301
|
Weissenstein U, Schumann A, Reif M, Link S, Toffol-Schmidt UD, Heusser P. Detection of circulating tumor cells in blood of metastatic breast cancer patients using a combination of cytokeratin and EpCAM antibodies. BMC Cancer 2012; 12:206. [PMID: 22646670 PMCID: PMC3404911 DOI: 10.1186/1471-2407-12-206] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 05/30/2012] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are detectable in peripheral blood of metastatic breast cancer patients (MBC). In this paper we evaluate a new CTC separation method based on a combination of anti-EpCAM- and anti-cytokeratin magnetic cell separation with the aim to improve CTC detection with low target antigen densities. METHODS Blood samples of healthy donors spiked with breast cancer cell line HCC1937 were used to determine accuracy and precision of the method. 10 healthy subjects were examined to evaluate specificity. CTC counts in 59 patients with MBC were measured to evaluate the prognostic value on overall survival. RESULTS Regression analysis of numbers of recovered vs. spiked HCC1937 cells yielded a coefficient of determination of R(2) = 0.957. The average percentage of cell recovery was 84%. The average within-run coefficient of variation for spiking of 185, 85 and 30 cells was 14%. For spiking of 10 cells the within-run CV was 30%. No CTCs were detected in blood of 10 healthy subjects examined. A standard threshold of 5 CTC/7.5 ml blood as a cut-off point between risk groups led to a highly significant prognostic marker (p < 0.001). To assess the prognostic value of medium CTC levels we additionally considered a low (CTC-L: 0 CTC), a medium (CTC-M: 1-4 CTC) and a high risk group (CTC-H: ≥5 CTC). The effect of this CTC-LMH marker on overall survival was significant as well (p < 0.001). A log-ratio test performed to compare the model with 3 vs. the model with 2 risk groups rejected the model with 2 risk groups (p = 0.026). For CTC as a count variable, we propose an offset reciprocal transformation 1/(1 + x) for overall survival prediction (p < 0.001). CONCLUSIONS We show that our CTC detection method is feasible and leads to accurate and reliable results. Our data suggest that a refined differentiation between patients with different CTC levels is reasonable.
Collapse
Affiliation(s)
| | | | - Marcus Reif
- Institute for Clinical Research, Berlin, Germany
| | | | | | - Peter Heusser
- Center for Integrative Medicine, University of Witten/Herdecke, Witten, Germany
| |
Collapse
|
302
|
Heine M, Freund B, Nielsen P, Jung C, Reimer R, Hohenberg H, Zangemeister-Wittke U, Wester HJ, Lüers GH, Schumacher U. High interstitial fluid pressure is associated with low tumour penetration of diagnostic monoclonal antibodies applied for molecular imaging purposes. PLoS One 2012; 7:e36258. [PMID: 22590529 PMCID: PMC3348149 DOI: 10.1371/journal.pone.0036258] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 03/30/2012] [Indexed: 11/19/2022] Open
Abstract
The human epithelial cell adhesion molecule (EpCAM) is highly expressed in a variety of clinical tumour entities. Although an antibody against EpCAM has successfully been used as an adjuvant therapy in colon cancer, this therapy has never gained wide-spread use. We have therefore investigated the possibilities and limitations for EpCAM as possible molecular imaging target using a panel of preclinical cancer models. Twelve human cancer cell lines representing six tumour entities were tested for their EpCAM expression by qPCR, flow cytometry analysis and immunocytochemistry. In addition, EpCAM expression was analyzed in vivo in xenograft models for tumours derived from these cells. Except for melanoma, all cell lines expressed EpCAM mRNA and protein when grown in vitro. Although they exhibited different mRNA levels, all cell lines showed similar EpCAM protein levels upon detection with monoclonal antibodies. When grown in vivo, the EpCAM expression was unaffected compared to in vitro except for the pancreatic carcinoma cell line 5072 which lost its EpCAM expression in vivo. Intravenously applied radio-labelled anti EpCAM MOC31 antibody was enriched in HT29 primary tumour xenografts indicating that EpCAM binding sites are accessible in vivo. However, bound antibody could only be immunohistochemically detected in the vicinity of perfused blood vessels. Investigation of the fine structure of the HT29 tumour blood vessels showed that they were immature and prone for higher fluid flux into the interstitial space. Consistent with this hypothesis, a higher interstitial fluid pressure of about 12 mbar was measured in the HT29 primary tumour via "wick-in-needle" technique which could explain the limited diffusion of the antibody into the tumour observed by immunohistochemistry.
Collapse
Affiliation(s)
- Markus Heine
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
303
|
Szathmari A, Champier J, Ghersi-Egea JF, Jouvet A, Watrin C, Wierinckx A, Fèvre Montange M. Molecular characterization of circumventricular organs and third ventricle ependyma in the rat: potential markers for periventricular tumors. Neuropathology 2012; 33:17-29. [PMID: 22537279 DOI: 10.1111/j.1440-1789.2012.01321.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Circumventricular organs (CVOs) are specialized ventricular structures around the third and fourth ventricles of the brain. In humans, these structures are present during the fetal period and some become vestigial after birth. Some of these organs, such as the pineal gland (PG), subcommissural organ (SCO), and organum vasculosum of the lamina terminalis, might be the sites of origin of periventricular tumors, notably pineal parenchymal tumors, papillary tumor of the pineal region and chordoid glioma. In contrast to the situation in humans, CVOs are present in the adult rat and can be dissected by laser capture microdissection (LCM). In this study, we used LCM and microarrays to analyze the transcriptomes of three CVOs, the SCO, the subfornical organ (SFO), and the PG and the third ventricle ependyma in the adult rat, in order to better characterize these organs at the molecular level. Several genes were expressed only, or mainly, in one of these structures, for example, Erbb2 and Col11a1 in the ependyma, Epcam and Claudin-3 (CLDN3) in the SCO, Ren1 and Slc22a3 in the SFO and Tph, Aanat and Asmt in the PG. The expression of these genes in periventricular tumors should be examined as evidence for a possible origin from the CVOs. Furthermore, we performed an immunohistochemical study of CLDN3, a membrane protein involved in forming cellular tight junctions and found that CLDN3 expression was restricted to the apical pole of ependymocytes in the SCO. This microarray study provides new evidence regarding the possible origin of some rare periventricular tumors.
Collapse
Affiliation(s)
- Alexandru Szathmari
- Fac Med RTH Laennec, Inserm U1028, CNRS UMR5292, Centre de Recherche en Neurosciences, Equipe Neurooncologie et Neuroinflammation, Université de Lyon, Lyon, France
| | | | | | | | | | | | | |
Collapse
|
304
|
Sithambaram D, Palanivelu S, Subramanian K, Sahoo S, Verma RS. Specific targeting of Ep-CAM in various carcinomas by novel monoclonal antibodies. Hybridoma (Larchmt) 2012; 30:511-8. [PMID: 22149275 DOI: 10.1089/hyb.2011.0069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Epithelial cell adhesion molecule (Ep-CAM) is a 40 kDa transmembrane glycoprotein overexpressed in majority of tumor epithelial cells and has a major morphoregulatory function, relevant not only to epithelial tissue development, but also in carcinogenesis and tumor progression. Since Ep-CAM localizes at the cell surface of most carcinomas, the molecule is an attractive target for immunotherapy and several strategies have been deployed to treat cancer using Ep-CAM targeting, including MAb therapy. For improving effective targeting of this protein for diagnostics in various clinical samples, we generated and characterized an anti-Ep-CAM MAb (C4) using recombinant Ep-CAM protein, comprising the highly immunogenic domain. The specificity of C4-MAb was characterized in Ep-CAM positive cell lines (PC3 and MCF-7) by flow cytometry and immunofluorescence. The immunohistochemistry analysis in clinical tissue samples showed specific detection of epithelial antigens in breast, colon, stomach, and prostate carcinomas. Thus, this Ep-CAM MAb (C4-MAb) could be used for both diagnostic and therapeutic applications due to its specificity.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/immunology
- Antibodies, Neoplasm/immunology
- Antibodies, Neoplasm/metabolism
- Antibody Specificity
- Antigens, Neoplasm/immunology
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/diagnosis
- Breast Neoplasms/immunology
- Cell Adhesion Molecules/immunology
- Culture Media
- Electrophoresis, Polyacrylamide Gel
- Epithelial Cell Adhesion Molecule
- Female
- Flow Cytometry
- HL-60 Cells
- Humans
- Hybridomas/immunology
- Immunohistochemistry
- Male
- Mice
- Mice, Inbred BALB C
- Molecular Diagnostic Techniques/methods
- Neoplasms, Glandular and Epithelial/diagnosis
- Neoplasms, Glandular and Epithelial/immunology
- Prostatic Neoplasms/diagnosis
- Prostatic Neoplasms/immunology
- Recombinant Proteins/immunology
- Recombinant Proteins/metabolism
- U937 Cells
Collapse
|
305
|
Hekimian K, Meisezahl S, Trompelt K, Rabenstein C, Pachmann K. Epithelial cell dissemination and readhesion: analysis of factors contributing to metastasis formation in breast cancer. ISRN ONCOLOGY 2012; 2012:601810. [PMID: 22530147 PMCID: PMC3317055 DOI: 10.5402/2012/601810] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 11/30/2011] [Indexed: 12/31/2022]
Abstract
Although considerable progress has been achieved in breast cancer diagnosis and treatment, the live-saving effect of mammography has hardly been measurable and the benefit of taxanes regarded as highly active is still a matter of debate, possibly because treatment effects have hitherto been mainly determined from the solid part of the tumor, due to lack of measurability of the systemic part of the disease. Here, we have quantified the influence on the systemic disease, cells mobilized from the solid tumor. Increased numbers of circulating epithelial cells were observed in screened individuals and still higher numbers in breast cancer patients with repeated mammograms as compared to mammogram naïve individuals. Taxanes as part of the subsequent systemic treatment led to mobilization of tumor suspect cells in up to 78% cases and the majority of relapses have occurred in these patients. Surgery-induced activation of disseminated cells may additionally contribute to metastasis formation.
Collapse
Affiliation(s)
- Katya Hekimian
- Department of Experimental Heamatology and Oncology, Clinic for Internal Medicine II, Friedrich-Schiller-University Jena, 07747 Jena, Germany
| | | | | | | | | |
Collapse
|
306
|
EpCAM regulates cell cycle progression via control of cyclin D1 expression. Oncogene 2012; 32:641-50. [PMID: 22391566 DOI: 10.1038/onc.2012.75] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The epithelial cell adhesion molecule (EpCAM) is an integral transmembrane protein that is frequently overexpressed in embryonic stem cells, tissue progenitors, carcinomas and cancer-initiating cells. In cancer cells, expression of EpCAM is associated with enhanced proliferation and upregulation of target genes including c-myc. However, the exact molecular mechanisms underlying the observed EpCAM-dependent cell proliferation remained unexplored. Here, we show that EpCAM directly affects cell cycle progression via its capacity to regulate the expression of cyclin D1 at the transcriptional level and depending on the direct interaction partner FHL2 (four-and-a-half LIM domains protein 2). As a result, downstream events such as phosphorylation of the retinoblastoma protein (Rb) and expression of cyclins E and A are similarly affected. In vivo, EpCAM expression strength and pattern are both positively correlated with the proliferation marker Ki67, high expression and nuclear localisation of cyclin D1, and Rb phosphorylation. Thus, EpCAM enhances cell cycle progression via the classical cyclin-regulated pathway.
