Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 229
Filter
1.
Gut ; 57(10): 1413-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18436576

ABSTRACT

OBJECTIVE: To compare the independent prognostic effect of a panel of immunohistochemical protein markers in colorectal cancer (CRC) and determine their ranking among the established prognostic factors T stage, N stage, vascular invasion, tumour budding and tumour grade. DESIGN: A tissue microarray of 1420 CRCs was immunostained for 23 markers and mismatch repair (MMR) proteins. Immunoreactivity was assessed semi-quantitatively. Receiver operating characteristic (ROC) curves were used to determine cut-off scores for tumour marker positivity. Survival time was investigated for each marker in multivariable analysis with T stage, N stage, vascular invasion, tumour budding and tumour grade. The hazard ratio (HR) was used to compare the prognostic effect of each marker on 5 year survival. RESULTS: To the standard prognostic features, only six markers added independent prognostic information including receptor for hyaluronic acid mediated motility (RHAMM) (HR = 2.39 (1.88 to 3.05)), epidermal growth factor receptor (HR = 1.65 (1.31 to 2.09)), tumour infiltrating lymphocytes (HR = 0.7 (0.54 to 0.92)), urokinase plasminogen activator (HR = 1.38 (1.09 to 1.75)), Raf-1 kinase inhibitor protein (HR = 0.75 (0.58 to 0.96)) and mammalian sterile 20-like kinase 1 (MST1) (HR = 0.75 (0.58 to 0.95). Diffuse (>90% staining) expression of RHAMM ranked above T stage, vascular invasion, tumour budding and tumour grade in terms of adverse prognostic significance and was associated with distant metastasis (p = 0.012) and with worse outcome in patients with metastatic disease (p = 0.031). CONCLUSIONS: The strong adverse effect of RHAMM on outcome in addition to its position within the hierarchy of well-established prognostic factors suggest that RHAMM should be considered a more important prognosticator than tumour grade, tumour budding and vascular invasion in patients with CRC.


Subject(s)
Biomarkers, Tumor/analysis , Colorectal Neoplasms/chemistry , Extracellular Matrix Proteins/analysis , Hyaluronan Receptors/analysis , Liver Neoplasms/pathology , Chronic Disease , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Liver Neoplasms/secondary , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , ROC Curve , Survival Analysis , Tissue Array Analysis
2.
Br J Cancer ; 98(2): 450-6, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18182986

ABSTRACT

The ability to predict complete pathologic response or sensitivity to radiation before treatment would have a significant impact on the selection of patients for preoperative radiotherapy or chemo-radiation therapy schedules. The aim of this study was to determine the value of epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), p53, Bcl-2 and apoptosis protease-activating factor-1 (APAF-1) as predictors of complete pathologic tumour regression in patients undergoing preoperative radiotherapy for advanced rectal cancer. Pretreatment tumour biopsies from predominantly cT3 patients undergoing a preoperative high-dose-rate brachytherapy protocol were immunostained for EGFR, VEGF, p53, Bcl-2 and APAF-1. Immunoreactivity was evaluated by three pathologists. Cut-off scores for tumour marker positivity were obtained by receiver-operating characteristic (ROC) curve analysis. The association of marker expression with complete pathologic response was analysed in univariate and multivariable analysis. Multi-marker phenotypes of the independent protein markers were evaluated. In multivariable analysis, loss of VEGF (P-value=0.009; odds ratio (OR) (95% CI)=0.24 (0.08-0.69)) and positive EGFR (P-value=0.01; OR (95% CI)=3.82 (1.37-10.6)) both demonstrated independent predictive value for complete pathologic response. The odds of complete response were 12.8 for the multi-marker combination of VEGF-negative and EGFR-positive tumours. Of the 34 EGFR-negative- and VEGF-positive cases, 32 (94.1%) had no complete pathologic response. The combined analysis of VEGF and EGFR is predictive of complete pathologic response in patients undergoing preoperative radiotherapy. In addition, the findings of this study have identified a subgroup of simultaneous EGFR-negative and VEGF-positive patients who are highly resistant to radiotherapy and should perhaps be considered candidates for innovative neoadjuvant combined modalities.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/radiotherapy , Adenocarcinoma/surgery , ErbB Receptors/analysis , Rectal Neoplasms/diagnosis , Rectal Neoplasms/radiotherapy , Rectal Neoplasms/surgery , Vascular Endothelial Growth Factor A/analysis , Adenocarcinoma/pathology , Algorithms , Biomarkers, Tumor/analysis , Brachytherapy , Combined Modality Therapy , Female , Humans , Male , Neoplasm Staging , Preoperative Care , Prognosis , ROC Curve , Radiotherapy, Adjuvant , Rectal Neoplasms/pathology , Remission Induction
3.
J Pathol ; 213(3): 257-65, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17893910

