 463, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22029821

ABSTRACT

BACKGROUND: Epigenetic regulation is an important mechanism leading to cancer initiation and promotion. Histone acetylation by histone deacetylases (HDACs) represents an important part of it. The development of HDAC inhibitors has identified the utility of HDACs as a therapeutic target. Little is known about the epigenetic regulation of vulvar intraepithelial neoplasia (VIN) and vulvar squamous cell cancer (VSCC). In this study, the expression of class I HDACs (HDAC 1, 2 and 3) was compared in a series of VIN and VSCC tissues. METHODS: A tissue micro array (TMA) with specimens from 106 patients with high-grade VIN and 59 patients with vulvar cancer was constructed. The expression of HDACs 1, 2 and 3 were analyzed with immunohistochemistry (IHC). The nuclear expression pattern was evaluated in terms of intensity and percentage of stained nuclei and was compared between vulvar preinvasive lesions and vulvar cancer. RESULTS: HDAC 2 expression was significantly higher in VIN than in VSCC (p < 0.001, Fisher's test). Also, 88.7% (n = 94/106) of VIN samples and only 54.5% (n = 31/57) of VSCC samples were scored at the maximum level. Conversely, HDAC 3 expression was significantly higher in VSCC (93%, 53/57) compared to VIN (73.6%, 78/106, p = 0.003), whereas only a small difference in the expression of HDAC 1 was found between these two entities of vulvar neoplasia. CONCLUSIONS: These results suggest that epigenetic regulation plays a considerable role in the transformation of VIN to invasive vulvar neoplasia.


Subject(s)
Carcinoma in Situ/enzymology , Carcinoma, Squamous Cell/enzymology , Histone Deacetylase 1/metabolism , Neoplasm Proteins/metabolism , Vulvar Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Invasiveness , Tissue Array Analysis/methods , Vulvar Neoplasms/pathology , Young Adult
14.
Zhonghua Zhong Liu Za Zhi ; 33(6): 421-5, 2011 Jun.
Article in Zh | MEDLINE | ID: mdl-21875481

ABSTRACT

OBJECTIVE: To investigate the expression variation and significance of ERK1/2 MAPK signaling transduction pathway in the pathogenesis of esophageal squamous cell carcinoma (ESCC) in Kazakh patients. METHODS: The expression level of p-ERK1/2 after serum starvation and treatment with U0126 inhibitor was detected in esophageal cancer cell line EC9706 by Western blot assay. The mRNA level of total ERK1/2 (t-ERK1/2) and expression level of t-ERK1/2 and p-ERK1/2 proteins of 25 pairs of ESCC and adjacent normal esophageal mucosal tissues of Kazakh patients were examined and identified by real-time quantitative PCR (qRT-PCR) and Western blotting, respectively. The expression of p-ERK1/2 protein was verified by immunohistochemistry in 126 paraffin-embeded specimens, including 19 normal esophageal mucosa, 55 esophageal carcinomas in situ and 52 invasive carcinomas. RESULTS: ERK1/2 MAPK signaling transduction pathway was in an active status in the EC9706 cells. The expression level of p-ERK1/2 in Ec9706 cells reached a peak at 10 min after transient serum stimulation, and p-ERK1/2 expression was totally restrained after the treatment with 50 µmol/L U0126. In the 25 pairs of ESCC and adjacent normal mucosa, the t-ERK1 mRNA level was 1.92 ± 3.49 in the ESCC tissues and 3.67 ± 7.47 in the adjacent normal mucosa. The t-ERK1 mRNA level in ESCC tissues was significantly lower than that in adjacent normal mucosa (P < 0.05), whereas there was no significant difference of t-ERK2 mRNA level between them(P > 0.05). The expression levels of p-ERK1 and p-ERK2 proteins were 0.87 ± 0.14 and 0.79 ± 0.10 in the ESCC tissues, and 1.10 ± 0.13 and 1.32 ± 0.12 in the adjacent normal mucosae. p-ERK1/2 protein in the ESCC tissues was significantly lower than that in the adjacent normal tissue (P < 0.01). However, there was no significant difference between their t-ERK1/2 protein levels (P > 0.05). In the 126 cases of paraffin-embeded specimens, positive expressions of both p-ERK1 and p-ERK2 in esophageal cancer tissues were 7.7% (4/52), significantly lower than those in adjacent normal mucosa (31.6%, 6/19) and carcinoma in situ (85.5%, 47/55, P < 0.05). CONCLUSIONS: ERK1/2 MAPK signaling pathway is in an active status in esophageal cancer and adjacent normal mucosa. Our results imply that the activation of p-ERK1/2 MAPK signaling transduction pathway plays a role in the early pathogenesis of ESCC in Kazakh patients.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Esophageal Neoplasms/enzymology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Butadienes/pharmacology , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , China/ethnology , Enzyme Inhibitors/pharmacology , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Nitriles/pharmacology , Phosphorylation , RNA, Messenger/metabolism
15.
Cancer Metastasis Rev ; 28(3-4): 305-16, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20013032

