51
|
Theocharis S, Klijanienko J, Giaginis C, Alexandrou P, Patsouris E, Sastre-Garau X. Ephrin receptor (Eph) -A1, -A2, -A4 and -A7 expression in mobile tongue squamous cell carcinoma: associations with clinicopathological parameters and patients survival. Pathol Oncol Res 2013; 20:277-84. [PMID: 24022400 DOI: 10.1007/s12253-013-9692-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 08/30/2013] [Indexed: 10/26/2022]
Abstract
Ephrin receptors (Ephs) are frequently overexpressed in a wide variety of human malignant tumors, being associated with tumor growth, invasion, metastasis and angiogenesis. The present study aimed to evaluate the clinical significance of Eph-A1, -A2, -A4 and -A7 protein expression in mobile tongue squamous cell carcinoma (SCC). Eph-A1, -A2, -A4 and -A7 protein expression was assessed immunohistochemically on 37 mobile tongue SCC tissue samples and was analyzed in relation with clinicopathological characteristics, overall and disease-free patients' survival. All the examined mobile tongue SCC cases were found positive for Eph-A1, -A2, -A4 and -A7. Significant associations were noted between high Eph-A1, -A4 and -A7 expression and absence of lymph node metastases (p = 0.0263, p = 0.0461 and p = 0.0461, respectively). High Eph-A1, -A2 and -A7 expression was significantly more frequently observed in patients presenting absence of vascular invasion (p = 0.0444), dense stromal inflammatory reaction (p = 0.0063) and female gender (p = 0.0327), respectively. Mobile tongue SCC patients with high Eph-A7 expression presented longer overall and disease-free survival compared to those with low Eph-A7 expression (log-rank test, p = 0.0093 and p = 0.0164, respectively). In multivariate analysis, Eph-A7 expression was identified as independent prognostic factor of overall survival (Cox-regression analysis, p = 0.0426). The present study supported evidence that Ephs may participate in the malignant transformation of mobile tongue SCC, reinforcing their utility as clinical markers for patients' management and prognosis, as also as targets for potential therapeutic intervention in tongue chemoprevention.
Collapse
|
52
|
Nishimura M, Jung EJ, Shah MY, Lu C, Spizzo R, Shimizu M, Han HD, Ivan C, Rossi S, Zhang X, Nicoloso MS, Wu SY, Almeida MI, Bottsford-Miller J, Pecot CV, Zand B, Matsuo K, Shahzad MM, Jennings NB, Rodriguez-Aguayo C, Lopez-Berestein G, Sood AK, Calin GA. Therapeutic synergy between microRNA and siRNA in ovarian cancer treatment. Cancer Discov 2013; 3:1302-15. [PMID: 24002999 DOI: 10.1158/2159-8290.cd-13-0159] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
UNLABELLED Development of improved RNA interference-based strategies is of utmost clinical importance. Although siRNA-mediated silencing of EphA2, an ovarian cancer oncogene, results in reduction of tumor growth, we present evidence that additional inhibition of EphA2 by a microRNA (miRNA) further "boosts" its antitumor effects. We identified miR-520d-3p as a tumor suppressor upstream of EphA2, whose expression correlated with favorable outcomes in two independent patient cohorts comprising 647 patients. Restoration of miR-520d-3p prominently decreased EphA2 protein levels, and suppressed tumor growth and migration/invasion both in vitro and in vivo. Dual inhibition of EphA2 in vivo using 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) nanoliposomes loaded with miR-520d-3p and EphA2 siRNA showed synergistic antitumor efficiency and greater therapeutic efficacy than either monotherapy alone. This synergy is at least in part due to miR-520d-3p targeting EphB2, another Eph receptor. Our data emphasize the feasibility of combined miRNA-siRNA therapy, and will have broad implications for innovative gene silencing therapies for cancer and other diseases. SIGNIFICANCE This study addresses a new concept of RNA inhibition therapy by combining miRNA and siRNA in nanoliposomal particles to target oncogenic pathways altered in ovarian cancer. Combined targeting of the Eph pathway using EphA2-targeting siRNA and the tumor suppressor miR-520d-3p exhibits remarkable therapeutic synergy and enhanced tumor suppression in vitro and in vivo compared with either monotherapy alone.
Collapse
Affiliation(s)
- Masato Nishimura
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Obstetrics and Gynecology, The University of Tokushima, Graduate School; Japan
| | - Eun-Jung Jung
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Surgery, School of Medicine, Gyeongsang National University, Jin-ju, South Korea
| | - Maitri Y Shah
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Chunhua Lu
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Riccardo Spizzo
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Masayoshi Shimizu
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Hee Dong Han
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Cristina Ivan
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,The Center for RNA Interference and Non-coding RNAs, The University of Texas M.D. Anderson Center, Houston, TX; USA
| | - Simona Rossi
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Batiment Genopode, Lausanne, Switzerland
| | - Xinna Zhang
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,The Center for RNA Interference and Non-coding RNAs, The University of Texas M.D. Anderson Center, Houston, TX; USA
| | - Milena S Nicoloso
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Sherry Y Wu
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Maria Ines Almeida
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Justin Bottsford-Miller
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Chad V Pecot
- Department of Thoracic, Head & Neck Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Behrouz Zand
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Koji Matsuo
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Mian M Shahzad
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Division of Gynecologic Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nicholas B Jennings
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,The Center for RNA Interference and Non-coding RNAs, The University of Texas M.D. Anderson Center, Houston, TX; USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,The Center for RNA Interference and Non-coding RNAs, The University of Texas M.D. Anderson Center, Houston, TX; USA.,Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,The Center for RNA Interference and Non-coding RNAs, The University of Texas M.D. Anderson Center, Houston, TX; USA.,Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,The Center for RNA Interference and Non-coding RNAs, The University of Texas M.D. Anderson Center, Houston, TX; USA
| |
Collapse
|
53
|
Kipps E, Tan DSP, Kaye SB. Meeting the challenge of ascites in ovarian cancer: new avenues for therapy and research. Nat Rev Cancer 2013; 13:273-82. [PMID: 23426401 PMCID: PMC4673904 DOI: 10.1038/nrc3432] [Citation(s) in RCA: 422] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Malignant ascites presents a considerable clinical challenge to the management of ovarian cancer, but also provides a wealth of opportunities for translational research. The accessibility of ascitic fluid and its cellular components make it an excellent source of tumour tissue for the investigation of prognostic and predictive biomarkers, pharmacodynamic markers and for molecular profiling analysis. In this Opinion article, we discuss recent advances in our understanding of its pathophysiology, the development of new methods to characterize its molecular features and how these findings can be used to improve the treatment of malignant ascites, particularly in the context of ovarian cancer.