ABSTRACT

Disruptions to the TGFbeta signalling pathway have been implicated in most human adenocarcinomas. As cancers progress, many acquire resistance to the growth-suppressing properties of TGFbeta while retaining sensitivity to its tumour-promoting effects. Microsatellite unstable colorectal cancers (MSI-H CRCs) possess truncating mutations in the type II TGFbeta receptor (TGFbetaRII) gene that have been assumed to render these tumours insensitive to TGFbeta. However, numerous reports of TGFbetaRII bypass exist and this study was thus undertaken in order to clarify the true extent of TGFbeta sensitivity in MSI-H CRCs. Using stimulation with exogenous TGFbeta, we demonstrated that, while MSI-H CRCs are capable of binding soluble TGFbeta, two out of three cell lines examined remain refractory to its signalling effects. In contrast, use of a specific inhibitor of the type I TGFbeta receptor (TGFbetaRI) revealed that all remain sensitive to signalling by endogenously produced TGFbeta. Specifically, autocrine signalling via TGFbetaRI mediates constitutive activation of Smad2 as well as repression of Erk signalling. Real-time PCR confirmed that these effects are sufficient to affect the expression level of various TGFbeta-modulated genes. An invasion assay revealed that autocrine TGFbetaRI signalling also promotes the invasion capacity of MSI-H CRCs to an extent similar to that seen in their non-MSI-H counterparts. Independent TGFbetaRI signalling, however, has no effect on the rate of proliferation of MSI-H CRC cells. Together, these results demonstrate that MSI-H CRC cell lines are not completely refractory to TGFbeta, despite lacking functional TGFbetaRII. In addition to clarifying the true consequences of natural TGFbetaRII loss and the independent function of TGFbetaRI, our results highlight the selective nature of TGFbeta resistance developed by cancers.


Subject(s)
Colorectal Neoplasms/genetics , Microsatellite Instability , Mutation , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Autocrine Communication , Benzamides/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dioxoles/pharmacology , Drug Resistance, Neoplasm/genetics , Humans , Loss of Heterozygosity , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction
4.
J Pathol ; 212(3): 260-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17516584

ABSTRACT

Tumour budding or dedifferentiation at the invasive margin of colorectal cancer (CRC) is an important prognostic marker and linked mechanistically to dysregulation of Wnt pathway signalling. Since budding is observed in only 40% of CRCs, we hypothesized that Wnt pathway dysregulation may be a necessary but insufficient explanation for budding and that buds may be destroyed selectively by tumour immune mechanisms. Twenty potential markers of tumour budding were evaluated in tissue microarrays (TMAs) obtained from the main tumour body of 1164 DNA mismatch repair-proficient CRCs and the findings were correlated with tumour budding, lymphocytic infiltration and survival. Loss of expression of E-cadherin and APAF-1 were independent predictors of budding (sensitivity 70.3% and specificity 48.2% when one or the other was lost). Peritumoral lymphocytes (PTLs) were observed more frequently in CRCs with loss of either E-cadherin or APAF-1 that were budding-negative. PTLs and tumour-infiltrating lymphocytes (TILs) were strongly correlated. The absence of TILs increased the adverse prognostic impact of E-cadherin and APAF-1 loss. Co-occurrence of E-cadherin loss, APAF-1 loss and low TIL counts in CRCs was an independent prognostic factor. The findings were verified in whole tissue sections from 88 CRCs with known KRAS mutation status (which was not associated with budding). Loss of E-cadherin and APAF-1 within the main body of CRCs are independent predictors of tumour budding. The prognostic benefit of lymphocytic infiltration may be explained by the immune destruction of budding cells.


Subject(s)
Adenoma/pathology , Apoptotic Protease-Activating Factor 1/genetics , Biomarkers, Tumor/analysis , Cadherins/genetics , Colorectal Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Adenoma/immunology , Adenoma/mortality , Apoptotic Protease-Activating Factor 1/analysis , Cadherins/analysis , Case-Control Studies , Colon/immunology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , DNA Mismatch Repair , Gene Deletion , Humans , Immunohistochemistry , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , ROC Curve , Survival Analysis
5.
J Pathol ; 212(2): 124-33, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17427169

ABSTRACT

There is increasing evidence for an alternative pathway of sporadic colorectal tumourigenesis that is associated with DNA microsatellite instability (MSI), due to methylation and loss of expression of the mismatch repair gene MLH1. Recent studies have highlighted a serrated pathway of colorectal cancer (CRC) in which serrated polyps with activating mutations in BRAF progress to CRCs with MSI following methylation and silencing of MLH1. The present study provides a novel mechanistic experimental model for these clinical observations. We investigated the role of BRAF activating mutation (BRAF-V600E) in colorectal tumourigenesis by studying the effects of forced expression of BRAF-V600E in the 'normal' colon epithelial NCM460 cell line and by targeting endogenous BRAF-V600E in MSI-High (MSI-H) colon cancer cell lines. The findings indicate that BRAF mutation in colon epithelial cells contributes to a gain in resistance towards apoptotic stimuli, which is likely to be an important characteristic of pre-malignant serrated lesions. BRAF-V600E also plays a role in the development and maintenance of transformed and invasive phenotypes in colon epithelial cells. Our findings also suggest that BRAF mutation potentiates promoter hypermethylation of the MLH1 gene promoter. Together, these results highlight BRAF as a potential target for therapeutic intervention in sporadic MSI-H colorectal cancers.


Subject(s)
Colorectal Neoplasms/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Colon/pathology , Colorectal Neoplasms/pathology , CpG Islands/genetics , Epithelial Cells/pathology , Humans , Intestinal Mucosa/pathology , Methylation , Microsatellite Instability , Models, Biological , MutL Protein Homolog 1 , Mutation , Neoplasm Invasiveness , Nuclear Proteins/genetics , Phenotype , Promoter Regions, Genetic/genetics
6.
Histopathology ; 50(4): 453-64, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17448021

ABSTRACT

AIMS: To investigate dysregulation of the wnt signalling pathway by assessing beta-catenin expression/increasing expression and loss of cytoplasmic adenomatous polyposis coli (APC) and membranous E-cadherin in colorectal cancer (CRC) and determining the prognostic significance of these variables. METHODS AND RESULTS: Unselected, non-consecutive CRC resections (n = 1420) were subdivided into three groups: mismatch repair (MMR)-proficient, MLH1- and presumed hereditary non-polyposis colonic cancer (HNPCC). Immunohistochemical analysis of beta-catenin expression (0% versus > 0%) and increasing expression (increasing percentage-positivity) and loss of APC and E-cadherin was performed using the tissue microarray technique. In MMR-proficient CRC, increased nuclear beta-catenin expression and loss of membranous E-cadherin were independently associated with higher N stage (P = 0.03 and < 0.0001), vascular invasion (P < 0.01 and < 0.001) and worse survival (P < 0.01 and < 0.001). Additionally, there was an association between loss of membranous E-cadherin and higher T stage (P = 0.03). In MLH1- CRC, loss of membranous E-cadherin was associated with higher N stage (P = 0.05) and worse survival (P = 0.03). In presumed HNPCC CRC nuclear beta-catenin and membranous E-cadherin were not associated with tumour progression or worse survival. In all CRC subsets loss of cytoplasmic APC was not associated with clinicopathological features. CONCLUSIONS: Increasing nuclear beta-catenin expression and loss of membranous E-cadherin are independent, adverse prognostic factors in MMR-proficient and MLH1- CRC.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , Cadherins/physiology , Colorectal Neoplasms/diagnosis , Wnt Proteins/physiology , beta Catenin/physiology , Adaptor Proteins, Signal Transducing , Adenomatous Polyposis Coli Protein/biosynthesis , Cadherins/biosynthesis , Carrier Proteins/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , Cytoplasm/metabolism , DNA Mismatch Repair , Female , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , MutL Protein Homolog 1 , Nuclear Proteins/genetics , Prognosis , Regression Analysis , Signal Transduction , Tissue Array Analysis , beta Catenin/biosynthesis
8.
Histopathology ; 50(1): 113-30, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17204026

ABSTRACT

Over the last 20 years it has become clear that colorectal cancer (CRC) evolves through multiple pathways. These pathways may be defined on the basis of two molecular features: (i) DNA microsatellite instability (MSI) status stratified as MSI-high (MSI-H), MSI-low (MSI-L) and MS stable (MSS), and (ii) CpG island methylator phenotype (CIMP) stratified as CIMP-high, CIMP-low and CIMP-negative (CIMP-neg). In this review the morphological correlates of five molecular subtypes are outlined: Type 1 (CIMP-high/MSI-H/BRAF mutation), Type 2 (CIMP-high/MSI-L or MSS/BRAF mutation), Type 3 (CIMP-low/MSS or MSI-L/KRAS mutation), Type 4 (CIMP-neg/MSS) and Type 5 or Lynch syndrome (CIMP-neg/MSI-H). The molecular pathways are determined at an early evolutionary stage and are fully established within precancerous lesions. Serrated polyps are the precursors of Types 1 and 2 CRC, whereas Types 4 and 5 evolve through the adenoma-carcinoma sequence. Type 3 CRC may arise within either type of polyp. Types 1 and 4 are conceived as having few, if any, molecular overlaps with each other, whereas Types 2, 3 and 5 combine the molecular features of Types 1 and 4 in different ways. This approach to the classification of CRC should accelerate understanding of causation and will impact on clinical management in the areas of both prevention and treatment.


Subject(s)
Adenocarcinoma/classification , Colorectal Neoplasms/classification , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , CpG Islands/genetics , DNA Methylation , DNA, Neoplasm/analysis , Humans , Microsatellite Instability , Mutation , Precancerous Conditions/genetics , Precancerous Conditions/pathology
9.
Histopathology ; 50(1): 163-74, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17204029

ABSTRACT

Poorly differentiated malignancies affecting the anal canal are uncommon but pose diagnostic difficulties because of the wide range of normal cell types that may occur within a limited anatomical region. The range of lesions that may present as poorly differentiated tumours includes squamous cell carcinoma, adenocarcinoma, small and large cell neuroendocrine carcinoma, neuroendocrine carcinoma expressing epithelial cytokeratins and other patterns of mixed differentiation, undifferentiated carcinoma, malignant melanoma, lymphoma and secondary tumours. This review discusses the differential diagnosis of these neoplasms with the aid of short illustrative case studies.


Subject(s)
Anal Canal/pathology , Anus Neoplasms/pathology , Adenocarcinoma/pathology , Aged , Carcinoma, Squamous Cell/pathology , Diagnosis, Differential , Female , Humans , Lymphoma/pathology , Male , Melanoma/pathology , Middle Aged , Pathology, Surgical/methods
10.
Oncogene ; 26(30): 4435-41, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17260021

ABSTRACT

The mutated in colorectal cancer (MCC) gene is in close linkage with the adenomatous polyposis coli (APC) gene on chromosome 5, in a region of frequent loss of heterozygosity in colorectal cancer. The role of MCC in carcinogenesis, however, has not been extensively analysed, and functional studies are emerging, which implicate it as a candidate tumor suppressor gene. The aim of this study was to examine loss of MCC expression due to promoter hypermethylation and its clinicopathologic significance in colorectal cancer. Correspondence of MCC methylation with gene silencing was demonstrated using bisulfite sequencing, reverse transcription-polymerase chain reaction and Western blotting. MCC methylation was detected in 45-52% of 187 primary colorectal cancers. There was a striking association with CDKN2A methylation (P<0.0001), the CpG island methylator phenotype (P<0.0001) and the BRAF V600E mutation (P<0.0001). MCC methylation was also more common (P=0.0084) in serrated polyps than in adenomas. In contrast, there was no association with APC methylation or KRAS mutations. This study demonstrates for the first time that MCC methylation is a frequent change during colorectal carcinogenesis. Furthermore, MCC methylation is significantly associated with a distinct spectrum of precursor lesions, which are suggested to give rise to cancers via the serrated neoplasia pathway.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Genes, MCC , Promoter Regions, Genetic , Adenoma/genetics , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , CpG Islands , Humans , Intestinal Polyps/genetics , Mutation , Phenotype , Proto-Oncogene Proteins B-raf/genetics
11.
J Clin Pathol ; 60(5): 534-9, 2007 May.
Article in English | MEDLINE | ID: mdl-16816167

ABSTRACT

BACKGROUND: Expression of mucin antigen MUC1 and down regulation of MUC2 are associated with adverse prognosis in colorectal cancer (CRC), but their prognostic significance with respect to differing DNA mis- match repair (MMR) status is poorly understood. OBJECTIVE: To determine the prognostic significance of MUC1 and MUC2 in CRC with different MMR statuses. METHODS: Using the tissue microarray (TMA) technique, a series of 1420 unselected, non-consecutive CRC resections was subdivided into three groups: (1) MMR-proficient; (2) MLH1-negative; and (3) presumed hereditary non-polyposis colon cancer (HNPCC). Immunohistochemical analysis of MUC1 and MUC2 expression (>0%) and loss (0%) was performed, and the results were correlated with clinicopathological parameters. RESULTS: In MMR-proficient CRC, MUC1 expression was more frequently found in tumours with higher tumour stage (p=0.004) and higher tumour grade (p=0.041) and loss of MUC2 was associated with higher tumour stage (p=0.028), node stage (p=0.001), presence of vascular invasion (p=0.028) and worse survival (p=0.034). In MLH1-negative CRC, MUC2 loss was associated with the presence of lymph node metastasis (p=0.028) and worse survival (p=0.015), but there was no association between MUC1 expression and clinicopathological features. In presumed HNPCC, MUC1 expression and MUC2 loss were not associated with clinicopathological parameters. CONCLUSIONS: Mucins have a prognostic significance in sporadic CRC, but not in hereditary CRC. Loss of MUC2 is an adverse prognostic factor in MMR-proficient and MLH1-negative CRC, whereas MUC1 expression is associated with tumour progression in MMR-proficient CRC only.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , DNA Mismatch Repair , Mucins/metabolism , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA, Neoplasm/genetics , Disease Progression , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Mucin-1 , Mucin-2 , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Neoplasm Staging , Prognosis , Protein Array Analysis/methods , Survival Analysis
12.
Gut ; 56(1): 13-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16785283

ABSTRACT

BACKGROUND: Oesophageal squamous cell carcinoma (OSCC) often arises from preceding dysplastic lesions in the oesophageal epithelium. However, the molecular changes occurring in premalignant lesions are not well understood. An epigenetic change is an example of OSCC that may occur within the epithelium. AIM: To investigate the methylation status of multiple promoters in cancer-derived DNA, as well as in the background epithelium of OSCC, including dysplastic lesions and non-neoplastic mucosa. The normal epithelium from patients without cancer was also examined. The findings were correlated with the mutational status of p53. PATIENTS AND METHODS: 56 patients with advanced OSCC, 21 patients with intraepithelial neoplasia (IEN), 56 patients with a background of non-neoplastic epithelium, adjacent to the OSCC, and 42 normal control epithelia from healthy volunteers were studied. The promoter methylation status of SFRP1, SFRP2, DCC, APC, p16(INK4a), p14(ARF), MINT1, MINT2, MINT31, CACNA1G, COX2, DAPK, hMLH1 and MGMT was examined by methylation-specific single polymerase chain reaction or combined bisulphite restriction analysis. The mutation of p53 by direct sequencing was assessed. RESULTS: DNA methylation was observed in OSCC and in its background epithelium. The frequency of CpG island methylation increased from a baseline level in the background non-neoplastic epithelium, through IEN, to advanced OSCC. However, mutations in p53 were almost exclusively observed in IEN and OSCC. More extensive DNA methylation was seen in the neoplastic lesions (OSCC or IEN) having a p53 mutation than in those with wild-type p53. CONCLUSION: DNA methylation is present at low levels in the non-neoplastic oesophageal epithelium and appears to contribute to the progression of the dysplasia-carcinoma sequence in OSCC carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Methylation , DNA, Neoplasm/genetics , Esophageal Neoplasms/genetics , Adult , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , CpG Islands/genetics , DNA, Neoplasm/metabolism , Disease Progression , Epithelium/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Humans , Male , Mucous Membrane/metabolism , Mutation , Promoter Regions, Genetic/genetics , Tumor Suppressor Protein p53/genetics
14.
Histopathology ; 49(4): 371-80, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16978200

ABSTRACT

AIMS: Colorectal cancers deficient in DNA mismatch repair (MMR) are often characterized by the presence of numerous intraepithelial lymphocytes (IELs). These CD8+ T cells selectively express CD103, which is upregulated locally by transforming growth factor (TGF)-beta, and adhere to E-cadherin expressed by mucosal epithelia. Many of these cancers also possess inactivating mutations in the type II TGF-beta receptor and are believed to be insensitive to TGF-beta. The present study aimed to explore whether such refractoriness to TGF-beta is an independently contributing factor to IEL retention. METHODS AND RESULTS: A panel of colorectal cancers enriched for DNA MMR deficiency was examined by immunohistochemistry to explore the expression levels and localization of various components in the TGF-beta signalling pathway. Logistic regression was then carried out in order to identify predictors of elevated lymphocytic infiltration independent of DNA MMR status. Increases in Smad4 expression, tumour cell proliferation and TGF-beta secretion each emerged as independent predictors of marked lymphocyte infiltration. CONCLUSIONS: These results strongly support the hypothesis that refractoriness to normal TGF-beta signalling in colorectal cancers plays a role in the retention of lymphocytes within tumour epithelium. Since IEL infiltration is an independent predictor of favourable prognosis, the TGF-beta pathway may represent an important therapeutic target.


Subject(s)
Adenocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Aged , Aged, 80 and over , Base Pair Mismatch , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , DNA Repair , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Smad4 Protein/metabolism
15.
Histopathology ; 49(2): 121-31, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16879389

ABSTRACT

AIM: To establish and explain the pattern of molecular signatures across colorectal polyps. METHODS AND RESULTS: Thirty-two sessile serrated adenomas (SSA), 10 mixed polyps (MP), 15 traditional serrated adenomas (SA), 49 hyperplastic polyps (HP) and 84 adenomas were assessed for mutation of KRAS and BRAF and aberrant expression of p53. The findings were correlated with loss of expression of O-6-methylguanine DNA methyltransferase (MGMT). KRAS mutation occurred more frequently (26.5%) than BRAF mutation (4.8%) in adenomas (P < 0.001) and particularly in adenomas with villous architecture (50%). Loss of expression of MGMT correlated with KRAS mutation in small tubular adenomas (P < 0.04). BRAF mutation was frequent in HPs (67%) and SSAs (81%), while KRAS mutation was infrequent (4% and 3%, respectively). Of MPs and SAs, 72% had either BRAF or KRAS mutation. Aberrant expression of p53 was uncommon overall, but occurred more frequently in MPs and SAs (12%) than adenomas (1%) (P < 0.04) and there was concordant loss of expression of MGMT. CONCLUSIONS: Molecular alterations that are characteristic of the serrated pathway and adenoma-carcinoma sequence can co-occur in a minority of advanced colorectal polyps that then show morphological features of both pathways. These lesions account for only 2% of colorectal polyps, but may be relatively aggressive.


Subject(s)
Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Mutation , Proto-Oncogene Proteins B-raf/genetics , ras Proteins/genetics , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Colonic Polyps/genetics , Colonic Polyps/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA Mutational Analysis , Humans , Immunohistochemistry , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Tumor Suppressor Protein p53/metabolism
16.
J Pathol ; 210(2): 137-40, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16917802

ABSTRACT

Interest in the role of oncogene-induced senescence in tumorigenesis is mounting. Raf-associated senescence in cutaneous nevi has been advanced as an example of this process occurring in the context of a human tumour. In this model, conversion from a senescent nevus to a malignant melanoma is accompanied by loss of expression of p16. Serrated polyps of the colorectum may provide a further example of oncogene-induced senescence. BRAF and KRAS mutation may initiate different pathways of senescence-associated serrated neoplasia in the colorectum, the former linked to CpG island methylator phenotype (CIMP)-high (CIMP1) and microsatellite instability (MSI)-high status and the latter with CIMP-low (CIMP2) and MSI-low status. The role of methylation in both Raf- and Ras-associated pathways is to drive tumorigenesis by silencing pro-apoptotic and cell cycle inhibitory genes. Both pathways are associated with mutation of Ras-induced senescence 1 (RIS1), but the biological role of RIS1 requires further elucidation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cellular Senescence/genetics , Intestinal Polyps/genetics , Colorectal Neoplasms/genetics , Genes, cdc , Humans , Mutation , Oncogenes/genetics
18.
Gut ; 55(10): 1467-74, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16469793

ABSTRACT

BACKGROUND: Hyperplastic polyposis of the colorectum is a precancerous condition that has been linked with DNA methylation. The polyps in this condition have been distinguished from typical small hyperplastic polyps and renamed sessile serrated adenomas. Sessile serrated adenomas also occur sporadically and appear to be indistinguishable from their counterparts in hyperplastic polyposis. AIMS AND METHODS: The existence of distinguishing molecular features was explored in a series of serrated polyps and matched normal mucosa from patients with and without hyperplastic polyposis by assessing mutation of BRAF, DNA methylation in 14 markers (MINTs 1, 2 and 31, p16, MGMT, MLH1, RASSF1, RASSF2, NORE1 (RASSF5), RKIP, MST1, DAPK, FAS, and CHFR), and immunoexpression of MLH1. RESULTS: There was more extensive methylation in sessile serrated adenomas from subjects with hyperplastic polyposis (p<0.0001). A more clearcut difference in patients with hyperplastic polyposis was the finding of extensive DNA methylation in normal mucosa from the proximal colon. CONCLUSIONS: A genetic predisposition may underlie at least some forms of hyperplastic polyposis in which the earliest manifestation may be hypermethylation of multiple gene promoters in normal colorectal mucosa. Additionally, some of the heterogeneity within hyperplastic polyposis may be explained by different propensities for MLH1 inactivation within polyps.


Subject(s)
Colonic Polyps/genetics , DNA Methylation , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Biomarkers, Tumor/metabolism , CpG Islands , DNA, Neoplasm/metabolism , Female , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Male , Middle Aged , MutL Protein Homolog 1 , Nuclear Proteins/metabolism , Polymerase Chain Reaction/methods
19.
J Pathol ; 208(3): 381-7, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16315333

ABSTRACT

The development of colorectal cancer is a major complication for patients with chronic idiopathic colitis. Colitis-associated tumours tend to occur at a younger age and be more aggressive than sporadic colorectal cancers. While we have previously associated the presence of tumour-infiltrating lymphocytes (TILs) and increased apoptosis in sporadic colorectal cancer with high-level microsatellite instability and improved prognosis, little is known of the relationship between these variables in colitis-associated colorectal cancer. The aim of this study was to correlate TILs and tumour cell apoptosis in colitis-associated neoplasms stratified according to microsatellite instability. Twenty tumour and 11 dysplastic samples resected from 21 patients with long-standing colitis were analysed for microsatellite instability at 10 microsatellite markers. TIL distribution (CD3, CD8) and function (granzyme B) were quantified by immunohistochemistry. Neoplastic cell apoptosis was assessed using the M30 CytoDEATH antibody. These findings were compared with 40 microsatellite stable (MSS) sporadic colorectal cancers previously evaluated for TILs and neoplastic apoptosis. Low-level microsatellite instability was found in 1/20 colitis-associated tumours. All other colitis-associated lesions were designated MSS. CD3(+) and CD8(+) TIL counts were significantly higher in colitis-associated lesions compared with MSS sporadic colorectal cancer (p < 0.0001, p = 0.001 respectively). Despite their higher TIL density, colitis-associated tumours were more likely to present late (Dukes' stage C or D) (p = 0.02). Functionally, colitis-associated TILs demonstrated significantly less granzyme B expression compared to sporadic cancers (p = 0.002). The level of tumour cell apoptosis was similar between the two groups (sporadic, 1.53%; colitis cancers, 1.45%). In conclusion, MSS colitis-associated tumours have a higher prevalence of CD3(+)/CD8(+) TILs but no associated increase in tumour cell killing by apoptosis. Unlike cytotoxic T cells in sporadic colorectal cancer, TILs do not appear to enhance the prognosis of colitis-associated colorectal cancer. This may be related to an impairment of granzyme B expression within these lesions.


Subject(s)
Colitis, Ulcerative/immunology , Colorectal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Aged , Apoptosis , CD2 Antigens/immunology , CD8 Antigens/immunology , Case-Control Studies , Colitis, Ulcerative/complications , Colitis, Ulcerative/pathology , Colorectal Neoplasms/complications , Colorectal Neoplasms/pathology , Epithelial Cells/pathology , Female , Granzymes , Humans , Immunohistochemistry/methods , Lymphocytes, Tumor-Infiltrating/enzymology , Male , Microsatellite Repeats , Middle Aged , Neoplasm Staging , Prognosis , Serine Endopeptidases/analysis
20.
Verh Dtsch Ges Pathol ; 90: 18-24, 2006.
Article in English | MEDLINE | ID: mdl-17867575

ABSTRACT

While most colorectal polyps can be classified as either adenomas (AD) or hyperplastic polyps (HP), approximately 5 % have some of the features of these lesions but are distinguishable from both. These lesions include sessile serrated adenoma or polyp (SSP), mixed polyp (MP), and traditional serrated adenoma (SA). These relatively recently described entities account for only about 3%, 1% and 1% of colorectal polyps respectively. Nevertheless, they may serve as the precursor lesions of the subset of colorectal cancer (15-20%) with extensive DNA methylation, mutation of BRAF, and DNA microsatellite instability. This overview summarises the key morphological features of traditional and newer types of colorectal polyps. It also discusses the differing molecular signatures of polyps, focusing on mutation of BRAF and KRAS and alterations of TP53 and the DNA repair genes O-6-Methylguanine DNA Methyltransferase (MGMT) and MLH1. A more detailed description of the features of MPs and SA is then developed and it is shown that these polyps are highly heterogeneous lesions in terms of both morphology and molecular pathology. Finally, a simple working nomenclature for the diagnostic reporting of colorectal polyps is suggested. In this system, MPs and SAs are combined as 'serrated polyps with dysplasia'. It is likely that the recognition and diagnosis of serrated polyps of the colorectum will assume increasing importance in the coming years and that their complex morphology and molecular heterogeneity will present interesting challenges for pathologists, scientists and clinicians.


Subject(s)
Adenoma/classification , Adenoma/pathology , Colorectal Neoplasms/classification , Colorectal Neoplasms/pathology , Colonic Polyps/pathology , Disease Progression , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...