Collapse
|
307
|
Glait-Santar C, Pasmanik-Chor M, Oron-Karni V, Benayahu D. Molecular profiling of functional interactions between pre-osteoblastic and breast carcinoma cells. Genes Cells 2012; 17:302-15. [DOI: 10.1111/j.1365-2443.2012.01590.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
308
|
Stübke K, Wicklein D, Herich L, Schumacher U, Nehmann N. Selectin-deficiency reduces the number of spontaneous metastases in a xenograft model of human breast cancer. Cancer Lett 2012; 321:89-99. [PMID: 22366582 DOI: 10.1016/j.canlet.2012.02.019] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 02/16/2012] [Accepted: 02/16/2012] [Indexed: 01/02/2023]
Abstract
Metastasis formation is a complex process still poorly understood. Previous work in a colon cancer xenograft model showed that E(ndothelial) and P(latelet) selectins mediate spontaneous metastasis to the lungs. To investigate the functional role of selectins in breast cancer, human DU4475 breast cancer cells were injected subcutaneously into pfp-/-rag2-/- mice and in all their selectin-deficient variants (EP-/-, E-/- and P-/-). Pfp-/-rag2-/- mice as well as all their selectin-deficient variants developed primary tumours and spontaneous metastases. Compared with the wild-type mice, disseminated tumours cells were significantly lower (74% reduction, P=0.046) in the bone marrow of selectin-deficient mice. Pfp-/-rag2-/- mice developed significantly higher numbers of lung metastases (6644.83±741.77) than the E-/- (4053.33±112.58; P=0.002) and the EP-/- pfp-/-rag2-/- mice (4665.65±754.50; P<0.001). The results indicate that E- and P-selectins play a role in spontaneous metastasis formation both into bone marrow and lungs. However, spontaneous metastasis was not completely abrogated, hence additional cell adhesion molecules must be involved in the metastatic spread.
Collapse
Affiliation(s)
- Katrin Stübke
- University Medical Center Hamburg-Eppendorf, Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, Hamburg, Germany.
| | | | | | | | | |
Collapse
|
309
|
de Albuquerque A, Kaul S, Breier G, Krabisch P, Fersis N. Multimarker Analysis of Circulating Tumor Cells in Peripheral Blood of Metastatic Breast Cancer Patients: A Step Forward in Personalized Medicine. ACTA ACUST UNITED AC 2012; 7:7-12. [PMID: 22553466 DOI: 10.1159/000336548] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
AIM: To develop an immunomagnetic assay for the isolation of circulating tumor cells (CTCs) followed by the analysis of a multimarker panel, which will enable the characterization of these malignant cells with high accuracy. PATIENTS AND METHODS: Peripheral blood (PB) was collected from 32 metastatic breast cancer patients and 42 negative controls. The antibodies BM7 and VU1D9 were used for immunomagnetic tumor cell enrichment. A real-time reverse transcription-polymerase chain reaction (RT-PCR) approach for the markers KRT19, SCGB2A2, MUC1, EPCAM, BIRC5 and ERBB2 was used for CTC detection and characterization. RESULTS: THE POSITIVITY RATES FOR EACH MARKER WERE AS FOLLOWS: 46.9% for KRT19, 25.0% for SCGB2A2, 28.1% for MUC1, 28.1% for EPCAM, 21.9% for BIRC5, and 15.6% for ERBB2. After the creation of individualized cutoffs, the sensitivity and specificity of the combined marker gene panel increased to 56.3% and 100%, respectively. Interestingly, 27.0% of the HER2-negative tumor patients showed ERBB2 mRNA-positive CTCs. CONCLUSIONS: The described technique can be used to measure CTCs with great accuracy. The use of a multimarker panel for the characterization of CTCs may provide real-time information and be of great value in therapy monitoring.
Collapse
Affiliation(s)
- Andreia de Albuquerque
- Department of Pathology, Faculty of Medicine Carl Gustav Carus, University of Dresden, Klinikum Chemnitz, Germany
| | | | | | | | | |
Collapse
|
310
|
Functional polymorphism in the EpCAM gene is associated with occurrence and advanced disease status of cervical cancer in Chinese population. Mol Biol Rep 2012; 39:7303-9. [PMID: 22322561 DOI: 10.1007/s11033-012-1560-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 01/24/2012] [Indexed: 10/14/2022]
Abstract
The epithelial cell adhesion molecule (EpCAM) was originally identified as a tumor associated antigen, attributable to its high expression on rapidly proliferating tumors of epithelial origin. EpCAM plays vital roles in carcinogenesis, tumor progression and metastasis in most tumors. A non-synonymous polymorphism (rs1126497 C/T) was found in exon 3 of EpCAM, which cause a transition from 115 Met to 115 Thr. Another polymorphism (rs1421 A/G) in the 3'UTR causes loss of has-miR-1183 binding. We performed a multiple independent case-control analysis to assess the association between EpCAM genotypes and cervical cancer risk. Genotyping a total of 518 patients with cervical cancer and 723 control subjects in a Chinese population, we observed that the variant EpCAM genotypes (rs1126497 CT, and TT) were associated with substantially increased risk of cervical cancer. Compared with the rs1126497 CC genotype, CT genotype had a significantly increased risk of cervical cancer (Crude OR = 1.70; 95% CI = 1.33-2.20; adjusted OR = 1.72; 95% CI = 1.33-2.22), the TT carriers had a further increased risk of cervical cancer (Crude OR = 1.94; 95% CI = 1.01-3.72; adjusted OR = 1.96; 95%CI = 1.01-3.81), and there was a trend for an allele dose effect on risk of cervical cancer (P < 0.001). Moreover, the allele T increases the risk for invasive disease or metastatic disease, compared with C allele. However, there exists no significant difference in genotype frequencies of rs1421 A/G site between cases and controls (P = 0.798). These findings suggest that rs1126497 C/T polymorphism in EpCAM may be a genetic modifier for developing cervical cancer.
Collapse
|
311
|
Hekimian K, Stein EL, Pachmann U, Pachmann K. Demasking of epithelial cell adhesion molecule (EpCAM) on circulating epithelial tumor cells by Tween®20 treatment in breast cancer patients. Clin Chem Lab Med 2012; 50:701-8. [DOI: 10.1515/cclm.2011.812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 11/15/2011] [Indexed: 11/15/2022]
Abstract
AbstractThe epithelial cell adhesion molecule (EpCAM) embedded in the plasma membrane of circulating epithelial tumor cells (CETC) is used for detection and enrichment of circulating tumor cells in peripheral blood and as a target for anti-epithelial antibodies elicited during immune response in anti-tumor immunization. Although an efficient immune response against EpCAM can be generated, the clinical application of such approaches has not been successful so far and the detection of circulating epithelial cells is highly variable. One reason for these discrepancies may be that not all circulating tumor cells are equally accessible for the specific antibody. A possible reason might be masking of EpCAM by glycoproteins or membrane lipoproteins preventing antibody binding.We have tested the application of detergents as demasking agents known to be successful in demasking red blood cell epitopes and determined how and in which way they affect integral membrane proteins and membrane lipids.The results showed that the polysorbate TweenThe data presented in this study suggest that EpCAM is present on part of circulating tumor cells in a masked form and that it is possible to demask EpCAM on CETC of breast cancer patients using Tween
Collapse
|
312
|
Yen TY, Macher BA, McDonald CA, Alleyne-Chin C, Timpe LC. Glycoprotein profiles of human breast cells demonstrate a clear clustering of normal/benign versus malignant cell lines and basal versus luminal cell lines. J Proteome Res 2011; 11:656-67. [PMID: 22106898 DOI: 10.1021/pr201041j] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Gene expression profiling has defined molecular subtypes of breast cancer including those identified as luminal and basal. To determine if glycoproteins distinguish various subtypes of breast cancer, we obtained glycoprotein profiles from 14 breast cell lines. Unsupervised hierarchical cluster analysis demonstrated that the glycoprotein profiles obtained can serve as molecular signatures to classify subtypes of breast cancer, as well as to distinguish normal and benign breast cells from breast cancer cells. Statistical analyses were used to identify glycoproteins that are overexpressed in normal versus cancer breast cells, and those that are overexpressed in luminal versus basal breast cancer. Among the glycoproteins distinguishing normal breast cells from cancer cells are several proteins known to be involved in cell adhesion, including proteins previously identified as being altered in breast cancer. Basal breast cancer cell lines overexpressed a number of CD antigens, including several integrin subunits, relative to luminal breast cancer cell lines, whereas luminal breast cancer cells overexpressed carbonic anhydrase 12, clusterin, and cell adhesion molecule 1. The differential expression of glycoproteins in these breast cancer cell lines readily allows the classification of the lines into normal, benign, malignant, basal, and luminal groups.
Collapse
Affiliation(s)
- Ten-Yang Yen
- Department of Chemistry and Biochemistry, San Francisco State University , San Francisco, California 94132, United States
| | | | | | | | | |
Collapse
|
313
|
Gires O. Lessons from common markers of tumor-initiating cells in solid cancers. Cell Mol Life Sci 2011; 68:4009-22. [PMID: 21786143 PMCID: PMC11114982 DOI: 10.1007/s00018-011-0772-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 07/04/2011] [Accepted: 07/07/2011] [Indexed: 12/14/2022]
Abstract
Tumor-initiating cells (TICs) have emerged as the driving force of carcinomas, which appear as hierarchically structured. TICs as opposed to the tumor bulk display tumor forming potential, which is linked to a certain degree of self-renewal and differentiation, both major features of stem cells. Markers such as CD44, CD133, CD24, EpCAM, CD166, Lgr5, CD47, and ALDH have been described, which allow for the prospective enrichment of TICs. It is conspicuous that the same markers allow for an enrichment of TICs in various entities and, on the other hand, that different combinations of these markers were independently reported for the same tumor entity. Potential functions of these markers in the regulation of TIC phenotypes remained somewhat neglected although they might give insights in common molecular themes of TICs. The present review discusses major TIC markers with respect to their function and potential contributions to the tumorigenic phenotype of TICs.
Collapse
MESH Headings
- AC133 Antigen
- Aldehyde Dehydrogenase/metabolism
- Aldehyde Dehydrogenase/physiology
- Antigens, CD/metabolism
- Antigens, CD/physiology
- Antigens, Neoplasm/metabolism
- Antigens, Neoplasm/physiology
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/physiology
- CD24 Antigen/metabolism
- CD24 Antigen/physiology
- CD47 Antigen/metabolism
- CD47 Antigen/physiology
- Cell Adhesion Molecules/metabolism
- Cell Adhesion Molecules/physiology
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Adhesion Molecules, Neuronal/physiology
- Epithelial Cell Adhesion Molecule
- Fetal Proteins/metabolism
- Fetal Proteins/physiology
- Glycoproteins/metabolism
- Glycoproteins/physiology
- Humans
- Hyaluronan Receptors/metabolism
- Hyaluronan Receptors/physiology
- Models, Biological
- Neoplasms/metabolism
- Neoplasms/pathology
- Peptides/metabolism
- Peptides/physiology
- Phenotype
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/physiology
Collapse
Affiliation(s)
- Olivier Gires
- Department of Otorhinolaryngology, Head and Neck Surgery, Grosshadern Medical Center, Ludwig-Maximilians-University of Munich, Marchioninistr. 15, 81377 Munich, Germany.
| |
Collapse
|
314
|
Sankpal NV, Mayfield JD, Willman MW, Fleming TP, Gillanders WE. Activator protein 1 (AP-1) contributes to EpCAM-dependent breast cancer invasion. Breast Cancer Res 2011; 13:R124. [PMID: 22132731 PMCID: PMC3326566 DOI: 10.1186/bcr3070] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 11/02/2011] [Accepted: 12/01/2011] [Indexed: 01/24/2023] Open
Abstract
Introduction EpCAM is a cell-surface glycoprotein that is overexpressed in the majority of epithelial carcinomas. However, the functional role of EpCAM in regulating cancer invasion remains controversial, and the mechanism(s) underlying EpCAM-mediated regulation of breast cancer invasion remain to be defined. Methods EpCAM expression was manipulated in breast cancer cell lines using RNA interference and cDNA expression constructs. Recombinant EpCAM was used to rescue EpCAM signaling following specific ablation of EpCAM. Protein and gene expression, invasion, transcription factor activity, and protein phosphorylation were measured using standard molecular biology techniques. Results In loss-of-function, and gain-of-function experiments we demonstrate that EpCAM expression is associated with increased breast cancer invasion in vitro and in vivo. We demonstrate further that specific ablation of EpCAM expression is associated with decreased activator protein-1 (AP-1) transcription factor activity. Phosphoprotein analyses confirm that specific ablation of EpCAM is associated with decreased phosphorylation of the AP-1 subunit c-Jun. Recombinant soluble extracellular EpCAM (rEpCAM) is able to rescue invasion, AP-1 transcription factor activity, and c-Jun phosphorylation in a dose-dependent fashion. Pharmacologic inhibitors, and constitutively active constructs of the c-Jun N-terminal kinase (JNK) signal transduction pathway, suggest that the impact of EpCAM expression on AP-1 transcription factor activity is mediated through the JNK pathway. In functional rescue experiments, forced expression of c-Jun rescues invasion in breast cancer cells following specific ablation of EpCAM. Conclusions These data demonstrate for the first time that EpCAM expression can influence the JNK/AP-1 signal transduction pathway, and suggest that modulation of AP-1 transcription factor activity contributes to EpCAM-dependent breast cancer invasion. These data have important implications for the design and application of molecular therapies targeting EpCAM.
