Abstract
Pre-clinical studies provide compelling evidence that members of the Eph family of receptor tyrosine kinases and their ephrin ligands promote tumor growth, invasion and metastasis, and neovascularization. Tumor suppressive roles have also been reported for the receptors, and ligand-dependent versus ligand-independent signaling has emerged as one key mechanism underlying tumor suppressive function as opposed to oncogenic effects. Determining how these observations relate to clinical outcome is a crucial step for translating the biological and mechanistic data into new molecularly targeted therapies. Expression profiling in human patient samples bridges this gap and provides valuable clinical relevance to laboratory observations. In addition to analyses performed using privately assembled patient tumor samples, publically available microarray datasets and tissue microarrays linked to clinical data have emerged as tractable tools for addressing the clinical relevance of specific molecules and families of related molecules. This review summarizes the clinical relevance of specific Eph and ephrin molecules in human breast, colorectal, and lung cancers.
Keywords: Eph, ephrin, cancer, patient, profiling, clinical relevance
Introduction
Tumorigenesis and malignant progression are complex processes that are regulated in part by activation of oncogenic signaling pathways and inhibition of tumor suppressor pathways [reviewed in [1–3]]. Oncogenic conversion, amplification, and/or overexpression of proto-oncogenes, such as those encoding receptor tyrosine kinases (RTKs), contribute to tumorigenesis [4]. Loss of tumor suppressor pathways that negatively regulate cell proliferation also contributes to tumorigenesis [5]. In addition, a third class of molecules displays dual roles in both tumor suppression and tumor promotion. The Eph family of RTKs belongs to this dual regulatory category. This large family is subdivided into class A and class B receptors based on sequence homology and binding affinity for two distinct types of membrane-anchored ephrin ligands. Originally characterized as axon guidance regulators, ephrins and Eph RTKs regulate physiologic and pathologic processes in development and disease [reviewed in {Brantley-Sieders, 2004 #7;Pasquale, 2010 #9].
The Eph family enhances tumor growth, invasion and metastasis, and neovascularization [reviewed in {Brantley-Sieders, 2004 #7;Pasquale, 2010 #9]. Indeed, data derived from laboratory research suggest that EphA2 and EphB4 function as oncogenes. Other studies, however, provide evidence that Eph receptors, including EphA2 and EphB4, inhibit tumor growth and progressson [reviewed in {Noren, 2007 #11}[6]]. These opposing functions appear to be influenced by tissue type, oncogenic context, and ligand-independent versus ligand-dependent signaling. Moreover, ‘reverse’ signaling that occurs downstream of membrane-tethered ephrin ligands upon receptor binding adds to the complexity of Eph and ephrin function [reviewed in [7]. Finally, the fact that this family is the largest RTK family in the genome, encompassing at least 14 receptors and 8 ligands that often display overlapping expression patterns in both tumor cells and the surrounding host stroma, also presents a potential barrier in designing molecularly targeted Eph therapies in human cancer [reviewed in {Pasquale, 2010 #9]].
Nonetheless, Eph receptors are very attractive therapeutic targets. They are expressed in a broad range of human cancer types in both tumor and its stromal microenvironment. In fact, several family members are known to simultaneously regulate tumor growth and neovascularization [reviewed in [6, 8]], enabling a single anti-Eph inhibitor to potentially disrupt at least two key processes in tumor progression. Translating the biological and mechanistic data from the laboratory into novel clinical therapeutic strategies will require a solid understanding of the expression patterns of individual Eph receptors, particularly in the context of relevant ligands, within molecularly defined tumor subsets. Identification of those patients who will receive the maximum therapeutic benefit can be achieved using expression profiling of large tumor datasets. In this review, we summarize expression profiles derived from a broad spectrum of human patient samples with an emphasis on how publically available datasets and samples linked to clinical outcome data have emerged as tractable tools for addressing the clinical relevance of Eph RTKs and ephrins to cancer. Due to space constraints, we limit our discussion to profiling efforts in human breast, colorectal, and lung cancers. Information and references for these data and a broader range of human epithelial malignancies are provided in Table 1 (Class A Eph family members) and in Table 2 (Class B Eph family members).
Table 1.
Eph A Class Family Member |
Cancer Type | ↑/↓ Relative to Normal Tissue |
References |
---|---|---|---|
EphA1 | Ovarian Colon, Gliobastoma Gastric Pancreatic Hepatocellular |
↑ ↓ (↑associated with necrosis) ↑ ↑(associated with tumor size) ↑(associated with angiogenesis) |
[77] [53, 55, 56, 78] [79, 80] [81] [82, 83] |
EphA2 | Breast, Lung, Gastric, Colon, Kidney; Breast Breast Lung (NSCLC) Ovarian Colon Gastric Esophageal Gliobastoma Pancreatic Hepatocellular Prostate Urinary Bladder |
↑ (tumor and endothelium) ↑ ↑ (HER2+ samples) ↑(correlates with smoking history, brain metastasis, EGFR somatic mutation); mutation G391R associated with invasion ↑(correlates with MVD) ↑(correlates with MVD) ↑ ↑ ↑(PS897-EphA2) ↑(associated with patient age) ↑ ↑ ↑ |
[10–12, 14, 84] (Brantley-Sieders et al., in preparation) [15] [66–70] [77, 85] [57, 60] [58, 59, 86] [87, 88] [89–91] [81, 92] [93] [94] [95] |
EphA3 | Lung (NSCLC) Hepatocellular Glioblastoma Melanoma |
↓(and/or mutated) mutated mutated mutated |
[[71, 72, 96, 97]; Zhuang et al., submitted] [98] [99] [99] |
EphA4 | Breast Colon Glioblastoma Pancreatic |
↑ ↑(correlates with liver metastases) ↑ ↑(associated with proliferation) |
(Brantley-Sieders et al., in preparation) [64] [100] [81, 101] |
EphA5 | Breast Pancreatic |
↓ ↑(associated with prognosis) |
[26] [81] |
EphA6 | Colon Kidney |
↓ ↓ |
[56] [56] |
EphA7 | Breast Colon Glioblastoma Pancreatic Prostate |
↑ ↓ ↑ ↑(associated with prognosis) ↓(promoter hypermethylation) |
(Brantley-Sieders et al., in preparation) [54] [102] [81] [103] |
EphA8 | Colon, Glioblastotma | ↓ | [56] |
Ephrin-A1 | Breast, Lung, Gastric, Colon, Kidney Ovarian Gastric Esophageal Melanoma Urinary Bladder Glioblastoma |
↑(tumor and endothelium) ↑ ↑ ↑ ↑ ↑ ↓ |
[10] [77] [58, 86] [87] [104] [95] [91] |
Ephrin-A2 | Hepatocellular | ↑ | [105] |
Ephrin-A3 | Lung | ↑ | [56] |
Ephrin-A5 | Ovarian Glioblastoma |
↑ ↓ |
[77] [106] |
Table 2.