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) pathway is a critical signal transduction pathway that regulates multiple cellular functions. Aberrant activation of this pathway has been identified in a wide range of cancers. Several pathway components including AKT, PI3K and mTOR represent potential therapeutic targets and many small molecule inhibitors are in development or early clinical trials. The complex regulation of the pathway, together with the multiple mechanisms by which it can be activated, make this a highly challenging pathway to target. For successful inhibition, detailed molecular information on individual tumours will be required and it is already clear that different tumour types show distinct combinations of alterations. Recent results have identified alterations in pathway components PIK3CA, PTEN, AKT1 and TSC1 in bladder cancer, some of which are significantly related to tumour phenotype and clinical behaviour. Co-existence of alterations to several PI3K pathway genes in some bladder tumours indicates that these proteins may have functions that are not related solely to the known canonical pathway.


Subject(s)
Carcinoma, Papillary/enzymology , Carcinoma, Transitional Cell/enzymology , Cell Transformation, Neoplastic/pathology , Neoplasm Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology , Urinary Bladder Neoplasms/enzymology , Antineoplastic Agents/pharmacology , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Cell Transformation, Neoplastic/genetics , Drug Delivery Systems , Enzyme Activation , Humans , Hyperplasia , Models, Biological , Mutation , Neoplasm Proteins/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/physiology , Receptor Protein-Tyrosine Kinases/physiology , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urothelium/pathology
16.
Mod Pathol ; 23(6): 844-55, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20228782

ABSTRACT

Serous tubal intraepithelial carcinoma (STIC) has been proposed as a precursor for many pelvic high-grade serous carcinomas. Our previous analysis of the ovarian cancer genome identified several genes with oncogenic potential that are amplified and/or overexpressed in the majority of high-grade serous carcinomas. Determining whether these genes are upregulated in STICs is important in further elucidating the relationship of STICs to high-grade serous carcinomas and is fundamental in understanding the molecular pathogenesis of high-grade serous carcinomas. In this study, 37 morphologically defined STICs were obtained from 23 patients with stage IIIC/IV high-grade serous carcinomas. Both STICs and the high-grade serous carcinomas were analyzed for expression of Rsf-1 (HBXAP), cyclin E, fatty acid synthase (FASN) and mucin-4. In addition, they were examined for expression of established markers including p53, Ki-67 and p16. We found that diffuse nuclear p53 and p16 immunoreactivity was observed in 27 (75%) of 36 and 18 (55%) of 33 STICs, respectively, whereas an elevated Ki-67 labeling index (>or=10%) was detected in 29 (78%) of 37 STICs. Cyclin E nuclear staining was seen in 24 (77%) of 35 STICs, whereas normal tubal epithelial cells were all negative. Increased Rsf-1 and FASN immunoreactivity occurred in 63%, and 62% of STICs, respectively, compared with adjacent normal-appearing tubal epithelium. Interestingly, only one STIC showed increased mucin-4 immunoreactivity. Carcinomas, when compared with STICs, overexpressed p16, Rsf-1, cyclin E and FASN in a higher proportion of cases. In conclusion, STICs express several markers including Rsf-1, cyclin E and FASN in high-grade serous carcinomas. In contrast, mucin-4 immunoreactivity either did not change or was reduced in most STICs. These results suggest that overexpression of Rsf-1, cyclin E and FASN occurs early in tumor progression.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma in Situ/chemistry , Cyclin E/analysis , Fallopian Tube Neoplasms/chemistry , Fatty Acid Synthase, Type I/analysis , Neoplasms, Cystic, Mucinous, and Serous/chemistry , Nuclear Proteins/analysis , Ovarian Neoplasms/chemistry , Trans-Activators/analysis , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Cyclin-Dependent Kinase Inhibitor p16/analysis , Fallopian Tube Neoplasms/enzymology , Fallopian Tube Neoplasms/pathology , Female , Humans , Ki-67 Antigen/analysis , Mucin-4/analysis , Neoplasms, Cystic, Mucinous, and Serous/enzymology , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Tumor Suppressor Protein p53/analysis
17.
Histopathology ; 57(1): 90-100, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20653782