Collapse
Affiliation(s)
- Emma Kipps
- The Institute of Cancer Research/Royal Marsden Hospital, Medicine, Downs Road, Sutton SM2 5PT, UK
| | | | | |
Collapse
|
54
|
Epigenetic silencing of HOPX promotes cancer progression in colorectal cancer. Neoplasia 2013; 14:559-71. [PMID: 22904674 DOI: 10.1593/neo.12330] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 06/12/2012] [Accepted: 06/14/2012] [Indexed: 11/18/2022]
Abstract
Homeodomain-only protein X (HOPX)-β promoter methylation was recently shown to be frequent in human cancers and was suggested as tumor suppressor gene in esophageal and gastric cancer. The aim of this study was to investigate the mechanistic roles of HOPX-β promoter methylation and its clinical relevance in colorectal cancer (CRC). HOPX-β promoter methylation was assessed in human CRC cell lines and 294 CRC tissues. HOPX mRNA and protein levels were measured in relation to HOPX-β promoter methylation. The effects of forced HOPX expression on tumorigenesis were studied using in vitro and in vivo assays. The association between HOPX-β promoter methylation and clinical relevance of CRC patients was determined. HOPX-β promoter methylation is cancer-specific and frequently found in CRC cell lines and tissues, resulting in the down-regulation of HOPX mRNA and protein levels. In CRC cell lines, forced expression of HOPX suppressed proliferation, invasion, and anchorage-independent growth. DNA microarray analyses suggested critical downstream genes that are associated with cancer cell proliferation, invasion or angiogenesis. In a mouse xenograft model, HOPX inhibited tumorigenesis and angiogenesis. Finally, HOPX-β promoter methylation was associated with worse prognosis of stage III CRC patients (hazard ratio= 1.40, P = .035) and also with poor differentiation (P = .014). In conclusion, HOPX-β promoter methylation is a frequent and cancer-specific event in CRC progression. This epigenetic alteration may have clinical ramifications in the diagnosis and treatment of CRC patients.
Collapse
|
55
|
Shen H, Rodriguez-Aguayo C, Xu R, Gonzalez-Villasana V, Mai J, Huang Y, Zhang G, Guo X, Bai L, Qin G, Deng X, Li Q, Erm DR, Aslan B, Liu X, Sakamoto J, Chavez-Reyes A, Han HD, Sood AK, Ferrari M, Lopez-Berestein G. Enhancing chemotherapy response with sustained EphA2 silencing using multistage vector delivery. Clin Cancer Res 2013; 19:1806-15. [PMID: 23386691 DOI: 10.1158/1078-0432.ccr-12-2764] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE RNA interference has the potential to specifically knockdown the expression of target genes and thereby transform cancer therapy. However, lack of effective delivery of siRNA has dramatically limited its in vivo applications. We have developed a multistage vector (MSV) system, composed of discoidal porous silicon particles loaded with nanotherapeutics, that directs effective delivery and sustained release of siRNA in tumor tissues. In this study, we evaluated therapeutic efficacy of MSV-loaded EphA2 siRNA (MSV/EphA2) with murine orthotopic models of metastatic ovarian cancers as a first step toward development of a new class of nanotherapeutics for the treatment of ovarian cancer. EXPERIMENTAL DESIGN Tumor accumulation of MSV/EphA2 and sustained release of siRNA from MSV were analyzed after intravenous administration of MSV/siRNA. Nude mice with metastatic SKOV3ip2 tumors were treated with MSV/EphA2 and paclitaxel, and therapeutic efficacy was assessed. Mice with chemotherapy-resistant HeyA8 ovarian tumors were treated with a combination of MSV/EphA2 and docetaxel, and enhanced therapeutic efficacy was evaluated. RESULTS Treatment of SKOV3ip2 tumor mice with MSV/EphA2 biweekly for 6 weeks resulted in dose-dependent (5, 10, and 15 μg/mice) reduction of tumor weight (36%, 64%, and 83%) and number of tumor nodules compared with the control groups. In addition, tumor growth was completely inhibited when mice were treated with MSV/EphA2 in combination with paclitaxel. Furthermore, combination treatment with MSV/EphA2 and docetaxel inhibited growth of HeyA8-MDR tumors, which were otherwise resistant to docetaxel treatment. CONCLUSION These findings indicate that MSV/EphA2 merits further development as a novel therapeutic agent for ovarian cancer.