Collapse
Affiliation(s)
- Narendra V Sankpal
- Department of Surgery, Washington University School of Medicine, Campus Box 8109, 660 South Euclid Avenue, St, Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
315
|
Andreopoulou E, Yang LY, Rangel KM, Reuben JM, Hsu L, Krishnamurthy S, Valero V, Fritsche HA, Cristofanilli M. Comparison of assay methods for detection of circulating tumor cells in metastatic breast cancer: AdnaGen AdnaTest BreastCancer Select/Detect™ versus Veridex CellSearch™ system. Int J Cancer 2011; 130:1590-7. [PMID: 21469140 DOI: 10.1002/ijc.26111] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 02/03/2011] [Indexed: 12/17/2022]
Abstract
The detection of CTCs prior to and during therapy is an independent and strong prognostic marker, and it is predictive of poor treatment outcome. A major challenge is that different technologies are available for isolation and characterization of CTCs in peripheral blood (PB). We compare the CellSearch system and AdnaTest BreastCancer Select/Detect, to evaluate the extent that these assays differ in their ability to detect CTCs in the PB of MBC patients. CTCs in 7.5 ml of PB were isolated and enumerated using the CellSearch, before new treatment. Two cutoff values of ≥2 and ≥5 CTCs/7.5 ml were used. AdnaTest requires 5 ml of PB to detect gene transcripts of tumor markers (GA733-2, MUC-1, and HER2) by RT-PCR. AdnaTest was scored positive if ≥1 of the transcript PCR products for the 3 markers were detected at a concentration ≥0.15 ng/μl. A total of 55 MBC patients were enrolled. 26 (47%) patients were positive for CTCs by the CellSearch (≥2 cutoff), while 20 (36%) were positive (≥5 cutoff). AdnaTest was positive in 29 (53%) with the individual markers being positive in 18% (GA733-2), 44% (MUC-1), and 35% (HER2). Overall positive agreement was 73% for CTC≥2 and 69% for CTC≥5. These preliminary data suggest that the AdnaTest has equivalent sensitivity to that of the CellSearch system in detecting 2 or more CTCs. While there is concordance between these 2 methods, the AdnaTest complements the CellSearch system by improving the overall CTC detection rate and permitting the assessment of genomic markers in CTCs.
Collapse
Affiliation(s)
- E Andreopoulou
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
316
|
Horio M, Sato M, Takeyama Y, Elshazley M, Yamashita R, Hase T, Yoshida K, Usami N, Yokoi K, Sekido Y, Kondo M, Toyokuni S, Gazdar AF, Minna JD, Hasegawa Y. Transient but not stable ZEB1 knockdown dramatically inhibits growth of malignant pleural mesothelioma cells. Ann Surg Oncol 2011; 19 Suppl 3:S634-45. [PMID: 22086445 DOI: 10.1245/s10434-011-2142-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Indexed: 01/08/2023]
Abstract
BACKGROUND The role of ZEB1, a master epithelial-to-mesenchymal transition gene, in malignant pleural mesothelioma (MPM) is unclear. METHODS The expression of ZEB1, E-cadherin, vimentin, and epithelial cell adhesion molecule (EpCAM) in 18 MPM cell lines and a normal pleural mesothelial cell line MeT-5A was determined by quantitative real-time polymerase chain reaction and Western blot testing. RNA interference-mediated transient and/or stable knockdown of ZEB1 and EpCAM was performed. Microarray expression analysis was performed with a TORAY-3D gene chip. Growth was evaluated by colorimetric proliferation and colony formation assays. Luciferase reporter assay was performed to access the effects of ZEB1 knockdown on EpCAM promoter activity. RESULTS Most MPM cell lines exhibited mesenchymal phenotype and expressed ZEB1. Transient ZEB1 knockdown suppressed growth in all four cell lines studied (ACC-MESO-1, H2052, Y-MESO-8A, Y-MESO-29) while stable ZEB1 knockdown suppressed growth only in Y-MESO-29. Genome-wide gene expression analysis revealed that EpCAM was the most prominently up-regulated gene by both transient and stable ZEB1 knockdown in ACC-MESO-1, with more marked up-regulation in stable knockdown. We hypothesized that EpCAM up-regulation counteracts the stable ZEB1 knockdown-induced growth inhibition in ACC-MESO-1. Transient EpCAM knockdown suppressed growth dramatically in ACC-MESO-1 cells expressing shZEB1 but only modestly in those expressing shGFP, supporting our hypothesis. Luciferase reporter assay showed that ZEB1 knockdown resulted in increased EpCAM promoter activity. EpCAM was also up-regulated in Y-MESO-29 expressing shZEB1, but this EpCAM up-regulation did not counteract ZEB1knockdown-induced growth suppression, suggesting that the counteracting effects of EpCAM may be cellular context dependent. CONCLUSIONS RNA interference-mediated ZEB1 knockdown may be a promising therapeutic strategy for MPM, but one has to consider the possibility of diminished growth inhibitory effects of long-term ZEB1 knockdown, possibly as a result of EpCAM up-regulation and/or other gene expression changes resulting from ZEB1 knockdown.
Collapse
Affiliation(s)
- Mihoko Horio
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
317
|
Han ME, Jeon TY, Hwang SH, Lee YS, Kim HJ, Shim HE, Yoon S, Baek SY, Kim BS, Kang CD, Oh SO. Cancer spheres from gastric cancer patients provide an ideal model system for cancer stem cell research. Cell Mol Life Sci 2011; 68:3589-605. [PMID: 21448722 PMCID: PMC11114917 DOI: 10.1007/s00018-011-0672-z] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 02/06/2011] [Accepted: 02/24/2011] [Indexed: 12/14/2022]
Abstract
Cancer stem cells have been hypothesized to drive the growth and metastasis of tumors. Because they need to be targeted for cancer treatment, they have been isolated from many solid cancers. However, cancer stem cells from primary human gastric cancer tissues have not been isolated as yet. For the isolation, we used two cell surface markers: the epithelial cell adhesion molecule (EpCAM) and CD44. When analyzed by flow cytometry, the EpCAM(+)/CD44(+) population accounts for 4.5% of tumor cells. EpCAM(+)/CD44(+) gastric cancer cells formed tumors in immunocompromised mice; however, EpCAM(-)/CD44(-), EpCAM(+)/CD44(-) and EpCAM(-)/CD44(+) cells failed to do so. Xenografts of EpCAM(+)/CD44(+) gastric cancer cells maintained a differentiated phenotype and reproduced the morphological and phenotypical heterogeneity of the original gastric tumor tissues. The tumorigenic subpopulation was serially passaged for several generations without significant phenotypic alterations. Moreover, EpCAM(+)/CD44(+), but not EpCAM(-)/CD44(-), EpCAM(+)/CD44(-) or EpCAM(-)/CD44(+) cells grew exponentially in vitro as cancer spheres in serum-free medium, maintaining the tumorigenicity. Interestingly, a single cancer stem cell generated a cancer sphere that contained various differentiated cells, supporting multi-potency and self-renewal of a cancer stem cell. EpCAM(+)/CD44(+) cells had greater resistance to anti-cancer drugs than other subpopulation cells. The above in vivo and in vitro results suggest that cancer stem cells, which are enriched in the EpCAM(+)/CD44(+) subpopulation of gastric cancer cells, provide an ideal model system for cancer stem cell research.
Collapse
Affiliation(s)
- Myoung-Eun Han
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Republic of Korea
| | - Tae-Yong Jeon
- Department of Surgery, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sun-Hwi Hwang
- Department of Surgery, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Young-Suk Lee
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
| | - Hyun-Jung Kim
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Republic of Korea
| | - Hye-Eun Shim
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Republic of Korea
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
| | - Sun-Yong Baek
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
| | - Bong-Seon Kim
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
| | - Chi-Dug Kang
- Department of Biochemistry, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, Gyeongsangnam-do 626-870 Republic of Korea
- Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
318
|
Zheng X, Cheung LSL, Schroeder JA, Jiang L, Zohar Y. Cell receptor and surface ligand density effects on dynamic states of adhering circulating tumor cells. LAB ON A CHIP 2011; 11:3431-9. [PMID: 21853194 PMCID: PMC6765388 DOI: 10.1039/c1lc20455f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Dynamic states of cancer cells moving under shear flow in an antibody-functionalized microchannel are investigated experimentally and theoretically. The cell motion is analyzed with the aid of a simplified physical model featuring a receptor-coated rigid sphere moving above a solid surface with immobilized ligands. The motion of the sphere is described by the Langevin equation accounting for the hydrodynamic loadings, gravitational force, receptor-ligand bindings, and thermal fluctuations; the receptor-ligand bonds are modeled as linear springs. Depending on the applied shear flow rate, three dynamic states of cell motion have been identified: (i) free motion, (ii) rolling adhesion, and (iii) firm adhesion. Of particular interest is the fraction of captured circulating tumor cells, defined as the capture ratio, via specific receptor-ligand bonds. The cell capture ratio decreases with increasing shear flow rate with a characteristic rate. Based on both experimental and theoretical results, the characteristic flow rate increases monotonically with increasing either cell-receptor or surface-ligand density within certain ranges. Utilizing it as a scaling parameter, flow-rate dependent capture ratios for various cell-surface combinations collapse onto a single curve described by an exponential formula.