ABSTRACT

AIMS: Metaplastic changes secondary to chronic inflammation at the gastro-oesophageal junction and at the pyloric antrum are recognized as the premalignant conditions of Barrett's oesophageal adenocarcinoma and intestinal-type gastric carcinoma (GC), respectively. Heparanase (HPSE) and cyclooxygenase (COX)-2 have been proved to play critical roles in inflammation as well as in cancer. The aim was to examine the meaning of their expression in inflammation-related carcinogenesis. METHODS AND RESULTS: First, expression of HPSE and COX-2 in 78 clinical tissues of Barrett's oesophagus was examined by immunohistochemistry and in situ hybridization. Their expression was increased during the metaplasia-dysplasia sequence with increased neovascularization. Successively, their expression in Barrett's dysplasia was compared with that of GC (22 cases of diffuse-type and 10 of intestinal-type). Interestingly, the expression pattern in Barrett's dysplasia was similar to that in intestinal-type GC, which mainly arises from chronic inflammation. Furthermore, cultured cell lines isolated from differentiated GC tissues, which are often found to be of intestinal-type, revealed up-regulated mRNA expression of HPSE and COX-2. CONCLUSIONS: HPSE and COX-2 are preferentially up-regulated in Barrett's oesophagus and intestinal-type GC. These molecules may play an important role during the development of inflammation-related adenocarcinoma of the upper gastrointestinal tract.


Subject(s)
Adenocarcinoma/enzymology , Barrett Esophagus/enzymology , Cyclooxygenase 2/metabolism , Esophageal Neoplasms/enzymology , Glucuronidase/metabolism , Stomach Neoplasms/enzymology , Adenocarcinoma/blood supply , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Base Sequence , Carcinoma in Situ/enzymology , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Cell Line, Tumor , Cyclooxygenase 2/genetics , DNA Primers/genetics , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Glucuronidase/genetics , Humans , Microvessels/pathology , Neovascularization, Pathologic , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Stomach Neoplasms/pathology , Up-Regulation
18.
J Oral Pathol Med ; 39(7): 565-70, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20412400

ABSTRACT

BACKGROUND: An inflammatory component consisting of cells and chemical mediators may influence the proliferation and dissemination of the oral squamous cell carcinoma (OSCC). In the present study, we evaluated the possible relationship between Ki-67, tumor-associated macrophages (TAMs), and COX-2 in OSCCs. In addition, the immunodetection of these proteins was associated with different histological grades of malignancy, including invasive and in situ tumors. METHODS: Twenty-seven OSCC cases were examined by light microscopy using criteria adopted WHO, and immunohistochemistry for Ki-67, CD68, and COX-2 using EnVision System in invasive and in situ lesions. Immunohistochemical detection of these proteins was assessed and scored for COX-2, and results were compared with their histological grades of malignancy. RESULTS: A correlation between Ki-67, COX-2, and CD68 was not found. Histological grade of malignancy (HDM) was associated with the Ki-67 immunostaining (P = 0.00), but this was not observed regarding both CD68 (P = 0.51) and COX-2 (P = 0.89). Furthermore, there was a COX-2 overexpression in 62.96% of the sample, and a high density of TAMs in both OSCCs and in situ carcinomas. CONCLUSIONS: Imunolabeling for Ki-67 was directly correlated with less-differentiated tumors, suggesting that this marker may contribute to understand the biological behavior of OSCC, and help to distinguish risk groups of OSCC. Furthermore, the lack of correlation between Ki-67, COX-2, and CD68 indicates that the latter two markers may play a pivotal role in oral carcinogenesis. However, further studies are needed to clarify their contribution for cell proliferation and tumor differentiation.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cyclooxygenase 2/analysis , Ki-67 Antigen/analysis , Macrophages/pathology , Mouth Neoplasms/pathology , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Biomarkers, Tumor/analysis , Carcinoma in Situ/enzymology , Carcinoma in Situ/immunology , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/immunology , Cell Nucleus/ultrastructure , Coloring Agents , Cytoplasm/ultrastructure , Epithelial Cells/enzymology , Epithelial Cells/pathology , Humans , Immunohistochemistry , Mouth Neoplasms/enzymology , Mouth Neoplasms/immunology , Neoplasm Invasiveness
19.
Int J Colorectal Dis ; 25(9): 1079-85, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20549218