Collapse
Affiliation(s)
- Haifa Shen
- Department of Nanomedicine, The Methodist Hospital Research Institute, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Thanapprapasr D, Hu W, Sood AK, Coleman RL. Moving beyond VEGF for anti-angiogenesis strategies in gynecologic cancer. Curr Pharm Des 2012; 18:2713-9. [PMID: 22390757 DOI: 10.2174/138161212800626201] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 01/06/2012] [Indexed: 02/03/2023]
Abstract
Gynecologic cancer is a major burden in both developed and developing countries. Almost a half million deaths from gynecologic cancer are reported each year. Understanding the molecular biology of cancer is a principle resource leading to the identification of new potential therapeutic targets, which may be parlayed into novel therapeutic options in gynecologic cancer. Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase, which plays a pivotal role in many aspects of malignant growth including cancer cell survival, migration, invasion, angiogenesis and metastasis. Various human cancer tissues have demonstrated high expression of FAK or activated FAK, which has been correlated with survival of cancer patients. Among gynecologic cancers, reports have emerged demonstrating that FAK is involved in the pathogenesis of ovarian, endometrial, and cervical cancers. In addition, the polycomb group protein enhancer of Zeste homologue 2 (EZH2), Dll4/notch and EphA2 has also emerged as important regulators of endothelial cell biology and angiogenesis. Herein, we review the role of these new targets in tumor angiogenesis and the rationale for further clinical development.
Collapse
Affiliation(s)
- Duangmani Thanapprapasr
- Department of Gynecologic Oncology, University of Texas, M.D. Anderson Cancer Center, 1155 Herman Pressler Dr. CPB 6.3271, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
57
|
Abstract
Epithelial ovarian cancer (EOC) remains the most lethal gynecological malignancy despite several decades of progress in diagnosis and treatment. Taking advantage of the robust development of discovery and utility of prognostic biomarkers, clinicians and researchers are developing personalized and targeted treatment strategies. This review encompasses recently discovered biomarkers of ovarian cancer, the utility of published prognostic biomarkers for EOC (especially biomarkers related to angiogenesis and key signaling pathways), and their integration into clinical practice.
Collapse
Affiliation(s)
- Jie Huang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | |
Collapse
|
58
|
Tan P, Liu Y, Yu C, Su Z, Li G, Zhou X, Huang D, Zhang X, Qiu Y, Tian Y. EphA2 silencing in nasopharyngeal carcinoma leads to decreased proliferation, invasion and increased sensitization to paclitaxel. Oncol Lett 2012; 4:429-434. [PMID: 23741245 DOI: 10.3892/ol.2012.746] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 05/22/2012] [Indexed: 11/06/2022] Open
Abstract
EphA2 is frequently overexpressed and functionally altered in a variety of human cancers. However, its roles in human nasopharyngeal carcinoma (NPC) remain unclear. To investigate the roles of EphA2 in the development and progression of NPC, we initially evaluated the expression pattern of EphA2 protein in NPC tissues using western blotting and CCK-8 assay. Fluorescence-activated cell sorting analysis and invasion assay were conducted to observe the effects of EphA2 inhibition in vivo. Our results demonstrated that EphA2 was overexpressed in NPC specimens and the expression of EphA2 was significantly associated with T classification, advanced clinical stage and lymph node metastasis. Moreover, human NPC 5-8F cells were infected with lentiviral vector-mediated EphA2-specific shRNA, which resulted in the significant inhibition of cell growth, invasion of 5-8F cells and markedly enhanced the sensitivity of 5-8F cells to the chemotherapeutic agent paclitaxel in vitro. Collectively, our results demonstrate that EphA2 is involved in malignant cell behavior and is a potential therapeutic target in human NPC.
Collapse
Affiliation(s)
- Pingqing Tan
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008 ; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, Hunan 410008
| | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Correlation of clinicopathological features and expression of molecular markers with prognosis after ¹³¹I treatment of differentiated thyroid carcinoma. Clin Nucl Med 2012; 37:e40-6. [PMID: 22310269 DOI: 10.1097/rlu.0b013e31823905e4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE The aim of this study was to identify molecular markers associated with differentiated thyroid carcinoma (DTC) patients and to determine whether there was a correlation between clinicopathological features or molecular markers and the outcome of radioiodine therapy. PATIENTS AND METHODS We retrospectively reviewed the records of 68 patients with papillary thyroid carcinoma (PTC) and 6 patients with follicular thyroid carcinoma (FTC), who underwent iodine treatment. All patients were divided into a cured group (no evidence for disease) and a noncured group (presence of residual or recurrent disease). Immunohistochemical analysis of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor (HER)-2 (C-erbB-2), ephrin receptor (EphA-2), and sodium-iodide symporter (NIS) was performed in all DTC specimens (68 PTCs, 6 FTCs) and in 14 follicular adenoma specimens. RESULTS Male patients and patients with lymph node and distant metastases had poorer prognosis. Overexpression of ER, PR, HER-2, EphA-2, and NIS was observed in 50.0% (37), 73.0% (54), 52.7% (39), 67.5% (50), and 70.3% (52) of DTC patients, respectively, and was significantly higher than in follicular adenoma patients (0%, 7.14%, 14.29%, 35.7%, and 35.7%, respectively). However, in patients with DTC, no significant difference in the expression of any marker was seen between the cured group and noncured group. CONCLUSIONS Male gender, and lymph node and distant metastases were found to be poor prognostic factors for patients with DTC. Higher expressions of ER, PR, HER-2, EphA-2, and NIS were associated with DTC, but were not correlated with the effectiveness of radioiodine treatment.