Collapse
Affiliation(s)
- Xiangjun Zheng
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
| | - Luthur Siu Lun Cheung
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
| | - Joyce A. Schroeder
- Department of Molecular and Cellular Biology, the University of Arizona, Tucson, AZ, USA
- Arizona Cancer Center, the University of Arizona, Tucson, AZ, USA
- BIO5 Innstitute, the University of Arizona, Tucson, AZ, USA
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
- College of Optical Science, the University of Arizona, Tucson, AZ, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
- Arizona Cancer Center, the University of Arizona, Tucson, AZ, USA
- BIO5 Innstitute, the University of Arizona, Tucson, AZ, USA
- Department of Biomedical Engineering, the University of Arizona, Tucson, AZ, USA
| |
Collapse
|
319
|
Agboola AJ, Paish EC, Rakha EA, Powe DG, Macmillan RD, Ellis IO, Green AR. EpCAM expression is an indicator of recurrence in basal-like breast cancer. Breast Cancer Res Treat 2011; 133:575-82. [DOI: 10.1007/s10549-011-1813-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 09/29/2011] [Indexed: 02/06/2023]
|
320
|
Zheng X, Cheung LSL, Schroeder JA, Jiang L, Zohar Y. A high-performance microsystem for isolating circulating tumor cells. LAB ON A CHIP 2011; 11:3269-76. [PMID: 21837324 PMCID: PMC6765387 DOI: 10.1039/c1lc20331b] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
A unique flow field pattern in a bio-functional microchannel is utilized to significantly enhance the performance of a microsystem developed for selectively isolating circulating tumor cells from cell suspensions. For high performance of such systems, disposal of maximum non-target species is just as important as retention of maximum target species; unfortunately, most studies ignore or fail to report this aspect. Therefore, sensitivity and specificity are introduced as quantitative criteria to evaluate the system performance enabling a direct comparison among systems employing different techniques. The newly proposed fluidic scheme combines a slow flow field, for maximum target-cell attachment, followed by a faster flow field, for maximum detachment of non-target cells. Suspensions of homogeneous or binary mixtures of circulating breast tumor cells, with varying relative concentrations, were driven through antibody-functionalized microchannels. Either EpCAM or cadherin-11 transmembrane receptors were targeted to selectively capture target cells from the suspensions. Cadherin-11-expressing MDA-MB-231 cancer cells were used as target cells, while BT-20 cells were used as non-target cells as they do not express cadherin-11. The attachment and detachment of these two cell lines are characterized, and a two-step attachment/detachment flow field pattern is implemented to enhance the system performance in capturing target cells from binary mixtures. While the system sensitivity remains high, above 0.95, the specificity increases from about 0.85 to 0.95 solely due to the second detachment step even for a 1 : 1000 relative concentration of the target cells.
Collapse
Affiliation(s)
- Xiangjun Zheng
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
| | - Luthur Siu Lun Cheung
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
| | - Joyce A. Schroeder
- Department of Molecular and Cellular Biology, the University of Arizona, Tucson, AZ, USA
- Arizona Cancer Center, the University of Arizona, Tucson, AZ, USA
- BIO5 Innstitute, the University of Arizona, Tucson, AZ, USA
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
- College of Optical Science, the University of Arizona, Tucson, AZ, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
- Arizona Cancer Center, the University of Arizona, Tucson, AZ, USA
- BIO5 Innstitute, the University of Arizona, Tucson, AZ, USA
- Department of Biomedical Engineering, the University of Arizona, Tucson, AZ, USA
| |
Collapse
|
321
|
Abstract
Malignant ascites are the cancer-associated accumulation of fluids in the peritoneal cavity. The neoplasms most frequently associated with ascites are ovarian, breast, colon, stomach and pancreas adenocarcinomas. Symptoms are abdominal distention, nausea, vomiting, anorexia, dyspnea and limbs oedemas. Several pathophysiological mechanisms might be implicated such as peritoneal carcinomatosis, lymphatic vessels' obstruction, portal hypertension or heart failure. Its diagnosis is most often performed in a context of already known neoplasia. Malignant ascites are associated with a pejorative evolution. Ascites which cannot be mobilized or show early recurrence and cannot be prevented by medical treatment are defined as refractory ascites. Therefore, management of refractory malignant ascites takes place in the context of palliative care and aims at improving the quality of life of these patients. This review lists the current data reported on the pathophysiology of malignant ascites and describes the present and future options for refractory malignant ascites management.
Collapse
|
322
|
Palmer TD, Ashby WJ, Lewis JD, Zijlstra A. Targeting tumor cell motility to prevent metastasis. Adv Drug Deliv Rev 2011; 63:568-81. [PMID: 21664937 PMCID: PMC3132821 DOI: 10.1016/j.addr.2011.04.008] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2011] [Revised: 03/31/2011] [Accepted: 04/05/2011] [Indexed: 01/15/2023]
Abstract
Mortality and morbidity in patients with solid tumors invariably result from the disruption of normal biological function caused by disseminating tumor cells. Tumor cell migration is under intense investigation as the underlying cause of cancer metastasis. The need for tumor cell motility in the progression of metastasis has been established experimentally and is supported empirically by basic and clinical research implicating a large collection of migration-related genes. However, there are few clinical interventions designed to specifically target the motility of tumor cells and adjuvant therapy to specifically prevent cancer cell dissemination is severely limited. In an attempt to define motility targets suitable for treating metastasis, we have parsed the molecular determinants of tumor cell motility into five underlying principles including cell autonomous ability, soluble communication, cell-cell adhesion, cell-matrix adhesion, and integrating these determinants of migration on molecular scaffolds. The current challenge is to implement meaningful and sustainable inhibition of metastasis by developing clinically viable disruption of molecular targets that control these fundamental capabilities.
Collapse
Affiliation(s)
- Trenis D. Palmer
- Department of Pathology, Vanderbilt University, C2104A Medical Center North 1161 21 Ave. S., Nashville TN, 37232
| | - William J. Ashby
- Department of Pathology, Vanderbilt University, C2104A Medical Center North 1161 21 Ave. S., Nashville TN, 37232
| | - John D. Lewis
- London Regional Cancer Program, London Health Science Centre, A4-823 790 Commissioners Rd E London ON, N6A 4L6
| | - Andries Zijlstra
- Department of Pathology, Vanderbilt University, C2104A Medical Center North 1161 21 Ave. S., Nashville TN, 37232
| |
Collapse
|
323
|
van der Gun BTF, de Groote ML, Kazemier HG, Arendzen AJ, Terpstra P, Ruiters MHJ, McLaughlin PMJ, Rots MG. Transcription factors and molecular epigenetic marks underlying EpCAM overexpression in ovarian cancer. Br J Cancer 2011; 105:312-9. [PMID: 21694727 PMCID: PMC3142811 DOI: 10.1038/bjc.2011.231] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/19/2011] [Accepted: 05/24/2011] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The epithelial cell adhesion molecule (EpCAM) is overexpressed on carcinomas, and its downregulation inhibits the oncogenic potential of multiple tumour types. Here, we investigated underlying mechanisms of epcam overexpression in ovarian carcinoma. METHODS Expression of EpCAM and DNA methylation (bisulphite sequencing) was determined for ovarian cancer cell lines. The association of histone modifications and 16 transcription factors with the epcam promoter was analysed by chromatin immunoprecipitation. Treatment with 5-Aza-2'-deoxycytidine (5-AZAC) was used to induce EpCAM expression. RESULTS Expression of EpCAM was correlated with DNA methylation and histone modifications. Treatment with 5-AZAC induced EpCAM expression in negative cells. Ten transcription factors were associated with the epcam gene in EpCAM expressing cells, but not in EpCAM-negative cells. Methylation of an Sp1 probe inhibited the binding of nuclear extract proteins in electromobility shift assays; such DNA methylation sensitivity was not observed for an NF-κB probe. CONCLUSION This study provides insights in transcriptional regulation of epcam in ovarian cancer. Epigenetic parameters associated with EpCAM overexpression are potentially reversible, allowing novel strategies for sustained silencing of EpCAM expression.
Collapse
Affiliation(s)
- B T F van der Gun
- Epigenetic Editing, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - M L de Groote
- Epigenetic Editing, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - H G Kazemier
- Epigenetic Editing, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - A J Arendzen
- Epigenetic Editing, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - P Terpstra
- Epidemiology, Department of Genetic Epidemiology and Bioinformatics, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - M H J Ruiters
- Epigenetic Editing, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Synvolux Therapeutics Inc., LJ. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - P M J McLaughlin
- Epigenetic Editing, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - M G Rots
- Epigenetic Editing, Department of Pathology and Medical Biology, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
324
|
Hase T, Sato M, Yoshida K, Girard L, Takeyama Y, Horio M, Elshazley M, Oguri T, Sekido Y, Shames DS, Gazdar AF, Minna JD, Kondo M, Hasegawa Y. Pivotal role of epithelial cell adhesion molecule in the survival of lung cancer cells. Cancer Sci 2011; 102:1493-500. [PMID: 21535318 DOI: 10.1111/j.1349-7006.2011.01973.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is overexpressed in a wide variety of human cancers including lung cancer, and its contribution to increased proliferation through upregulation of cell cycle accelerators such as cyclins A and E has been well established in breast and gastric cancers. Nevertheless, very little is known about its role in supporting the survival of cancer cells. In addition, the functional role of EpCAM in the pathogenesis of lung cancer remains to be explored. In this study, we show that RNAi-mediated knockdown of EpCAM suppresses proliferation and clonogenic growth of three EpCAM-expressing lung cancer cell lines (H3255, H358, and HCC827), but does not induce cell cycle arrest in any of these. In addition, EpCAM knockdown inhibits invasion in the highly invasive H358 but not in less invasive H3255 cells in a Transwell assay. Of note, the EpCAM knockdown induces massive apoptosis in the three cell lines as well as in another EpCAM-expressing lung cancer cell line, HCC2279, but to a much lesser extent in a cdk4/hTERT immortalized normal human bronchial epithelial cell line, HBEC4, suggesting that EpCAM could be a therapeutic target for lung cancer. Finally, EpCAM knockdown partially restores contact inhibition in HCC827, in association with p27(Kip1) upregulation. These results indicate that EpCAM could contribute substantially to the pathogenesis of lung cancer, especially cancer cell survival, and suggest that EpCAM targeted therapy for lung cancer may have potential.
Collapse
Affiliation(s)
- Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Epithelial cell adhesion molecule expression (CD326) in cancer: a short review. Cancer Treat Rev 2011; 38:68-75. [PMID: 21576002 DOI: 10.1016/j.ctrv.2011.04.002] [Citation(s) in RCA: 337] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 04/01/2011] [Accepted: 04/16/2011] [Indexed: 12/14/2022]
Abstract
Epithelial cell adhesion molecule (EpCAM, CD326) is a pleiotropic molecule that potentially offers therapeutic applications in cancer treatment. Initially described as a dominant surface antigen on human colon carcinoma, it is a transmembrane glycoprotein mediating epithelial-specific intercellular cell-adhesion. Recent data suggest that EpCAM is also involved in cell signaling, migration, proliferation and differentiation. Since EpCAM is expressed exclusively in epithelia and epithelial-derived neoplasms, EpCAM can be used as diagnostic marker. Testing for EpCAM is based on morphology and phenotypical staining and can be performed with primary carcinoma tissue and cells harvested from malignant effusions. Stable or highly expressed EpCAM has been detected in most adenocarcinomas and has also been found in metastases, malignant effusions, and cancer stem cells. EpCAM may thus be an ideal tumor antigen candidate to detect circulating and metastasizing cancer cells by microchip technologies. In certain tumor types overexpression was linked to advanced stage of disease and worse overall survival, suggesting EpCAM as a potential prognostic marker. In addition to its diagnostic and prognostic role, EpCAM's broad expression and apparent involvement in tumorigenesis and metastasis point to its potential as a target for immunotherapeutic strategies. The first EpCAM targeting, trifunctional antibody catumaxomab (Removab®) has shown clear clinical benefits in treatment of malignant ascites associated with EpCAM positive carcinomas. Further research and clinical studies should unravel EpCAM's complex role in oncological processes, and expand potential therapeutic applications of EpCAM targeted strategies.