Eph B Class Family Member |
Cancer Type | ↑/↓ Relative to Normal Tissue |
References |
---|---|---|---|
EphB1 | Colon Gastric |
↓(transition to invasive cancer) ↓(transition to invasive cancer) |
[46] [47] |
EphB2 | Breast Colon Gastric Glioblastoma Neuroblastoma Hepatocellular |
↑ ↑/↓(transition from adenoma to carcinoma) ↓ ↑tyrosine phosphorylation ↑(higher levels in low stage) ↑ |
[22] [42]/[43, 44, 48, 49] [50] [107] [108] [56] |
EphB3 | Colon Lung (NSCLC) |
↑/↓ (transition from adenoma to carcinoma)* ↑ |
[42]/[43, 45] [75] |
EphB4 | Breast Ovarian Cervix Endometrium Colon Prostate |
↓/↑ ↑ ↑(correlates with MVD) ↑ ↑/↓(transition from adenoma to carcinoma) ↑ |
[21–23]/(Brantley- Sieders et al., in preparation) [73] [109, 110] [111, 112] [40–42]/[43, 51] [113] |
EphB6 | Breast Glioma Neuroblastoma Melanoma |
↓/↑ ↑variant protein ↑(higher levels in low stage) ↓ |
[27–31] /(Brantley- Sieders et al., in preparation) [114, 115] [116–118] [119] |
Ephrin-B1 | Ovarian Glioblastoma Hepatocellular |
↑ ↑total and tyrosine phosphorylated ↑associated with tumor angiogenesis |
[120] [121] [122] |
Ephrin-B2 | Ovarian Cervix Endometrium Colon Esophageal Glioblastoma Melanoma |
↑ ↑(correlates with MVD) ↑ ↑ ↑ ↑total and tyrosine phosphorylated ↑ |
[73, 120] [109, 110] [111, 112] [41, 42] [123] [121] [124] |
Ephrin-B3 | Ovarian Glioblastoma Neuroblastoma |
↑ ↑/increased tyrosine phosphorylation ↑(higher levels in low stage) |
[120] [125]/[126] [116–118] |
1.1 Eph expression profiles in human breast cancer
EphA2 and EphB4 are the two Eph RTK family members that have been most extensively studied in breast cancer [reviewed in [9]]. Ogawa et al. first reported expression of EphA2 and its primary ligand, ephrin-A1, in both tumor epithelium and associated vascular endothelium in human breast cancers, [10], and EphA2 overexpression in human breast cancer relative to benign human breast epithelium was also reported by Zelinksi et al. [11], and may be associated with estrogen receptor (ER) expression [12]. Subsequent studies reported that increased ephA2 mRNA expression levels, which are relatively low in normal human breast tissue, correlated with poor patient prognosis in two independent breast cancer microarray datasets [13, 14]. These data are consistent with cell culture, biochemical, and genetic laboratory studies that support the oncogenic function of EphA2 in human breast cancer [reviewed in [9]]. In addition, EphA2 appears to play a role in both intrinsic and acquired resistance to trastuzumab (Herceptin), a monoclonal anti-HER2 antibody used as a first line of treatment for HER2 amplified breast cancer [15]. These data are consistent with laboratory studies showing a physical and functional interaction with ErbB/EGFR RTKs [16–19]. Indeed, elevated ephA2 mRNA expression correlated significantly with decreased overall and recurrence-free survival in HER2-positive patients in a microarray dataset [15], providing clinically relevant evidence to support this association. These data suggest that expression profiling for ephA2 and/or other resistance-associated gene products in resistant versus sensitive patient samples could be used to identify patients who may benefit from EphA2-targeted therapies. In addition, expression profiling in host endothelium may also provide valuable diagnostic information. A recent study reported that EphA2-positive human breast tumor endothelium correlated with diminished expression of Slit2, a tumor suppressive angiocrine signal. Negative regulation of endothelial Slit2 by EphA2 in murine endothelial cells enables increased tumor growth [20]. These data support a novel role for endothelial EphA2 in regulation of tumor growth and motility independent of its angiogenic function, suggesting additional therapeutic benefits for EphA2 inhibition in human breast cancer.
Elevated mRNA and protein expression of EphB4 and EphB2 have also been reported in human breast cancer [21, 22]. While elevated EphB2 expression was associated with poorer overall and disease-free survival, EphB4 protein expression increased with grade and stage but showed no clear association with survival. Indeed, stronger EphB2 and EphB4 staining was observed in normal breast glandular epithelium than in malignant tumor epithelium [22]. An independent profiling study reported similar trends, with higher expression in normal breast tissue and low histologic grade tumors relative to invasive human breast carcinomas [23]. These expression data illustrate the often paradoxical findings regarding Eph RTKs in tumor promotion versus tumor suppression [8]. For example, laboratory studies demonstrated that systemic delivery of ephrin-B2-Fc inhibits the growth of MDA-MB-435 tumor xenografts [24]. EphB4 forward signaling activates the Abl/Crk pathway, inhibiting tumor cell growth and motility in breast cancer cells [24]. In contrast, a more recent expression analysis of multiple large patient datasets correlated elevated ephB4 mRNA expression with reduced overall and recurrence-free survival (Brantley-Sieders et al., in preparation). Together, these data suggest that further analysis of EphB4 expression, in both tumor parenchyma and the surrounding stroma, should be performed using large sets of human patient samples carefully stratified by stage and grade, as well as by molecular subtype and treatment regimen. Particular attention should be paid to expression profiles in tumor endothelium, given the role of B class receptors like EphB4 in angiogenesis and tumor neovascularization [8], as well as vessel maturation and vascular integrity [25].
EphA5 was recently identified as a putative tumor suppressor through expression profiling, as mRNA expression was significantly downregulated in human breast cancer samples relative to normal human breast tissue, likely due to aberrant promoter methylation [26]. While several laboratory studies present evidence supporting EphB6 promoter methylation and tumor suppressor function [27–31], our analysis of mRNA expression in patient datasets revealed a significant association between elevated ephB6 and poorer overall and recurrence-free survival in breast cancer (Brantley-Sieders et al., in preparation). In addition, we also observed negative associations between survival/recurrence and elevated mRNA expression of ephA2, ephA4, ephA7, and ephB4 (Brantley-Sieders et al., in preparation). These observations are consistent with laboratory data for some Eph family members (e.g. EphA2, EphA7), but not others (e.g. EphA4, EphB4, EphB6), in human breast cancer cell lines [27], suggesting that cell line models must be carefully selected and multiple cell lines should be used so that they accurately recapitulate trends in human disease. At least one explanation for these conflicting data may reside in ligand-independent versus dependent signaling. For example, though we found no clear positive or negative correlations between expression of ephrin ligands and clinical outcome, we did observe an inverse correlation between EphA2 and ephrin-A1 protein expression in a significant number of invasive ductal carcinoma samples in lymph node relative to normal breast and ductal carcinomas confined to the breast, which co-express both (Brantley-Sieders et al., in preparation). This observation is consistent with profiling studies in breast cancer cell lines [16] and with the laboratory observations that ephrin-A1 ligand inhibits tumor cell growth and invasion [16, 32, 33]. Thus, future profiling efforts should include the full spectrum of relevant ephrin-ligands as well as Eph RTKs in order to elucidate potential differences in clinical outcome associated with the presence or absence of ligand. Moreover, soluble, monomeric ephrin-A1 has been detected in human breast cancer line supernatants [34, 35]. Soluble ligand can impose alternate biological outcomes compared to membrane-tethered ligands [35–37]. Therefore, assessing the localization (membrane versus soluble) of these ligands in situ may provide insight into the behavior of receptors detected in human cancers.
Together, these profiling efforts identified several A and B class Eph RTKs that may serve as tractable targets for human breast malignancies. Further analysis of how ligand expression profiles track with RTK expression will help elucidate the function of these receptors in specific malignancies. In addition, protein expression profiling, particularly studies geared toward detection of soluble ligands and post-translational modifications (e.g. serine/threonine versus tyrosine phosphorylation) of Eph RTKs and ephrin-B ligands, will no doubt shed light on the complex function of these molecules in cancer.
1.2 Eph expression profiles in human colorectal cancer
In addition to roles in normal gastrointestinal homeostasis and cell sorting in the gut, several B class Eph RTKs have been implicated in colorectal cancer [reviewed in [38, 39]]. Using commercial cDNA arrays coupled with immunoblot and immunohistochemical methods, Stephenson et al. reported elevated EphB4 expression in 82% of colon cancer tissues relative to matched normal tissue from the same patients, with protein expression localizing to tumor epithelium [40]. Martiny-Baron et al. reported elevated mRNA expression of ephB4 in human colon carcinomas relative to adjacent normal tissue controls [41]. Expression of ephB2, ephB3, and ephB4 mRNA was reported in human colon cancer tissue, along with B class ligand ephrin-B2. Interestingly, these tumors, as well as a panel of colorectal cancer (CRC) cell lines, were negative for ephB1, ephrin-B1, and ephrin-B3 [41, 42]. While these early studies suggested that several EphB family members play a role in human CRC, the relative number of patient samples analyzed was low [n=15 to 60; [40–42]] and the samples were not stratified by stage, grade, or level of invasiveness.
Although increased EphB RTK expression was detected in colorectal tumors, subsequent expression analyses coupled with genetically engineered mouse models suggest tumor suppressive functions for EphB receptors. Reduction or loss of EphB2 and EphB4 expression correlated with the shift from adenoma to invasive carcinoma in a panel of 108 human CRC samples, and loss of ephB3 mRNA was also observed in a smaller panel of tumors [43]. Similar results were reported in independent EphB2 [44] and EphB3 [45] profiling studies for human CRC. Reduced expression of EphB1 was also reported in poorly-differentiated, invasive CRCs [46] and in invasive gastric carcinomas [47]. In normal gut epithelium, the Wnt/β-catenin/TCF pathway regulates expression of EphB receptors in the Paneth/progenitor cell domain within the lower regions of crypts in a counter gradient to ephrin-B ligands that are expressed in differentiating cells higher in the crypt and in the villus. Thus, loss of EphB receptor expression may disrupt the normal cues that restrict tumor cell movement and enable unrestricted repopulation of the epithelial compartment as tumor progression proceeds [reviewed in [38]]. Indeed, a positive correlation between EphB2 expression and better overall and recurrence-free survival in human CRC patients has been identified in three independent studies [48–50]. Similar trends were reported for EphB4 expression in human CRC patient samples [51]. Moreover, EphB4 may have prognostic value for risk of relapse in human CRC [52]. Together, these data suggest that EphB receptors function as tumor suppressors in human CRC.
Relative to B class receptors and ligands, EphA RTKs have not been investigated extensively in human CRC. EphA1, EphA7, and EphA8 are reported to be downregulated in human CRC [53–56], and reduced EphA1 expression correlated with poor overall survival [53, 55]. These data suggest that, like their B class counterparts, these A class receptors may function as tumor suppressors in human CRC. Elevated EphA2 expression, however, correlated with liver/lymph node metastasis, lymphatic vessel infiltration, and clinical stage, as opposed to E-cadherin [57], and a similar trend was also reported for EphA2 [58, 59] and E-cadherin [59] in gastric cancer. EphA2 and ephrin-A1 expression were also correlated with MVD in human CRC samples [60], suggesting they might regulate neovascularization as well as tumorigenesis. These clinical observations are consistent with data derived from cell culture and animal studies [61–63]. Elevated EphA4 expression was reported for CRCs in the presence of liver metastasis, whereas lower EphB2 levels correlated with liver metastasis [64].
As with breast carcinoma, several Eph RTKs display expression profiles suggesting complex roles in CRC tumor progression. An emerging theme from analysis of clinical specimens and laboratory models is that Eph RTKs differentially regulate tumor growth versus tumor suppression in a stage-dependent manner. Thus, expression must be scored in the context of stage and grade. In addition, future profiling efforts should be geared toward looking at RTK expression in the context of ligand. In the case of CRC, this may be particularly important in determining the invasive potential of individual tumors, as localization of ligand relative to receptor may restrict invasion until receptor expression is lost, similar to what has been observed in development and normal gut homeostasis [reviewed in [38, 65]].
1.3 Eph expression profiles in human lung cancers
Recent studies suggest that Eph RTKs, particularly of the A class, play important roles in tumor progression or tumor suppression, depending on the individual family member. Initial presentation with high levels of EphA2 expression in non-small cell lung cancer (NSCLC) patients correlates with a history of smoking and is prognostic for metastasis, particularly brain metastasis, whereas low levels in the primary tumor correlate positively with disease-free survival or contralateral lung metastasis [66–68]. Subsequent studies demonstrated that ephA2 is part of a gene signature in NSCLC patients harboring somatic mutations in EGFR [69], an interesting parallel to the interaction between EphA2 and EGFR family members in breast cancer [reviewed in [9]]. EphA2 is also mutated in human NSCLC, as reported by Faoro et al. who demonstrated that an activating mutation G391R in EphA2 confers constitutive activation and activated signaling pathways that promote invasion [70]. Ephrin-A3 is also reportedly upregulated in human lung cancer [56]. In addition to changes in receptor levels, somatic mutations have been found in nearly all Eph receptors. Notably, 11 somatic mutations in EphA3 receptor were identified in 5–10% of lung cancer, placing EphA3 among 27 most frequently mutated genes in human lung adenocarcinoma [[52, 71–73]; Zhuang et al., submitted]. However, as these mutations are scattered throughout the receptor, and it is unclear whether they are "driver" or biologically neutral "passenger" genetic mutations. Elucidating the effects of these mutations will greatly improve our understanding of how Eph receptor functions in cancer. Indeed, insight into the function of individual domains, particularly putative and confirmed phosphotyrosine residues, will no doubt be provided by studies such as those conducted by Shi et al. [74]. We did observe, however, that overexpression of wild-type EphA3, but not several variants harboring mutations found in human cancers, significantly inhibited tumor cell survival in culture and in xenograft models, suggesting that they do play an important, possibly tumor suppressive role, in NSCLC (Zhuang et al., submitted).
Class B family members have also been detected in human lung cancer. EphB3 overexpression in human NSCLC samples correlated with tumor size, differentiation, and metastasis, in agreement with laboratory studies showing tumor promoting effects in culture and in mouse models [75]. Recent phosphoproteomic profiling analysis suggests that interplay between ephrin-B3 silencing in NSCLC lines and stabilization of EphA2 by phosphorylation Akt target Ser-897 may promote stability of EphA2 to support tumor cell survival [76]. It would be of great interest to determine if these observations are relevant to human disease, as they suggest that cross-talk between class A and class B Eph family molecules may also play a critical role in tumor progression.
Conclusions
In summary, Eph RTKs are altered in several types of human cancers and many represent promising targets for novel, molecularly targeted therapies, particularly in breast, colon, and lung carcinomas. While the studies discussed here demonstrate the relevance of Eph RTKs and their ligands to human malignancies, several questions remain. In spite of the wealth of information regarding expression profiles in human cancer, gaps remain in our knowledge for some family members, as well as for the spectrum of tumor progression (e.g. stage and grade). Given the complexity of signaling regulated by this RTK family, as well as extensive cross-talk with other RTK families involved in cancer, future efforts should be aimed at understanding how Eph receptor expression and function is modulated in the context of relevant cancer pathways.
Acknowledgments
I thank Drs. Jin Chen and Rebecca Cook at Vanderbilt University for helpful discussions and proofreading. Dana M. Brantley-Sieders is supported by NIH/NCI grants CA1179151 and CA148934.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 2.Hahn WC, Weinberg RA. Rules for making human tumor cells. N Engl J Med. 2002;347:1593–1603. doi: 10.1056/NEJMra021902. [DOI] [PubMed] [Google Scholar]
- 3.Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10:789–799. doi: 10.1038/nm1087. [DOI] [PubMed] [Google Scholar]
- 4.Perona R. Cell signalling: growth factors and tyrosine kinase receptors. Clin Transl Oncol. 2006;8:77–82. doi: 10.1007/s12094-006-0162-1. [DOI] [PubMed] [Google Scholar]
- 5.Blume-Jensen P, Hunter T. Oncogenic kinase signalling. Nature. 2001;411:355–365. doi: 10.1038/35077225. [DOI] [PubMed] [Google Scholar]
- 6.Wykosky J, Debinski W. The EphA2 receptor and ephrinA1 ligand in solid tumors: function and therapeutic targeting. Mol Cancer Res. 2008;6:1795–1806. doi: 10.1158/1541-7786.MCR-08-0244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Himanen JP, Saha N, Nikolov DB. Cell-cell signaling via Eph receptors and ephrins. Curr Opin Cell Biol. 2007;19:534–542. doi: 10.1016/j.ceb.2007.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Noren NK, Pasquale EB. Paradoxes of the EphB4 receptor in cancer. Cancer Res. 2007;67:3994–3997. doi: 10.1158/0008-5472.CAN-07-0525. [DOI] [PubMed] [Google Scholar]
- 9.Vaught D, Brantley-Sieders DM, Chen J. Eph receptors in breast cancer: roles in tumor promotion and tumor suppression. Breast Cancer Res. 2008;10:217. doi: 10.1186/bcr2207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ogawa K, Pasqualini R, Lindberg RA, Kain R, Freeman AL, Pasquale EB. The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene. 2000;19:6043–6052. doi: 10.1038/sj.onc.1204004. [DOI] [PubMed] [Google Scholar]
- 11.Zelinski DP, Zantek ND, Stewart JC, Irizarry AR, Kinch MS. EphA2 overexpression causes tumorigenesis of mammary epithelial cells. Cancer Res. 2001;61:2301–2306. [PubMed] [Google Scholar]
- 12.Pan M. Overexpression of EphA2 gene in invasive human breast cancer and its association with hormone receptor statues [ASCO annual meeting proceedings] J Clin Oncol. 2005;23:9583. [Google Scholar]
- 13.Fournier MV, Martin KJ, Kenny PA, Xhaja K, Bosch I, Yaswen P, et al. Gene expression signature in organized and growth-arrested mammary acini predicts good outcome in breast cancer. Cancer Res. 2006;66:7095–7102. doi: 10.1158/0008-5472.CAN-06-0515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Martin KJ, Patrick DR, Bissell MJ, Fournier MV. Prognostic breast cancer signature identified from 3D culture model accurately predicts clinical outcome across independent datasets. PLoS One. 2008;3:e2994. doi: 10.1371/journal.pone.0002994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhuang G, Brantley-Sieders DM, Vaught D, Yu J, Xie L, Wells S, et al. Elevation of receptor tyrosine kinase EphA2 mediates resistance to trastuzumab therapy. Cancer Res. 2010;70:299–308. doi: 10.1158/0008-5472.CAN-09-1845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Macrae M, Neve RM, Rodriguez-Viciana P, Haqq C, Yeh J, Chen C, et al. A conditional feedback loop regulates Ras activity through EphA2. Cancer Cell. 2005;8:111–118. doi: 10.1016/j.ccr.2005.07.005. [DOI] [PubMed] [Google Scholar]
- 17.Brantley-Sieders DM, Zhuang G, Hicks D, Fang WB, Hwang Y, Cates JM, et al. The receptor tyrosine kinase EphA2 promotes mammary adenocarcinoma tumorigenesis and metastatic progression in mice by amplifying ErbB2 signaling. J Clin Invest. 2008;118:64–78. doi: 10.1172/JCI33154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Larsen AB, Pedersen MW, Stockhausen MT, Grandal MV, van Deurs B, Poulsen HS. Activation of the EGFR gene target EphA2 inhibits epidermal growth factor-induced cancer cell motility. Mol Cancer Res. 2007;5:283–293. doi: 10.1158/1541-7786.MCR-06-0321. [DOI] [PubMed] [Google Scholar]
- 19.Pedersen K, Angelini PD, Laos S, Bach-Faig A, Cunningham MP, Ferrer-Ramon C, et al. A naturally occurring HER2 carboxy-terminal fragment promotes mammary tumor growth and metastasis. Mol Cell Biol. 2009;29:3319–3331. doi: 10.1128/MCB.01803-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Brantley-Sieders DM, Dunaway CM, Rao M, Short S, Hwang Y, Gao Y, et al. Angiocrine factors modulate tumor proliferation and motility through EphA2 repression of Slit2 tumor suppressor function in endothelium. Cancer Res. 2011;71:976–987. doi: 10.1158/0008-5472.CAN-10-3396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Berclaz G, Andres AC, Albrecht D, Dreher E, Ziemiecki A, Gusterson BA, et al. Expression of the receptor protein tyrosine kinase myk-1/htk in normal and malignant mammary epithelium. Biochem Biophys Res Commun. 1996;226:869–875. doi: 10.1006/bbrc.1996.1442. [DOI] [PubMed] [Google Scholar]
- 22.Wu Q, Suo Z, Risberg B, Karlsson MG, Villman K, Nesland JM. Expression of Ephb2 and Ephb4 in breast carcinoma. Pathol Oncol Res. 2004;10:26–33. doi: 10.1007/BF02893405. [DOI] [PubMed] [Google Scholar]
- 23.Berclaz G, Flutsch B, Altermatt HJ, Rohrbach V, Djonov V, Ziemiecki A, et al. Loss of EphB4 receptor tyrosine kinase protein expression during carcinogenesis of the human breast. Oncol Rep. 2002;9:985–989. [PubMed] [Google Scholar]
- 24.Noren NK, Foos G, Hauser CA, Pasquale EB. The EphB4 receptor suppresses breast cancer cell tumorigenicity through an Abl-Crk pathway. Nat Cell Biol. 2006;8:815–825. doi: 10.1038/ncb1438. [DOI] [PubMed] [Google Scholar]
- 25.Salvucci O, Maric D, Economopoulou M, Sakakibara S, Merlin S, Follenzi A, et al. EphrinB reverse signaling contributes to endothelial and mural cell assembly into vascular structures. Blood. 2009;114:1707–1716. doi: 10.1182/blood-2008-12-192294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Fu DY, Wang ZM, Wang BL, Chen L, Yang WT, Shen ZZ, et al. Frequent epigenetic inactivation of the receptor tyrosine kinase EphA5 by promoter methylation in human breast cancer. Hum Pathol. 2010;41:48–58. doi: 10.1016/j.humpath.2009.06.007. [DOI] [PubMed] [Google Scholar]
- 27.Fox BP, Kandpal RP. Invasiveness of breast carcinoma cells and transcript profile: Eph receptors and ephrin ligands as molecular markers of potential diagnostic and prognostic application. Biochem Biophys Res Commun. 2004;318:882–892. doi: 10.1016/j.bbrc.2004.04.102. [DOI] [PubMed] [Google Scholar]
- 28.Fox BP, Tabone CJ, Kandpal RP. Potential clinical relevance of Eph receptors and ephrin ligands expressed in prostate carcinoma cell lines. Biochem Biophys Res Commun. 2006;342:1263–1272. doi: 10.1016/j.bbrc.2006.02.099. [DOI] [PubMed] [Google Scholar]
- 29.Fox BP, Kandpal RP. EphB6 receptor significantly alters invasiveness and other phenotypic characteristics of human breast carcinoma cells. Oncogene. 2009;28:1706–1713. doi: 10.1038/onc.2009.18. [DOI] [PubMed] [Google Scholar]
- 30.Kandpal RP. Tyrosine kinase-deficient EphB6 receptor-dependent alterations in proteomic profiles of invasive breast carcinoma cells as determined by difference gel electrophoresis. Cancer Genomics Proteomics. 2010;7:253–260. [PubMed] [Google Scholar]
- 31.Truitt L, Freywald T, DeCoteau J, Sharfe N, Freywald A. The EphB6 receptor cooperates with c-Cbl to regulate the behavior of breast cancer cells. Cancer Res. 2010;70:1141–1153. doi: 10.1158/0008-5472.CAN-09-1710. [DOI] [PubMed] [Google Scholar]
- 32.Miao H, Wei BR, Peehl DM, Li Q, Alexandrou T, Schelling JR, et al. Activation of EphA receptor tyrosine kinase inhibits the Ras/MAPK pathway. Nat Cell Biol. 2001;3:527–530. doi: 10.1038/35074604. [DOI] [PubMed] [Google Scholar]
- 33.Guo H, Miao H, Gerber L, Singh J, Denning MF, Gilliam AC, et al. Disruption of EphA2 receptor tyrosine kinase leads to increased susceptibility to carcinogenesis in mouse skin. Cancer Res. 2006;66:7050–7058. doi: 10.1158/0008-5472.CAN-06-0004. [DOI] [PubMed] [Google Scholar]
- 34.Wykosky J, Palma E, Gibo DM, Ringler S, Turner CP, Debinski W. Soluble monomeric EphrinA1 is released from tumor cells and is a functional ligand for the EphA2 receptor. Oncogene. 2008;27:7260–7273. doi: 10.1038/onc.2008.328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Alford S, Watson-Hurthig A, Scott N, Carette A, Lorimer H, Bazowski J, et al. Soluble ephrin a1 is necessary for the growth of HeLa and SK-BR3 cells. Cancer Cell Int. 2010;10:41. doi: 10.1186/1475-2867-10-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Stein E, Lane AA, Cerretti DP, Schoecklmann HO, Schroff AD, Van Etten RL, et al. Eph receptors discriminate specific ligand oligomers to determine alternative signaling complexes, attachment, and assembly responses. Genes Dev. 1998;12:667–678. doi: 10.1101/gad.12.5.667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Alford SC, Bazowski J, Lorimer H, Elowe S, Howard PL. Tissue transglutaminase clusters soluble A-type ephrins into functionally active high molecular weight oligomers. Exp Cell Res. 2007;313:4170–4179. doi: 10.1016/j.yexcr.2007.07.019. [DOI] [PubMed] [Google Scholar]
- 38.Clevers H, Batlle E. EphB/EphrinB receptors and Wnt signaling in colorectal cancer. Cancer Res. 2006;66:2–5. doi: 10.1158/0008-5472.CAN-05-3849. [DOI] [PubMed] [Google Scholar]
- 39.Herath NI, Boyd AW. The role of Eph receptors and ephrin ligands in colorectal cancer. Int J Cancer. 2010;126:2003–2011. doi: 10.1002/ijc.25147. [DOI] [PubMed] [Google Scholar]
- 40.Stephenson SA, Slomka S, Douglas EL, Hewett PJ, Hardingham JE. Receptor protein tyrosine kinase EphB4 is up-regulated in colon cancer. BMC Mol Biol. 2001;2:15. doi: 10.1186/1471-2199-2-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Martiny-Baron G, Korff T, Schaffner F, Esser N, Eggstein S, Marme D, et al. Inhibition of tumor growth and angiogenesis by soluble EphB4. Neoplasia. 2004;6:248–257. doi: 10.1593/neo.3457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Liu W, Ahmad SA, Jung YD, Reinmuth N, Fan F, Bucana CD, et al. Coexpression of ephrin-Bs and their receptors in colon carcinoma. Cancer. 2002;94:934–939. doi: 10.1002/cncr.10122. [DOI] [PubMed] [Google Scholar]
- 43.Batlle E, Bacani J, Begthel H, Jonkheer S, Gregorieff A, van de Born M, et al. EphB receptor activity suppresses colorectal cancer progression. Nature. 2005;435:1126–1130. doi: 10.1038/nature03626. [DOI] [PubMed] [Google Scholar]
- 44.Guo DL, Zhang J, Yuen ST, Tsui WY, Chan AS, Ho C, et al. Reduced expression of EphB2 that parallels invasion and metastasis in colorectal tumours. Carcinogenesis. 2006;27:454–464. doi: 10.1093/carcin/bgi259. [DOI] [PubMed] [Google Scholar]
- 45.Chiu ST, Chang KJ, Ting CH, Shen HC, Li H, Hsieh FJ. Over-expression of EphB3 enhances cell-cell contacts and suppresses tumor growth in HT-29 human colon cancer cells. Carcinogenesis. 2009;30:1475–1486. doi: 10.1093/carcin/bgp133. [DOI] [PubMed] [Google Scholar]
- 46.Sheng Z, Wang J, Dong Y, Ma H, Zhou H, Sugimura H, et al. EphB1 is underexpressed in poorly differentiated colorectal cancers. Pathobiology. 2008;75:274–280. doi: 10.1159/000151707. [DOI] [PubMed] [Google Scholar]
- 47.Wang JD, Dong YC, Sheng Z, Ma HH, Li GL, Wang XL, et al. Loss of expression of EphB1 protein in gastric carcinoma associated with invasion and metastasis. Oncology. 2007;73:238–245. doi: 10.1159/000127421. [DOI] [PubMed] [Google Scholar]
- 48.Jubb AM, Zhong F, Bheddah S, Grabsch HI, Frantz GD, Mueller W, et al. EphB2 is a prognostic factor in colorectal cancer. Clin Cancer Res. 2005;11:5181–5187. doi: 10.1158/1078-0432.CCR-05-0143. [DOI] [PubMed] [Google Scholar]
- 49.Lugli A, Spichtin H, Maurer R, Mirlacher M, Kiefer J, Huusko P, et al. EphB2 expression across 138 human tumor types in a tissue microarray: high levels of expression in gastrointestinal cancers. Clin Cancer Res. 2005;11:6450–6458. doi: 10.1158/1078-0432.CCR-04-2458. [DOI] [PubMed] [Google Scholar]
- 50.Yu G, Gao Y, Ni C, Chen Y, Pan J, Wang X, et al. Reduced expression of EphB2 is significantly associated with nodal metastasis in Chinese patients with gastric cancer. J Cancer Res Clin Oncol. 2011;137:73–80. doi: 10.1007/s00432-010-0861-4. [DOI] [PubMed] [Google Scholar]
- 51.Davalos V, Dopeso H, Castano J, Wilson AJ, Vilardell F, Romero-Gimenez J, et al. EPHB4 and survival of colorectal cancer patients. Cancer Res. 2006;66:8943–8948. doi: 10.1158/0008-5472.CAN-05-4640. [DOI] [PubMed] [Google Scholar]
- 52.Lloyd JM, McIver CM, Stephenson SA, Hewett PJ, Rieger N, Hardingham JE. Identification of early-stage colorectal cancer patients at risk of relapse post-resection by immunobead reverse transcription-PCR analysis of peritoneal lavage fluid for malignant cells. Clin Cancer Res. 2006;12:417–423. doi: 10.1158/1078-0432.CCR-05-1473. [DOI] [PubMed] [Google Scholar]
- 53.Dong Y, Wang J, Sheng Z, Li G, Ma H, Wang X, et al. Downregulation of EphA1 in colorectal carcinomas correlates with invasion and metastasis. Mod Pathol. 2009;22:151–160. doi: 10.1038/modpathol.2008.188. [DOI] [PubMed] [Google Scholar]
- 54.Wang J, Kataoka H, Suzuki M, Sato N, Nakamura R, Tao H, et al. Downregulation of EphA7 by hypermethylation in colorectal cancer. Oncogene. 2005;24:5637–5647. doi: 10.1038/sj.onc.1208720. [DOI] [PubMed] [Google Scholar]
- 55.Herath NI, Doecke J, Spanevello MD, Leggett BA, Boyd AW. Epigenetic silencing of EphA1 expression in colorectal cancer is correlated with poor survival. Br J Cancer. 2009;100:1095–1102. doi: 10.1038/sj.bjc.6604970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Hafner C, Schmitz G, Meyer S, Bataille F, Hau P, Langmann T, et al. Differential gene expression of Eph receptors and ephrins in benign human tissues and cancers. Clin Chem. 2004;50:490–499. doi: 10.1373/clinchem.2003.026849. [DOI] [PubMed] [Google Scholar]
- 57.Saito T, Masuda N, Miyazaki T, Kanoh K, Suzuki H, Shimura T, et al. Expression of EphA2 and E-cadherin in colorectal cancer: correlation with cancer metastasis. Oncol Rep. 2004;11:605–611. [PubMed] [Google Scholar]
- 58.Yuan WJ, Ge J, Chen ZK, Wu SB, Shen H, Yang P, et al. Over-expression of EphA2 and EphrinA-1 in human gastric adenocarcinoma and its prognostic value for postoperative patients. Dig Dis Sci. 2009;54:2410–2417. doi: 10.1007/s10620-008-0649-4. [DOI] [PubMed] [Google Scholar]
- 59.Yuan W, Chen Z, Wu S, Ge J, Chang S, Wang X, et al. Expression of EphA2 and E-cadherin in gastric cancer: correlated with tumor progression and lymphogenous metastasis. Pathol Oncol Res. 2009;15:473–478. doi: 10.1007/s12253-008-9132-y. [DOI] [PubMed] [Google Scholar]
- 60.Kataoka H, Igarashi H, Kanamori M, Ihara M, Wang JD, Wang YJ, et al. Correlation of EPHA2 overexpression with high microvessel count in human primary colorectal cancer. Cancer Sci. 2004;95:136–141. doi: 10.1111/j.1349-7006.2004.tb03194.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Potla L, Boghaert ER, Armellino D, Frost P, Damle NK. Reduced expression of EphrinA1 (EFNA1) inhibits three-dimensional growth of HT29 colon carcinoma cells. Cancer Lett. 2002;175:187–195. doi: 10.1016/s0304-3835(01)00613-9. [DOI] [PubMed] [Google Scholar]
- 62.Li Z, Tanaka M, Kataoka H, Nakamura R, Sanjar R, Shinmura K, et al. EphA2 up-regulation induced by deoxycholic acid in human colon carcinoma cells, an involvement of extracellular signal-regulated kinase and p53-independence. J Cancer Res Clin Oncol. 2003;129:703–708. doi: 10.1007/s00432-003-0493-z. [DOI] [PubMed] [Google Scholar]
- 63.Bogan C, Chen J, O'Sullivan MG, Cormier RT. Loss of EphA2 receptor tyrosine kinase reduces ApcMin/+ tumorigenesis. Int J Cancer. 2009;124:1366–1371. doi: 10.1002/ijc.24083. [DOI] [PubMed] [Google Scholar]
- 64.Oshima T, Akaike M, Yoshihara K, Shiozawa M, Yamamoto N, Sato T, et al. Overexpression of EphA4 gene and reduced expression of EphB2 gene correlates with liver metastasis in colorectal cancer. Int J Oncol. 2008;33:573–577. [PubMed] [Google Scholar]
- 65.Pasquale EB. Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nat Rev Cancer. 2010;10:165–180. doi: 10.1038/nrc2806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kinch MS, Moore MB, Harpole DH., Jr Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin Cancer Res. 2003;9:613–618. [PubMed] [Google Scholar]
- 67.Brannan JM, Sen B, Saigal B, Prudkin L, Behrens C, Solis L, et al. EphA2 in the early pathogenesis and progression of non-small cell lung cancer. Cancer Prev Res (Phila) 2009;2:1039–1049. doi: 10.1158/1940-6207.CAPR-09-0212. [DOI] [PubMed] [Google Scholar]
- 68.Brannan JM, Dong W, Prudkin L, Behrens C, Lotan R, Bekele BN, et al. Expression of the receptor tyrosine kinase EphA2 is increased in smokers and predicts poor survival in non-small cell lung cancer. Clin Cancer Res. 2009;15:4423–4430. doi: 10.1158/1078-0432.CCR-09-0473. [DOI] [PubMed] [Google Scholar]
- 69.Choi K, Creighton CJ, Stivers D, Fujimoto N, Kurie JM. Transcriptional profiling of non-small cell lung cancer cells with activating EGFR somatic mutations. PLoS One. 2007;2:e1226. doi: 10.1371/journal.pone.0001226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Faoro L, Singleton PA, Cervantes GM, Lennon FE, Choong NW, Kanteti R, et al. EphA2 mutation in lung squamous cell carcinoma promotes increased cell survival, cell invasion, focal adhesions, and mammalian target of rapamycin activation. J Biol Chem. 2010;285:18575–18585. doi: 10.1074/jbc.M109.075085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Davies H, Hunter C, Smith R, Stephens P, Greenman C, Bignell G, et al. Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res. 2005;65:7591–7595. doi: 10.1158/0008-5472.CAN-05-1855. [DOI] [PubMed] [Google Scholar]
- 72.Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, et al. Patterns of somatic mutation in human cancer genomes. Nature. 2007;446:153–158. doi: 10.1038/nature05610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Alam SM, Fujimoto J, Jahan I, Sato E, Tamaya T. Coexpression of EphB4 and ephrinB2 in tumour advancement of ovarian cancers. Br J Cancer. 2008;98:845–851. doi: 10.1038/sj.bjc.6604216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Shi G, Yue G, Zhou R. EphA3 functions are regulated by collaborating phosphotyrosine residues. Cell Res. 2010;20:1263–1275. doi: 10.1038/cr.2010.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ji XD, Li G, Feng YX, Zhao JS, Li JJ, Sun ZJ, et al. EphB3 is overexpressed in non-small-cell lung cancer and promotes tumor metastasis by enhancing cell survival and migration. Cancer Res. 2011;71:1156–1166. doi: 10.1158/0008-5472.CAN-10-0717. [DOI] [PubMed] [Google Scholar]
- 76.Stahl S, Mm Branca R, Efazat G, Ruzzene M, Zhivotovsky B, Lewensohn R, et al. Phosphoproteomic Profiling of NSCLC Cells Reveals that Ephrin B3 Regulates Pro-survival Signaling through Akt1-Mediated Phosphorylation of the EphA2 Receptor. J Proteome Res. 2011 doi: 10.1021/pr200037u. [DOI] [PubMed] [Google Scholar]
- 77.Herath NI, Spanevello MD, Sabesan S, Newton T, Cummings M, Duffy S, et al. Over-expression of Eph and ephrin genes in advanced ovarian cancer: ephrin gene expression correlates with shortened survival. BMC Cancer. 2006;6:144. doi: 10.1186/1471-2407-6-144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Raza SM, Fuller GN, Rhee CH, Huang S, Hess K, Zhang W, et al. Identification of necrosis-associated genes in glioblastoma by cDNA microarray analysis. Clin Cancer Res. 2004;10:212–221. doi: 10.1158/1078-0432.ccr-0155-3. [DOI] [PubMed] [Google Scholar]
- 79.Wang J, Dong Y, Wang X, Ma H, Sheng Z, Li G, et al. Expression of EphA1 in gastric carcinomas is associated with metastasis and survival. Oncol Rep. 2010;24:1577–1584. doi: 10.3892/or_00001020. [DOI] [PubMed] [Google Scholar]
- 80.Kiyokawa E, Takai S, Tanaka M, Iwase T, Suzuki M, Xiang YY, et al. Overexpression of ERK, an EPH family receptor protein tyrosine kinase, in various human tumors. Cancer Res. 1994;54:3645–3650. [PubMed] [Google Scholar]
- 81.Giaginis C, Tsourouflis G, Zizi-Serbetzoglou A, Kouraklis G, Chatzopoulou E, Dimakopoulou K, et al. Clinical significance of ephrin (eph)-A1, -A2, -a4, -a5 and -a7 receptors in pancreatic ductal adenocarcinoma. Pathol Oncol Res. 2010;16:267–276. doi: 10.1007/s12253-009-9221-6. [DOI] [PubMed] [Google Scholar]
- 82.Maru Y, Hirai H, Yoshida MC, Takaku F. Evolution, expression, and chromosomal location of a novel receptor tyrosine kinase gene, eph. Mol Cell Biol. 1988;8:3770–3776. doi: 10.1128/mcb.8.9.3770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Chen G, Wang Y, Zhou M, Shi H, Yu Z, Zhu Y, et al. EphA1 receptor silencing by small interfering RNA has antiangiogenic and antitumor efficacy in hepatocellular carcinoma. Oncol Rep. 2010;23:563–570. [PubMed] [Google Scholar]
- 84.Fournier MV, Martin KJ. Transcriptome profiling in clinical breast cancer: from 3D culture models to prognostic signatures. J Cell Physiol. 2006;209:625–630. doi: 10.1002/jcp.20787. [DOI] [PubMed] [Google Scholar]
- 85.Lin YG, Han LY, Kamat AA, Merritt WM, Landen CN, Deavers MT, et al. EphA2 overexpression is associated with angiogenesis in ovarian cancer. Cancer. 2007;109:332–340. doi: 10.1002/cncr.22415. [DOI] [PubMed] [Google Scholar]
- 86.Nakamura R, Kataoka H, Sato N, Kanamori M, Ihara M, Igarashi H, et al. EPHA2/EFNA1 expression in human gastric cancer. Cancer Sci. 2005;96:42–47. doi: 10.1111/j.1349-7006.2005.00007.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Xu F, Zhong W, Li J, Shanshen Z, Cui J, Nesland JM, et al. Predictive value of EphA2 and EphrinA-1 expression in oesophageal squamous cell carcinoma. Anticancer Res. 2005;25:2943–2950. [PubMed] [Google Scholar]
- 88.Miyazaki T, Kato H, Fukuchi M, Nakajima M, Kuwano H. EphA2 overexpression correlates with poor prognosis in esophageal squamous cell carcinoma. Int J Cancer. 2003;103:657–663. doi: 10.1002/ijc.10860. [DOI] [PubMed] [Google Scholar]
- 89.Wykosky J, Gibo DM, Stanton C, Debinski W. EphA2 as a novel molecular marker and target in glioblastoma multiforme. Mol Cancer Res. 2005;3:541–551. doi: 10.1158/1541-7786.MCR-05-0056. [DOI] [PubMed] [Google Scholar]
- 90.Miao H, Li DQ, Mukherjee A, Guo H, Petty A, Cutter J, et al. EphA2 mediates ligand-dependent inhibition and ligand-independent promotion of cell migration and invasion via a reciprocal regulatory loop with Akt. Cancer Cell. 2009;16:9–20. doi: 10.1016/j.ccr.2009.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Li X, Wang L, Gu JW, Li B, Liu WP, Wang YG, et al. Up-regulation of EphA2 and down-regulation of EphrinA1 are associated with the aggressive phenotype and poor prognosis of malignant glioma. Tumour Biol. 2010;31:477–488. doi: 10.1007/s13277-010-0060-6. [DOI] [PubMed] [Google Scholar]
- 92.Mudali SV, Fu B, Lakkur SS, Luo M, Embuscado EE, Iacobuzio-Donahue CA. Patterns of EphA2 protein expression in primary and metastatic pancreatic carcinoma and correlation with genetic status. Clin Exp Metastasis. 2006;23:357–365. doi: 10.1007/s10585-006-9045-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yang P, Yuan W, He J, Wang J, Yu L, Jin X, et al. Overexpression of EphA2, MMP-9, and MVD-CD34 in hepatocellular carcinoma: Implications for tumor progression and prognosis. Hepatol Res. 2009;39:1169–1177. doi: 10.1111/j.1872-034X.2009.00563.x. [DOI] [PubMed] [Google Scholar]
- 94.Walker-Daniels J, Coffman K, Azimi M, Rhim JS, Bostwick DG, Snyder P, et al. Overexpression of the EphA2 tyrosine kinase in prostate cancer. Prostate. 1999;41:275–280. doi: 10.1002/(sici)1097-0045(19991201)41:4<275::aid-pros8>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
- 95.Abraham S, Knapp DW, Cheng L, Snyder PW, Mittal SK, Bangari DS, et al. Expression of EphA2 and Ephrin A-1 in carcinoma of the urinary bladder. Clin Cancer Res. 2006;12:353–360. doi: 10.1158/1078-0432.CCR-05-1505. [DOI] [PubMed] [Google Scholar]
- 96.Wood LD, Calhoun ES, Silliman N, Ptak J, Szabo S, Powell SM, et al. Somatic mutations of GUCY2F, EPHA3, and NTRK3 in human cancers. Hum Mutat. 2006;27:1060–1061. doi: 10.1002/humu.9452. [DOI] [PubMed] [Google Scholar]
- 97.Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–1075. doi: 10.1038/nature07423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bae HJ, Song JH, Noh JH, Kim JK, Jung KH, Eun JW, et al. Low frequency mutation of the Ephrin receptor A3 gene in hepatocellular carcinoma. Neoplasma. 2009;56:331–334. doi: 10.4149/neo_2009_04_331. [DOI] [PubMed] [Google Scholar]
- 99.Balakrishnan A, Bleeker FE, Lamba S, Rodolfo M, Daniotti M, Scarpa A, et al. Novel somatic and germline mutations in cancer candidate genes in glioblastoma, melanoma, and pancreatic carcinoma. Cancer Res. 2007;67:3545–3550. doi: 10.1158/0008-5472.CAN-07-0065. [DOI] [PubMed] [Google Scholar]
- 100.Fukai J, Yokote H, Yamanaka R, Arao T, Nishio K, Itakura T. EphA4 promotes cell proliferation and migration through a novel EphA4-FGFR1 signaling pathway in the human glioma U251 cell line. Mol Cancer Ther. 2008;7:2768–2778. doi: 10.1158/1535-7163.MCT-07-2263. [DOI] [PubMed] [Google Scholar]
- 101.Iiizumi M, Hosokawa M, Takehara A, Chung S, Nakamura T, Katagiri T, et al. EphA4 receptor, overexpressed in pancreatic ductal adenocarcinoma, promotes cancer cell growth. Cancer Sci. 2006;97:1211–1216. doi: 10.1111/j.1349-7006.2006.00313.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wang LF, Fokas E, Juricko J, You A, Rose F, Pagenstecher A, et al. Increased expression of EphA7 correlates with adverse outcome in primary and recurrent glioblastoma multiforme patients. BMC Cancer. 2008;8:79. doi: 10.1186/1471-2407-8-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Guan M, Xu C, Zhang F, Ye C. Aberrant methylation of EphA7 in human prostate cancer and its relation to clinicopathologic features. Int J Cancer. 2009;124:88–94. doi: 10.1002/ijc.23890. [DOI] [PubMed] [Google Scholar]
- 104.Easty DJ, Hill SP, Hsu MY, Fallowfield ME, Florenes VA, Herlyn M, et al. Up-regulation of ephrin-A1 during melanoma progression. Int J Cancer. 1999;84:494–501. doi: 10.1002/(sici)1097-0215(19991022)84:5<494::aid-ijc8>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
- 105.Feng YX, Zhao JS, Li JJ, Wang T, Cheng SQ, Yuan Y, et al. Liver cancer: EphrinA2 promotes tumorigenicity through Rac1/Akt/NF-kappaB signaling pathway 120. Hepatology. 2010;51:535–544. doi: 10.1002/hep.23313. [DOI] [PubMed] [Google Scholar]
- 106.Li JJ, Liu DP, Liu GT, Xie D. EphrinA5 acts as a tumor suppressor in glioma by negative regulation of epidermal growth factor receptor. Oncogene. 2009;28:1759–1768. doi: 10.1038/onc.2009.15. [DOI] [PubMed] [Google Scholar]
- 107.Nakada M, Niska JA, Tran NL, McDonough WS, Berens ME. EphB2/R-Ras signaling regulates glioma cell adhesion, growth, and invasion. Am J Pathol. 2005;167:565–576. doi: 10.1016/S0002-9440(10)62998-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Tang XX, Evans AE, Zhao H, Cnaan A, Brodeur GM, Ikegaki N. Association among EPHB2, TrkA, and MYCN expression in low-stage neuroblastomas. Med Pediatr Oncol. 2001;36:80–82. doi: 10.1002/1096-911X(20010101)36:1<80::AID-MPO1019>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
- 109.Zhang S, Jiang T, Liang M. Expression of EphB4 and Ephrin B2 in cervical cancer tissues and angiogenesis. Int J Gynaecol Obstet. 2007;96:46–47. doi: 10.1016/j.ijgo.2006.08.017. [DOI] [PubMed] [Google Scholar]
- 110.Alam SM, Fujimoto J, Jahan I, Sato E, Tamaya T. Coexpression of EphB4 and ephrinB2 in tumor advancement of uterine cervical cancers. Gynecol Oncol. 2009;114:84–88. doi: 10.1016/j.ygyno.2009.03.017. [DOI] [PubMed] [Google Scholar]
- 111.Takai N, Miyazaki T, Fujisawa K, Nasu K, Miyakawa I. Expression of receptor tyrosine kinase EphB4 and its ligand ephrin-B2 is associated with malignant potential in endometrial cancer. Oncol Rep. 2001;8:567–573. doi: 10.3892/or.8.3.567. [DOI] [PubMed] [Google Scholar]
- 112.Berclaz G, Karamitopoulou E, Mazzucchelli L, Rohrbach V, Dreher E, Ziemiecki A, et al. Activation of the receptor protein tyrosine kinase EphB4 in endometrial hyperplasia and endometrial carcinoma. Ann Oncol. 2003;14:220–226. doi: 10.1093/annonc/mdg072. [DOI] [PubMed] [Google Scholar]
- 113.Lee YC, Perren JR, Douglas EL, Raynor MP, Bartley MA, Bardy PG, et al. Investigation of the expression of the EphB4 receptor tyrosine kinase in prostate carcinoma. BMC Cancer. 2005;5:119. doi: 10.1186/1471-2407-5-119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Jin M, Komohara Y, Shichijo S, Harada M, Yamanaka R, Miyamoto S, et al. Identification of EphB6 variant-derived epitope peptides recognized by cytotoxic T-lymphocytes from HLA-A24+ malignant glioma patients. Oncol Rep. 2008;19:1277–1283. [PubMed] [Google Scholar]
- 115.Jin M, Komohara Y, Shichijo S, Yamanaka R, Nikawa J, Itoh K, et al. Erythropoietin-producing hepatocyte B6 variant-derived peptides with the ability to induce glioma-reactive cytotoxic T lymphocytes in human leukocyte antigen-A2+ glioma patients. Cancer Sci. 2008;99:1656–1662. doi: 10.1111/j.1349-7006.2008.00866.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Tang XX, Evans AE, Zhao H, Cnaan A, London W, Cohn SL, et al. High-level expression of EPHB6, EFNB2, and EFNB3 is associated with low tumor stage and high TrkA expression in human neuroblastomas. Clin Cancer Res. 1999;5:1491–1496. [PubMed] [Google Scholar]
- 117.Tang XX, Zhao H, Robinson ME, Cohen B, Cnaan A, London W, et al. Implications of EPHB6, EFNB2, and EFNB3 expressions in human neuroblastoma. Proc Natl Acad Sci U S A. 2000;97:10936–10941. doi: 10.1073/pnas.190123297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Tang XX, Zhao H, Robinson ME, Cnaan A, London W, Cohn SL, et al. Prognostic significance of EPHB6, EFNB2, and EFNB3 expressions in neuroblastoma. Med Pediatr Oncol. 2000;35:656–658. doi: 10.1002/1096-911x(20001201)35:6<656::aid-mpo37>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
- 119.Hafner C, Bataille F, Meyer S, Becker B, Roesch A, Landthaler M, et al. Loss of EphB6 expression in metastatic melanoma. Int J Oncol. 2003;23:1553–1559. [PubMed] [Google Scholar]
- 120.Castellvi J, Garcia A, de la Torre J, Hernandez J, Gil A, Xercavins J, et al. Ephrin B expression in epithelial ovarian neoplasms correlates with tumor differentiation and angiogenesis. Hum Pathol. 2006;37:883–889. doi: 10.1016/j.humpath.2006.02.021. [DOI] [PubMed] [Google Scholar]
- 121.Nakada M, Anderson EM, Demuth T, Nakada S, Reavie LB, Drake KL, et al. The phosphorylation of ephrin-B2 ligand promotes glioma cell migration and invasion. Int J Cancer. 2010;126:1155–1165. doi: 10.1002/ijc.24849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Sawai Y, Tamura S, Fukui K, Ito N, Imanaka K, Saeki A, et al. Expression of ephrin-B1 in hepatocellular carcinoma: possible involvement in neovascularization. J Hepatol. 2003;39:991–996. doi: 10.1016/s0168-8278(03)00498-7. [DOI] [PubMed] [Google Scholar]
- 123.Tachibana M, Tonomoto Y, Hyakudomi R, Hyakudomi M, Hattori S, Ueda S, et al. Expression and prognostic significance of EFNB2 and EphB4 genes in patients with oesophageal squamous cell carcinoma. Dig Liver Dis. 2007;39:725–732. doi: 10.1016/j.dld.2007.05.013. [DOI] [PubMed] [Google Scholar]
- 124.Vogt T, Stolz W, Welsh J, Jung B, Kerbel RS, Kobayashi H, et al. Overexpression of Lerk-5/Eplg5 messenger RNA: a novel marker for increased tumorigenicity and metastatic potential in human malignant melanomas. Clin Cancer Res. 1998;4:791–797. [PubMed] [Google Scholar]
- 125.Hoelzinger DB, Mariani L, Weis J, Woyke T, Berens TJ, McDonough WS, et al. Gene expression profile of glioblastoma multiforme invasive phenotype points to new therapeutic targets. Neoplasia. 2005;7:7–16. doi: 10.1593/neo.04535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Nakada M, Drake KL, Nakada S, Niska JA, Berens ME. Ephrin-B3 ligand promotes glioma invasion through activation of Rac1. Cancer Res. 2006;66:8492–8500. doi: 10.1158/0008-5472.CAN-05-4211. [DOI] [PubMed] [Google Scholar]