Collapse
|
60
|
Liu Y, Yu C, Qiu Y, Huang D, Zhou X, Zhang X, Tian Y. Downregulation of EphA2 expression suppresses the growth and metastasis in squamous-cell carcinoma of the head and neck in vitro and in vivo. J Cancer Res Clin Oncol 2012; 138:195-202. [PMID: 22086185 DOI: 10.1007/s00432-011-1087-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 11/01/2011] [Indexed: 12/28/2022]
Abstract
PURPOSE Our previous study has revealed that EphA2 overexpression is significantly associated with aggressive behavior and poor prognosis in patients with squamous-cell carcinoma of the head and neck (SCCHN). However, the function of EphA2 in tumorigenesis and cervical lymph node metastasis of SCCHN has never been elucidated in vivo. METHODS EphA2 was knocked down in SCCHN cell lines. CCK-8 assays, fluorescence-activated cell sorting analysis, invasion and migration assays were performed in vitro. In vivo tumorigenicity assays were performed, and the impact on cervical lymph node metastasis was evaluated. RESULTS The present investigation demonstrated that suppression of EphA2 resulted in a significant inhibition of proliferation, migration, invasion of SCCHN cells in vitro and markedly diminished their tumorigenicity and lymph node metastasis in vivo. CONCLUSIONS These results suggest that EphA2 plays a critical role in SCCHN growth and metastasis and may be a promising therapeutic target to prevent the progression of SCCHN.
Collapse
Affiliation(s)
- Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, Hunan Province, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
61
|
Brun JL, Cortez A, Lesieur B, Uzan S, Rouzier R, Daraï E. Expression of MMP-2, -7, -9, MT1-MMP and TIMP-1 and -2 has no prognostic relevance in patients with advanced epithelial ovarian cancer. Oncol Rep 2011; 27:1049-57. [PMID: 22200690 PMCID: PMC3583568 DOI: 10.3892/or.2011.1608] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2011] [Accepted: 09/21/2011] [Indexed: 12/24/2022] Open
Abstract
Matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) are involved in tumor invasion, but their prognostic significance is still under discussion. We set out to analyze the epithelial and stromal expression of MMP-2, MMP-7, MMP-9, MT1-MMP, TIMP-1 and TIMP-2 in advanced epithelial ovarian cancers and to assess their prognostic value. A tissue microarray of malignant ovarian tumors from 69 patients was constructed. Immunostaining results were scored using the HSCORE and assessed by univariate analysis with Bonferroni correction and classical multidimensional scaling (CMDS). Kaplan-Meier survival curves calculated with regard to patient and tumor characteristics were compared by the log-rank test. Patients treated by primary surgery (n=43) had a higher tumor size and a trend toward higher epithelial MMP and TIMP expression than those treated by interval surgery (n=26). Optimal cytoreduction (residue ≤ 1 cm) was obtained in 27 and 18 patients, respectively. Clinical and histological characteristics were not different in patients with optimal cytoreduction and those with suboptimal cytoreduction. The expression of epithelial MMP-9 (P=0.002) and TIMP-2 (P=0.026) were higher in the latter group. CMDS failed to demonstrate any influence of MMP and TIMP expression with regard to cytoreduction outcome. MMP and TIMP expression did not influence survival. Their prognostic values were outweighed by histological type, lymph node involvement and cytoreduction. Standard statistical analysis adjusted after Bonferroni correction and CMDS reduced the relevance of MMPs and TIMPs in the prognosis of patients with advanced ovarian cancer.
Collapse
Affiliation(s)
- Jean-Luc Brun
- Department of Obstetrics, Hôpital Tenon, AP-HP, F-75571 Paris, France.
| | | | | | | | | | | |
Collapse
|
62
|
Hou F, Yuan W, Huang J, Qian L, Chen Z, Ge J, Wu S, Chen J, Wang J, Chen Z. Overexpression of EphA2 correlates with epithelial-mesenchymal transition-related proteins in gastric cancer and their prognostic importance for postoperative patients. Med Oncol 2011; 29:2691-700. [PMID: 22189617 DOI: 10.1007/s12032-011-0127-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Accepted: 11/24/2011] [Indexed: 12/19/2022]
Abstract
The expression of EphA2 and three epithelial-mesenchymal transition-related proteins (E-cadherin, β-catenin and vimentin) was detected by immunohistochemistry in human gastric cancer and normal gastric mucosa. The expression of EphA2 and vimentin was significantly higher in gastric cancer tissues than in normal gastric mucosa tissues, and similar results were found for negative E-cadherin expression and ectopic β-catenin expression. Further analysis showed that the expression of EphA2 was closely correlated with the depth of tumor invasion, tumor-node-metastasis (TNM) stages and lymph node metastasis. Down-regulated expression of the epithelial protein E-cadherin, overexpression of the mesenchymal protein vimentin and ectopic expression of β-catenin were associated with the depth of tumor invasion, tumor differentiation, TNM stages and lymph node metastasis. The Spearman rank test indicated that the positive expression of EphA2 was negatively associated with E-cadherin expression and was positively correlated with β-catenin ectopic expression and vimentin expression. In addition, the Kaplan-Meier survival analysis showed that the overexpression of EphA2 and vimentin, ectopic expression of β-catenin and down-regulation of E-cadherin indicate a poor outcome. Moreover, multivariate Cox analysis showed that TNM stages, lymph node metastasis, EphA2 expression, E-cadherin expression and β-catenin ectopic expression were independent prognostic factors for postoperative gastric cancer. These findings indicate that the overexpression of EphA2 correlates with the loss of epithelial proteins and the appearance of mesenchymal proteins. Therefore, EphA2 may play a role in epithelial-mesenchymal transition in gastric cancer.
Collapse
Affiliation(s)
- Futao Hou
- Department of General Surgery, Xiangya Hospital, Central South University, Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Brantley-Sieders DM. Clinical relevance of Ephs and ephrins in cancer: lessons from breast, colorectal, and lung cancer profiling. Semin Cell Dev Biol 2011; 23:102-8. [PMID: 22040912 DOI: 10.1016/j.semcdb.2011.10.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 10/17/2011] [Indexed: 01/04/2023]
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.