Collapse
|
326
|
Signaling mechanism of cell adhesion molecules in breast cancer metastasis: potential therapeutic targets. Breast Cancer Res Treat 2011; 128:7-21. [PMID: 21499686 DOI: 10.1007/s10549-011-1499-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 03/31/2011] [Indexed: 01/13/2023]
Abstract
Metastasis is responsible for the majority of breast cancer-related deaths. The metastatic spread of cancer cells is a complicated process that requires considerable flexibility in the adhesive properties of both tumor cells and other interacting cells. Cell adhesion molecules (CAMs) are membrane receptors that mediate cell-cell and cell-matrix interactions, and are essential for transducing intracellular signals responsible for adhesion, migration, invasion, angiogensis, and organ-specific metastasis. This review will discuss the recent advances in our understanding on the biological functions, signaling mechanisms, and therapeutic potentials of important CAMs involved in breast cancer metastasis.
Collapse
|
327
|
Dhankhar R, Vyas SP, Jain AK, Arora S, Rath G, Goyal AK. Advances in novel drug delivery strategies for breast cancer therapy. ACTA ACUST UNITED AC 2011; 38:230-49. [PMID: 20677900 DOI: 10.3109/10731199.2010.494578] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Breast cancer remains one of the world's most devastating diseases. However, better understanding of tumor biology and improved diagnostic devices could lead to improved therapeutic outcomes. Nanotechnology has the potential to revolutionize cancer diagnosis and therapy. Various nanocarriers have been introduced to improve the therapeutic efficacy of anticancer drugs, including liposomes, polymeric micelles, quantum dots, nanoparticles, and dendrimers. Recently, targeted drug delivery systems for anti-tumor drugs have demonstrated great potential to lower cytotoxicity and increase therapeutic effects. Various ligands/approaches have been explored for targeting breast cancer. This paper provides an overview of breast cancer, conventional therapy, potential of nanotechnology in management of breast cancer, and rational approaches for targeting breast cancer.
Collapse
Affiliation(s)
- Ritu Dhankhar
- Nanomedicine Research Centre, Department of Pharmaceutics, Indo-Soviet Friendship College of Pharmacy, GT Road, Moga, Punjab, India
| | | | | | | | | | | |
Collapse
|
328
|
Asati A, Kaittanis C, Santra S, Perez JM. pH-tunable oxidase-like activity of cerium oxide nanoparticles achieving sensitive fluorigenic detection of cancer biomarkers at neutral pH. Anal Chem 2011; 83:2547-53. [PMID: 21370817 PMCID: PMC3069142 DOI: 10.1021/ac102826k] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The reliable and sensitive detection of cancer-specific biomarkers is important for the diagnosis and treatment of cancer. Hence, detection of these biomarkers has to be reliably and rapidly performed in diverse settings. A limitation of the conventional biomarker-screening method of enzyme-linked immunosorbent assay (ELISA) is the employment of labile components, such as hydrogen peroxide and horseradish peroxidase. Previously, we reported that nanoceria is able to oxidize various colorimertic dyes at acidic pH, such as 3,3',5,5'-tetramethylbenzydine (TMB) and 2,2-azinobis-(3-ethylbenzothizoline-6-sulfonic acid) (AzBTS), and an assay was designed for screening the folate receptor. Herein, we show that the ability of nanoceria to oxidize a substrate can be tuned by modulating the pH. Results showed that nanoceria can oxidize the nonfluorescent substrate ampliflu, either to the very stable fluorescent product resorufin at pH 7.0 or to the nonfluorescent resazurin at pH 4.0. On the basis of these findings, we conjugated Protein G to immobilize antibodies on the surface of nanoceria, in order to detect the expression of prototypic cancer biomarkers at pH 7.0, such as the folate receptor and EpCAM. We found that within 3 h, nanoceria identified the expression of the folate receptor and EpCAM on lung carcinoma and breast adenocarcinoma cells, respectively. Traditional ELISA had a readout time of 15 h and a higher detection threshold, while requiring multiple washing steps. Considering these results and nanoceria's ability to oxidize ampliflu to its stable fluorescent product at neutral pH, the use of antibody-carrying nanoceria in the lab and point-of-care molecular diagnostics is anticipated.
Collapse
Affiliation(s)
- Atul Asati
- NanoScience Technology Center, Suite 400, 12424 Research Parkway, Orlando, Florida 32826
| | - Charalambos Kaittanis
- NanoScience Technology Center, Suite 400, 12424 Research Parkway, Orlando, Florida 32826
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32826
| | - Santimukul Santra
- NanoScience Technology Center, Suite 400, 12424 Research Parkway, Orlando, Florida 32826
| | - J Manuel Perez
- NanoScience Technology Center, Suite 400, 12424 Research Parkway, Orlando, Florida 32826
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida 32826
- Department of Chemistry, University of Central Florida
| |
Collapse
|
329
|
Spizzo G, Fong D, Wurm M, Ensinger C, Obrist P, Hofer C, Mazzoleni G, Gastl G, Went P. EpCAM expression in primary tumour tissues and metastases: an immunohistochemical analysis. J Clin Pathol 2011; 64:415-20. [PMID: 21415054 PMCID: PMC3088404 DOI: 10.1136/jcp.2011.090274] [Citation(s) in RCA: 211] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aims Epithelial cell adhesion molecule (EpCAM) is a cell surface protein with oncogenic features that is expressed on healthy human epithelia and corresponding malignant tumours. EpCAM expression frequently correlates with more aggressive tumour behaviour and new EpCAM-specific therapeutic agents have recently been approved for clinical use in patients with cancer. However, no consensus exists on how and when to evaluate EpCAM expression in patients with cancer. Material and methods EpCAM expression was assessed by a well-established immunohistochemical staining protocol in 2291 primary tumour tissues and in 108 metastases using the EpCAM-specific antibody clone VU1D9. A total immunostaining score was calculated as the product of a proportion score and an intensity score. Four expression subgroups (no, weak, moderate and intense) were defined. As described previously, the term ‘EpCAM overexpression’ was reserved for tissues showing a total immunostaining score >4. Results EpCAM was highly expressed in most tumours of gastrointestinal origin and in some carcinomas of the genitourinary tract. However, hepatocellular carcinomas, clear cell renal cell cancer, urothelial cancer and squamous cell cancers were frequently EpCAM negative. EpCAM expression in breast cancer depended on the histological subtype; lobular histology usually showed no or weak expression. Most metastases were EpCAM positive and they frequently reflected the expression phenotype of the primary tumour. Conclusion EpCAM expression was detected on adenocarcinomas of various primary sites. If EpCAM-specific antibodies are intended to be used in patients with cancer, we recommend prior immunohistochemical evaluation of EpCAM expression, particularly in patients with renal cell cancer, hepatocellular carcinoma, urothelial carcinoma, breast cancer and squamous cell carcinomas.
Collapse
Affiliation(s)
- Gilbert Spizzo
- Laboratory for Experimental Oncology (Tyrolean Cancer Research Institute), Innsbruck Medical University, Innsbruck, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
330
|
Akita H, Nagano H, Takeda Y, Eguchi H, Wada H, Kobayashi S, Marubashi S, Tanemura M, Takahashi H, Ohigashi H, Tomita Y, Ishikawa O, Mori M, Doki Y. Ep-CAM is a significant prognostic factor in pancreatic cancer patients by suppressing cell activity. Oncogene 2011; 30:3468-76. [PMID: 21399662 DOI: 10.1038/onc.2011.59] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The identification of molecular markers useful for predicting prognosis in pancreatic cancer patients is crucial for advances in disease management. The epithelial cell adhesion molecule (Ep-CAM) is known to express in most epithelial malignancies and was reported as a tumor marker or a candidate of molecular targeting therapy. However, the clinical significance of Ep-CAM expression in pancreatic cancer is not well-known. We determined the difference of malignant potential between parental and Ep-CAM-transfected pancreatic cancer cell lines by using proliferation, invasion and migration assay. Furthermore, we determined the relationship between tumoral Ep-CAM expression of resected specimens and clinical prognosis in 95 pancreatic cancer patients receiving radical surgery at two different cancer centers. One of the three Ep-CAM-transfected cell lines showed significantly low proliferation rate compared with the parental cell, while there was no difference in the other two cell lines. In invasion and migration assays, Ep-CAM-transfected cells showed significantly lower malignant potential than parental in all of the three cell lines. In 95 pancreatic cancer patients, 47 patients showed high-Ep-CAM expression, while 48 patients showed low, and there was no difference of clinicopathological features between Ep-CAM high and low-expression group. High-Ep-CAM expression group showed significantly good prognosis in overall survival (3-year survival; 56.2 versus 19.2%, P=0.0018) as well as in disease-free survival (3-year survival; 40.3 versus 14.4%, P=0.038) compared with low-expression group. In addition, the impact of Ep-CAM was observed strongly in LN-negative group when the influence of Ep-CAM was examined with dividing patients into LN-positive and negative group. In multivariate analysis, Ep-CAM expression was one of the independent prognostic factors as well as histology and lymph node metastasis. Ep-CAM expression was found to be related to the suppression of pancreatic cancer cell activity and the good prognosis in pancreatic cancer patients receiving the curative resection.
Collapse
Affiliation(s)
- H Akita
- Department of Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
331
|
Ortiz-Plata A, Moreno-Leyva K, López-Gómez M, Santos-Salinas S, Sánchez-García A, Tena-Suck ML. Epithelial cell adhesion molecule expression in pituitary adenomas: an immunohistochemical study. Ann Diagn Pathol 2011; 14:418-24. [PMID: 21074690 DOI: 10.1016/j.anndiagpath.2010.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 06/25/2010] [Indexed: 10/18/2022]
Abstract
An important amount of data correlating the expression of epithelial cell adhesion molecule (Ep-CAM) with cellular proliferation and de-differentiation could directly contribute to carcinogenesis. The aim of this study is to evaluate prognosis relevance of Ep-CAM expression in a group of pituitary adenomas. Epithelial cell adhesion molecule, proliferating cell nuclear antigen, and microvascular density labeling indices in pituitary adenomas were determined by immunohistochemistry on tissue samples obtained from each adenoma after surgery. We evaluated 45 adenomas. Sixty-two percent were nonsecretor adenomas and 37.8% were secretor tumors. Immunohistochemistry was scored for immunoexpression of Ep-CAM (cytoplasmic, membrane, and mixed pattern). Proliferating cell nuclear antigen and vascular density (CD34) labeling indices were assessed. Statistical significance was observed between Ep-CAM cytoplasmic immunoreactions (P = .000) and higher proliferating cell nuclear antigen (P = .001) in secretor adenomas compared with nonsecretor tumors. Vascular density labeling indices did not show statistical significance. Therefore, Ep-CAM could be evaluated to distinguish secretor and nonsecretor pituitary adenomas. These suggest that the markers could predict the growth potential of individual pituitary adenomas.
Collapse
Affiliation(s)
- Alma Ortiz-Plata
- Laboratory of Experimental Neuropathology, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, México, DF 14269, Mexico
| | | | | | | | | | | |
Collapse
|
332
|
Zöller M, Jung T. The Colorectal Cancer Initiating Cell: Markers and Their Role in Liver Metastasis. ACTA ACUST UNITED AC 2011. [DOI: 10.1007/978-94-007-0292-9_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
333
|
|
334
|
Gostner JM, Fong D, Wrulich OA, Lehne F, Zitt M, Hermann M, Krobitsch S, Martowicz A, Gastl G, Spizzo G. Effects of EpCAM overexpression on human breast cancer cell lines. BMC Cancer 2011; 11:45. [PMID: 21281469 PMCID: PMC3042418 DOI: 10.1186/1471-2407-11-45] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 01/31/2011] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Recently, EpCAM has attracted major interest as a target for antibody- and vaccine-based cancer immunotherapies. In breast cancer, the EpCAM antigen is overexpressed in 30-40% of all cases and this increased expression correlates with poor prognosis. The use of EpCAM-specific monoclonal antibodies is a promising treatment approach in these patients. METHODS In order to explore molecular changes following EpCAM overexpression, we investigated changes of the transcriptome upon EpCAM gene expression in commercially available human breast cancer cells lines Hs578T and MDA-MB-231. To assess cell proliferation, a tetrazolium salt based assay was performed. A TCF/LEF Reporter Kit was used to measure the transcriptional activity of the Wnt/β-catenin pathway. To evaluate the accumulation of β-catenin in the nucleus, a subcellular fractionation assay was performed. RESULTS For the first time we could show that expression profiling data of EpCAM transfected cell lines Hs578TEpCAM and MDA-MB-231EpCAM indicate an association of EpCAM overexpression with the downregulation of the Wnt signaling inhibitors SFRP1 and TCF7L2. Confirmation of increased Wnt signaling was provided by a TCF/LEF reporter kit and by the finding of the nuclear accumulation of ß-catenin for MDA-MB-231 EpCAM but not Hs578T EpCAM cells. In Hs578T cells, an increase of proliferation and chemosensitivity to Docetaxel was associated with EpCAM overexpression. CONCLUSIONS These data show a cell type dependent modification of Wnt signaling components after EpCAM overexpression in breast cancer cell lines, which results in marginal functional changes. Further investigations on the interaction of EpCAM with SFRP1 and TCF7L2 and on additional factors, which may be causal for changes upon EpCAM overexpression, will help to characterize unique molecular properties of EpCAM-positive breast cancer cells.
Collapse
Affiliation(s)
- Johanna M Gostner
- Laboratory for Experimental Oncology, Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck Austria
| | - Dominic Fong
- Laboratory for Experimental Oncology, Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck Austria
- Department of Haematology and Oncology, Innsbruck Medical University Anichstrasse 35, 6020 Innsbruck, Austria
| | - Oliver A Wrulich
- Division of Medical Biochemistry, Biocenter Innsbruck Fritz-Pregl Strasse 3, 6020 Innsbruck, Austria
| | - Florian Lehne
- Laboratory for Experimental Oncology, Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck Austria
| | - Marion Zitt
- Laboratory for Experimental Oncology, Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck Austria
| | | | - Sylvia Krobitsch
- Max Planck Institute for Molecular Genetics, Ihnestrasse 73, 14195 Berlin, Germany
| | - Agnieszka Martowicz
- Laboratory for Experimental Oncology, Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck Austria
| | - Guenther Gastl
- Laboratory for Experimental Oncology, Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck Austria
- Department of Haematology and Oncology, Innsbruck Medical University Anichstrasse 35, 6020 Innsbruck, Austria
| | - Gilbert Spizzo
- Laboratory for Experimental Oncology, Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck Austria
- Department of Haematology and Oncology, Innsbruck Medical University Anichstrasse 35, 6020 Innsbruck, Austria
- Department of Haematology and Oncology, Franz Tappeiner Hospital, Via Rossini 5, 39012 Merano, Italy
| |
Collapse
|
335
|
Ströhlein MA, Heiss MM. The trifunctional antibody catumaxomab in treatment of malignant ascites and peritoneal carcinomatosis. Future Oncol 2011; 6:1387-94. [PMID: 20919824 DOI: 10.2217/fon.10.111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Peritoneal carcinomatosis remains an unsolved medical problem in modern oncologic treatment. Excruciating symptoms such as malignant ascites, ileus, nausea, vomiting, dyspnoea and pain deteriorate the quality of life for affected patients. There is still no effective standard treatment for peritoneal carcinomatosis. The trifunctional antibody catumaxomab (antiepithelial cell adhesion molecule x anti-CD3) is able to direct T lymphocytes and Fcg-receptor-positive accessory cells to epithelial cell adhesion molecule-positive tumor cells. Intraperitoneal catumaxomab therapy was shown to be the first effective therapy against accumulation of malignant ascites in patients with peritoneal carcinomatosis of epithelial cancer, reducing the need of paracentesis and prolonging puncture-free survival. This paper reviews the mode of action of catumaxomab and analyzes different fields of local immunotherapy in patients with peritoneal carcinomatosis. A summary of completed and ongoing studies is included. Catumaxomab is discussed to be an outstanding option for local control and therapy of peritoneal carcinomatosis, which could be an optimal modular therapy in addition to systemic chemotherapy and surgical tumor resection.
Collapse
Affiliation(s)
- Michael A Ströhlein
- Department of Abdominal, Vascular & Transplant Surgery, Cologne-Merheim Medical Center, University of Witten/Herdecke, Ostmerheimer Strasse 200, Cologne, Germany.
| | | |
Collapse
|
336
|
Kawashima R, Abei M, Fukuda K, Nakamura K, Murata T, Wakayama M, Seo E, Hasegawa N, Mizuguchi H, Obata Y, Hyodo I, Hamada H, Yokoyama KK. EpCAM- and EGFR-targeted selective gene therapy for biliary cancers using Z33-fiber-modified adenovirus. Int J Cancer 2011; 129:1244-53. [PMID: 21710497 DOI: 10.1002/ijc.25758] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Accepted: 09/08/2010] [Indexed: 12/14/2022]
Abstract
A critical issue in adenovirus (Ad)-based cancer gene therapy is to improve the specificity of gene delivery to cancer cells for better efficacy and safety. We explored methods of retargeting Ad vectors for selective gene therapy of human biliary cancers using the Ad incorporating an IgG Fc-binding motif (Z33) from the Staphylococcus protein A (Ad-FZ33) combined with tumor-specific antibodies. Flow cytometry analysis revealed high-expression levels of epithelial cell adhesion molecule (EpCAM) and epidermal growth factor receptor (EGFR) on human biliary cancer cells. Ad-FZ33 expressing LacZ combined with antibodies against EpCAM or EGFR, followed by β-gal assay, demonstrated highly efficient gene transduction in these biliary cancer cells, compared to the treatment with control antibody or without antibody. Ad-FZ33 expressing uracil phosphoribosyl transferase (UPRT), an enzyme which greatly enhances the toxicity of 5-fluorouracil (FU), combined with antibodies against EpCAM or EGFR, remarkably enhanced the sensitivity of biliary cancer cells to 5-FU. By contrast, the treatment did not affect the 5-FU sensitivity of the cells not expressing EpCAM or EGFR including normal hepatocytes. Finally, treatments with the UPRT-expressing Ad-FZ33 with antibodies against EpCAM or EGFR, followed by 5-FU administration, significantly suppressed the growth of biliary cancer xenografts in nude mice. These results indicate that the gene therapy mediated by the Z33 fiber modified Ad with anti-EpCAM or anti-EGFR antibodies offers a potentially effective therapeutic modality against biliary cancers.
Collapse
Affiliation(s)
- Rei Kawashima
- Division of Gastroenterology, University of Tsukuba Graduate School of Comprehensive Human Sciences, Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
337
|
Richter CE, Cocco E, Bellone S, Silasi DA, Rüttinger D, Azodi M, Schwartz PE, Rutherford TJ, Pecorelli S, Santin AD. High-grade, chemotherapy-resistant ovarian carcinomas overexpress epithelial cell adhesion molecule (EpCAM) and are highly sensitive to immunotherapy with MT201, a fully human monoclonal anti-EpCAM antibody. Am J Obstet Gynecol 2010; 203:582.e1-7. [PMID: 20870202 PMCID: PMC2993821 DOI: 10.1016/j.ajog.2010.07.041] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 06/02/2010] [Accepted: 07/22/2010] [Indexed: 01/30/2023]
Abstract
OBJECTIVE We evaluated the expression of epithelial cell adhesion molecule (EpCAM) and the potential of MT201 (adecatumumab), a human-monoclonal-antibody that targets EpCAM against chemotherapy-resistant ovarian disease. STUDY DESIGN EpCAM expression was evaluated by real-time polymerase chain reaction and flow cytometry. Sensitivity to MT201 antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity was tested in 4-hour chromium-release assays. The effect of interleukin-2 on MT201 ADCC was also studied. RESULTS High messenger RNA expression by real-time polymerase chain reaction and high EpCAM surface expression by flow cytometry was detected in 71% of ovarian cancers (5 of 7 cell lines). Although these cell lines were highly resistant to complement-dependent cytotoxicity and natural killer-dependent cytotoxicity in vitro (range of killing, 0-7%), EpCAM-positive cell lines showed high sensitivity to MT201 ADCC (range of killing, 27-66%). Incubation with interleukin-2 further increased the cytotoxic activity against EpCAM-positive ovarian cancer cell lines. CONCLUSION MT201 may represent a novel, potentially highly effective treatment option for patients with ovarian carcinoma whose body is harboring disease refractory to chemotherapy.
Collapse
Affiliation(s)
- Christine E. Richter
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Emiliano Cocco
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Stefania Bellone
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Dan-Arin Silasi
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | | | - Masoud Azodi
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Peter E. Schwartz
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Thomas J. Rutherford
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Sergio Pecorelli
- Division of Gynecologic Oncology, University of Brescia, Brescia, Italy
| | - Alessandro D. Santin
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
338
|
Ralhan R, He HCH, So AKC, Tripathi SC, Kumar M, Hasan MR, Kaur J, Kashat L, MacMillan C, Chauhan SS, Freeman JL, Walfish PG. Nuclear and cytoplasmic accumulation of Ep-ICD is frequently detected in human epithelial cancers. PLoS One 2010; 5:e14130. [PMID: 21152431 PMCID: PMC2994724 DOI: 10.1371/journal.pone.0014130] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Accepted: 10/24/2010] [Indexed: 12/30/2022] Open
Abstract
Background We previously demonstrated that nuclear and cytoplasmic accumulation of the intracellular domain (Ep-ICD) of epithelial cell adhesion molecule (EpCAM) accompanied by a reciprocal reduction of its extracellular domain (EpEx), occurs in aggressive thyroid cancers. This study was designed to determine whether similar accumulation of Ep-ICD is a common event in other epithelial cancers. Methodology and Results Ten epithelial cancers were immunohistochemically analyzed using Ep-ICD and EpEx domain-specific antibodies. The subcellular localization of EpEx and Ep-ICD in the human colon adenocarcinoma cell line CX-1 was observed using immunofluorescence. Nuclear and cytoplasmic Ep-ICD expression was increased in cancers of the breast (31 of 38 tissues, 82%), prostate (40 of 49 tissues, 82%), head and neck (37 of 57 tissues, 65%) and esophagus (17 of 46 tissues, 37%) compared to their corresponding normal tissues that showed membrane localization of the protein. Importantly, Ep-ICD was not detected in the nuclei of epithelial cells in most normal tissues. High nuclear and cytoplasmic Ep-ICD accumulation also occurred in the other six epithelial cancer types analyzed - lung, colon, liver, bladder, pancreatic, and ovarian. A concomitant reduction in membrane EpEx expression was observed in a subset of all cancer types. Receiver operating characteristic curve analysis revealed nuclear Ep-ICD distinguished breast cancers with 82% sensitivity and 100% specificity and prostate cancers with 82% sensitivity and 78% specificity. Similar findings were observed for cytoplasmic accumulation of Ep-ICD in these cancers. We provide clinical evidence of increased nuclear and cytoplasmic Ep-ICD accumulation and a reduction in membranous EpEx in these cancers. Conclusions Increased nuclear and cytoplasmic Ep-ICD was observed in all epithelial cancers analyzed and distinguished them from normal tissues with high-sensitivity, specificity, and AUC. Development of a robust high throughput assay for Ep-ICD will facilitate the determination of its diagnostic, prognostic and therapeutic relevance in epithelial cancers.