ABSTRACT

PURPOSE: We aimed to study the intracellular expression of 5-lipoxygenase (5-LOX), the primary competitor with cyclooxygenase-2 in arachidonic acid metabolism, as inflammatory enzymes may be involved in blocking apoptosis and promoting cancer growth by changing arachidonic acid metabolism within cells. Our purpose was to investigate the possible connection between 5-LOX expression and colon carcinogenesis by characterizing 5-LOX expression in histologically different colonic adenomas, determining the relationship between high expression of 5-LOX and various conventional clinicopathological features of adenomas, and finally characterizing the histological localization of cells with 5-LOX overexpression. METHODS: A total of 111 patients were examined and 120 histologically different colonic adenomas analyzed (including four cases of intramucosal adenocarcinoma in a polyp). Immunohistochemical staining with polyclonal anti-5-LOX antibodies was performed. RESULTS: There was a significant correlation between high 5-LOX expression and patient age, increased polyp size, high grade of intraepithelial neoplasia, villous and tubulovillous adenoma, and histological epithelial localization. CONCLUSIONS: We observed a strong positive correlation between 5-LOX overexpression and the appearance of typical high-risk factors for malignant transformation in adenomatous polyps. The results support the role of 5-LOX in early stages of colon carcinogenesis.


Subject(s)
Adenoma/enzymology , Adenoma/pathology , Arachidonate 5-Lipoxygenase/metabolism , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Adult , Aged , Aged, 80 and over , Carcinoma in Situ/enzymology , Carcinoma in Situ/pathology , Colonic Polyps/enzymology , Colonic Polyps/pathology , Female , Humans , Male , Middle Aged
20.
BMC Cancer ; 9: 185, 2009 Jun 16.
Article in English | MEDLINE | ID: mdl-19531212

ABSTRACT

BACKGROUND: New, third-generation aromatase inhibitors (AIs) have proven comparable or superior to the anti-estrogen tamoxifen for treatment of estrogen receptor (ER) and/or progesterone receptor (PR) positive breast cancer. AIs suppress total body and intratumoral estrogen levels. It is unclear whether in situ carcinoma cell aromatization is the primary source of estrogen production for tumor growth and whether the aromatase expression is predictive of response to endocrine therapy. Due to methodological difficulties in the determination of the aromatase protein, COX-2, an enzyme involved in the synthesis of aromatase, has been suggested as a surrogate marker for aromatase expression. METHODS: Primary tumor material was retrospectively collected from 88 patients who participated in a randomized clinical trial comparing the AI letrozole to the anti-estrogen tamoxifen for first-line treatment of advanced breast cancer. Semi-quantitative immunohistochemical (IHC) analysis was performed for ER, PR, COX-2 and aromatase using Tissue Microarrays (TMAs). Aromatase was also analyzed using whole sections (WS). Kappa analysis was applied to compare association of protein expression levels. Univariate Wilcoxon analysis and the Cox-analysis were performed to evaluate time to progression (TTP) in relation to marker expression. RESULTS: Aromatase expression was associated with ER, but not with PR or COX-2 expression in carcinoma cells. Measurements of aromatase in WS were not comparable to results from TMAs. Expression of COX-2 and aromatase did not predict response to endocrine therapy. Aromatase in combination with high PR expression may select letrozole treated patients with a longer TTP. CONCLUSION: TMAs are not suitable for IHC analysis of in situ aromatase expression and we did not find COX-2 expression in carcinoma cells to be a surrogate marker for aromatase. In situ aromatase expression in tumor cells is associated with ER expression and may thus point towards good prognosis. Aromatase expression in cancer cells is not predictive of response to endocrine therapy, indicating that in situ estrogen synthesis may not be the major source of intratumoral estrogen. However, aromatase expression in combination with high PR expression may select letrozole treated patients with longer TTP. TRIAL REGISTRATION: Sub-study of trial P025 for advanced breast cancer.


Subject(s)
Aromatase/biosynthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Nitriles/therapeutic use , Receptors, Estrogen/biosynthesis , Tamoxifen/therapeutic use , Triazoles/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/enzymology , Carcinoma in Situ/drug therapy , Carcinoma in Situ/enzymology , Carcinoma in Situ/metabolism , Cyclooxygenase 2/biosynthesis , Female , Humans , Immunohistochemistry , Letrozole , Receptors, Progesterone/biosynthesis , Retrospective Studies , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL