Next Article in Journal
Behavioral Impairment and Oxidative Damage Induced by Chronic Application of Nonylphenol
Next Article in Special Issue
IL8 and Cathepsin B as Melanoma Serum Biomarkers
Previous Article in Journal
Toxic Compound, Anti-Nutritional Factors and Functional Properties of Protein Isolated from Detoxified Jatropha curcas Seed Cake
Previous Article in Special Issue
Biomarkers in Rare Disorders: The Experience with Spinal Muscular Atrophy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cell Surface Markers in Colorectal Cancer Prognosis

School of Molecular Bioscience, University of Sydney, Sydney, NSW 2006, Australia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2011, 12(1), 78-113; https://doi.org/10.3390/ijms12010078
Submission received: 30 November 2010 / Revised: 16 December 2010 / Accepted: 20 December 2010 / Published: 28 December 2010
(This article belongs to the Special Issue Biomarkers)

Abstract

:
The classification of colorectal cancers (CRC) is currently based largely on histologically determined tumour characteristics, such as differentiation status and tumour stage, i.e., depth of tumour invasion, involvement of regional lymph nodes and the occurrence of metastatic spread to other organs. These are the conventional prognostic factors for patient survival and often determine the requirement for adjuvant therapy after surgical resection of the primary tumour. However, patients with the same CRC stage can have very different disease-related outcomes. For some, surgical removal of early-stage tumours leads to full recovery, while for others, disease recurrence and metastasis may occur regardless of adjuvant therapy. It is therefore important to understand the molecular processes that lead to disease progression and metastasis and to find more reliable prognostic markers and novel targets for therapy. This review focuses on cell surface proteins that correlate with tumour progression, metastasis and patient outcome, and discusses some of the challenges in finding prognostic protein markers in CRC.

Graphical Abstract

1. Introduction

Colorectal cancer (CRC) is a cancer of epithelial origin, localized to the large intestine and rectum. It is one of the most common cancers and will occur at some stage in approximately 5% of the population of the western world. The conventional prognostic factors for patient survival are histologic tumour grade (differentiation) and tumour stage (TNM, tumors/nodes/metastases, stages I–IV) [1,2], which is based on depth of tumour invasion, involvement of regional lymph nodes and metastatic spread to other organs [3]. If metastasis has occurred, patient 5-year survival after surgery falls dramatically from 90% to less than 10% [4]. It is therefore important to increase our understanding of the molecular changes leading to development, spread and metastasis of CRC and to identify potentially prognostic and predictive markers for the disease.
Despite the discovery of a range of intra- and extra-cellular protein biomarkers for CRC and continuing efforts to discover potentially prognostic and predictive markers using various approaches [512], the translation of this increasing volume of differential proteomic data into patient care remains a major challenge. This is partly due to the heterogeneous nature of CRC [13,14] and the complexity of processes involved in its development and spread, but also to challenges presented in detecting and characterising glycoproteins and low abundance proteins [15,16] in complex mixtures. In addition, validation of candidate proteins as prognostic markers is time-consuming and expensive, and recent critical reviews of differentially expressed proteins from comparative proteomic studies have suggested that some reported proteins may be associated with general cellular stress responses, rather than being disease-specific biomarkers [17,18]. The development of a reliable assay for determining prognosis and guiding post-surgical therapy therefore remains elusive.
The metastatic potential of CRC may be already encoded in the primary tumour [19], and molecular staging using a classifier based on 43 genes has identified patient prognosis more accurately than the traditional clinical staging, particularly for intermediate stage II and III patients [20]. It may be possible to identify disease signatures based on protein expression profiles in primary CRC tissues that reflect potential for disease progression and metastasis, and subsequently use these signatures to predict patient recovery and survival after surgical removal of the primary CRC tissue. With this goal in mind, we have developed a 122-antibody microarray (DotScan™ CRC microarray; Medsaic Pty Ltd, NSW, Australia) for immunophenotyping live cells from fresh disaggregated CRC tissues [21,22]. Microarrays prepared as 10 nl antibody dots on nitrocellulose-coated microscope slides (FAST; Grace Bio-labs, Bend, OR, USA) bind cells with corresponding surface molecules, producing a binding pattern that reflects the surface immunophenotype of the mixed cell population. Bound cells are then fixed to the microarray with formalin, and specific sub-populations are profiled by multiplexing with mixtures of soluble fluorescently-labelled antibodies. Hierarchical clustering of binding patterns for these sub-populations of cells yields patient clusters that may correlate with disease stage, tumour invasiveness, differentiation, drug susceptibility or patient outcome. This antibody-based multiple marker approach has already been validated for the classification of a range of common human leukaemias and lymphomas [23].
The evolution of CRC from an adenomatous polyp to metastatic disease is dependent not only on progressive accumulation of genetic and epigenetic abnormalities [24], but also on the complex interactions of different sub-populations of cells within the tumour microenvironment (cancer cells, normal stromal cells, infiltrating leukocytes and the soluble factors they produce) [25]. The importance of cell surface molecules for cell-cell adhesion and communication between cancer cells and non-malignant cells has been recently reviewed [26,27], and the significance of signalling pathways involved in CRC progression as a result of these interactions has also been described [2832]. Since many drugs used in cancer therapy target cell membrane proteins [33], this review seeks to summarise current knowledge on cell surface molecules that may serve as therapeutic targets, have prognostic significance or provide important information about the invasive, metastatic and proliferative potential of cancers of the large intestine and rectum. Antibodies to some of these proteins have been incorporated into the current DotScan™ CRC antibody microarray; others could be added in the future. Readers are also referred to several previous reviews on markers of metastasis and prognosis in CRC [34,35].

2. Tumour Stroma

Recently, the importance of the tumour microenvironment for the development, promotion and spread of cancer has become clear [36], and the potential for using tumour stroma-derived biomarkers in prognostication and therapy of cancer patients has been realised [37]. The stroma consists of the non-malignant cells of the tumour—fibroblasts, infiltrating leukocytes and cells of the blood vessels. However, despite experimental evidence for the importance of tumour stroma for cancer development and progression, clinical applications for stromal biomarkers remain limited. Down-regulation of TGF-beta receptor 2 (TGF-β-R2) in tumour-associated stroma correlates with a worse prognosis [38], while expression of platelet-derived growth factor receptor (PDGFR) on stromal cells correlates directly with metastatic potential and advanced-stage disease in CRC [39,40].
Stromal CXCR4 (CD184) and CXCL12 expression is associated with distant recurrence and poor prognosis in rectal cancer after chemotherapy [41]. Patients whose colon tumours have high levels of stromal fibroblast activation protein (FAP) are more likely to have aggressive disease progression and potential development of metastases or recurrence [42]. Urokinase receptor (uPAR; CD87) expression in stromal fibroblasts has been implicated in the occurrence of haematogenous metastasis (dissemination via the blood) of CRC [43]. The presence of high levels of stroma and down-regulation of Deleted in Pancreatic Cancer 4 (SMAD4) predict worse survival for early-stage CRC patients [44]. Expression of CD10 (a cell surface metalloprotease) on stromal cells adjacent to cancer cells within the area of invasive growth is significantly higher in invasive CRC than in non-invasive CRC or adenomas [45]. The stromal expression of CD10 also appears to be significantly associated with abnormal accumulation of nuclear p53 in tumour cells and a larger tumour size, suggesting that CD10 expression may contribute to tumour invasion and possibly facilitate metastasis [45].
Thrombospondin-1 (TSP-1), mainly localised in fibroblasts of the CRC stroma [46], has been considered as an important negative-regulator of tumour angiogenesis. TSP-1 expression in primary CRC tumours has been correlated inversely with metastatic potential and prognosis [4751]. In CRC liver metastases, however, TSP-1 expression has been linked to significantly poorer survival, suggesting that TSP-1 in liver metastases may have an alternative mode of action, facilitating tumour invasion rather than acting as an anti-angiogenic growth factor [51].

3. Inflammation and Infiltrating Leukocytes

Inflammatory reactions in tumours can have a positive influence on survival [52,53] or alternatively can be associated with development of metastases and disease progression [5456]. The role of tumour-associated macrophages (TAM) in tumourigenesis is complex because TAMs can either prevent or promote tumour development. While Forssell et al. [52] showed that a dense TAM infiltration at the tumour front positively influenced prognosis in colon cancer, strong pro-tumourigenic effects of TAMs in CRC have also been reported [5457]. It has been suggested that the balance between pro- and anti-tumourigenic properties of TAMs may depend on their interaction with cancer cells, other stromal cells, and the tumour microenvironment [54] or be influenced by the degree of cell-cell contact [52].
The presence of high numbers of mature CD208+ infiltrating dendritic cells (DC) in the tumour epithelium has been associated with shorter overall patient survival, while patients with high numbers of CD1a+ DCs in the advancing margin of the tumour appear to have a shorter disease-free survival [58]. These findings are controversial, however, as a strong correlation has also been found between the presence of DCs together with high density T-cells and no detectable metastases [59] and better patient survival [60].
The presence of large numbers of tumour infiltrating lymphocytes (TILs) at the invasive tumour front has generally been associated with a good prognosis for CRC patients [6164]. Patients with high levels of CD3+, CD8+, CD25+, CD45RO+ or CD95L(CD154)+ TILs appear to have a better clinical course [6569] than those with low levels, particularly in early disease stages [70]. The importance of the intra-tumoural location, immunophenotype and density of these infiltrates has been demonstrated [7173].
It has been shown that CD3+ TILs located in the tumour stroma are not as strong predictors of prognosis as T-cells located within cancer cell nests (clusters), consistent with the requirement for direct contact between target and effector cells [7174]. In addition, although the presence of high density CD3+ TILS in primary tumours has been associated with good prognosis in patients with lymph node-negative CRC, there was no significant correlation between high density CD3+ TILs and absence of post-surgical metastases in patients with lymph node-positive CRC [75].
Patients with T-cells showing low CD4+/CD8+ ratios have a better clinical course than those with high ratios [59,76]. The infiltration of cancer cell nests by CD8+ T-cells has been associated with better survival [67], supporting the importance of activated cytotoxic CD8+ T-cells in the anti-tumour response to CRC [77,78].
The better overall survival of CRC patients with high infiltrates of CD3+ (total) and CD8+ (cytotoxic) T-cells within cancer cell nests, was recently confirmed [70]. Interestingly, however, this correlation was not significant for the rectal cancer patient sub-group when it was analysed separately from the colon cancer group. Furthermore, in patients whose tumours were positive for microsatellite instability (MSI+), a hallmark of DNA mismatch repair (MMR) deficiency [79], no association was found between high levels of activated cytotoxic T-cells and a favourable prognosis. Although these findings are in agreement with those of several other groups, they are in conflict with a number of studies in which high infiltrates of CD8+ TILs correlated with good prognosis regardless of MSI-status [70].
Marked infiltration of CD8+ and CD57+ natural killer (NK) or NK-like T-cells in the advancing tumour margin has also been associated with longer disease-free survival, and this was most evident in MSI+ tumours [80]. Metastatic spread of CRC is associated with decreased NK-cell activity [81], but it remains speculative whether decreased NK-cell activity precedes the development of metastases and may therefore be prognostic.
Koch et al. [82] reported strong and significant enrichment for CD4+ T helper cells in CRC compared to corresponding normal mucosa, i.e., decreased proportions of CD8+ cells in total T-cell populations of CRC. However, a significantly higher proportion of the CD8+ TILs in CRC expressed markers of activation (CD69+) and cytotoxic activity (CD107a+) compared with normal mucosa. Significantly, the proportion of activated CD8+ TILs decreased with tumour stage [82].
If a tumour exhibits a high T-cell infiltrate, the prognosis for patients with strong tumour expression of major histocompatibility antigen class I (MHC 1) is better than for those who have weak MHC 1 expression [74], because the immune response mounted against the tumour cells is weaker in these patients.
TILs recovered from primary tumours showing no sign of metastasis differ immunophenotypically from those showing early signs of tumour metastasis (presence of vascular emboli, lymphatic invasion and perineural infiltration). Using flow cytometry, Pages et al. [68] found that TILs from tumours with no sign of metastasis showed higher expression of markers of cell adhesion (CD62L, CCR7, CD103, CD49d and CXCR3), activation (HLA-DR, CD98, CD80, CD86 and CD134) and differentiation (CD45RO, CD45RA, CD27, CD28, CCR7 and CD127). Primary CRC tumours without early signs of metastatic invasion such as vascular emboli (i.e., infiltration of cancer tissue into blood vessels) presented with significant increases in T-cell sub-populations from early memory (CD45RO+CCR7- CD28+CD27+) to effector memory CD8+ T-cells (CD45RO+CCR7-CD28+CD27-). Using immunohistochemistry-based tissue microarray analysis of 415 tumours, they confirmed that a high density of memory T-cells (CD3+CD45RO+) in the primary tumour correlated with the absence of early signs of metastatic invasion, confirming previous findings [83]. In a comparison of patients with high T-cell infiltration, primary tumours from patients with metastases had significantly decreased densities of CD8+ T-cells and effector memory T-cells (CD27-CD45RA-) than patients without metastases [59].
The surface antigen expression of total TILs differs from that of T-cells in peripheral blood, with markedly lower expression of CD29, CD49d and CD49f, and slightly higher expression of CD49a and CD49b [84]. CD11a and CD18 are reduced on CD8+, but not CD4+, sub-populations [84]. TILs also differ from lymphocytes within normal colon interstitium by lacking CD28, CD56, CD98 and CD154 [85] and showing altered expression of CD3, CD29, LFA-1 (CD11a/CD18) and LFA-3 (CD58) [86].
The function and relative importance of different CD4+ sub-populations in CRC is still only partially understood. Although CD4+ TILs are observed in CRC [59,80,87], the density of this subInt. population has shown little prognostic value in CRC to date, and is not significantly different in primary tumours of patients with metastatic or non-metastatic disease [59]. A high density of CD4+ TILs in CRC liver metastases has correlated with poorer patient outcome [63].
Recent attention has focussed on CD4+CD25+ regulatory T-cells (Treg), which are characterised by expression of nuclear transcription factor forkhead box P3 (FoxP3). The CD3+/FOXP3+ cell ratio [88] and the CD8+/FOXP3+ cell ratio [89] are both predictive markers for disease-free survival time and overall survival time in patients with CRC, supporting the down-modulatory role of Treg in the presence of protective cytotoxic T-cells. Controversially, the presence of a high density of FoxP3+ Treg in CRC has also been associated with improved survival in stage II and stage III CRC patients [66,83] and in relapsed CRC patients undergoing chemotherapy or chemoimmunotherapy [90]. Although this result is unexpected and counterintuitive, it has been suggested that the increase in Tregs in tumour tissue may represent a feedback response to a pre-existing cancer cell-directed immune response [90]. Frey et al. [91] found that high FOXP3(+) Treg density was associated with early tumour stage and independently predicted improved disease-specific survival in MMR-proficient CRCs, but not in MMR-deficient CRCs, though high Treg density correlated with absence of lymph node involvement and vascular invasion in the latter. They suggested that the role of Treg may differ according to the clinical stage and the MMR status of CRC.
We have prepared live cell suspensions from enzymically disaggregated primary tumour tissue from CRC patients and corresponding normal mucosa. DotScan™ CRC antibody microarrays and fluorescence multiplexing were used to profile the cells, and the CD3+ subset showed differential expression of HLA-DR, TCRα/β, CD49d, CD52, CD49e, CD5, CD95, CD28, CD38 and CD71 in descending order of difference [21].

4. Cancer Cells

4.1. Tetraspanins and Potential Tetraspanin-Associated Proteins

Tetraspanins are small transmembrane proteins, such as CD9, CD37, CD53, CD63, CD81, CD82, CD151 and tetraspanin 8, that are involved in a multitude of biological processes, including cell-cell adhesion, metastasis suppression and tumour progression [9296]. Clinical studies have established a link between tetraspanin levels and prognosis and/or metastasis in cancer. The tetraspanins CD82 and CD9 mostly suppress tumour progression; their expression is often reduced in late-stage human tumours and their down-regulation during CRC progression is associated with poor prognosis [97]. CD151 and tetraspanin 8 support tumour progression; increased CD151 is an indicator of poor prognosis.
Tetraspanins interact with each other and other proteins that are required for their function, to form complexes in tetraspanin-enriched membrane domains (TEM) [95]. This has been referred to as the ‘tetraspanin web’ [93,94]. The most prominent non-tetraspanin partners are integrins, e.g., β1 (CD29), α3 (CD49c), α4 (CD49d), α6 (CD49f) and β4 (CD104); matrix metalloproteinases (MMP), e.g., MT1- MMP (MMP14); members of the Ig superfamily, e.g., ICAM-1 (CD54) and VCAM-1 (CD106); as well as CD26, CD44, EpCAM (CD326), E-cadherin (CD324), beta-catenin (β-catenin) and ADAM10. Some of these molecules have been implicated in tumour metastasis and poor patient prognosis.
Down-regulation of E-cadherin, the prime mediator of epithelial cell-cell adhesion [98], correlates with a strong invasive potential and poor prognosis in CRC [99101]. High CRC cell staining for β-catenin, a binding partner of E-cadherin [102], is independently associated with better patient survival [103]. In contrast, membranous expression of β-catenin, together with low E-cadherin, is associated with CRC tumour progression, and may be an independent prognostic factor for poor patient outcome [104].
Absence of CD49c correlates with reduced overall and disease-free survival compared to patients with CD49c+ CRC [105]. CD29 is commonly down-regulated in CRC compared to normal intestinal mucosa [22,106,107], and this has been correlated with invasion and metastasis [108]. However, there is little evidence for correlation between CD29 levels and patient outcome, possibly because up-regulation of CD29 has also been correlated with lymph node metastasis and depth of invasion in CRC [106]. The association of CD104 with members of the tetraspanin web also appears to support tumour cell motility and liver metastasis [109].
CD44 over-expression in CRC is generally considered to be an unfavourable prognostic factor for overall survival, though this may depend on the isoform. CD44 variant 6 (CD44v6) expression correlates with poor prognosis [110112]. However, loss of CD44 significantly correlates with poor survival when associated with a corresponding loss in E-cadherin, especially in Stage II CRC, suggesting that CD44 and E-cadherin may have an inter-dependent role in suppressing invasion and metastasis [113]. The worst prognosis was associated with low E-cadherin coupled with high MT1-MMP (MMP14) [113], itself associated with tumour invasiveness in CRC [114116].
Low numbers of CD54-expressing tumour cells may be associated with a shorter disease-free survival [117]. The prognosis of patients with CD54-negative tumours is significantly poorer than those with CD54-positive tumours, suggesting that low CD54 expression is closely associated with metastasis and may be a useful indicator of prognosis [118]. Infiltration by TILs has been more frequently observed in the CD54-positive than in CD54 negative tumours.

4.2. Epithelial-Specific Cell Adhesion Activation Molecule (EpCAM, TROP-1, CD326)

EpCAM is a pan-epithelial differentiation antigen which promotes cell adhesion [119]. In normal epithelial tissues, increased EpCAM expression is associated with enhanced proliferation and a lower differentiation grade of epithelial cells. In the proliferative phase, EpCAM expression is associated with epithelial tissue remodelling. After cell proliferation, EpCAM expression declines and cellular differentiation commences [120].
The regulation of EpCAM in tumour invasion and metastasis has previously been reviewed [121125]. In carcinogenesis, EpCAM over-expression is present during the early stages, but dramatically decreases during malignant transformation and progression. Over-expression of EpCAM reportedly correlates with biological aggressiveness and poor prognosis in CRC [126], possibly by promoting tumour immune evasion by strongly favouring T-helper cell type 2 (Th2) development that promotes growth of EpCAM-expressing tumors [127]. However, a reduction in surface expression of EpCAM has also been associated with aggressive cancers and poor prognosis in CRC [120]. The hypothesis that loss of EpCAM expression may be involved in cancer metastasis is further supported by the finding that EpCAM protein expression on circulating tumour cells (CTC) is reduced compared with primary and metastatic tumours [128].
Using a monoclonal antibody to detect an extra-cellular epitope of EpCAM in rectal tumours, Gosens et al. [120] found that decreased membranous EpCAM staining in the “budding” cells at the invasive tumour front was always accompanied by a focally infiltrating growth pattern, correlated with a significantly higher risk of local recurrence and indicated an elevated risk of distant recurrence. Importantly, loss of membranous EpCAM coincided with increasing cytoplasmic staining at the tumour front (detected by polyclonal antibody), suggesting altered cellular localisation due to abnormal post-translational processing of EpCAM rather than reduced expression of this antigen. An alternative view is that regulated intra-membrane proteolysis results in the shedding of EpCAM’s ectodomain and the nuclear translocation of its intracellular domain, resulting in mitogenic signal transduction [129].
This decline in membranous EpCAM expression at the tumour front was associated with nuclear β-catenin localization [120], which is associated with reduced cell–cell adhesion and increased migratory potential [130]. It did not correlate with disease stage, but did correlate with lower tumour differentiation grade, and was strongly prognostic [120]. It was predominantly observed in budding tumour cells or clusters. Tumour budding is a term used to describe the appearance of foci of de-differentiated cancer cells at the invasive tumour front of differentiated adenocarcinomas, i.e., epithelial-to-mesenchymal transition (EMT) [131133], and is frequently linked to high-grade tumours, lymph node positivity, vascular and lymphatic invasion and local and distant metastases [134].
Decreased membranous EpCAM staining throughout the tumour rather than just at the invasive margin also correlates with increased risk of local recurrence, but is not indicative of distant recurrence [120]. Interestingly, increased EpCAM expression has been linked to CRC progression when it is associated with claudin-7 and recruited into TEM where it complexes with CD44v6 and tetraspanin 8 [135,136]. This complex formation may facilitate metastasis by preventing EpCAM-mediated cell-cell adhesion and promoting migration, proliferation, apoptosis resistance and tumourigenicity.

4.3. Mucins, Carbohydrates, Lectins

Cancer cells, especially adenocarcinomas, tend to over-express mucins or express aberrant forms of mucins, the function and significance of which have been reviewed [137139]. In CRC, high mucin content has been associated with more aggressive phenotype and poorer prognosis [140]. CD227 (MUC1), a membrane-associated mucin that displays altered O-glycosylation patterns in carcinomas, presents as an independent prognostic factor of CRC [141,142]. MUC1-Tn epitope becomes exposed during malignant transformation to adenocarcinoma and is associated with poor clinical outcome [143]. CD175s (sialosyl-Tn) can modulate a malignant phenotype inducing a more aggressive cell behaviour, and is associated with poor clinical outcome [144]. MUC2 has been associated with less aggressive tumour phenotypes and improved survival in some studies [61,145], although reverse findings have also been reported [146].
CD15s (Sialyl-Lewisx, sLex) has an important role in defining the invasion and metastasis of human CRC; high level expression has been significantly correlated with metachronous metastases after surgery and poor prognosis [147150]. The incidence of CD15s positive primary tumours was correlated with the depth of tumour invasion, lymph node metastasis, lymphatic invasion, and the disease stage [150]; but Baldus et al. [141] did not find CD15s to be an independent prognostic factor of CRC.
Sialyl-Lewisa (sLea; CA 19-9), a mucin epitope whose levels in CRC correlate with overall survival and disease free survival, may serve as an indicator of metastatic potential [141,151]. It is believed that tumour metastasis and cancer cell arrest in the vasculature is facilitated through the tethering of circulating cancer cells to vascular endothelium through the interaction of molecules such as CD15s, CA 19-9 and CD44 variants on the tumour cell surface and selectins (CD62-P, -L and –E) expressed on endothelial cells, neutrophils, monocytes, NK cells or platelets [152155].
Galectin-3, a lectin, is involved in neoplastic transformation and tumour progression and has demonstrated significant prognostic value, particularly in stages I and II CRC [141,156,157]. Low levels of galectin-3 expression in the tumour epithelium are associated with significantly better prognoses than those characterized by high levels [158]. Galectin-3 up-regulates MUC-2 expression [159], and a functional link has been demonstrated between galectin-3, MUC-2 and metastasis in animal models [160,161]. However, these findings contradict other reports of an inverse relationship between MUC2 expression and tumour aggressiveness [61,162].

4.4. Carcinoembryonic Antigen (CEA) Family

The best known member of this group is carcinoembryonic antigen (CEA; CD66e), which is up-regulated relative to normal intestinal mucosa in 94% of CRC samples [163]. However, the significance of CEA over-expression in CRC as a prognostic marker remains controversial. Despite several reports supporting a possible correlation between CEA expression in CRC and tumour spread or patient survival [164166], no definitive correlation has been established [163,167] and CEA has found little utility as a prognostic marker. However, the expression of CEA by disseminated tumour cells isolated from intra-peritoneal lavage was found to strongly correlate with advanced tumour stages and an extremely poor prognosis [168], consistent with the suggestion that CEA may enhance metastasis by functioning as an attachment factor for disseminated CRC cells [169].
CD66a (CEACAM1) and CD66c (CEACAM6) are also over-expressed in 58% and 55% of patients, respectively [163]. While no prognostic significance has been associated with CD66a, the over-expression of CD66c was found to be significantly associated with worse prognosis in a group of patients who received peri-operative FU-based chemotherapy [163]. CD66c has also demonstrated a role in tumour cell migration, invasion and adhesion, and formation of distant metastases [170]. CEA-related cell adhesion molecule 7 (CEACAM-7) is down-regulated in rectal cancers relative to normal mucosa and independently predicts recurrence-free survival for stage II rectal cancers [171].

4.5. Annexins

Annexins A1, A2, A4 and A11 are expressed at higher levels in CRC than in normal colon, and have been implicated in tumour development and progression [172]. Over-expression of Annexins A4 and A11 [172], Annexin A2 (Annexin II) [173] and Annexin A5 [174] may serve as markers of poor prognosis in patients with CRC.

4.6. Claudins

Claudin-1 and Claudin-4, transmembrane proteins important for tight junction formation in normal mucosal epithelial cells [175], are both up-regulated in CRC compared to normal mucosa, and this up-regulation is associated with down-regulation of E-cadherin and significant disorganisation of tight junction fibrils [176]. The over-expression of claudin-1 and -4 occurs not only in the plasma membrane, but also in the cytoplasm [176], and its function in CRC is not yet defined. However, loss of claudin-1 expression is a strong predictor of disease recurrence and poor patient survival in stage II CRC [177], while a decrease in claudin-4 at the invasive front in CRC has been associated with cancer invasion and metastasis [178].

4.7. Chemokine Receptors

Expression of the chemokine receptors CXCR3 [179,180] and CXCR4 (CD184) [181,182] has been associated with metastasis and poorer patient prognosis in CRC. Stroma-derived chemokines such as stromal cell derived factor-1 (a ligand of CXCR-4), may induce growth and metastatic progression of tumours by modulating proliferation, survival and homing, through chemokine receptors expressed on cancer initiating cells [183]. In contrast, chemokine receptor 5 (CCR5) expression has been associated with non-metastatic CRC and increased CD8+ T-cell infiltration [184].

4.8. Growth Factor Receptors

Epidermal growth factor receptor (EGFR) plays an important role in the biology of CRC [185] and is being used as a target for therapeutic antibodies in CRC patients with unmutated KRAS genes, but not in those with KRAS gene mutations which cause resistance [186,187]. However, despite reports indicating its value as a prognostic marker [188,189], its potential has not been realised [190]. Although patients with EGFR expression have a higher risk for disease recurrence compared to those with EGFR negative tumours, there appears to be no relationship between EGFR expression and overall survival [191,192].
A significant correlation has been demonstrated between insulin growth factor receptor (IGFR) expression levels (membrane plus cytoplasmic) and lymph node metastasis in CRC patients, especially when analysed in combination with vascular endothelial growth factor (VEGF) and VEGF-C [193].
Although cytoplasmic over-expression of human epidermal growth factor receptor 2 (CD340, HER-2/neu, ErbB2) is reported to be an independent prognostic indicator in CRC [194], membranous expression of this protein shows no prognostic significance [195]. Membranous expression of receptor tyrosine-protein kinase ErbB4, however, has recently been reported as an independent prognostic factor for recurrence in CRC patients after radical surgery [196].
Hepatocyte growth factor receptor (Met) expression correlates with poor overall survival in advanced stages of CRC [197]. In stage I and II patients, however, membranous Met alone does not appear to be a significant predictor of patient outcome, although the membranous to cytoplasmic Met ratio correlates with survival [198].

4.9. Death Receptors (DR) and Ligands

The roles of death receptors and their ligands in cancer onset, progression and therapy have been reviewed [199]. Low expression of death receptor 4 (DR4; TRAILR1; CD261) has been linked to poor prognosis in CRC [200], consistent with a tumour suppressor role for DR4 and its ligand, tumour necrosis factor related apoptosis-inducing ligand (TRAIL). However, TRAIL has shown no prognostic significance in several studies [201,202] and has even correlated with worse overall survival in stage II and III CRC patients [203]. Interestingly, high DR4 expression has also been associated with worse disease-free and overall survival in stage III adjuvant-treated CRC patients [202]. These findings are consistent with the proposed existence of an alternative TRAIL signalling pathway in TRAIL-resistant cells, which leads to cancer promotion rather than inhibition [204,205]. The prognostic significance of the up- or down- regulation of these molecules may therefore vary in different patient sub-groups.
Loss of CD95 (Fas) in tumour cells is related to adverse prognosis and maybe an independent prognostic factor in CRC [206]. Fas-ligand (CD178; CD95L; FasL) expression, however, correlates with lymph node involvement and distant metastases in CRC [207]. It has been suggested that the up-regulation of FasL in CRC cells can induce apoptosis of Fas-expressing T lymphocytes in the tumour [208]. Low Fas and high FasL maybe a useful indicator of lymph node metastases and poor prognosis in CRC [209].

4.10. Major Histocompatibility Complex (MHC)

The major histocompatibility complex (MHC) codes for human leukocyte antigens Class I (HLA-A, -B, -C) and Class II (HLA-DR, -DQ, -DP). Better prognosis correlates with the down-regulation of Class I antigens [210,211] and strong HLA-DR expression [212,213] on CRC cells.

4.11. Other Protein Markers in CRC

The A33 antigen is found in 95% of primary and metastatic CRC and also in the normal intestinal mucosa [214]. A33 is a promising target for immunotherapy [215,216] and radioimmunotherapy [217,218] of CRC, but A33 levels have not been associated with prognosis.
CD10 expression in CRC correlates with liver metastasis [219221] and with the development and progression of CRC [162], but its prognostic significance in CRC has not been confirmed [222]. CD13 (aminopeptidase N; APN) is involved in cell motility and angiogenesis, and CD13 expression may be a useful indicator of a poor prognosis for node-positive patients with colon cancer [223]. Membrane complement resistance factors CD55 and CD59 have been reported as markers of tumour aggression, and have been associated with reduced survival in CRC patients [224,225]. CD55 acts as a cellular ligand for CD97; strong CD97 expression by tumour cells at the invasive front has been correlated with a higher clinical stage and increased lymph vessel invasion compared to cases with uniform CD97 staining throughout the tumour [226].
Membranous expression of the cell adhesion molecule CD166 (ALCAM), a member of the immunoglobulin superfamily, has been correlated with significantly shorter survival time in CRC patients [227]. Cell adhesion molecule CD171 (L1) expression is also associated with tumour progression and poor prognosis in CRC patients [99,228], and has been used, in combination with E-cadherin and membranous β-catenin, to identify stage II patients who may benefit from adjuvant chemotherapy [104].
Deleted in colorectal cancer protein (DCC) has been proposed as a putative tumour suppressor, and loss of DCC expression has been associated with poor prognosis and risk of metastasis [229]. Expression of DCC is a strong positive predictive factor for survival in CRC [230,231].
Several novel membrane proteins have recently been identified in CRC. ATP-binding cassette sub-family G member 5 (ABCG5) is a membrane protein involved in efflux transport of cholesterol. ABCG5 expression in tumour buds has recently been associated with significantly poorer prognosis in node-negative CRC patients [134]. ATP11A is an integral membrane ATPase, probably involved with the transport of ions such as calcium across membranes. Patients with high ATP11A expression have shown poorer disease-free survival compared to those with low expression, indicating that an increase in ATP11A expression may be an independent predictor of metastasis in CRC patients [232].

5. Colorectal Cancer Stem Cells (CSC)

Cancer stem cells have been identified in several human malignancies, including CRC, and are believed to drive tumour initiation, progression and metastasis [233238]. Tumour buds, discussed above, appear to have many features of CSC, and the possibility that a sub-population of tumour buds may represent malignant stem cells is an open question requiring further investigation [133].
The presence of a sub-population of CD26+ CSC in primary CRC tumours was found to predict distant metastases on follow-up [239]. These cells were associated with enhanced invasiveness and chemoresistance. High EpCAM expression was also associated with oncogenic potential in CSC [122], and CD166 (ALCAM) was found to be differentially expressed on EpCAMhigh/CD44+ CSC [240]. Other CSC markers, CD133 and CD24, correlated with invasiveness and differentiation in CRC, while CD44 expression was related to tumour burden [241]. No correlation was found between the expression of these three markers and patient survival [241], but others have shown that the combined evaluation of CD133, CD166 and CD44 could be used to identify high and low survival among stage I and II CRC patients [242]. Although CD133+ cells have demonstrated CRC initiating potential [243], CD133 expression may not be restricted to cancer-initiating cells [244]. In addition, highly metastatic CD133- sub-populations of cancer-initiating cells have been identified in a human CRC xenograft model, suggesting that CD133+ tumour cells might give rise to a more aggressive CD133- subset during the metastatic transition [244].
CD200 (OX2) is a membrane glycoprotein that induces an immuno-regulatory signal through its receptor (CD200R), leading to the suppression of T-cell-mediated immune responses. CD200 has been associated with tumour progression and poor prognosis in several cancers [245,246] and is over-expressed in CRC with markers for CSC [247], suggesting that tumour-initiating CSC may evade immune system detection through CD200-induced suppression of T-cell-mediated immune responses.

6. CRC Translational Proteomics Research

A number of proteomic studies on CRC tissues have identified differentially expressed proteins from whole tissue samples of CRC and paired normal mucosa [8,16,248252]. In other studies, enrichment for specific sub-populations of cells has been performed using techniques such as laser micro-dissection [253,254], fluorescence activated cell sorting (FACS) [240] or antibody-conjugated magnetic beads [240,255]. Interpretation of results is complicated by the fact that different methods have been employed for sample preparation, antigen detection and protein quantification. For antibody-based detection methods, the choice of monoclonal antibody is important, as differences in staining may reflect changes in epitope structure due to mutations, alternative splicing or post translational modifications, rather than up- or down-regulation of protein expression. In addition, some changes in protein activities during disease progression may correlate with altered sub-cellular localisation of proteins [120]. Thus the use of different methods of protein quantification may lead to conflicting results.
Methods used for membrane proteomics in cancer biomarker discovery have been previously reviewed [26]. Due to the hydrophobicity of membrane glycoproteins and their relatively low levels compared to soluble intracellular proteins [256], the identification and validation of novel differentially expressed surface markers in CRC has been limited. Membrane fractions from patient CRCs have been analysed using two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) saturation labelling followed by MALDI-TOF-TOF mass spectrometry, with identification of 23 proteins, many classified as membrane or membrane-associated [5]. Others have been identified from membrane CRC fractions by iTRAQ [248]. Although approaches such as 2D-DIGE and mass spectrometry have led to the discovery of a number of differentially expressed proteins in CRC [58,1012,257,258], the prognostic significance of these molecules and their contribution to cancer growth and metastasis has not always been easy to establish. Hence very few of these proteins have found clinical utility either as prognostic markers or therapeutic targets.
Proteomic methodologies and their application in CRC research have been reviewed, and the limitations of traditional approaches for cancer biomarker marker discovery and future challenges have been discussed [259261]. It has been suggested that multiple markers will be required to enable reliable sub-classification of cancers [262264]. The use of immunohistochemistry for analysis of CRC tissue sections for multiple potentially prognostic/predictive markers [141,265,266] has the advantage of revealing antigen location; however the method is relatively low through-put, expensive, labour-intensive, and quantification is difficult. The use of tissue microarrays enables analysis of a large number of samples simultaneously, but the number of antigens that can be detected in a single assay is limited [267].
The DotScan™ CRC antibody microarray allows rapid immunophenotyping for 122 surface antigens on a population of disaggregated live tumour cells in suspension, requiring 4 × 106 cells per assay. Density of cell binding per antibody dot depends on the proportion of positive cells in the population, level of antigen expression and affinity of the antibody. Figure 1 shows optical and fluorescent binding profiles for a CRC sample from a stage II patient. The optical binding pattern reflects the immunophenotype of the mixed cell population, while the CD3-PE and EpCAM-Alexa Fluor 647 staining detect T-cells and cancer/epithelial cells, respectively. The limit of detection is approximately 1 cell in 10 for the optical scans, while the sensitivity of fluorescent scans is four times higher. We have demonstrated sub-types of CRC based on hierarchical clustering of microarray patterns [22] that may provide prognostic information when data are retrospectively analysed with patient outcomes. This study is ongoing. With the addition of antibodies against newly discovered markers of CRC, this method could be used to correlate cell binding densities with disease stage, invasion, metastasis and patient outcome, and also for finding multiple-marker signatures allowing sub-classification of CRC.
Cell surface proteins are logical candidates as biomarkers and therapeutic targets; they enable a cell population to interact with other cells and soluble factors in the tumour microenvironment and reflect important genomic, epigenetic, transcriptional and translational changes during disease progression. As an alternative to the use of viable cells for analysis with the DotScan CRC antibody microarray, tumour cell-derived membrane fragments, microvesicles or microparticles could be used, as demonstrated with plasma-derived microparticles from patients with cardiovascular disease [268]. This alternative approach may facilitate the detection of potential prognostic biomarkers shed from cells located at the invasive front. Samples of the invasive front are required for routine pathology analysis and are not readily available for research purposes.
Zlobec and Lugli [133] have described the invasive front of CRC as a dynamic interface between pro- and anti-tumour factors. Tumour budding at the invasive front promotes progression and dissemination of CRC cells into the vasculature and lymph nodes, while infiltrating immune cells, particularly cytotoxic T-cells, mount an immune response. The CD8+ lymphocyte/tumour-budding index appears to be an independent prognostic factor in CRC [269]. E-cadherin expression is lower in tumour budding sites than in the centre of the tumour [270], possibly contributing to reduced cell-cell adhesion at the tumour front. It has been suggested that a comparison of the pro- and anti-tumour factors at the invasive front could contribute to development of a prognostic score for patients with CRC [133], with proteomic analysis providing new prognostic markers for CRC.
An alternative proteomic approach is the analysis of patient blood plasma or other biological fluids for CRC biomarkers [271], which include cancer-associated soluble factors, as well as tumour-derived microvesicles [272275]. Prefractionation of biological fluids is required to detect low abundance proteins [276]. Lectin glycoarrays have been used to profile plasma for CRC biomarkers, with the identification of significant glycosylation changes associated with tumour progression [277281].
In summary, the search continues for a reliable method of predicting metastatic spread and patient outcome after surgical resection of primary CRC. There is a general consensus that patterns of multiple biomarkers will be required; innovative approaches are needed to translate accumulating data into a useful method for sub-classifying CRC patients for disease stage and those likely to benefit from available treatments. Further progress in cell surface protein discovery will supplement new discoveries provided by other innovative technologies, such as metabolomics [282,283], study of oncogenic pathways [30,31], anti-sense therapeutic strategies [9,284], and combined proteomics/genomics strategies [285].

Acknowledgement

This review was based on studies supported by a Cancer Institute New South Wales Translational Program Grant.

Abbreviations

CEA
carcinoembryonic antigen
CD44v6
CD44 variant 6
CCR
chemokine receptor
CRC
colorectal cancer
CSC
colorectal cancer stem cell
CXCR
chemokine CXC motif receptor
DC
dendritic cells
2D-DIGE
two-dimensional difference gel electrophoresis
DR
death receptor
EpCAM
epithelial-specific cell adhesion activation molecule
FoxP3
forkhead box P3
MHC
major histocompatibility complex
HLA
human leukocyte antigen
Met
hepatocyte growth factor receptor
MMP
matrix metalloproteinase
MMR
DNA mismatch repair
MSI
microsatellite instability
NK
natural killer
TAM
tumour-associated macrophage
TEM
tetraspanin-enriched membrane domains
TIL
tumour infiltrating lymphocyte
Treg
regulatory T-cell

References

  1. Puppa, G; Sonzogni, A; Colombari, R; Pelosi, G. TNM staging system of colorectal carcinoma: A critical appraisal of challenging issues. Arch. Pathol. Lab. Med 2010, 134, 837–852. [Google Scholar]
  2. Zinkin, LD. A critical review of the classifications and staging of colorectal cancer. Dis. Colon. Rectum 1983, 26, 37–43. [Google Scholar]
  3. Compton, CC. Colorectal carcinoma: Diagnostic, prognostic, and molecular features. Mod. Pathol 2003, 16, 376–388. [Google Scholar]
  4. O’Connell, JB; Maggard, MA; Ko, CY. Colon cancer survival rates with the new American Joint Committee on Cancer sixth edition staging. J. Natl. Cancer Inst 2004, 96, 1420–1425. [Google Scholar]
  5. Alfonso, P; Canamero, M; Fernandez-Carbonie, F; Nunez, A; Casal, JI. Proteome analysis of membrane fractions in colorectal carcinomas by using 2D-DIGE saturation labeling. J. Proteome Res 2008, 7, 4247–4255. [Google Scholar]
  6. Andre, M; Le Caer, JP; Greco, C; Planchon, S; El Nemer, W; Boucheix, C; Rubinstein, E; Chamot-Rooke, J; Le Naour, F. Proteomic analysis of the tetraspanin web using LC-ESI-MS/MS and MALDI-FTICR-MS. Proteomics 2006, 6, 1437–1449. [Google Scholar]
  7. Barderas, R; Babel, I; Casal, JI. Colorectal cancer proteomics, molecular characterization and biomarker discovery. Proteomics-Clin. Appl. Rev 2010, 4, 159–178. [Google Scholar]
  8. Friedman, DB; Hill, S; Keller, JW; Merchant, NB; Levy, SE; Coffey, RJ; Caprioli, RM. Proteome analysis of human colon cancer by two-dimensional difference gel electrophoresis and mass spectrometry. Proteomics 2004, 4, 793–811. [Google Scholar]
  9. Gil-Bazo, I. Novel translational strategies in colorectal Cancer Res.earch. World J. Gastroenterol 2007, 13, 5902–5910. [Google Scholar]
  10. Habermann, JK; Bader, FG; Franke, C; Zimmermann, K; Gemoll, T; Fritzsche, B; Ried, T; Auer, G; Bruch, HP; Roblick, UJ. From the genome to the proteome--biomarkers in colorectal cancer. Langenbecks Arch. Surg 2008, 393, 93–104. [Google Scholar]
  11. Jimenez, CR; Knol, JC; Meijer, GA; Fijneman, RJ. Proteomics of colorectal cancer: Overview of discovery studies and identification of commonly identified cancer-associated proteins and candidate CRC serum markers. J Proteomics 2010, 73, 1873–1895. [Google Scholar]
  12. Luque-Garcia, JL; Martinez-Torrecuadrada, JL; Epifano, C; Canamero, M; Babel, I; Casal, JI. Differential protein expression on the cell surface of colorectal cancer cells associated to tumor metastasis. Proteomics 2010, 10, 940–952. [Google Scholar]
  13. Hlubek, F; Brabletz, T; Budczies, J; Pfeiffer, S; Jung, A; Kirchner, T. Heterogeneous expression of Wnt/beta-catenin target genes within colorectal cancer. Int. J Cancer 2007, 121, 1941–1948. [Google Scholar]
  14. Li, JQ; Xu, BJ; Shakhtour, B; Deane, N; Merchant, N; Heslin, MJ; Washington, K; Coffey, RJ; Beauchamp, RD; Shyr, Y; Billheimer, D. Variability of in situ proteomic profiling and implications for study design in colorectal tumors. Int. J. Oncol 2007, 31, 103–111. [Google Scholar]
  15. Ahn, YH; Lee, JY; Kim, YS; Ko, JH; Yoo, JS. Quantitative analysis of an aberrant glycoform of TIMP1 from colon cancer serum by L-PHA-enrichment and SISCAPA with MRM mass spectrometry. J. Proteome Res 2009, 8, 4216–4224. [Google Scholar]
  16. Rho, JH; Qin, S; Wang, JY; Roehrl, MH. Proteomic expression analysis of surgical human colorectal cancer tissues: Up-regulation of PSB7, PRDX1, and SRP9 and hypoxic adaptation in cancer. J. Proteome Res 2008, 7, 2959–2572. [Google Scholar]
  17. Petrak, J; Ivanek, R; Toman, O; Cmejla, R; Cmejlova, J; Vyoral, D; Zivny, J; Vulpe, CD. Deja vu in proteomics. A hit parade of repeatedly identified differentially expressed proteins. Proteomics 2008, 8, 1744–1749. [Google Scholar]
  18. Wang, P; Bouwman, FG; Mariman, EC. Generally detected proteins in comparative proteomics--a matter of cellular stress response? Proteomics 2009, 9, 2955–2566. [Google Scholar]
  19. Yamasaki, M; Takemasa, I; Komori, T; Watanabe, S; Sekimoto, M; Doki, Y; Matsubara, K; Monden, M. The gene expression profile represents the molecular nature of liver metastasis in colorectal cancer. Int. J. Oncol 2007, 30, 129–138. [Google Scholar]
  20. Eschrich, S; Yang, I; Bloom, G; Kwong, KY; Boulware, D; Cantor, A; Coppola, D; Kruhoffer, M; Aaltonen, L; Orntoft, TF; Quackenbush, J; Yeatman, TJ. Molecular staging for survival prediction of colorectal cancer patients. J. Clin. Oncol 2005, 23, 3526–3535. [Google Scholar]
  21. Ellmark, P; Belov, L; Huang, P; Lee, CS; Solomon, MJ; Morgan, DK; Christopherson, RI. Multiplex detection of surface molecules on colorectal cancers. Proteomics 2006, 6, 1791–1802. [Google Scholar]
  22. Zhou, J; Belov, L; Huang, PY; Shin, JS; Solomon, MJ; Chapuis, PH; Bokey, L; Chan, C; Clarke, C; Clarke, SJ; Christopherson, RI. Surface antigen profiling of colorectal cancer using antibody microarrays with fluorescence multiplexing. J. Immunol Methods 2010, 355, 40–51. [Google Scholar]
  23. Belov, L; Mulligan, SP; Barber, N; Woolfson, A; Scott, M; Stoner, K; Chrisp, JS; Sewell, WA; Bradstock, KF; Bendall, L; Pascovici, DS; Thomas, M; Erber, W; Huang, P; Sartor, M; Young, GA; Wiley, JS; Juneja, S; Wierda, WG; Green, AR; Keating, MJ; Christopherson, RI. Analysis of human leukaemias and lymphomas using extensive immunophenotypes from an antibody microarray. Br. J. Haematol 2006, 135, 184–197. [Google Scholar]
  24. Moyret-Lalle, C; Falette, N; Grelier, G; Puisieux, A. Tumour genomics: An unstable landscape. Bull Cancer 2008, 95, 923–930. [Google Scholar]
  25. McAllister, SS; Weinberg, RA. Tumor-Host Interactions: A Far-Reaching Relationship. J. Clin. Oncol 2010, 28, 4022–4028. [Google Scholar]
  26. Kaufman, KL; Mactier, S; Kohnke, P; Christopherson, RI. Davis, JR, Ed.; Cell surface oncoproteomics: Cancer biomarker discovery and clinical applications. In Oncoproteins: Types and Detection; Nova Science Publishes, Inc: Hauppauge, NY, USA, 2010. [Google Scholar]
  27. Leth-Larsen, R; Lund, RR; Ditzel, HJ. Plasma membrane proteomics and its application in clinical cancer biomarker discovery. Mol Cell Proteomics 2010, 9, 1369–1382. [Google Scholar]
  28. Baldus, SE; Schaefer, KL; Engers, R; Hartleb, D; Stoecklein, NH; Gabbert, HE. Prevalence and heterogeneity of KRAS, BRAF, and PIK3CA mutations in primary colorectal adenocarcinomas and their corresponding metastases. Clin. Cancer Res 2010, 16, 790–799. [Google Scholar]
  29. Fang, JY; Richardson, BC. The MAPK signalling pathways and colorectal cancer. Lancet Oncol 2005, 6, 322–327. [Google Scholar]
  30. Markowitz, SD; Bertagnolli, MM. Molecular origins of cancer: Molecular basis of colorectal cancer. N. Engl. J. Med 2009, 361, 2449–2460. [Google Scholar]
  31. Saif, MW; Chu, E. Biology of colorectal cancer. Cancer J 2010, 16, 196–201. [Google Scholar]
  32. Sancho, E; Batlle, E; Clevers, H. Signaling pathways in intestinal development and cancer. 2004, 20, 695–723. [Google Scholar]
  33. Wu, CC; Yates, JR, III. The application of mass spectrometry to membrane proteomics. Nat. Biotechnol 2003, 21, 262–267. [Google Scholar]
  34. Bird, NC; Mangnall, D; Majeed, AW. Biology of colorectal liver metastases: A review. J. Surg. Oncol 2006, 94, 68–80. [Google Scholar]
  35. Haier, J; Nasralla, M; Nicolson, GL. Cell surface molecules and their prognostic values in assessing colorectal carcinomas. Ann. Surg 2000, 231, 11–24. [Google Scholar]
  36. De Wever, O; Mareel, M. Role of tissue stroma in cancer cell invasion. J. Pathol 2003, 200, 429–447. [Google Scholar]
  37. Sund, M; Kalluri, R. Tumor stroma derived biomarkers in cancer. Cancer Metastasis Rev 2009, 28, 177–183. [Google Scholar]
  38. Bacman, D; Merkel, S; Croner, R; Papadopoulos, T; Brueckl, W; Dimmler, A. TGF-beta receptor 2 downregulation in tumour-associated stroma worsens prognosis and high-grade tumours show more tumour-associated macrophages and lower TGF-beta1 expression in colon carcinoma: A retrospective study. BMC Cancer 2007, 7, 156. [Google Scholar]
  39. Kitadai, Y; Sasaki, T; Kuwai, T; Nakamura, T; Bucana, CD; Fidler, IJ. Targeting the expression of platelet-derived growth factor receptor by reactive stroma inhibits growth and metastasis of human colon carcinoma. Am. J. Pathol 2006, 169, 2054–2065. [Google Scholar]
  40. Kitadai, Y; Sasaki, T; Kuwai, T; Nakamura, T; Bucana, CD; Hamilton, SR; Fidler, IJ. Expression of activated platelet-derived growth factor receptor in stromal cells of human colon carcinomas is associated with metastatic potential. Int. J Cancer 2006, 119, 2567–2574. [Google Scholar]
  41. Saigusa, S; Toiyama, Y; Tanaka, K; Yokoe, T; Okugawa, Y; Kawamoto, A; Yasuda, H; Inoue, Y; Miki, C; Kusunoki, M. Stromal CXCR4 and CXCL12 expression is associated with distant recurrence and poor prognosis in rectal cancer after chemoradiotherapy. Ann. Surg. Oncol 2010, 17, 2051–2058. [Google Scholar]
  42. Henry, LR; Lee, HO; Lee, JS; Klein-Szanto, A; Watts, P; Ross, EA; Chen, WT; Cheng, JD. Clinical implications of fibroblast activation protein in patients with colon cancer. Clin. Cancer Res 2007, 13, 1736–1741. [Google Scholar]
  43. Saito, K; Takeha, S; Shiba, K; Matsuno, S; Sorsa, T; Nagura, H; Ohtani, H. Clinicopathologic significance of urokinase receptor- and MMP-9-positive stromal cells in human colorectal cancer: Functional multiplicity of matrix degradation on hematogenous metastasis. Int. J Cancer 2000, 86, 24–29. [Google Scholar]
  44. Mesker, WE; Liefers, GJ; Junggeburt, JM; van Pelt, GW; Alberici, P; Kuppen, PJ; Miranda, NF; van Leeuwen, KA; Morreau, H; Szuhai, K; Tollenaar, RA; Tanke, HJ. Presence of a high amount of stroma and downregulation of SMAD4 predict for worse survival for stage I-II colon cancer patients. Cell Oncol 2009, 31, 169–178. [Google Scholar]
  45. Ogawa, H; Iwaya, K; Izumi, M; Kuroda, M; Serizawa, H; Koyanagi, Y; Mukai, K. Expression of CD10 by stromal cells during colorectal tumor development. Hum. Pathol 2002, 33, 806–811. [Google Scholar]
  46. Miyanaga, K; Kato, Y; Nakamura, T; Matsumura, M; Amaya, H; Horiuchi, T; Chiba, Y; Tanaka, K. Expression and role of thrombospondin-1 in colorectal cancer. AntiCancer Res 2002, 22, 3941–3948. [Google Scholar]
  47. Kaio, E; Tanaka, S; Oka, S; Hiyama, T; Kitadai, Y; Haruma, K; Chayama, K. Clinical significance of thrombospondin-1 expression in relation to vascular endothelial growth factor and interleukin-10 expression at the deepest invasive tumor site of advanced colorectal carcinoma. Int. J. Oncol 2003, 23, 901–911. [Google Scholar]
  48. Maeda, K; Nishiguchi, Y; Kang, SM; Yashiro, M; Onoda, N; Sawada, T; Ishikawa, T; Hirakawa, K. Expression of thrombospondin-1 inversely correlated with tumor vascularity and hematogenous metastasis in colon cancer. Oncol. Rep 2001, 8, 763–766. [Google Scholar]
  49. Maeda, K; Nishiguchi, Y; Yashiro, M; Yamada, S; Onoda, N; Sawada, T; Kang, SM; Hirakawa, K. Expression of vascular endothelial growth factor and thrombospondin-1 in colorectal carcinoma. Int. J. Mol. Med 2000, 5, 373–378. [Google Scholar]
  50. Roberts, DD. Regulation of tumor growth and metastasis by thrombospondin-1. FASEB J 1996, 10, 1183–1191. [Google Scholar]
  51. Sutton, CD; O’Byrne, K; Goddard, JC; Marshall, LJ; Jones, L; Garcea, G; Dennison, AR; Poston, G; Lloyd, DM; Berry, DP. Expression of thrombospondin-1 in resected colorectal liver metastases predicts poor prognosis. Clin. Cancer Res 2005, 11, 6567–6573. [Google Scholar]
  52. Forssell, J; Oberg, A; Henriksson, ML; Stenling, R; Jung, A; Palmqvist, R. High macrophage infiltration along the tumor front correlates with improved survival in colon cancer. Clin. Cancer Res 2007, 13, 1472–1479. [Google Scholar]
  53. Klintrup, K; Makinen, JM; Kauppila, S; Vare, PO; Melkko, J; Tuominen, H; Tuppurainen, K; Makela, J; Karttunen, TJ; Makinen, MJ. Inflammation and prognosis in colorectal cancer. Eur. J Cancer 2005, 41, 2645–2654. [Google Scholar]
  54. Jedinak, A; Dudhgaonkar, S; Sliva, D. Activated macrophages induce metastatic behavior of colon cancer cells. Immunobiology 2010, 215, 242–249. [Google Scholar]
  55. Kaler, P; Galea, V; Augenlicht, L; Klampfer, L. Tumor associated macrophages protect colon cancer cells from TRAIL-induced apoptosis through IL-1beta-dependent stabilization of Snail in tumor cells. PLoS One 2010, 5, e11700. [Google Scholar]
  56. Yuan, A; Chen, JJ; Yang, PC. Pathophysiology of tumor-associated macrophages. Adv Clin. Chem 2008, 45, 199–223. [Google Scholar]
  57. Jedinak, A; Dudhgaonkar, S; Sliva, D. Activated macrophages induce metastatic behavior of colon cancer cells. Immunobiology 2010, 215, 242–249. [Google Scholar]
  58. Sandel, MH; Dadabayev, AR; Menon, AG; Morreau, H; Melief, CJ; Offringa, R; van der Burg, SH; Janssen-van Rhijn, CM; Ensink, NG; Tollenaar, RA; van de Velde, CJ; Kuppen, PJ. Prognostic value of tumor-infiltrating dendritic cells in colorectal cancer: Role of maturation status and intratumoral localization. Clin. Cancer Res 2005, 11, 2576–2582. [Google Scholar]
  59. Camus, M; Tosolini, M; Mlecnik, B; Pages, F; Kirilovsky, A; Berger, A; Costes, A; Bindea, G; Charoentong, P; Bruneval, P; Trajanoski, Z; Fridman, WH; Galon, J. Coordination of intratumoral immune reaction and human colorectal cancer recurrence. Cancer Res 2009, 69, 2685–2693. [Google Scholar]
  60. Nagorsen, D; Voigt, S; Berg, E; Stein, H; Thiel, E; Loddenkemper, C. Tumor-infiltrating macrophages and dendritic cells in human colorectal cancer: Relation to local regulatory T cells, systemic T-cell response against tumor-associated antigens and survival. J. Transl. Med 2007, 5, 62. [Google Scholar]
  61. Adams, H; Tzankov, A; Lugli, A; Zlobec, I. New time-dependent approach to analyse the prognostic significance of immunohistochemical biomarkers in colon cancer and diffuse large B-cell lymphoma. J. Clin. Pathol 2009, 62, 986–997. [Google Scholar]
  62. Jass, JR; Ajioka, Y; Allen, JP; Chan, YF; Cohen, RJ; Nixon, JM; Radojkovic, M; Restall, AP; Stables, SR; Zwi, LJ. Assessment of invasive growth pattern and lymphocytic infiltration in colorectal cancer. Histopathology 1996, 28, 543–548. [Google Scholar]
  63. Katz, SC; Pillarisetty, V; Bamboat, ZM; Shia, J; Hedvat, C; Gonen, M; Jarnagin, W; Fong, Y; Blumgart, L; D’Angelica, M; DeMatteo, RP. T cell infiltrate predicts long-term survival following resection of colorectal cancer liver metastases. Ann. Surg. Oncol 2009, 16, 2524–2530. [Google Scholar]
  64. Ohtani, H. Focus on TILs: Prognostic significance of tumor infiltrating lymphocytes in human colorectal cancer. Cancer Immun 2007, 7, 4. [Google Scholar]
  65. Galon, J; Costes, A; Sanchez-Cabo, F; Kirilovsky, A; Mlecnik, B; Lagorce-Pages, C; Tosolini, M; Camus, M; Berger, A; Wind, P; Zinzindohoue, F; Bruneval, P; Cugnenc, PH; Trajanoski, Z; Fridman, WH; Pages, F. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 2006, 313, 1960–1964. [Google Scholar]
  66. Lee, WS; Park, S; Lee, WY; Yun, SH; Chun, HK. Clinical impact of tumor-infiltrating lymphocytes for survival in stage II colon cancer. Cancer 2010, 116, 5188–5199. [Google Scholar]
  67. Naito, Y; Saito, K; Shiiba, K; Ohuchi, A; Saigenji, K; Nagura, H; Ohtani, H. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 1998, 58, 3491–3494. [Google Scholar]
  68. Pages, F; Galon, J; Dieu-Nosjean, MC; Tartour, E; Sautes-Fridman, C; Fridman, WH. Immune infiltration in human tumors: A prognostic factor that should not be ignored. Oncogene 2010, 29, 1093–1102. [Google Scholar]
  69. Strater, J; Herter, I; Merkel, G; Hinz, U; Weitz, J; Moller, P. Expression and prognostic significance of APAF-1, caspase-8 and caspase-9 in stage II/III colon carcinoma: Caspase-8 and caspase-9 is associated with poor prognosis. Int. J Cancer 2010, 127, 873–880. [Google Scholar]
  70. Deschoolmeester, V; Baay, M; Specenier, P; Lardon, F; Vermorken, JB. A review of the most promising biomarkers in colorectal cancer: One step closer to targeted therapy. Oncologist 2010, 15, 699–731. [Google Scholar]
  71. Galon, J; Fridman, WH; Pages, F. The adaptive immunologic microenvironment in colorectal cancer: A novel perspective. Cancer Res 2007, 67, 1883–1886. [Google Scholar]
  72. Halama, N; Michel, S; Kloor, M; Zoernig, I; Pommerencke, T; von Knebel Doeberitz, M; Schirmacher, P; Weitz, J; Grabe, N; Jager, D. The localization and density of immune cells in primary tumors of human metastatic colorectal cancer shows an association with response to chemotherapy. Cancer Immun 2009, 9, 1. [Google Scholar]
  73. Pages, F; Galon, J; Fridman, WH. The essential role of the in situ immune reaction in human colorectal cancer. J. Leukoc Biol 2008, 84, 981–987. [Google Scholar]
  74. Simpson, JA; Al-Attar, A; Watson, NF; Scholefield, JH; Ilyas, M; Durrant, LG. Intratumoral T cell infiltration, MHC class I and STAT1 as biomarkers of good prognosis in colorectal cancer. Gut 2010, 59, 926–933. [Google Scholar]
  75. Laghi, L; Bianchi, P; Miranda, E; Balladore, E; Pacetti, V; Grizzi, F; Allavena, P; Torri, V; Repici, A; Santoro, A; Mantovani, A; Roncalli, M; Malesci, A. CD3+ cells at the invasive margin of deeply invading (pT3-T4) colorectal cancer and risk of post-surgical metastasis: A longitudinal study. Lancet Oncol 2009, 10, 877–884. [Google Scholar]
  76. Diederichsen, AC; Hjelmborg, JB; Christensen, PB; Zeuthen, J; Fenger, C. Prognostic value of the CD4+/CD8+ ratio of tumour infiltrating lymphocytes in colorectal cancer and HLA-DR expression on tumour cells. Cancer Immunol. Immunother 2003, 52, 423–428. [Google Scholar]
  77. Dolcetti, R; Viel, A; Doglioni, C; Russo, A; Guidoboni, M; Capozzi, E; Vecchiato, N; Macri, E; Fornasarig, M; Boiocchi, M. High prevalence of activated intraepithelial cytotoxic T lymphocytes and increased neoplastic cell apoptosis in colorectal carcinomas with microsatellite instability. Am. J. Pathol 1999, 154, 1805–1813. [Google Scholar]
  78. Prall, F; Duhrkop, T; Weirich, V; Ostwald, C; Lenz, P; Nizze, H; Barten, M. Prognostic role of CD8+ tumor-infiltrating lymphocytes in stage III colorectal cancer with and without microsatellite instability. Hum. Pathol 2004, 35, 808–816. [Google Scholar]
  79. Clark, AJ; Barnetson, R; Farrington, SM; Dunlop, MG. Prognosis in DNA mismatch repair deficient colorectal cancer: Are all MSI tumours equivalent? Fam Cancer 2004, 3, 85–91. [Google Scholar]
  80. Menon, AG; Janssen-van Rhijn, CM; Morreau, H; Putter, H; Tollenaar, RA; van de Velde, CJ; Fleuren, GJ; Kuppen, PJ. Immune system and prognosis in colorectal cancer: A detailed immunohistochemical analysis. Lab. Invest 2004, 84, 493–501. [Google Scholar]
  81. Nussler, NC; Strange, BJ; Petzold, M; Nussler, AK; Glanemann, M; Guckelberger, O. Reduced NK-cell activity in patients with metastatic colon cancer. EXCLI J 2007, 6, 1–9. [Google Scholar]
  82. Koch, M; Beckhove, P; Op den Winkel, J; Autenrieth, D; Wagner, P; Nummer, D; Specht, S; Antolovic, D; Galindo, L; Schmitz-Winnenthal, FH; Schirrmacher, V; Buchler, MW; Weitz, J. Tumor infiltrating T lymphocytes in colorectal cancer: Tumor-selective activation and cytotoxic activity in situ. Ann. Surg 2006, 244, 986–992. [Google Scholar]
  83. Salama, P; Phillips, M; Grieu, F; Morris, M; Zeps, N; Joseph, D; Platell, C; Iacopetta, B. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J. Clin. Oncol 2009, 27, 186–192. [Google Scholar]
  84. Kitayama, J; Nagawa, H; Nakayama, H; Tuno, N; Shibata, Y; Muto, T. Functional expression of beta1 and beta2 integrins on tumor infiltrating lymphocytes (TILs) in colorectal cancer. J. Gastroenterol 1999, 34, 327–333. [Google Scholar]
  85. Kruger, K; Buning, C; Schriever, F. Activated T lymphocytes bind in situ to stromal tissue of colon carcinoma but lack adhesion to tumor cells. Eur. J. Immunol 2001, 31, 138–145. [Google Scholar]
  86. Berndt, U; Philipsen, L; Bartsch, S; Wiedenmann, B; Baumgart, DC; Hammerle, M; Sturm, A. Systematic high-content proteomic analysis reveals substantial immunologic changes in colorectal cancer. Cancer Res 2008, 68, 880–888. [Google Scholar]
  87. Diederichsen, AC; Zeuthen, J; Christensen, PB; Kristensen, T. Characterisation of tumour infiltrating lymphocytes and correlations with immunological surface molecules in colorectal cancer. Eur. J Cancer 1999, 35, 721–726. [Google Scholar]
  88. Sinicrope, FA; Rego, RL; Ansell, SM; Knutson, KL; Foster, NR; Sargent, DJ. Intraepithelial effector (CD3+)/regulatory (FoxP3+) T-cell ratio predicts a clinical outcome of human colon carcinoma. Gastroenterology 2009, 137, 1270–1279. [Google Scholar]
  89. Suzuki, H; Chikazawa, N; Tasaka, T; Wada, J; Yamasaki, A; Kitaura, Y; Sozaki, M; Tanaka, M; Onishi, H; Morisaki, T; Katano, M. Intratumoral CD8(+) T/FOXP3 (+) cell ratio is a predictive marker for survival in patients with colorectal cancer. Cancer Immunol. Immunother 2010, 59, 653–661. [Google Scholar]
  90. Correale, P; Rotundo, MS; Del Vecchio, MT; Remondo, C; Migali, C; Ginanneschi, C; Tsang, KY; Licchetta, A; Mannucci, S; Loiacono, L; Tassone, P; Francini, G; Tagliaferri, P. Regulatory (FoxP3+) T-cell tumor infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing chemo or chemoimmunotherapy. J. Immunother 2010, 33, 435–441. [Google Scholar]
  91. Frey, DM; Droeser, RA; Viehl, CT; Zlobec, I; Lugli, A; Zingg, U; Oertli, D; Kettelhack, C; Terracciano, L; Tornillo, L. High frequency of tumor-infiltrating FOXP3(+) regulatory T cells predicts improved survival in mismatch repair-proficient colorectal cancer patients. Int. J Cancer 2010, 126, 2635–2643. [Google Scholar]
  92. Hashida, H; Takabayashi, A; Tokuhara, T; Hattori, N; Taki, T; Hasegawa, H; Satoh, S; Kobayashi, N; Yamaoka, Y; Miyake, M. Clinical significance of transmembrane 4 superfamily in colon cancer. Br. J Cancer 2003, 89, 158–167. [Google Scholar]
  93. Le Naour, F; Andre, M; Boucheix, C; Rubinstein, E. Membrane microdomains and proteomics: Lessons from tetraspanin microdomains and comparison with lipid rafts. Proteomics 2006, 6, 6447–6454. [Google Scholar]
  94. Le Naour, F; Andre, M; Greco, C; Billard, M; Sordat, B; Emile, JF; Lanza, F; Boucheix, C; Rubinstein, E. Profiling of the tetraspanin web of human colon cancer cells. Mol Cell Proteomics 2006, 5, 845–857. [Google Scholar]
  95. Yanez-Mo, M; Barreiro, O; Gordon-Alonso, M; Sala-Valdes, M; Sanchez-Madrid, F. Tetraspanin-enriched microdomains: A functional unit in cell plasma membranes. Trends Cell Biol 2009, 19, 434–446. [Google Scholar]
  96. Zoller, M. Gastrointestinal tumors: Metastasis and tetraspanins. Z Gastroenterol 2006, 44, 573–586. [Google Scholar]
  97. Mori, M; Mimori, K; Shiraishi, T; Haraguchi, M; Ueo, H; Barnard, GF; Akiyoshi, T. Motility related protein 1 (MRP1/CD9) expression in colon cancer. Clin. Cancer Res 1998, 4, 1507–1510. [Google Scholar]
  98. El-Bahrawy, M; Poulsom, R; Rowan, AJ; Tomlinson, IT; Alison, MR. Characterization of the E-cadherin/catenin complex in colorectal carcinoma cell lines. Int. J. Exp. Pathol 2004, 85, 65–74. [Google Scholar]
  99. Boo, YJ; Park, JM; Kim, J; Chae, YS; Min, BW; Um, JW; Moon, HY. L1 expression as a marker for poor prognosis, tumor progression, and short survival in patients with colorectal cancer. Ann. Surg. Oncol 2007, 14, 1703–1711. [Google Scholar]
  100. Mohri, Y. Prognostic significance of E-cadherin expression in human colorectal cancer tissue. Surg Today 1997, 27, 606–612. [Google Scholar]
  101. Tsanou, E; Peschos, D; Batistatou, A; Charalabopoulos, A; Charalabopoulos, K. The E-cadherin adhesion molecule and colorectal cancer. A global literature approach. AntiCancer Res 2008, 28, 3815–3826. [Google Scholar]
  102. Hayashida, Y; Honda, K; Idogawa, M; Ino, Y; Ono, M; Tsuchida, A; Aoki, T; Hirohashi, S; Yamada, T. E-cadherin regulates the association between beta-catenin and actinin-4. Cancer Res 2005, 65, 8836–8845. [Google Scholar]
  103. Wanitsuwan, W; Kanngurn, S; Boonpipattanapong, T; Sangthong, R; Sangkhathat, S. Overall expression of beta-catenin outperforms its nuclear accumulation in predicting outcomes of colorectal cancers. World J. Gastroenterol 2008, 14, 6052–6059. [Google Scholar]
  104. Fang, QX; Lu, LZ; Yang, B; Zhao, ZS; Wu, Y; Zheng, XC. L1, beta-catenin, and E-cadherin expression in patients with colorectal cancer: Correlation with clinicopathologic features and its prognostic significance. J. Surg. Oncol 2010, 102, 433–442. [Google Scholar]
  105. Hashida, H; Takabayashi, A; Tokuhara, T; Taki, T; Kondo, K; Kohno, N; Yamaoka, Y; Miyake, M. Integrin alpha3 expression as a prognostic factor in colon cancer: Association with MRP-1/CD9 and KAI1/CD82. Int. J Cancer 2002, 97, 518–525. [Google Scholar]
  106. Fujita, S; Watanabe, M; Kubota, T; Teramoto, T; Kitajima, M. Alteration of expression in integrin beta 1-subunit correlates with invasion and metastasis in colorectal cancer. Cancer Lett 1995, 91, 145–149. [Google Scholar]
  107. Gao, XQ; Han, JX; Xu, ZF; Zhang, WD; Zhang, HN; Huang, HY. Identification of the differential expressive tumor associated genes in rectal cancers by cDNA microarray. World J. Gastroenterol 2007, 13, 341–348. [Google Scholar]
  108. Okazaki, K; Nakayama, Y; Shibao, K; Hirata, K; Nagata, N; Itoh, H. Enhancement of metastatic activity of colon cancer as influenced by expression of cell surface antigens. J. Surg. Res 1998, 78, 78–84. [Google Scholar]
  109. Herlevsen, M; Schmidt, DS; Miyazaki, K; Zoller, M. The association of the tetraspanin D6.1A with the alpha6beta4 integrin supports cell motility and liver metastasis formation. J. Cell Sci 2003, 116, 4373–4390. [Google Scholar]
  110. Mulder, JW; Kruyt, PM; Sewnath, M; Oosting, J; Seldenrijk, CA; Weidema, WF; Offerhaus, GJ; Pals, ST. Colorectal cancer prognosis and expression of exon-v6-containing CD44 proteins. Lancet 1994, 344, 1470–1472. [Google Scholar]
  111. Ropponen, KM; Eskelinen, MJ; Lipponen, PK; Alhava, E; Kosma, VM. Expression of CD44 and variant proteins in human colorectal cancer and its relevance for prognosis. Scand. J. Gastroenterol 1998, 33, 301–309. [Google Scholar]
  112. Vizoso, FJ; Fernandez, JC; Corte, MD; Bongera, M; Gava, R; Allende, MT; Garcia-Muniz, JL; Garcia-Moran, M. Expression and clinical significance of CD44V5 and CD44V6 in resectable colorectal cancer. J. Cancer Res. Clin. Oncol 2004, 130, 679–686. [Google Scholar]
  113. Ngan, CY; Yamamoto, H; Seshimo, I; Ezumi, K; Terayama, M; Hemmi, H; Takemasa, I; Ikeda, M; Sekimoto, M; Monden, M. A multivariate analysis of adhesion molecules expression in assessment of colorectal cancer. J. Surg. Oncol 2007, 95, 652–662. [Google Scholar]
  114. Kikuchi, R; Noguchi, T; Takeno, S; Kubo, N; Uchida, Y. Immunohistochemical detection of membrane-type-1-matrix metalloproteinase in colorectal carcinoma. Br. J Cancer 2000, 83, 215–218. [Google Scholar]
  115. Malhotra, S; Newman, E; Eisenberg, D; Scholes, J; Wieczorek, R; Mignatti, P; Shamamian, P. Increased membrane type 1 matrix metalloproteinase expression from adenoma to colon cancer: A possible mechanism of neoplastic progression. Dis. Colon. Rectum 2002, 45, 537–543. [Google Scholar]
  116. Shiomi, T; Okada, Y. MT1-MMP and MMP-7 in invasion and metastasis of human cancers. Cancer Metastasis Rev 2003, 22, 145–152. [Google Scholar]
  117. Mulder, WM; Stern, PL; Stukart, MJ; de Windt, E; Butzelaar, RM; Meijer, S; Ader, HJ; Claessen, AM; Vermorken, JB; Meijer, CJ; Wagstaff, J; Scheper, RJ; Bloemena, E. Low intercellular adhesion molecule 1 and high 5T4 expression on tumor cells correlate with reduced disease-free survival in colorectal carcinoma patients. Clin. Cancer Res 1997, 3, 1923–1930. [Google Scholar]
  118. Maeda, K; Kang, SM; Sawada, T; Nishiguchi, Y; Yashiro, M; Ogawa, Y; Ohira, M; Ishikawa, T; Hirakawa, YSCK. Expression of intercellular adhesion molecule-1 and prognosis in colorectal cancer. Oncol. Rep 2002, 9, 511–514. [Google Scholar]
  119. Chaudry, MA; Sales, K; Ruf, P; Lindhofer, H; Winslet, MC. EpCAM an immunotherapeutic target for gastrointestinal malignancy: Current experience and future challenges. Br. J Cancer 2007, 96, 1013–1019. [Google Scholar]
  120. Gosens, MJ; van Kempen, LC; van de Velde, CJ; van Krieken, JH; Nagtegaal, ID. Loss of membranous Ep-CAM in budding colorectal carcinoma cells. Mod. Pathol 2007, 20, 221–232. [Google Scholar]
  121. Baeuerle, PA; Gires, O. EpCAM (CD326) finding its role in cancer. Br. J Cancer 2007, 96, 417–423. [Google Scholar]
  122. Munz, M; Baeuerle, PA; Gires, O. The emerging role of EpCAM in cancer and stem cell signaling. Cancer Res 2009, 69, 5627–5629. [Google Scholar]
  123. Shiah, S-G; Tai, K-Y; Wu, C-W. Epigenetic regulation of EpCAM in tumor invasion and metastasis. J. Cancer Mol 2008, 3, 165–168. [Google Scholar]
  124. Trzpis, M; McLaughlin, PM; de Leij, LM; Harmsen, MC. Epithelial cell adhesion molecule: More than a carcinoma marker and adhesion molecule. Am. J. Pathol 2007, 171, 386–395. [Google Scholar]
  125. Winter, MJ; Nagtegaal, ID; van Krieken, JH; Litvinov, SV. The epithelial cell adhesion molecule (Ep-CAM) as a morphoregulatory molecule is a tool in surgical pathology. Am. J. Pathol 2003, 163, 2139–2148. [Google Scholar]
  126. Ohmachi, T; Tanaka, F; Mimori, K; Inoue, H; Yanaga, K; Mori, M. Clinical significance of TROP2 expression in colorectal cancer. Clin. Cancer Res 2006, 12, 3057–3063. [Google Scholar]
  127. Ziegler, A; Heidenreich, R; Braumuller, H; Wolburg, H; Weidemann, S; Mocikat, R; Rocken, M. EpCAM, a human tumor-associated antigen promotes Th2 development and tumor immune evasion. Blood 2009, 113, 3494–3502. [Google Scholar]
  128. Rao, CG; Chianese, D; Doyle, GV; Miller, MC; Russell, T; Sanders, RA, Jr; Terstappen, LW. Expression of epithelial cell adhesion molecule in carcinoma cells present in blood and primary and metastatic tumors. Int. J. Oncol 2005, 27, 49–57. [Google Scholar]
  129. Maetzel, D; Denzel, S; Mack, B; Canis, M; Went, P; Benk, M; Kieu, C; Papior, P; Baeuerle, PA; Munz, M; Gires, O. Nuclear signalling by tumour-associated antigen EpCAM. Nat. Cell Biol 2009, 11, 162–171. [Google Scholar]
  130. Brabletz, T; Jung, A; Reu, S; Porzner, M; Hlubek, F; Kunz-Schughart, LA; Knuechel, R; Kirchner, T. Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc. Natl. Acad. Sci USA 2001, 98, 10356–10361. [Google Scholar]
  131. Thiery, JP. Epithelial-mesenchymal transitions in tumour progression. Nat. Rev Cancer 2002, 2, 442–454. [Google Scholar]
  132. Ueno, H; Murphy, J; Jass, JR; Mochizuki, H; Talbot, IC. Tumour ‘budding’ as an index to estimate the potential of aggressiveness in rectal cancer. Histopathology 2002, 40, 127–132. [Google Scholar]
  133. Zlobec, I; Lugli, A. Invasive front of colorectal cancer: Dynamic interface of pro-/anti-tumor factors. World J. Gastroenterol 2009, 15, 5898–5906. [Google Scholar]
  134. Hostettler, L; Zlobec, I; Terracciano, L; Lugli, A. ABCG5-positivity in tumor buds is an indicator of poor prognosis in node-negative colorectal cancer patients. World J. Gastroenterol 2010, 16, 732–739. [Google Scholar]
  135. Kuhn, S; Koch, M; Nubel, T; Ladwein, M; Antolovic, D; Klingbeil, P; Hildebrand, D; Moldenhauer, G; Langbein, L; Franke, WW; Weitz, J; Zoller, M. A complex of EpCAM, claudin-7, CD44 variant isoforms, and tetraspanins promotes colorectal cancer progression. Mol. Cancer Res 2007, 5, 553–567. [Google Scholar]
  136. Nubel, T; Preobraschenski, J; Tuncay, H; Weiss, T; Kuhn, S; Ladwein, M; Langbein, L; Zoller, M. Claudin-7 regulates EpCAM-mediated functions in tumor progression. Mol. Cancer Res 2009, 7, 285–299. [Google Scholar]
  137. Brockhausen, I. Mucin-type O-glycans in human colon and breast cancer: Glycodynamics and functions. EMBO Rep 2006, 7, 599–604. [Google Scholar]
  138. Hollingsworth, MA; Swanson, BJ. Mucins in cancer: Protection and control of the cell surface. Nat. Rev Cancer 2004, 4, 45–60. [Google Scholar]
  139. Kufe, DW. Mucins in cancer: Function, prognosis and therapy. Nat. Rev Cancer 2009, 9, 874–885. [Google Scholar]
  140. Farhat, MH; Barada, KA; Tawil, AN; Itani, DM; Hatoum, HA; Shamseddine, AI. Effect of mucin production on survival in colorectal cancer: A case-control study. World J. Gastroenterol 2008, 14, 6981–6985. [Google Scholar]
  141. Baldus, SE; Monig, SP; Hanisch, FG; Zirbes, TK; Flucke, U; Oelert, S; Zilkens, G; Madejczik, B; Thiele, J; Schneider, PM; Holscher, AH; Dienes, HP. Comparative evaluation of the prognostic value of MUC1, MUC2, sialyl-Lewis(a) and sialyl-Lewis(x) antigens in colorectal adenocarcinoma. Histopathology 2002, 40, 440–449. [Google Scholar]
  142. Hiraga, Y; Tanaka, S; Haruma, K; Yoshihara, M; Sumii, K; Kajiyama, G; Shimamoto, F; Kohno, N. Immunoreactive MUC1 expression at the deepest invasive portion correlates with prognosis of colorectal cancer. Oncology 1998, 55, 307–319. [Google Scholar]
  143. Saeland, E; van Vliet, SJ; Backstrom, M; van den Berg, VC; Geijtenbeek, TB; Meijer, GA; van Kooyk, Y. The C-type lectin MGL expressed by dendritic cells detects glycan changes on MUC1 in colon carcinoma. Cancer Immunol. Immunother 2007, 56, 1225–1236. [Google Scholar]
  144. Itzkowitz, SH; Yuan, M; Montgomery, CK; Kjeldsen, T; Takahashi, HK; Bigbee, WL; Kim, YS. Expression of Tn, sialosyl-Tn, and T antigens in human colon cancer. Cancer Res 1989, 49, 197–204. [Google Scholar]
  145. Iwasa, S; Okada, K; Chen, WT; Jin, X; Yamane, T; Ooi, A; Mitsumata, M. Increased expression of seprase, a membrane-type serine protease, is associated with lymph node metastasis in human colorectal cancer. Cancer Lett 2005, 227, 229–236. [Google Scholar]
  146. Perez, RO; Bresciani, BH; Bresciani, C; Proscurshim, I; Kiss, D; Gama-Rodrigues, J; Pereira, DD; Rawet, V; Cecconnello, I; Habr-Gama, A. Mucinous colorectal adenocarcinoma: Influence of mucin expression (Muc1, 2 and 5) on clinico-pathological features and prognosis. Int. J. Colorectal. Dis 2008, 23, 757–765. [Google Scholar]
  147. Doekhie, FS; Morreau, H; de Bock, GH; Speetjens, FM; Dekker-Ensink, NG; Putter, H; van de Velde, CJ; Tollenaar, RA; Kuppen, PJ. Sialyl Lewis X expression and lymphatic microvessel density in primary tumors of node-negative colorectal cancer patients predict disease recurrence. Cancer Microenviron 2008, 1, 141–151. [Google Scholar]
  148. Gunther, K; Dworak, O; Remke, S; Pfluger, R; Merkel, S; Hohenberger, W; Reymond, MA. Prediction of distant metastases after curative surgery for rectal cancer. J. Surg. Res 2002, 103, 68–78. [Google Scholar]
  149. Nakagoe, T; Fukushima, K; Hirota, M; Kusano, H; Ayabe, H; Tomita, M; Kamihira, S. Immunohistochemical expression of sialyl Lex antigen in relation to survival of patients with colorectal carcinoma. Cancer 1993, 72, 2323–2330. [Google Scholar]
  150. Nakamori, S; Kameyama, M; Imaoka, S; Furukawa, H; Ishikawa, O; Sasaki, Y; Kabuto, T; Iwanaga, T; Matsushita, Y; Irimura, T. Increased expression of sialyl Lewisx antigen correlates with poor survival in patients with colorectal carcinoma: Clinicopathological and immunohistochemical study. Cancer Res 1993, 53, 3632–3637. [Google Scholar]
  151. Matsui, T; Kojima, H; Suzuki, H; Hamajima, H; Nakazato, H; Ito, K; Nakao, A; Sakamoto, J. Sialyl Lewisa expression as a predictor of the prognosis of colon carcinoma patients in a prospective randomized clinical trial. Jpn. J. Clin. Oncol 2004, 34, 588–593. [Google Scholar]
  152. Borsig, L; Wong, R; Hynes, RO; Varki, NM; Varki, A. Synergistic effects of L- and P-selectin in facilitating tumor metastasis can involve non-mucin ligands and implicate leukocytes as enhancers of metastasis. Proc. Natl. Acad. Sci USA 2002, 99, 2193–2198. [Google Scholar]
  153. Kannagi, R. Carbohydrate antigen sialyl Lewis a--its pathophysiological significance and induction mechanism in cancer progression. Chang Gung Med. J 2007, 30, 189–209. [Google Scholar]
  154. Kohler, S; Ullrich, S; Richter, U; Schumacher, U. E-/P-selectins and colon carcinoma metastasis: First in vivo evidence for their crucial role in a clinically relevant model of spontaneous metastasis formation in the lung. Br. J Cancer 2010, 102, 602–609. [Google Scholar]
  155. Napier, SL; Healy, ZR; Schnaar, RL; Konstantopoulos, K. Selectin ligand expression regulates the initial vascular interactions of colon carcinoma cells: The roles of CD44v and alternative sialofucosylated selectin ligands. J. Biol. Chem 2007, 282, 3433–3441. [Google Scholar]
  156. Endo, K; Kohnoe, S; Tsujita, E; Watanabe, A; Nakashima, H; Baba, H; Maehara, Y. Galectin-3 expression is a potent prognostic marker in colorectal cancer. AntiCancer Res 2005, 25, 3117–3121. [Google Scholar]
  157. Nagy, N; Legendre, H; Engels, O; Andre, S; Kaltner, H; Wasano, K; Zick, Y; Pector, JC; Decaestecker, C; Gabius, HJ; Salmon, I; Kiss, R. Refined prognostic evaluation in colon carcinoma using immunohistochemical galectin fingerprinting. Cancer 2003, 97, 1849–1858. [Google Scholar]
  158. Legendre, H; Decaestecker, C; Nagy, N; Hendlisz, A; Schuring, MP; Salmon, I; Gabius, HJ; Pector, JC; Kiss, R. Prognostic values of galectin-3 and the macrophage migration inhibitory factor (MIF) in human colorectal cancers. Mod. Pathol 2003, 16, 491–504. [Google Scholar]
  159. Song, S; Byrd, JC; Mazurek, N; Liu, K; Koo, JS; Bresalier, RS. Galectin-3 modulates MUC2 mucin expression in human colon cancer cells at the level of transcription via AP-1 activation. Gastroenterology 2005, 129, 1581–1591. [Google Scholar]
  160. Bresalier, RS; Byrd, JC; Wang, L; Raz, A. Colon cancer mucin: A new ligand for the betagalactoside- binding protein galectin-3. Cancer Res 1996, 56, 4354–4357. [Google Scholar]
  161. Dudas, SP; Yunker, CK; Sternberg, LR; Byrd, JC; Bresalier, RS. Expression of human intestinal mucin is modulated by the beta-galactoside binding protein galectin-3 in colon cancer. Gastroenterology 2002, 123, 817–826. [Google Scholar]
  162. Iwase, T; Kushima, R; Mukaisho, K; Mitsufuji, S; Okanoue, T; Hattori, T. Overexpression of CD10 and reduced MUC2 expression correlate with the development and progression of colorectal neoplasms. Pathol. Res. Pract 2005, 201, 83–91. [Google Scholar]
  163. Jantscheff, P; Terracciano, L; Lowy, A; Glatz-Krieger, K; Grunert, F; Micheel, B; Brummer, J; Laffer, U; Metzger, U; Herrmann, R; Rochlitz, C. Expression of CEACAM6 in resectable colorectal cancer: A factor of independent prognostic significance. J. Clin. Oncol 2003, 21, 3638–3646. [Google Scholar]
  164. Cosimelli, M; De Peppo, F; Castelli, M; Giannarelli, D; Schinaia, G; Castaldo, P; Buttini, GL; Sciarretta, F; Bigotti, G; Di Filippo, F; et al. Multivariate analysis of a tissue CEA, TPA, and CA 19.9 quantitative study in colorectal cancer patients. A preliminary finding. Dis. Colon. Rectum 1989, 32, 389–397. [Google Scholar]
  165. Kim, JC; Han, MS; Lee, HK; Kim, WS; Park, SK; Park, KC; Bodmer, WF; Rowan, AJ; Kim, OJ. Distribution of carcinoembryonic antigen and biologic behavior in colorectal carcinoma. Dis. Colon. Rectum 1999, 42, 640–648. [Google Scholar]
  166. Lorenzi, M; Vindigni, C; Minacci, C; Tripodi, SA; Iroatulam, A; Petrioli, R; Francini, G. Histopathological and prognostic evaluation of immunohistochemical findings in colorectal cancer. Int. J. Biol Markers 1997, 12, 68–74. [Google Scholar]
  167. Nazato, DM; Matos, LL; Waisberg, DR; Souza, JR; Martins, LC; Waisberg, J. Prognostic value of carcinoembryonic antigen distribution in tumor tissue of colorectal carcinoma. Arq. Gastroenterol 2009, 46, 26–31. [Google Scholar]
  168. Vogel, I; Francksen, H; Soeth, E; Henne-Bruns, D; Kremer, B; Juhl, H. The carcinoembryonic antigen and its prognostic impact on immunocytologically detected intraperitoneal colorectal cancer cells. Am. J. Surg 2001, 181, 188–193. [Google Scholar]
  169. Hostetter, RB; Augustus, LB; Mankarious, R; Chi, KF; Fan, D; Toth, C; Thomas, P; Jessup, JM. Carcinoembryonic antigen as a selective enhancer of colorectal cancer metastasis. J. Natl. Cancer Inst 1990, 82, 380–385. [Google Scholar]
  170. Blumenthal, RD; Hansen, HJ; Goldenberg, DM. Inhibition of adhesion, invasion, and metastasis by antibodies targeting CEACAM6 (NCA-90) and CEACAM5 (Carcinoembryonic Antigen). Cancer Res 2005, 65, 8809–8817. [Google Scholar]
  171. Messick, CA; Sanchez, J; Dejulius, KL; Hammel, J; Ishwaran, H; Kalady, MF. CEACAM- 7: A predictive marker for rectal cancer recurrence. Surgery 2010, 147, 713–719. [Google Scholar]
  172. Duncan, R; Carpenter, B; Main, LC; Telfer, C; Murray, GI. Characterisation and protein expression profiling of annexins in colorectal cancer. Br. J Cancer 2008, 98, 426–433. [Google Scholar]
  173. Emoto, K; Yamada, Y; Sawada, H; Fujimoto, H; Ueno, M; Takayama, T; Kamada, K; Naito, A; Hirao, S; Nakajima, Y. Annexin II overexpression correlates with stromal tenascin-C overexpression: A prognostic marker in colorectal carcinoma. Cancer 2001, 92, 1419–1426. [Google Scholar]
  174. Xue, G; Hao, LQ; Ding, FX; Mei, Q; Huang, JJ; Fu, CG; Yan, HL; Sun, SH. Expression of annexin a5 is associated with higher tumor stage and poor prognosis in colorectal adenocarcinomas. J. Clin. Gastroenterol 2009, 43, 831–837. [Google Scholar]
  175. Tsukita, S; Yamazaki, Y; Katsuno, T; Tamura, A. Tight junction-based epithelial microenvironment and cell proliferation. Oncogene 2008, 27, 6930–6938. [Google Scholar]
  176. de Oliveira, SS; de Oliveira, IM; De Souza, W; Morgado-Diaz, JA. Claudins upregulation in human colorectal cancer. FEBS Lett 2005, 579, 6179–6185. [Google Scholar]
  177. Resnick, MB; Konkin, T; Routhier, J; Sabo, E; Pricolo, VE. Claudin-1 is a strong prognostic indicator in stage II colonic cancer: A tissue microarray study. Mod. Pathol 2005, 18, 511–518. [Google Scholar]
  178. Ueda, J; Semba, S; Chiba, H; Sawada, N; Seo, Y; Kasuga, M; Yokozaki, H. Heterogeneous expression of claudin-4 in human colorectal cancer: Decreased claudin-4 expression at the invasive front correlates cancer invasion and metastasis. Pathobiology 2007, 74, 32–41. [Google Scholar]
  179. Cambien, B; Karimdjee, BF; Richard-Fiardo, P; Bziouech, H; Barthel, R; Millet, MA; Martini, V; Birnbaum, D; Scoazec, JY; Abello, J; Al Saati, T; Johnson, MG; Sullivan, TJ; Medina, JC; Collins, TL; Schmid-Alliana, A; Schmid-Antomarchi, H. Organ-specific inhibition of metastatic colon carcinoma by CXCR3 antagonism. Br. J Cancer 2009, 100, 1755–1764. [Google Scholar]
  180. Kawada, K; Hosogi, H; Sonoshita, M; Sakashita, H; Manabe, T; Shimahara, Y; Sakai, Y; Takabayashi, A; Oshima, M; Taketo, MM. Chemokine receptor CXCR3 promotes colon cancer metastasis to lymph nodes. Oncogene 2007, 26, 4679–4688. [Google Scholar]
  181. Kim, J; Takeuchi, H; Lam, ST; Turner, RR; Wang, HJ; Kuo, C; Foshag, L; Bilchik, AJ; Hoon, DS. Chemokine receptor CXCR4 expression in colorectal cancer patients increases the risk for recurrence and for poor survival. J. Clin. Oncol 2005, 23, 2744–2753. [Google Scholar]
  182. Schimanski, CC; Galle, PR; Moehler, M. Chemokine receptor CXCR4-prognostic factor for gastrointestinal tumors. World J. Gastroenterol 2008, 14, 4721–4724. [Google Scholar]
  183. Gelmini, S; Mangoni, M; Serio, M; Romagnani, P; Lazzeri, E. The critical role of SDF- 1/CXCR4 axis in cancer and cancer stem cells metastasis. J. Endocrinol. Invest 2008, 31, 809–819. [Google Scholar]
  184. Zimmermann, T; Moehler, M; Gockel, I; Sgourakis, GG; Biesterfeld, S; Muller, M; Berger, MR; Lang, H; Galle, PR; Schimanski, CC. Low expression of chemokine receptor CCR5 in human colorectal cancer correlates with lymphatic dissemination and reduced CD8+ T-cell infiltration. Int. J. Colorectal. Dis 2010, 25, 417–4124. [Google Scholar]
  185. de Castro-Carpeno, J; Belda-Iniesta, C; Casado Saenz, E; Hernandez Agudo, E; Feliu Batlle, J; Gonzalez Baron, M. EGFR and colon cancer: A clinical view. Clin. Transl. Oncol 2008, 10, 6–13. [Google Scholar]
  186. Dasari, A; Messersmith, WA. New strategies in colorectal cancer: Biomarkers of response to epidermal growth factor receptor monoclonal antibodies and potential therapeutic targets in phosphoinositide 3-kinase and mitogen-activated protein kinase pathways. Clin. Cancer Res 2010, 16, 3811–3818. [Google Scholar]
  187. Markman, B; Capdevila, J; Elez, E; Tabernero, J. New trends in epidermal growth factor receptor-directed monoclonal antibodies. Immunotherapy 2009, 1, 965–982. [Google Scholar]
  188. Azria, D; Bibeau, F; Barbier, N; Zouhair, A; Lemanski, C; Rouanet, P; Ychou, M; Senesse, P; Ozsahin, M; Pelegrin, A; Dubois, JB; Thezenas, S. Prognostic impact of epidermal growth factor receptor (EGFR) expression on loco-regional recurrence after preoperative radiotherapy in rectal cancer. BMC Cancer 2005, 5, 62. [Google Scholar]
  189. Galizia, G; Lieto, E; Ferraraccio, F; De Vita, F; Castellano, P; Orditura, M; Imperatore, V; La Mura, A; La Manna, G; Pinto, M; Catalano, G; Pignatelli, C; Ciardiello, F. Prognostic significance of epidermal growth factor receptor expression in colon cancer patients undergoing curative surgery. Ann. Surg. Oncol 2006, 13, 823–835. [Google Scholar]
  190. Spano, JP; Lagorce, C; Atlan, D; Milano, G; Domont, J; Benamouzig, R; Attar, A; Benichou, J; Martin, A; Morere, JF; Raphael, M; Penault-Llorca, F; Breau, JL; Fagard, R; Khayat, D; Wind, P. Impact of EGFR expression on colorectal cancer patient prognosis and survival. Ann. Oncol 2005, 16, 102–108. [Google Scholar]
  191. Arkom, C; Preecha, R; Anant, K; Soisuda, S; Sangduan, P; Suleeporn, S. Determination of epidermal growth factor receptor (EGFR) in patients with colorectal cancer (Institutional series). Cancer Ther 2007, 5, 137–142. [Google Scholar]
  192. Doger, FK; Meteoglu, I; Tuncyurek, P; Okyay, P; Cevikel, H. Does the EGFR and VEGF expression predict the prognosis in colon cancer? Eur. Surg. Res 2006, 38, 540–544. [Google Scholar]
  193. Zhang, C; Hao, L; Wang, L; Xiao, Y; Ge, H; Zhu, Z; Luo, Y; Zhang, Y. Elevated IGFIR expression regulating VEGF and VEGF-C predicts lymph node metastasis in human colorectal cancer. BMC Cancer 2010, 10, 184. [Google Scholar]
  194. Osako, T; Miyahara, M; Uchino, S; Inomata, M; Kitano, S; Kobayashi, M. Immunohistochemical study of c-erbB-2 protein in colorectal cancer and the correlation with patient survival. Oncology 1998, 55, 548–555. [Google Scholar]
  195. Kavanagh, DO; Chambers, G; O’Grady, L; Barry, KM; Waldron, RP; Bennani, F; Eustace, PW; Tobbia, I. Is overexpression of HER-2 a predictor of prognosis in colorectal cancer? BMC Cancer 2009, 9, 1. [Google Scholar]
  196. Baiocchi, G; Lopes, A; Coudry, RA; Rossi, BM; Soares, FA; Aguiar, S; Guimaraes, GC; Ferreira, FO; Nakagawa, WT. ErbB family immunohistochemical expression in colorectal cancer patients with higher risk of recurrence after radical surgery. Int. J. Colorectal. Dis 2009, 24, 1059–1068. [Google Scholar]
  197. De Oliveira, AT; Matos, D; Logullo, AF; SR, DAS; Neto, RA; Filho, AL; Saad, SS. MET Is highly expressed in advanced stages of colorectal cancer and indicates worse prognosis and mortality. AntiCancer Res 2009, 29, 4807–4811. [Google Scholar]
  198. Ginty, F; Adak, S; Can, A; Gerdes, M; Larsen, M; Cline, H; Filkins, R; Pang, Z; Li, Q; Montalto, MC. The relative distribution of membranous and cytoplasmic met is a prognostic indicator in stage I and II colon cancer. Clin. Cancer Res 2008, 14, 3814–3822. [Google Scholar]
  199. Johnstone, RW; Frew, AJ; Smyth, MJ. The TRAIL apoptotic pathway in cancer onset, progression and therapy. Nat. Rev Cancer 2008, 8, 782–798. [Google Scholar]
  200. Strater, J; Hinz, U; Walczak, H; Mechtersheimer, G; Koretz, K; Herfarth, C; Moller, P; Lehnert, T. Expression of TRAIL and TRAIL receptors in colon carcinoma: TRAIL-R1 is an independent prognostic parameter. Clin. Cancer Res 2002, 8, 3734–3740. [Google Scholar]
  201. Bavi, P; Prabhakaran, SE; Abubaker, J; Qadri, Z; George, T; Al-Sanea, N; Abduljabbar, A; Ashari, LH; Alhomoud, S; Al-Dayel, F; Hussain, AR; Uddin, S; Al-Kuraya, KS. Prognostic significance of TRAIL death receptors in Middle Eastern colorectal carcinomas and their correlation to oncogenic KRAS alterations. Mol Cancer 2010, 9, 203. [Google Scholar]
  202. van Geelen, CM; Westra, JL; de Vries, EG; Boersma-van Ek, W; Zwart, N; Hollema, H; Boezen, HM; Mulder, NH; Plukker, JT; de Jong, S; Kleibeuker, JH; Koornstra, JJ. Prognostic significance of tumor necrosis factor-related apoptosis-inducing ligand and its receptors in adjuvantly treated stage III colon cancer patients. J. Clin. Oncol 2006, 24, 4998–5004. [Google Scholar]
  203. McLornan, DP; Barrett, HL; Cummins, R; McDermott, U; McDowell, C; Conlon, SJ; Coyle, VM; van Schaeybroeck, S; Wilson, R; Kay, EW; Longley, DB; Johnston, PG. Prognostic significance of TRAIL signaling molecules in stage II and III colorectal cancer. Clin. Cancer Res 2010, 16, 3442–3451. [Google Scholar]
  204. Ishimura, N; Isomoto, H; Bronk, SF; Gores, GJ. Trail induces cell migration and invasion in apoptosis-resistant cholangiocarcinoma cells. Am. J. Physiol. Gastrointest. Liver Physiol 2006, 290, G129–G136. [Google Scholar]
  205. Trauzold, A; Siegmund, D; Schniewind, B; Sipos, B; Egberts, J; Zorenkov, D; Emme, D; Roder, C; Kalthoff, H; Wajant, H. TRAIL promotes metastasis of human pancreatic ductal adenocarcinoma. Oncogene 2006, 25, 7434–7439. [Google Scholar]
  206. Strater, J; Hinz, U; Hasel, C; Bhanot, U; Mechtersheimer, G; Lehnert, T; Moller, P. Impaired CD95 expression predisposes for recurrence in curatively resected colon carcinoma: Cinical evidence for immunoselection and CD95L mediated control of minimal residual disease. Gut 2005, 54, 661–665. [Google Scholar]
  207. Nozoe, T; Yasuda, M; Honda, M; Inutsuka, S; Korenaga, D. Fas ligand expression is correlated with metastasis in colorectal carcinoma. Oncology 2003, 65, 83–88. [Google Scholar]
  208. Zhang, W; Ding, EX; Wang, Q; Zhu, DQ; He, J; Li, YL; Wang, YH. Fas ligand expression in colon cancer: A possible mechanism of tumor immune privilege. World J. Gastroenterol 2005, 11, 3632–3635. [Google Scholar]
  209. Lin, MH; Yao, YH; Yu, JH. Expression and clinical significance of nm23-H1, Fas and FasL in colorectal carcinoma tissues. J. Sichuan Univ. Med. Sci. Edn 2005, 36, 503–505. [Google Scholar]
  210. Menon, AG; Morreau, H; Tollenaar, RA; Alphenaar, E; van Puijenbroek, M; Putter, H; Janssen-Van Rhijn, CM; van De Velde, CJ; Fleuren, GJ; Kuppen, PJ. Down-regulation of HLA-A expression correlates with a better prognosis in colorectal cancer patients. Lab. Invest 2002, 82, 1725–1733. [Google Scholar]
  211. Speetjens, FM; de Bruin, EC; Morreau, H; Zeestraten, EC; Putter, H; van Krieken, JH; van Buren, MM; van Velzen, M; Dekker-Ensink, NG; van de Velde, CJ; Kuppen, PJ. Clinical impact of HLA class I expression in rectal cancer. Cancer Immunol. Immunother 2008, 57, 601–609. [Google Scholar]
  212. Lovig, T; Andersen, SN; Thorstensen, L; Diep, CB; Meling, GI; Lothe, RA; Rognum, TO. Strong HLA-DR expression in microsatellite stable carcinomas of the large bowel is associated with good prognosis. Br. J Cancer 2002, 87, 756–762. [Google Scholar]
  213. Matsushita, K; Takenouchi, T; Shimada, H; Tomonaga, T; Hayashi, H; Shioya, A; Komatsu, A; Matsubara, H; Ochiai, T. Strong HLA-DR antigen expression on cancer cells relates to better prognosis of colorectal cancer patients: Possible involvement of c-myc suppression by interferon-gamma in situ. Cancer Sci 2006, 97, 57–63. [Google Scholar]
  214. Sakamoto, J; Kojima, H; Kato, J; Hamashima, H; Suzuki, H. Organ-specific expression of the intestinal epithelium-related antigen A33, a cell surface target for antibody-based imaging and treatment in gastrointestinal cancer. Cancer Chemother. Pharmacol 2000, 46, S27–S32. [Google Scholar]
  215. Scott, AM; Lee, FT; Jones, R; Hopkins, W; MacGregor, D; Cebon, JS; Hannah, A; Chong, G; Paul, U; Papenfuss, A; Rigopoulos, A; Sturrock, S; Murphy, R; Wirth, V; Murone, C; Smyth, FE; Knight, S; Welt, S; Ritter, G; Richards, E; Nice, EC; Burgess, AW; Old, LJ. A phase I trial of humanized monoclonal antibody A33 in patients with colorectal carcinoma: Biodistribution, pharmacokinetics, and quantitative tumor uptake. Clin. Cancer Res 2005, 11, 4810–4817. [Google Scholar]
  216. Welt, S; Ritter, G; Williams, C, Jr; Cohen, LS; John, M; Jungbluth, A; Richards, EA; Old, LJ; Kemeny, NE. Phase I study of anticolon cancer humanized antibody A33. Clin. Cancer Res 2003, 9, 1338–1346. [Google Scholar]
  217. Chong, G; Lee, FT; Hopkins, W; Tebbutt, N; Cebon, JS; Mountain, AJ; Chappell, B; Papenfuss, A; Schleyer, P; Paul, U; Murphy, R; Wirth, V; Smyth, FE; Potasz, N; Poon, A; Davis, ID; Saunder, T; O’Keefe, GJ; Burgess, AW; Hoffman, EW; Old, LJ; Scott, AM. Phase I trial of 131I-huA33 in patients with advanced colorectal carcinoma. Clin. Cancer Res 2005, 11, 4818–4826. [Google Scholar]
  218. Welt, S; Divgi, CR; Kemeny, N; Finn, RD; Scott, AM; Graham, M; Germain, JS; Richards, EC; Larson, SM; Oettgen, HF; et al. Phase I/II study of iodine 131-labeled monoclonal antibody A33 in patients with advanced colon cancer. J. Clin. Oncol 1994, 12, 1561–1571. [Google Scholar]
  219. Fujimoto, Y; Nakanishi, Y; Sekine, S; Yoshimura, K; Akasu, T; Moriya, Y; Shimoda, T. CD10 expression in colorectal carcinoma correlates with liver metastasis. Dis. Colon. Rectum 2005, 48, 1883–1889. [Google Scholar]
  220. Ohji, Y; Yao, T; Eguchi, T; Yamada, T; Hirahashi, M; Iida, M; Tsuneyoshi, M. Evaluation of risk of liver metastasis in colorectal adenocarcinoma based on the combination of risk factors including CD10 expression: Multivariate analysis of clinicopathological and immunohistochemical factors. Oncol. Rep 2007, 17, 525–530. [Google Scholar]
  221. Yao, T; Takata, M; Tustsumi, S; Nishiyama, K; Taguchi, K; Nagai, E; Tsuneyoshi, M. Phenotypic expression of gastrointestinal differentiation markers in colorectal adenocarcinomas with liver metastasis. Pathology 2002, 34, 556–560. [Google Scholar]
  222. Fujita, S; Yamamoto, S; Akasu, T; Moriya, Y; Taniguchi, H; Shimoda, T. Quantification of CD10 mRNA in colorectal cancer and relationship between mRNA expression and liver metastasis. Anticancer Res 2007, 27, 3307–3311. [Google Scholar]
  223. Hashida, H; Takabayashi, A; Kanai, M; Adachi, M; Kondo, K; Kohno, N; Yamaoka, Y; Miyake, M. Aminopeptidase N is involved in cell motility and angiogenesis: Its clinical significance in human colon cancer. Gastroenterology 2002, 122, 376–386. [Google Scholar]
  224. Durrant, LG; Chapman, MA; Buckley, DJ; Spendlove, I; Robins, RA; Armitage, NC. Enhanced expression of the complement regulatory protein CD55 predicts a poor prognosis in colorectal cancer patients. Cancer Immunol. Immunother 2003, 52, 638–642. [Google Scholar]
  225. Watson, NF; Durrant, LG; Madjd, Z; Ellis, IO; Scholefield, JH; Spendlove, I. Expression of the membrane complement regulatory protein CD59 (protectin) is associated with reduced survival in colorectal cancer patients. Cancer Immunol. Immunother 2006, 55, 973–980. [Google Scholar]
  226. Steinert, M; Wobus, M; Boltze, C; Schutz, A; Wahlbuhl, M; Hamann, J; Aust, G. Expression and regulation of CD97 in colorectal carcinoma cell lines and tumor tissues. Am. J. Pathol 2002, 161, 1657–1667. [Google Scholar]
  227. Weichert, W; Knosel, T; Bellach, J; Dietel, M; Kristiansen, G. ALCAM/CD166 is overexpressed in colorectal carcinoma and correlates with shortened patient survival. J. Clin. Pathol 2004, 57, 1160–1164. [Google Scholar]
  228. Kaifi, JT; Reichelt, U; Quaas, A; Schurr, PG; Wachowiak, R; Yekebas, EF; Strate, T; Schneider, C; Pantel, K; Schachner, M; Sauter, G; Izbicki, JR. L1 is associated with micrometastatic spread and poor outcome in colorectal cancer. Mod. Pathol 2007, 20, 1183–1190. [Google Scholar]
  229. Mehlen, P; Fearon, ER. Role of the dependence receptor DCC in colorectal cancer pathogenesis. J. Clin. Oncol 2004, 22, 3420–3428. [Google Scholar]
  230. Saito, M; Yamaguchi, A; Goi, T; Tsuchiyama, T; Nakagawara, G; Urano, T; Shiku, H; Furukawa, K. Expression of DCC protein in colorectal tumors and its relationship to tumor progression and metastasis. Oncology 1999, 56, 134–141. [Google Scholar]
  231. Shibata, D; Reale, MA; Lavin, P; Silverman, M; Fearon, ER; Steele, G, Jr; Jessup, JM; Loda, M; Summerhayes, IC. The DCC protein and prognosis in colorectal cancer. N. Engl. J. Med 1996, 335, 1727–1732. [Google Scholar]
  232. Miyoshi, N; Ishii, H; Mimori, K; Tanaka, F; Nagai, K; Uemura, M; Sekimoto, M; Doki, Y; Mori, M. ATP11A is a novel predictive marker for metachronous metastasis of colorectal cancer. Oncol. Rep 2010, 23, 505–510. [Google Scholar]
  233. Korkaya, H; Wicha, MS. Cancer stem cells: Nature versus nurture. Nat. Cell Biol 2010, 12, 419–421. [Google Scholar]
  234. Papailiou, J; Bramis, KJ; Gazouli, M; Theodoropoulos, G. Stem cells in colon cancer. A new era in cancer theory begins. Int J Colorectal Dis 2010, 1022–6. [Google Scholar] [CrossRef]
  235. Shipitsin, M; Polyak, K. The cancer stem cell hypothesis: In search of definitions, markers, and relevance. Lab. Invest 2008, 88, 459–463. [Google Scholar]
  236. Tan, BT; Park, CY; Ailles, LE; Weissman, IL. The cancer stem cell hypothesis: A work in progress. Lab. Invest 2006, 86, 1203–1207. [Google Scholar]
  237. Vermeulen, L; De Sousa, EMF; van der Heijden, M; Cameron, K; de Jong, JH; Borovski, T; Tuynman, JB; Todaro, M; Merz, C; Rodermond, H; Sprick, MR; Kemper, K; Richel, DJ; Stassi, G; Medema, JP. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat. Cell Biol 2010, 12, 468–476. [Google Scholar]
  238. Wicha, MS; Liu, S; Dontu, G. Cancer stem cells: An old idea--a paradigm shift. Cancer Res 2006, 66, 1883–1890. [Google Scholar]
  239. Pang, R; Law, WL; Chu, AC; Poon, JT; Lam, CS; Chow, AK; Ng, L; Cheung, LW; Lan, XR; Lan, HY; Tan, VP; Yau, TC; Poon, RT; Wong, BC. A subpopulation of CD26+ cancer stem cells with metastatic capacity in human colorectal cancer. Cell Stem Cell 2010, 6, 603–615. [Google Scholar]
  240. Dalerba, P; Dylla, SJ; Park, IK; Liu, R; Wang, X; Cho, RW; Hoey, T; Gurney, A; Huang, EH; Simeone, DM; Shelton, AA; Parmiani, G; Castelli, C; Clarke, MF. Phenotypic characterization of human colorectal cancer stem cells. Proc. Natl. Acad. Sci USA 2007, 104, 10158–10163. [Google Scholar]
  241. Choi, D; Lee, HW; Hur, KY; Kim, JJ; Park, GS; Jang, SH; Song, YS; Jang, KS; Paik, SS. Cancer stem cell markers CD133 and CD24 correlate with invasiveness and differentiation in colorectal adenocarcinoma. World J. Gastroenterol 2009, 15, 2258–2264. [Google Scholar]
  242. Horst, D; Kriegl, L; Engel, J; Kirchner, T; Jung, A. Prognostic significance of the cancer stem cell markers CD133, CD44, and CD166 in colorectal cancer. Cancer Invest 2009, 27, 844–850. [Google Scholar]
  243. Ricci-Vitiani, L; Lombardi, DG; Pilozzi, E; Biffoni, M; Todaro, M; Peschle, C; De Maria, R. Identification and expansion of human colon-cancer-initiating cells. Nature 2007, 445, 111–115. [Google Scholar]
  244. Shmelkov, SV; Butler, JM; Hooper, AT; Hormigo, A; Kushner, J; Milde, T; St. Clair, R; Baljevic, M; White, I; Jin, DK; Chadburn, A; Murphy, AJ; Valenzuela, DM; Gale, NW; Thurston, G; Yancopoulos, GD; D’Angelica, M; Kemeny, N; Lyden, D; Rafii, S. CD133 expression is not restricted to stem cells, and both CD133+ and CD133- metastatic colon cancer cells initiate tumors. J. Clin. Invest 2008, 118, 2111–2120. [Google Scholar]
  245. Kawasaki, BT; Farrar, WL. Cancer stem cells, CD200 and immunoevasion. Trends Immunol 2008, 29, 464–468. [Google Scholar]
  246. Moreaux, J; Veyrune, JL; Reme, T; De Vos, J; Klein, B. CD200: A putative therapeutic target in cancer. Biochem. Biophys. Res. Commun 2008, 366, 117–122. [Google Scholar]
  247. Kawasaki, BT; Mistree, T; Hurt, EM; Kalathur, M; Farrar, WL. Co-expression of the toleragenic glycoprotein, CD200, with markers for cancer stem cells. Biochem. Biophys. Res. Commun 2007, 364, 778–782. [Google Scholar]
  248. Chen, JS; Chen, KT; Fan, CW; Han, CL; Chen, YJ; Yu, JS; Chang, YS; Chien, CW; Wu, CP; Hung, RP; Chan, EC. Comparison of membrane fraction proteomic profiles of normal and cancerous human colorectal tissues with gel-assisted digestion and iTRAQ labeling mass spectrometry. FEBS J 2010, 277, 3028–3038. [Google Scholar]
  249. Krasnov, GS; Oparina, N; Khankin, SL; Mashkova, TD; Ershov, AN; Zatsepina, OG; Karpov, VL; Beresten, SF. Colorectal cancer 2D-proteomics: Identification of altered protein expression. Mol. Biol (Mosk) 2009, 43, 348–356. [Google Scholar]
  250. Mazzanti, R; Solazzo, M; Fantappie, O; Elfering, S; Pantaleo, P; Bechi, P; Cianchi, F; Ettl, A; Giulivi, C. Differential expression proteomics of human colon cancer. Am. J. Physiol. Gastrointest. Liver Physiol 2006, 290, G1329–G1338. [Google Scholar]
  251. Pei, H; Zhu, H; Zeng, S; Li, Y; Yang, H; Shen, L; Chen, J; Zeng, L; Fan, J; Li, X; Gong, Y; Shen, H. Proteome analysis and tissue microarray for profiling protein markers associated with lymph node metastasis in colorectal cancer. J. Proteome Res 2007, 6, 2495–4501. [Google Scholar]
  252. Xing, XM; Wang, YH; Huang, Q; Lu, BJ; Lai, MD. Differential expression of secretagogin and glucose-related protein 78 in colorectal carcinoma: A proteome study. Chin. J. Pathol 2007, 36, 107–112. [Google Scholar]
  253. Lawrie, LC; Curran, S. Laser capture microdissection and colorectal cancer proteomics. Methods Mol. Biol 2005, 293, 245–253. [Google Scholar]
  254. Sheehan, KM; Gulmann, C; Eichler, GS; Weinstein, JN; Barrett, HL; Kay, EW; Conroy, RM; Liotta, LA; Petricoin, EF, III. Signal pathway profiling of epithelial and stromal compartments of colonic carcinoma reveals epithelial-mesenchymal transition. Oncogene 2008, 27, 323–331. [Google Scholar]
  255. Kaufman, KL; Belov, L; Huang, P; Mactier, S; Scolyer, RA; Mann, GJ; Christopherson, RI. An extended antibody microarray for surface profiling metastatic melanoma. J. Immunol Methods 2010, 358, 23–34. [Google Scholar]
  256. Ahram, M; Litou, ZI; Fang, R; Al-Tawallbeh, G. Estimation of membrane proteins in the human proteome. In Silico Biol 2006, 6, 379–386. [Google Scholar]
  257. Bitarte, N; Bandres, E; Zarate, R; Ramirez, N; Garcia-Foncillas, J. Moving forward in colorectal Cancer Res.earch, what proteomics has to tell. World J. Gastroenterol 2007, 13, 5813–5821. [Google Scholar]
  258. Yeoh, LC; Loh, CK; Gooi, BH; Singh, M; Gam, LH. Hydrophobic protein in colorectal cancer in relation to tumor stages and grades. World J. Gastroenterol 2010, 16, 2754–2763. [Google Scholar]
  259. Ikonomou, G; Samiotaki, M; Panayotou, G. Proteomic methodologies and their application in colorectal Cancer Res.earch. Crit. Rev. Clin. Lab. Sci 2009, 46, 319–342. [Google Scholar]
  260. Nibbe, RK; Chance, MR. Approaches to biomarkers in human colorectal cancer: Looking back, to go forward. Biomark Med 2009, 3, 385–396. [Google Scholar]
  261. Soreide, K; Nedrebo, BS; Knapp, JC; Glomsaker, TB; Soreide, JA; Korner, H. Evolving molecular classification by genomic and proteomic biomarkers in colorectal cancer: Potential implications for the surgical oncologist. Surg. Oncol 2009, 18, 31–50. [Google Scholar]
  262. Pepe, MS; Cai, T; Longton, G. Combining predictors for classification using the area under the receiver operating characteristic curve. Biometrics 2006, 62, 221–229. [Google Scholar]
  263. Zlobec, I; Lugli, A. Prognostic and predictive factors in colorectal cancer. J. Clin. Pathol 2008, 61, 561–569. [Google Scholar]
  264. Zlobec, I; Minoo, P; Baumhoer, D; Baker, K; Terracciano, L; Jass, JR; Lugli, A. Multimarker phenotype predicts adverse survival in patients with lymph node-negative colorectal cancer. Cancer 2008, 112, 495–502. [Google Scholar]
  265. Fernebro, E; Bendahl, PO; Dictor, M; Persson, A; Ferno, M; Nilbert, M. Immunohistochemical patterns in rectal cancer: Application of tissue microarray with prognostic correlations. Int. J Cancer 2004, 111, 921–928. [Google Scholar]
  266. Lyall, MS; Dundas, SR; Curran, S; Murray, GI. Profiling markers of prognosis in colorectal cancer. Clin. Cancer Res 2006, 12, 1184–1191. [Google Scholar]
  267. Eckel-Passow, JE; Lohse, CM; Sheinin, Y; Crispen, PL; Krco, CJ; Kwon, ED. Tissue microarrays: One size does not fit all. Diagn. Pathol 2010, 5, 48–57. [Google Scholar]
  268. Lal, S; Brown, A; Nguyen, L; Braet, F; Dyer, W; Dos Remedios, C. Using antibody arrays to detect microparticles from acute coronary syndrome patients based on cluster of differentiation (CD) antigen expression. Mol Cell Proteomics 2009, 8, 799–804. [Google Scholar]
  269. Lugli, A; Karamitopoulou, E; Panayiotides, I; Karakitsos, P; Rallis, G; Peros, G; Iezzi, G; Spagnoli, G; Bihl, M; Terracciano, L; Zlobec, I. CD8+ lymphocytes/tumour-budding index: An independent prognostic factor representing a ‘pro-/anti-tumour’ approach to tumour host interaction in colorectal cancer. Br. J Cancer 2009, 101, 1382–1392. [Google Scholar]
  270. Jang, TJ. Expression of E-cadherin and beta-catenin is altered at tumor budding sites, whose number is associated with the progression of colorectal carcinoma. Korean J. Pathol 2009, 43, 523–527. [Google Scholar]
  271. Hanash, SM; Pitteri, SJ; Faca, VM. Mining the plasma proteome for cancer biomarkers. Nature 2008, 452, 571–579. [Google Scholar]
  272. Hong, BS; Cho, JH; Kim, H; Choi, EJ; Rho, S; Kim, J; Kim, JH; Choi, DS; Kim, YK; Hwang, D; Gho, YS. Colorectal cancer cell-derived microvesicles are enriched in cell cyclerelated mRNAs that promote proliferation of endothelial cells. BMC Genomics 2009, 10, 556. [Google Scholar]
  273. Huber, V; Fais, S; Iero, M; Lugini, L; Canese, P; Squarcina, P; Zaccheddu, A; Colone, M; Arancia, G; Gentile, M; Seregni, E; Valenti, R; Ballabio, G; Belli, F; Leo, E; Parmiani, G; Rivoltini, L. Human colorectal cancer cells induce T-cell death through release of proapoptotic microvesicles: Role in immune escape. Gastroenterology 2005, 128, 1796–804. [Google Scholar]
  274. Mrvar-Brecko, A; Sustar, V; Jansa, V; Stukelj, R; Jansa, R; Mujagic, E; Kruljc, P; Iglic, A; Hagerstrand, H; Kralj-Iglic, V. Isolated microvesicles from peripheral blood and body fluids as observed by scanning electron microscope. Blood Cells Mol. Dis 2010, 44, 307–312. [Google Scholar]
  275. Valenti, R; Huber, V; Iero, M; Filipazzi, P; Parmiani, G; Rivoltini, L. Tumor-released microvesicles as vehicles of immunosuppression. Cancer Res 2007, 67, 2912–2915. [Google Scholar]
  276. Moritz, RL; Skandarajah, AR; Ji, H; Simpson, RJ. Proteomic analysis of colorectal cancer: Prefractionation strategies using two-dimensional free-flow electrophoresis. Comp. Funct Genomics 2005, 6, 236–243. [Google Scholar]
  277. Qiu, Y; Patwa, TH; Xu, L; Shedden, K; Misek, DE; Tuck, M; Jin, G; Ruffin, MT; Turgeon, DK; Synal, S; Bresalier, R; Marcon, N; Brenner, DE; Lubman, DM. Plasma glycoprotein profiling for colorectal cancer biomarker identification by lectin glycoarray and lectin blot. J. Proteome Res 2008, 7, 1693–1703. [Google Scholar]
  278. Drake, PM; Cho, W; Li, B; Prakobphol, A; Johansen, E; Anderson, NL; Regnier, FE; Gibson, BW; Fisher, SJ. Sweetening the pot: Adding glycosylation to the biomarker discovery equation. Clin. Chem 2010, 56, 223–236. [Google Scholar]
  279. Fuster, MM; Esko, JD. The sweet and sour of cancer: Glycans as novel therapeutic targets. Nat. Rev Cancer 2005, 5, 526–542. [Google Scholar]
  280. Reis, CA; Osorio, H; Silva, L; Gomes, C; David, L. Alterations in glycosylation as biomarkers for cancer detection. J. Clin. Pathol 2010, 63, 322–329. [Google Scholar]
  281. Taylor, AD; Hancock, WS; Hincapie, M; Taniguchi, N; Hanash, SM. Towards an integrated proteomic and glycomic approach to finding cancer biomarkers. Genome Med 2009, 1, 57. [Google Scholar]
  282. Wang, H; Tso, VK; Slupsky, CM; Fedorak, RN. Metabolomics and detection of colorectal cancer in humans: A systematic review. Future Oncol 2010, 6, 1395–1406. [Google Scholar]
  283. Nambiar, PR; Gupta, RR; Misra, V. An “Omics” based survey of human colon cancer. Mutat. Res 2010, 693, 3–18. [Google Scholar]
  284. Durai, R; Yang, SY; Seifalian, AM; Winslet, MC. Principles and applications of gene therapy in colon cancer. J. Gastrointest. Liver Dis 2008, 17, 59–67. [Google Scholar]
  285. Habermann, JK; Paulsen, U; Roblick, UJ; Upender, MB; McShane, LM; Korn, EL; Wangsa, D; Kruger, S; Duchrow, M; Bruch, HP; Auer, G; Ried, T. Stage-specific alterations of the genome, transcriptome, and proteome during colorectal carcinogenesis. Genes Chromosomes Cancer 2007, 46, 10–26. [Google Scholar]
Figure 1. Binding patterns of a disaggregated stage II, poorly differentiated, CRC sample on a DotScan™ CRC antibody microarray. (A) Antibody locations for duplicate arrays. Numbers refer to CD antigens; other abbreviations are TCR, T-cell receptor; κ, λ, immunoglobulin light chains; slg, surface immunoglobulin; DCC, deleted in colorectal cancer protein; EGFR, epidermal growth factor receptor; FAP, fibroblast activation protein; HLA-A,B,C and HLA-DR, human leukocyte antigens A,B,C and DR, respectively; MICA, MHC class I chain-related protein A; MMP-14, matrix metallopeptidase 14; PIGR, polymeric immunoglobulin receptor; TSP-1, thrombospondin-1; Mabthera, humanised anti-CD20. Alignment dots around microarray consist of a mixture of CD44 and CD29 antibodies. (B) Optical scan; (C) and (D) fluorescence multiplexing using PE-anti-CD3 and Alexa 647-EpCAM, respectively.
Figure 1. Binding patterns of a disaggregated stage II, poorly differentiated, CRC sample on a DotScan™ CRC antibody microarray. (A) Antibody locations for duplicate arrays. Numbers refer to CD antigens; other abbreviations are TCR, T-cell receptor; κ, λ, immunoglobulin light chains; slg, surface immunoglobulin; DCC, deleted in colorectal cancer protein; EGFR, epidermal growth factor receptor; FAP, fibroblast activation protein; HLA-A,B,C and HLA-DR, human leukocyte antigens A,B,C and DR, respectively; MICA, MHC class I chain-related protein A; MMP-14, matrix metallopeptidase 14; PIGR, polymeric immunoglobulin receptor; TSP-1, thrombospondin-1; Mabthera, humanised anti-CD20. Alignment dots around microarray consist of a mixture of CD44 and CD29 antibodies. (B) Optical scan; (C) and (D) fluorescence multiplexing using PE-anti-CD3 and Alexa 647-EpCAM, respectively.
Ijms 12 00078f1

Share and Cite

MDPI and ACS Style

Belov, L.; Zhou, J.; Christopherson, R.I. Cell Surface Markers in Colorectal Cancer Prognosis. Int. J. Mol. Sci. 2011, 12, 78-113. https://doi.org/10.3390/ijms12010078

AMA Style

Belov L, Zhou J, Christopherson RI. Cell Surface Markers in Colorectal Cancer Prognosis. International Journal of Molecular Sciences. 2011; 12(1):78-113. https://doi.org/10.3390/ijms12010078

Chicago/Turabian Style

Belov, Larissa, Jerry Zhou, and Richard I. Christopherson. 2011. "Cell Surface Markers in Colorectal Cancer Prognosis" International Journal of Molecular Sciences 12, no. 1: 78-113. https://doi.org/10.3390/ijms12010078

Article Metrics

Back to TopTop