Collapse
Affiliation(s)
- Ranju Ralhan
- Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Department of Otolaryngology-Head and Neck Surgery Program, Mount Sinai Hospital, Toronto, Ontario, Canada
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
- Department of Otolaryngology-Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- * E-mail: (PGW); (RR)
| | - Helen C.-H. He
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
| | - Anthony K.-C. So
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
| | - Satyendra C. Tripathi
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Md. Raghibul Hasan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Jatinder Kaur
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Lawrence Kashat
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Christina MacMillan
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
| | - Shyam Singh Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Jeremy L. Freeman
- Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Department of Otolaryngology-Head and Neck Surgery Program, Mount Sinai Hospital, Toronto, Ontario, Canada
- Endocrine Division, Department of Medicine, Mount Sinai Hospital and University of Toronto Medical School, Toronto, Ontario, Canada
| | - Paul G. Walfish
- Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Department of Otolaryngology-Head and Neck Surgery Program, Mount Sinai Hospital, Toronto, Ontario, Canada
- Alex and Simona Shnaider Laboratory in Molecular Oncology, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Joseph and Wolf Lebovic Health Complex, Toronto, Ontario, Canada
- Endocrine Division, Department of Medicine, Mount Sinai Hospital and University of Toronto Medical School, Toronto, Ontario, Canada
- Department of Otolaryngology-Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- * E-mail: (PGW); (RR)
| |
Collapse
|
339
|
Sebastian M. Review of catumaxomab in the treatment of malignant ascites. Cancer Manag Res 2010; 2:283-6. [PMID: 21188120 PMCID: PMC3004584 DOI: 10.2147/cmr.s14115] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Indexed: 11/25/2022] Open
Abstract
Malignant ascites is frequently found with various solid tumors, and no established treatment options exist, apart from symptomatic paracentesis. Catumaxomab, a trifunctional bispecific monoclonal antibody, has two binding specificities directed to epithelial cell adhesion molecule (EpCAM) and the T cell antigen CD3. With its Fc-fragment, catumaxomab additionally binds accessory cells, including dendritic cells, macrophages, and natural killer cells. The trifunctional approach thus leads to a major histocompatibility complex-unrestricted but specific killing of epithelial tumor cells without need for preactivation or external costimulation. Because EpCAM is expressed in most solid tumors, but not in tissue of mesothelial origin, intraperitoneal treatment with catumaxomab is tumor-specific. Intraperitoneal treatment with catumaxomab resulted in a significant prolongation of puncture-free survival in patients with malignant ascites due to epithelial cancer. Catumaxomab has been approved in Europe for the intraperitoneal treatment of malignant ascites in patients with EpCAM-positive epithelial tumors where standard therapy is not available or no longer feasible.
Collapse
Affiliation(s)
- Martin Sebastian
- Department of Internal Medicine III, Hematology, Medical Oncology, and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
340
|
Münz M, Murr A, Kvesic M, Rau D, Mangold S, Pflanz S, Lumsden J, Volkland J, Fagerberg J, Riethmüller G, Rüttinger D, Kufer P, Baeuerle PA, Raum T. Side-by-side analysis of five clinically tested anti-EpCAM monoclonal antibodies. Cancer Cell Int 2010; 10:44. [PMID: 21044305 PMCID: PMC2989956 DOI: 10.1186/1475-2867-10-44] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 11/02/2010] [Indexed: 12/12/2022] Open
Abstract
Background Epithelial cell adhesion molecule (EpCAM) is frequently and highly expressed on human carcinomas. The emerging role of EpCAM as a signalling receptor and activator of the wnt pathway, and its expression on tumor-initiating cells, further add to its attractiveness as target for immunotherapy of cancer. Thus far, five conventional monoclonal IgG antibodies have been tested in cancer patients. These are murine IgG2a edrecolomab and its murine/human chimeric IgG1 antibody version, and humanized, human-engineered and fully human IgG1 antibodies 3622W94, ING-1, and adecatumumab (MT201), respectively. Here we compared all anti-EpCAM antibodies in an attempt to explain differences in clinical activity and safety. Methods We recombinantly produced all antibodies but murine edrecolomab and investigated them for binding affinity, EpCAM epitope recognition, ADCC and CDC, and inhibition of breast cancer cell proliferation. Results ING-1 and 3622W94 bound to EpCAM with much higher affinity than adecatumumab and edrecolomab. Edrecolomab, ING-1, and 3622W94 all recognized epitopes in the exon 2-encoded N-terminal domain of EpCAM, while adecatumumab recognized a more membrane proximal epitope encoded by exon 5. All antibodies induced lysis of EpCAM-expressing cancer cell lines by both ADCC and CDC with potencies that correlated with their binding affinities. The chimeric version of edrecolomab with a human Fcγ1 domain was much more potent in ADCC than the murine IgG2a version. Only adecatumumab showed a significant inhibition of MCF-7 breast cancer cell proliferation in the absence of complement and immune cells. Conclusion A moderate binding affinity and recognition of a distinct domain of EpCAM may best explain why adecatumumab showed a larger therapeutic window in cancer patients than the two high-affinity IgG1 antibodies ING-1 and 3622W94, both of which caused acute pancreatitis.
Collapse
Affiliation(s)
- Markus Münz
- Micromet AG, Staffelseestr, 2, 81477 Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
341
|
Maghzal N, Vogt E, Reintsch W, Fraser JS, Fagotto F. The tumor-associated EpCAM regulates morphogenetic movements through intracellular signaling. ACTA ACUST UNITED AC 2010; 191:645-59. [PMID: 20974811 PMCID: PMC3003323 DOI: 10.1083/jcb.201004074] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Epithelial cell adhesion molecule (EpCAM) is best known as a tumor-associated protein highly expressed in carcinomas. The function of this cell surface protein during embryonic development and its potential role in cancer are still poorly understood. We identified EpCAM in a gain-of-function screen for inducers of abnormal tissue mixing during gastrulation. Elevated EpCAM levels in either the ectoderm or the mesoderm confer "invasive" properties to cells in both populations. We found that this phenotype represents an "overstimulation" of an essential activity of EpCAM in controlling cell movements during embryonic development. Surprisingly, this property is independent of the putative adhesive function of EpCAM, and rather relies on a novel signaling function that operates through down-regulation of PKC activity. We show that inhibition of novel PKCs accounts entirely for the invasive phenotype induced by abnormally high levels of EpCAM as well as for its normal function in regulating cell rearrangement during early development.
Collapse
Affiliation(s)
- Nadim Maghzal
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
342
|
Kimura O, Takahashi T, Ishii N, Inoue Y, Ueno Y, Kogure T, Fukushima K, Shiina M, Yamagiwa Y, Kondo Y, Inoue J, Kakazu E, Iwasaki T, Kawagishi N, Shimosegawa T, Sugamura K. Characterization of the epithelial cell adhesion molecule (EpCAM)+ cell population in hepatocellular carcinoma cell lines. Cancer Sci 2010; 101:2145-2155. [PMID: 20707805 PMCID: PMC11159121 DOI: 10.1111/j.1349-7006.2010.01661.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Accumulating evidence suggests that cancer stem cells (CSC) play an important role in tumorigenicity. Epithelial cell adhesion molecule (EpCAM) is one of the markers that identifies tumor cells with high tumorigenicity. The expression of EpCAM in liver progenitor cells prompted us to investigate whether CSC could be identified in hepatocellular carcinoma (HCC) cell lines. The sorted EpCAM(+) subpopulation from HCC cell lines showed a greater colony formation rate than the sorted EpCAM(-) subpopulation from the same cell lines, although cell proliferation was comparable between the two subpopulations. The in vivo evaluation of tumorigenicity, using supra-immunodeficient NOD/scid/γc(null) (NOG) mice, revealed that a smaller number of EpCAM(+) cells (minimum 100) than EpCAM(-) cells was necessary for tumor formation. The bifurcated differentiation of EpCAM(+) cell clones into both EpCAM(+) and EpCAM(-) cells was obvious both in vitro and in vivo, but EpCAM(-) clones sustained their phenotype. These clonal analyses suggested that EpCAM(+) cells may contain a multipotent cell population. Interestingly, the introduction of exogenous EpCAM into EpCAM(+) clones, but not into EpCAM(-) clones, markedly enhanced their tumor-forming ability, even though both transfectants expressed a similar level of EpCAM. Therefore, the difference in the tumor-forming ability between EpCAM(+) and EpCAM(-) cells is probably due to the intrinsic biological differences between them. Collectively, our results suggest that the EpCAM(+) population is biologically quite different from the EpCAM(-) population in HCC cell lines, and preferentially contains a highly tumorigenic cell population with the characteristics of CSC.
Collapse
Affiliation(s)
- Osamu Kimura
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
343
|
van der Gun BTF, Melchers LJ, Ruiters MHJ, de Leij LFMH, McLaughlin PMJ, Rots MG. EpCAM in carcinogenesis: the good, the bad or the ugly. Carcinogenesis 2010; 31:1913-21. [PMID: 20837599 DOI: 10.1093/carcin/bgq187] [Citation(s) in RCA: 250] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The epithelial cell adhesion molecule (EpCAM) is a membrane glycoprotein that is highly expressed on most carcinomas and therefore of potential use as a diagnostic and prognostic marker for a variety of carcinomas. Interestingly, EpCAM is explored as target in antibody-based therapies. Recently, EpCAM has been identified as an additional marker of cancer-initiating cells. In this review, we describe the controversial biological role of EpCAM with the focus on carcinogenesis: as an adhesion molecule, EpCAM mediates homophilic adhesion interactions, which in turn might prevent metastasis. On the other hand, EpCAM abrogates E-cadherin mediated cell-cell adhesion thereby promoting metastasis. Also, upon cleavage of EpCAM, the intracellular domain functions as a part of a transcriptional complex inducing c-myc and cyclin A and E. In line with these seemingly controversial roles, EpCAM overexpression has been associated with both decreased and increased survival of patients. Similarly, either induction or downregulation of EpCAM expression lowers the oncogenic potential depending on the cell type. As epigenetic dysregulation underlies aberrant EpCAM expression, we propose epigenetic editing as a novel approach to investigate the biological role of EpCAM, expanding the options for EpCAM as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Bernardina T F van der Gun
- Epigenetic Editing, Department of Medical Biology, University Medical Centre Groningen, Hanzeplein 1, Groningen, TheNetherlands
| | | | | | | | | | | |
Collapse
|
344
|
Kurtz JE, Dufour P. Adecatumumab: an anti-EpCAM monoclonal antibody, from the bench to the bedside. Expert Opin Biol Ther 2010; 10:951-8. [PMID: 20426706 DOI: 10.1517/14712598.2010.482098] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
IMPORTANCE OF THE FIELD In developing new anticancer drugs, the identification of relevant targets is a key issue of growing importance. Ideally, an anticancer drug target should be specific to cancer cells, in order to both increase efficacy and decrease toxicity of the compound. AREAS COVERED IN THIS REVIEW Epithelial cell adhesion molecule (EpCAM) is a membrane protein with proto-oncogenic properties that is expressed in a number of endothelium-derived cancers and is a promising anticancer drug target. Adecatumumab is a monoclonal, fully human IgG1 antibody that targets EpCAM, development of which is at present reaching Phase III trials. WHAT THE READER WILL GAIN From a review of literature, we here update the rationale for using EpCAM as an anticancer target for monoclonal antibodies, with a special focus on adecatumumab. The fully human nature of adecatumumab is also discussed to put the drug in perspective with other related anti-EpCAM monoclonal antibodies, such as edrecolomab and catumaxomab. Adecatumumab studies are recapitulated, in order to provide the reader with a comprehensive view of the development of this promising anticancer agent. TAKE HOME MESSAGE Adecatumumab is a promising fully human monoclonal antibody targeting EpCAM which is expressed in almost all adenocarcinomas and its activity is not dependent of K-Ras status.
Collapse
Affiliation(s)
- Jean-Emmanuel Kurtz
- Département d'Hématologie et d'Oncologie, Hôpitaux Universitaires de Strasbourg, Avenue Molière, 67200 Strasbourg, France
| | | |
Collapse
|
345
|
A non-synonymous polymorphism Thr115Met in the EpCAM gene is associated with an increased risk of breast cancer in Chinese population. Breast Cancer Res Treat 2010; 126:487-95. [DOI: 10.1007/s10549-010-1094-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 07/23/2010] [Indexed: 01/16/2023]
|
346
|
van der Gun BTF, Maluszynska-Hoffman M, Kiss A, Arendzen AJ, Ruiters MHJ, McLaughlin PMJ, Weinhold E, Rots MG. Targeted DNA methylation by a DNA methyltransferase coupled to a triple helix forming oligonucleotide to down-regulate the epithelial cell adhesion molecule. Bioconjug Chem 2010; 21:1239-45. [PMID: 20593890 PMCID: PMC2907751 DOI: 10.1021/bc1000388] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 05/20/2010] [Indexed: 02/05/2023]
Abstract
The epithelial cell adhesion molecule (EpCAM) is a membrane glycoprotein that has been identified as a marker of cancer-initiating cells. EpCAM is highly expressed on most carcinomas, and transient silencing of EpCAM expression leads to reduced oncogenic potential. To silence the EpCAM gene in a persistent manner via targeted DNA methylation, a low activity mutant (C141S) of the CpG-specific DNA methyltransferase M.SssI was coupled to a triple-helix-forming oligonucleotide (TFO-C141S) specifically designed for the EpCAM gene. Reporter plasmids encoding the green fluorescent protein under control of different EpCAM promoter fragments were treated with the TFO-C141S conjugate to determine the specificity of targeted DNA methylation in the context of a functional EpCAM promoter. Treatment of the plasmids with TFO-C141S resulted in efficient and specific methylation of the targeted CpG located directly downstream of the triple helix forming site (TFS). No background DNA methylation was observed neither in a 700 bp region of the EpCAM promoter nor in a 400 bp region of the reporter gene downstream of the TFS. Methylation of the target CpG did not have a detectable effect on promoter activity. This study shows that the combination of a specific TFO and a reduced activity methyltransferase variant can be used to target DNA methylation to predetermined sites with high specificity, allowing determination of crucial CpGs for promoter activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Marianne G. Rots
- To whom correspondence should be addressed. E-mail: . Phone: +31-50-361 0153. Fax: +31-50-361 9911
| |
Collapse
|
347
|
Andreopoulou E, Cristofanilli M. Circulating tumor cells as prognostic marker in metastatic breast cancer. Expert Rev Anticancer Ther 2010; 10:171-7. [PMID: 20131993 DOI: 10.1586/era.09.105] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Testing for circulating tumor cells has emerged as a new and promising tool for stratifying and monitoring patients with metastatic disease. Appropriate risk and biologic stratification in breast cancer is important for the development of more effectively tailored targeted therapies. To optimize patient care, it is important for the clinicians to rely on validated and robust tools able to provide accurate predictive and prognostic information for each patient at any time during treatment. The recent demonstration that the presence of circulating tumor cells predicts the prognosis at any time during the treatment of patients with metastatic breast cancer raises the possibility that this approach will allow for a true 'biologic staging' of the disease. Important questions regarding the biological characteristics of cells and the reasons for the reduced capacity of systemic treatments to arrest or eradicate the cancer were raised. A further study suggests that comprehensive analysis of circulating tumor cells is likely to provide new insights into the biology of breast cancer and contribute to defining novel treatments and better prediction of clinical benefit. Efforts are being made to genotype and phenotype micrometastatic cells. Considerable progress has been already accomplished which should lead to further noninvasive, real-time monitoring of these rare events in the adjuvant and metastatic settings.
Collapse
Affiliation(s)
- Eleni Andreopoulou
- University of Texas MD Anderson Cancer Center, Department of Breast Medical Oncology, 1155 Herman P Pressler, CPB5.3494 Houston, TX 77230, USA.
| | | |
Collapse
|
348
|
Schmidt M, Petry IB, Böhm D, Lebrecht A, von Törne C, Gebhard S, Gerhold-Ay A, Cotarelo C, Battista M, Schormann W, Freis E, Selinski S, Ickstadt K, Rahnenführer J, Sebastian M, Schuler M, Koelbl H, Gehrmann M, Hengstler JG. Ep-CAM RNA expression predicts metastasis-free survival in three cohorts of untreated node-negative breast cancer. Breast Cancer Res Treat 2010; 125:637-46. [PMID: 20352488 DOI: 10.1007/s10549-010-0856-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Accepted: 03/16/2010] [Indexed: 02/08/2023]
Abstract
Epithelial cell adhesion molecule (Ep-CAM) recently received increased attention as a prognostic factor in breast cancer. We aimed to validate the influence of Ep-CAM RNA expression in untreated node-negative breast cancer. Ep-CAM RNA expression was evaluated utilizing microarray-based gene-expression profiling in 194 consecutive node-negative breast cancer patients with long-term follow-up not treated in the adjuvant setting. The prognostic significance of Ep-CAM RNA expression for disease-free survival (DFS), metastasis-free survival (MFS), and breast cancer-specific overall survival (OS) was evaluated in univariate and multivariate analysis adjusted for age, grading, pTstage, ER as well as PR receptor and HER-2 status. Additionally, Ep-CAM RNA expression was compared with immunohistochemistry (IHC) for Ep-CAM in 194 patients. The prognostic impact of Ep-CAM gene expression was validated in further 588 node-negative breast cancer patients. Levels of Ep-CAM RNA expression showed a significant correlation with IHC (P = 0.001) and predicted in univariate analysis DFS (P = 0.001, HR = 2.4), MFS (P = 0.003, HR = 2.5), and OS (P = 0.002, HR = 3.1) accurately. The prognostic influence of Ep-CAM RNA was significant also in multivariate analysis for DFS (P = 0.017, HR = 2.0), MFS (P = 0.049, HR = 1.9), and OS (P = 0.042, HR = 2.3), respectively. The association with MFS was confirmed in an independent validation cohort in univariate (P = 0.006, HR = 1.9) and multivariate (P = 0.035, HR = 1.7) analysis. Ep-CAM RNA correlated with the proliferation metagene (P < 0.001, R=0.425) Nevertheless, in multivariate analysis, Ep-CAM was associated with MFS independent from the proliferation metagene (P = 0.030, HR = 1.8). In conclusion, Ep-CAM RNA expression is associated with poor MFS in three cohorts of untreated node-negative breast cancer.
Collapse
Affiliation(s)
- Marcus Schmidt
- Department of Obstetrics and Gynecology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131, Mainz, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
349
|
Osta WA, El-Osta MA, Pezhman EA, Raad RA, Ferguson K, McKelvey GM, Marsh HM, White M, Perov S. Nicotinic acetylcholine receptor gene expression is altered in burn patients. Anesth Analg 2010; 110:1355-9. [PMID: 20304984 DOI: 10.1213/ane.0b013e3181d41512] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Burn patients have been observed to be more susceptible to the hyperkalemic effect of the depolarizing muscle relaxant succinylcholine. Changes in nicotinic acetylcholine receptor (nAChR) subunit composition may alter electrophysiologic, pharmacologic, and metabolic characteristics of the receptor inducing hyperkalemia on exposure to succinylcholine. No studies have been performed that show the upregulation and/or alteration of nAChR subunit composition in human burn patients. The scarcity of studies performed on humans with burn injury is mainly attributable to the technical and ethical difficulties in obtaining muscle biopsies at different time frames of illness in these acutely injured patients. nAChRs are expressed in oral keratinocytes and are upregulated or altered in smokers. However, no studies have addressed the expression of nAChRs in the oral mucosa of burn patients. METHODS Buccal mucosal scrapings were collected from 9 burn patients and 6 control nonburn surgical intensive care unit patients. For burn and control patients, tissues were collected upon presentation (time: 0 hour) and at time points 12, 24, and 48 hours, 1 week, and 2 weeks. Gene expression of the nAChR subunits alpha1, alpha7, gamma, and epsilon were performed using real-time reverse transcriptase polymerase chain reaction. RESULTS alpha7 and gamma nAChR genes were significantly upregulated in burn patients, whereas alpha1 and epsilon nAChR genes were minimally affected, showing no significant changes over time. DISCUSSION Over the 2 weeks of measurement, an upregulation of the alpha7 and gamma genes occurred in both burn and control patients; however, the proportion of alpha7 and gamma subunit increases was significantly higher in burn patients than in control surgical intensive care unit patients. The relationship between the thermal injury and the observed alteration in gene expression suggests a possible cause/effect relationship. This effect was observed at a site not affected by the burn injury and in nonmuscle tissues, thus emphasizing the systemic nature of the effect caused by the thermal injury. Because gene expression is the basis of protein production, the upregulation of alpha7 and gamma genes might translate into more alpha7 and gamma protein subunits. These proteins can also combine with each other or with other types of subunits (alpha1, beta, epsilon . . .) to form nAChRs with altered electrophysiologic characteristics leading to the observed abnormal clinical outcomes. CONCLUSION Thermal injury may infer a systemic effect because upregulation/alteration of nAChRs occurs in nonmuscle tissues distant from the site of injury. The effect of thermal injury on nAChR gene subunits can be studied using a minimally invasive method (buccal mucosal scraping) and a highly sensitive technology (real-time reverse transcriptase polymerase chain reaction) obviating the need for more invasive methods.
Collapse
Affiliation(s)
- Walid A Osta
- Department of Anesthesia,Wayne State University/Detroit Medical Center, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
350
|
Ng VY, Ang SN, Chan JX, Choo ABH. Characterization of epithelial cell adhesion molecule as a surface marker on undifferentiated human embryonic stem cells. Stem Cells 2010; 28:29-35. [PMID: 19785009 DOI: 10.1002/stem.221] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human embryonic stem cells (hESCs) have the capacity to remain pluripotent and self-renew indefinitely. To discover novel players in the maintenance of hESCs, we have previously reported the generation of monoclonal antibodies that bind to cell surface markers on hESCs, and not to mouse embryonic stem cells or differentiated embryoid bodies. In this study, we have identified the antigen target of one such monoclonal antibody as the epithelial cell adhesion molecule (EpCAM). In undifferentiated hESCs, EpCAM is localized to Octamer 4 (OCT4)-positive pluripotent cells, and its expression is down-regulated upon differentiation. To further understand its biological function in hESCs, endogenous EpCAM expression was silenced using small interfering RNA. EpCAM knockdown had marginal negative effects on OCT4 and TRA-1-60 expression, however cell proliferation was decreased by >40%. Examination of lineage marker expression showed marked upregulation of endoderm and mesoderm genes in EpCAM-silenced cells, under both pluripotent and differentiating conditions. These results were validated using a hESC line whose EpCAM expression has been stably knocked down. Data from the stable line confirmed that downregulation of EpCAM decreases cell growth and increases gene expression in the endoderm and mesoderm lineages. In vivo, hESCs lacking EpCAM were able to form teratomas containing tissues representing the three germ layers, and gene expression analysis yielded marked increase in the endoderm marker alpha fetoprotein compared with control. Together these data demonstrate that EpCAM is a surface marker on undifferentiated hESCs and plays functional roles in proliferation and differentiation.
Collapse
Affiliation(s)
- Valerie Y Ng
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138668, Singapore.
| | | | | | | |
Collapse
|