Collapse
Affiliation(s)
- Dana M Brantley-Sieders
- Department of Medicine, Vanderbilt University School of Medicine, A-4323 MCN, 1161 21st Avenue South, Nashville, TN 37232-2363, USA.
| |
Collapse
|
64
|
Hu W, Lu C, Dong HH, Huang J, Shen DY, Stone RL, Nick AM, Shahzad MMK, Mora E, Jennings NB, Lee SJ, Roh JW, Matsuo K, Nishimura M, Goodman BW, Jaffe RB, Langley RR, Deavers MT, Lopez-Berestein G, Coleman RL, Sood AK. Biological roles of the Delta family Notch ligand Dll4 in tumor and endothelial cells in ovarian cancer. Cancer Res 2011; 71:6030-9. [PMID: 21795478 DOI: 10.1158/0008-5472.can-10-2719] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Emerging evidence suggests that the Notch/Delta-like ligand 4 (Dll4) pathway may offer important new targets for antiangiogenesis approaches. In this study, we investigated the clinical and biological significance of Dll4 in ovarian cancer. Dll4 was overexpressed in 72% of tumors examined in which it was an independent predictor of poor survival. Patients with tumors responding to anti-VEGF therapy had lower levels of Dll4 than patients with stable or progressive disease. Under hypoxic conditions, VEGF increased Dll4 expression in the tumor vasculature. Immobilized Dll4 also downregulated VEGFR2 expression in endothelial cells directly through methylation of the VEGFR2 promoter. RNAi-mediated silencing of Dll4 in ovarian tumor cells and tumor-associated endothelial cells inhibited cell growth and angiogenesis, accompanied by induction of hypoxia in the tumor microenvironment. Combining Dll4-targeted siRNA with bevacizumab resulted in greater inhibition of tumor growth, compared with control or treatment with bevacizumab alone. Together, our findings establish that Dll4 plays a functionally important role in both the tumor and endothelial compartments of ovarian cancer and that targeting Dll4 in combination with anti-VEGF treatment might improve outcomes of ovarian cancer treatment.
Collapse
Affiliation(s)
- Wei Hu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Tinelli A, Vergara D, Martignago R, Leo G, Malvasi A, Tinelli R, Marsigliante S, Maffia M, Lorusso V. Ovarian cancer biomarkers: a focus on genomic and proteomic findings. Curr Genomics 2011; 8:335-42. [PMID: 19384429 PMCID: PMC2652404 DOI: 10.2174/138920207782446142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 09/20/2007] [Accepted: 09/26/2007] [Indexed: 12/19/2022] Open
Abstract
Among the gynaecological malignancies, ovarian cancer is one of the neoplastic forms with the poorest prognosis and with the bad overall and disease-free survival rates than other gynaecological cancers; several studies, analyzing clinical data and pathological features on ovarian cancers, have focused on the identification of both diagnostic and prognostic markers for applications in clinical practice. High-throughput technologies have accelerated the process of biomarker discovery, but their validity should be still demonstrated by extensive researches on sensibility and sensitivity of ovarian cancer novel biomarkers, determining whether gene profiling and proteomics could help differentiate between patients with metastatic ovarian cancer and primary ovarian carcinomas, and their potential impact on management. Therefore, considerable interest lies in identifying molecular prognostic biomarkers and protein indicators to guide treatment decisions and clinical follow up; the current state of knowledge about the potential clinical value of gene expression profiling in ovarian cancer is discussed, focusing on three main areas: distinguishing normal ovarian tissue from ovarian tumors, identifying different subtypes of ovarian cancer and identifying cancer likely to be responsive to therapy. In this elaborate we discuss the use of novel molecules, discovered by proteomics and genomics approaches, as potential protein biomarkers in the management of ovarian cancer, to improve the anticancer therapy for malignant ovarian tumors and to monitor the clinical follow up.
Collapse
Affiliation(s)
- Andrea Tinelli
- Department of Obstetrics and Gynecology, "Vito Fazzi" Hospital, Lecce, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Tinelli A, Vergara D, Martignago R, Leo G, Pisanò M, Malvasi A. An outlook on ovarian cancer and borderline ovarian tumors: focus on genomic and proteomic findings. Curr Genomics 2011; 10:240-9. [PMID: 19949545 PMCID: PMC2709935 DOI: 10.2174/138920209788488553] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Revised: 03/16/2009] [Accepted: 03/18/2009] [Indexed: 01/16/2023] Open
Abstract
Among the gynaecological malignancies, ovarian cancer is one of the neoplastic forms with the poorest prognosis and with the bad overall and disease-free survival rates than other gynaecological cancers. Ovarian tumors can be classified on the basis of the cells of origin in epithelial, stromal and germ cell tumors. Epithelial ovarian tumors display great histological heterogeneity and can be further subdivided into benign, intermediate or borderline, and invasive tumors. Several studies on ovarian tumors, have focused on the identification of both diagnostic and prognostic markers for applications in clinical practice. High-throughput technologies have accelerated the process of biomolecular study and genomic discovery; unfortunately, validity of these should be still demonstrated by extensive researches on sensibility and sensitivity of ovarian cancer novel biomarkers, determining whether gene profiling and proteomics could help differentiate between patients with metastatic ovarian cancer and primary ovarian carcinomas, and their potential impact on management. Therefore, considerable interest lies in identifying molecular and protein biomarkers and indicators to guide treatment decisions and clinical follow up. In this review, the current state of knowledge about the genoproteomic and potential clinical value of gene expression profiling in ovarian cancer and ovarian borderline tumors is discussed, focusing on three main areas: distinguishing normal ovarian tissue from ovarian cancers and borderline tumors, identifying different genotypes of ovarian tissue and identifying proteins linked to cancer or tumor development. By these targets, authors focus on the use of novel molecules, developed on the proteomics and genomics researches, as potential protein biomarkers in the management of ovarian cancer or borderline tumor, overlooking on current state of the art and on future perspectives of researches.
Collapse
Affiliation(s)
- Andrea Tinelli
- Department of Obstetrics and Gynaecology, Vito Fazzi Hospital, Lecce, Italy
| | | | | | | | | | | |
Collapse
|
67
|
Abstract
IMPORTANCE OF THE FIELD High mortality rates with cancers warrant further development of earlier diagnostics and better treatment strategies. Membrane-bound erythropoietin-producing hepatocellular receptor tyrosine kinase class A2 (EphA2) is overexpressed in breast, prostate, urinary bladder, skin, lung, ovary and brain cancers. AREAS COVERED IN THIS REVIEW EphA2 overexpression in cancers, its signaling mechanisms and strategies to target its deregulation. WHAT THE READER WILL GAIN High EphA2 expression in cancer cells is correlated with a poor prognosis associated with recurrence due to enhanced metastasis. Interaction of the EphA2 receptor with its ligand (e.g., ephrinA1) triggers events that are deregulated and implicated in carcinogenesis. EphrinA1-independent oncogenic activity and ephrinA1-dependent tumor suppressor roles for EphA2 are described. Molecular interactions of EphA2 with signaling proteins are associated with the modulation of cytoskeleton dynamics, cell adhesion, proliferation, differentiation and metastasis. The deregulated signaling by EphA2 and its involvement in oncogenesis provide multiple avenues for the rational design of intervention approaches. TAKE HOME MESSAGE EphA2 has been tested as a drug target using multiple approaches such as agonist antibodies, RNA interference, immunotherapy, virus vector-mediated gene transfer, small-molecule inhibitors and nanoparticles. With over a decade of research, encouraging results with targeting of EphA2 expression in various pre-clinical cancer models necessitate further studies.
Collapse
Affiliation(s)
- Manish Tandon
- Department of Comparative Pathobiology, School of Veterinary Medicine, and Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Sai Vikram Vemula
- Department of Comparative Pathobiology, School of Veterinary Medicine, and Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, School of Veterinary Medicine, and Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
68
|
Abstract
Background Angiogenesis is a critical component of tumor development and proliferation, and increased angiogenesis has been associated with a worse clinical outcome in a number of solid tumors, including ovarian cancer. Therefore, agents that target the angiogenic process are of considerable interest in the treatment of ovarian cancer. Methods Studies evaluating the efficacy of antiangiogenic agents in ovarian cancer are reported. Antiangiogenic agents examined include vascular endothelial growth factor (VEGF) pathway inhibitors, including monoclonal antibodies, tyrosine kinase inhibitors (TKIs), and a soluble receptor decoy, as well as inhibitors of other angiogenic factors and vascular disrupting agents. Results The VEGF inhibitor bevacizumab has been shown to have efficacy in ovarian cancer in phase II trials and a progression-free survival advantage in one phase III trial. TKIs block the VEGF receptors and secondary angiogenic pathways and have shown activity in phase I and II trials. Alternative angiogenesis inhibitors include EphA2 inhibitors and a selective angiopoietin 1/2-neutralizing peptibody. Another strategy is to destroy the existing tumor vasculature, and a number of vascular disrupting agents are being studied in preclinical and phase I trials. Antiangiogenic agents have a unique side effect profile, likely due to inhibition of normal physiologic angiogenesis. Conclusions Phase II and early phase III trials have demonstrated that antiangiogenic therapies have significant activity in ovarian cancer. The results of phase III trials in the front-line and recurrent settings will determine the extent of clinical benefit of antiangiogenic therapies in combination with chemotherapy. Antiangiogenic agents have a distinct side effect profile, and further studies are necessary to evaluate how to minimize the incidence of these events and to identify women most likely to benefit from these therapies.
Collapse
Affiliation(s)
- Deanna G. K. Teoh
- Division of Gynecologic Oncology at Duke Comprehensive Cancer Center, Durham, North Carolina
| | - Angeles Alvarez Secord
- Division of Gynecologic Oncology at Duke Comprehensive Cancer Center, Durham, North Carolina
| |
Collapse
|
69
|
Merritt WM, Kamat AA, Hwang JY, Bottsford-Miller J, Lu C, Lin YG, Coffey D, Spannuth W, Nugent E, Han LY, Landen CN, Nick AM, Stone RL, Coffman KT, Bruckheimer E, Broaddus R, Gershenson DM, Coleman RL, Sood AK. Clinical and biological impact of EphA2 overexpression and angiogenesis in endometrial cancer. Cancer Biol Ther 2010; 10:1306-14. [PMID: 20948320 PMCID: PMC3047089 DOI: 10.4161/cbt.10.12.13582] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE EphA2 overexpression predicts poor prognosis in endometrial cancer. To explore mechanisms for this association and assess its potential as therapeutic target, the relationship of EphA2 expression to markers of angiogenesis was examined using patient samples and an orthotopic mouse model of uterine cancer. EXPERIMENTAL DESIGN Expression of EphA2, estrogen receptor (ER), progesterone receptor (PR), Ki-67, vascular endothelial growth factor (VEGF) and microvessel density (MVD) was evaluated using immunohistochemistry in 85 endometrioid endometrial adenocarcinomas (EEC) by two independent investigators. Results were correlated with clinicopathological characteristics. The effect of EphA2- agonist monoclonal antibody EA5, alone or in combination with docetaxel was studied in vitro and in vivo. Samples were analyzed for markers of angiogenesis, proliferation and apoptosis. RESULTS Of 85 EEC samples, EphA2 was overexpressed in 47% of tumors and was significantly associated with high VEGF expression (p=0.001) and high MVD counts (p=0.02). High EphA2 expression, high VEGF expression and high MVD counts were significantly associated with shorter disease-specific survival. EA5 led to decrease in EphA2 expression and phosphorylation in vitro. In the murine model, while EA5 (33-88%) and docetaxel (23-55%) individually led to tumor inhibition over controls, combination therapy had the greatest efficacy (78-92%, p.
Collapse
Affiliation(s)
- William M Merritt
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Aparna A Kamat
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Jee-Young Hwang
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA,Division of Gynecologic Oncology; Department of Obstetrics and Gynecology; Dongguk University College of Medicine; Kyung-ju, Korea
| | - Justin Bottsford-Miller
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Chunhua Lu
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Yvonne G Lin
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | | | - Whitney Spannuth
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Elizabeth Nugent
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Liz Y Han
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Charles N Landen
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Alpa M Nick
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Rebecca L Stone
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | | | | | - Russell Broaddus
- Department of Pathology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - David M Gershenson
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Robert L Coleman
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| | - Anil K Sood
- Department of Gynecologic Oncology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA,Department of Cancer Biology; University of Texas M.D. Anderson Cancer Center; Houston, TX USA,Center for RNA Interference and Non-Coding RNA; University of Texas M.D. Anderson Cancer Center; Houston, TX USA
| |
Collapse
|
70
|
Faoro L, Singleton PA, Cervantes GM, Lennon FE, Choong NW, Kanteti R, Ferguson BD, Husain AN, Tretiakova MS, Ramnath N, Vokes EE, Salgia R. 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; 285:18575-85. [PMID: 20360610 PMCID: PMC2881783 DOI: 10.1074/jbc.m109.075085] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 03/12/2010] [Indexed: 11/06/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has a poor prognosis and improved therapies are needed. Expression of EphA2 is increased in NSCLC metastases. In this study, we investigated EphA2 mutations in NSCLC and examined molecular pathways involved in NSCLC. Tumor and cell line DNA was sequenced. One EphA2 mutation was modeled by expression in BEAS2B cells, and functional and biochemical studies were conducted. A G391R mutation was detected in H2170 and 2/28 squamous cell carcinoma patient samples. EphA2 G391R caused constitutive activation of EphA2 with increased phosphorylation of Src, cortactin, and p130(Cas). Wild-type (WT) and G391R cells had 20 and 40% increased invasiveness; this was attenuated with knockdown of Src, cortactin, or p130(Cas). WT and G391R cells demonstrated a 70% increase in focal adhesion area. Mammalian target of rapamycin (mTOR) phosphorylation was increased in G391R cells with increased survival (55%) compared with WT (30%) and had increased sensitivity to rapamycin. A recurrent EphA2 mutation is present in lung squamous cell carcinoma and increases tumor invasion and survival through activation of focal adhesions and actin cytoskeletal regulatory proteins as well as mTOR. Further study of EphA2 as a therapeutic target is warranted.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Benjamin D. Ferguson
- From the Sections of Hematology and Oncology and
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois 60637, and
| | | | | | - Nithya Ramnath
- the Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Ravi Salgia
- From the Sections of Hematology and Oncology and
| |
Collapse
|
71
|
Tanaka T, Mangala LS, Vivas-Mejia PE, Nieves-Alicea R, Mann AP, Mora E, Han HD, Shahzad MMK, Liu X, Bhavane R, Gu J, Fakhoury JR, Chiappini C, Lu C, Matsuo K, Godin B, Stone RL, Nick AM, Lopez-Berestein G, Sood AK, Ferrari M. Sustained small interfering RNA delivery by mesoporous silicon particles. Cancer Res 2010; 70:3687-96. [PMID: 20430760 DOI: 10.1158/0008-5472.can-09-3931] [Citation(s) in RCA: 226] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
RNA interference (RNAi) is a powerful approach for silencing genes associated with a variety of pathologic conditions; however, in vivo RNAi delivery has remained a major challenge due to lack of safe, efficient, and sustained systemic delivery. Here, we report on a novel approach to overcome these limitations using a multistage vector composed of mesoporous silicon particles (stage 1 microparticles, S1MP) loaded with neutral nanoliposomes (dioleoyl phosphatidylcholine, DOPC) containing small interfering RNA (siRNA) targeted against the EphA2 oncoprotein, which is overexpressed in most cancers, including ovarian. Our delivery methods resulted in sustained EphA2 gene silencing for at least 3 weeks in two independent orthotopic mouse models of ovarian cancer following a single i.v. administration of S1MP loaded with EphA2-siRNA-DOPC. Furthermore, a single administration of S1MP loaded with-EphA2-siRNA-DOPC substantially reduced tumor burden, angiogenesis, and cell proliferation compared with a noncoding control siRNA alone (SKOV3ip1, 54%; HeyA8, 57%), with no significant changes in serum chemistries or in proinflammatory cytokines. In summary, we have provided the first in vivo therapeutic validation of a novel, multistage siRNA delivery system for sustained gene silencing with broad applicability to pathologies beyond ovarian neoplasms.
Collapse
Affiliation(s)
- Takemi Tanaka
- Department of Nanomedicine and Biomedical Engineering, University of Texas Health Science Center at Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Seiradake E, Harlos K, Sutton G, Aricescu AR, Jones EY. An extracellular steric seeding mechanism for Eph-ephrin signaling platform assembly. Nat Struct Mol Biol 2010; 17:398-402. [PMID: 20228801 PMCID: PMC3672960 DOI: 10.1038/nsmb.1782] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 01/29/2010] [Indexed: 01/27/2023]
Abstract
Erythropoetin-producing hepatoma (Eph) receptors are cell-surface protein tyrosine kinases mediating cell-cell communication. Upon activation, they form signaling clusters. We report crystal structures of the full ectodomain of human EphA2 (eEphA2) both alone and in complex with the receptor-binding domain of the ligand ephrinA5 (ephrinA5 RBD). Unliganded eEphA2 forms linear arrays of staggered parallel receptors involving two patches of residues conserved across A-class Ephs. eEphA2-ephrinA5 RBD forms a more elaborate assembly, whose interfaces include the same conserved regions on eEphA2, but rearranged to accommodate ephrinA5 RBD. Cell-surface expression of mutant EphA2s showed that these interfaces are critical for localization at cell-cell contacts and activation-dependent degradation. Our results suggest a 'nucleation' mechanism whereby a limited number of ligand-receptor interactions 'seed' an arrangement of receptors which can propagate into extended signaling arrays.
Collapse
Affiliation(s)
- Elena Seiradake
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
73
|
Yuan W, Chen Z, Wu S, Ge J, Chang S, Wang X, Chen J, Chen Z. Expression of EphA2 and E-cadherin in gastric cancer: correlated with tumor progression and lymphogenous metastasis. Pathol Oncol Res 2010; 15:473-8. [PMID: 19048396 DOI: 10.1007/s12253-008-9132-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Accepted: 11/14/2008] [Indexed: 12/11/2022]
Abstract
In this study, gastric cancer progression was correlated with the over-expression of erythropoietin-producing hepatocellular (Eph)A2 receptor and down-expression of epithelial cadherin (E-cadherin). Immunohistochemistry of EphA2 and E-cadherin were performed on these tumor samples from 165 primary lesions of gastric cancer. The results showed that expression of EphA2 was obviously increased in gastric cancer tissues (P < 0.01), which was positively correlated with the depth of cancer invasion, tumor-node-metastasis (TNM) stage and lymph node metastasis (P < 0.05). Meanwhile, the expression of E-cadherin was significantly reduced (P < 0.01), which was negatively correlated with the depth of cancer invasion, grade of tumor differentiation, TNM stage and lymph node metastasis (P < 0.05). The correlation between EphA2 and E-cadherin expression was negative (r = -0.198, P = 0.011). In conclusion, either the over-expression of EphA2 or the down-expression of E-cadherin is correlated with cancer progression and lymphogenous metastasis in gastric cancer, suggesting that both of them may play an important role in tumor progression and metastasis.
Collapse
Affiliation(s)
- Weijie Yuan
- Department of General Surgery, Xiangya Hospital, Central South University, Xiangya Road, Changsha 410008, Hunan Province, Peoples Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Yang P, Yuan W, He J, Wang J, Yu L, Jin X, Hu Y, Liao M, Chen Z, Zhang Y. Overexpression of EphA2, MMP-9, and MVD-CD34 in hepatocellular carcinoma: Implications for tumor progression and prognosis. Hepatol Res 2009; 39:1169-77. [PMID: 19788698 DOI: 10.1111/j.1872-034x.2009.00563.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM To investigate the expression of erythropoietin-producing hepatocellular (Eph)A2 receptor, matrix metalloproteinase (MMP)-9, and angiogenesis in hepatocellular carcinoma (HCC), in order to reveal their expression correlations with tumor invasion, metastasis, and prognosis. METHODS From January 2000 to June 2003, 129 specimens of resected tumors from the patients with HCC were obtained. Corresponding pericarcinomatous liver tissues were also obtained and selected as a control group. Expressions of EphA2, MMP-9, and CD34 were detected with immunohistochemical staining. Microvascular density (MVD) was calculated with counting of CD34-positive vascular endothelial cells. RESULTS The expressions of EphA2, MMP-9, and MVD in the HCC tissues were significantly higher than those in the pericarcinomatous liver tissues (P < 0.01). Statistical analysis showed there were significant correlations between the expressions of EphA2, MMP-9 and MVD in some classicclinicopathological parameters (i.e. tumor nodule, vein invasion, tumor, node, metastasis stages, extrahepatic metastasis; P < 0.05). The correlation between EphA2 and MMP-9 expression was positive (r = 0.625, P = 0.011). Tumor MVD was closely associated with EphA2 (r = 0.281, P = 0.01) and MMP-9 (r = 0.319, P < 0.01) expressions. In particular, EphA2, MMP-9, and MVD expressions levels were found to be independent prognostic factors after HCC resection. CONCLUSIONS Overexpressions of EphA2 and MMP-9 relate to tumor progression, metastasis, and prognosis in HCC. The present study suggests that EphA2 is associated with key mediators of angiogenesis and invasion.
Collapse
Affiliation(s)
- Pu Yang
- Hepatobiliary & Enteric Surgery Research Center, Xiangya Hospital, Central South University, Xiangya Road, Changsha 410008, Hunan Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Clinical Significance of Ephrin (Eph)-A1, -A2, -A4, -A5 and -A7 Receptors in Pancreatic Ductal Adenocarcinoma. Pathol Oncol Res 2009; 16:267-76. [DOI: 10.1007/s12253-009-9221-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 10/20/2009] [Indexed: 10/20/2022]
|
76
|
Yuan WJ, Ge J, Chen ZK, Wu SB, Shen H, Yang P, Hu B, Zhang GW, Chen ZH. Over-expression of EphA2 and EphrinA-1 in human gastric adenocarcinoma and its prognostic value for postoperative patients. Dig Dis Sci 2009; 54:2410-7. [PMID: 19101799 DOI: 10.1007/s10620-008-0649-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Accepted: 11/21/2008] [Indexed: 12/29/2022]
Abstract
This study aims to investigate the expression and significance of EphA2 and EphrinA-1 in human gastric adenocarcinoma progression and prognosis. The expression of EphA2 and EphrinA-1 was detected in the cell lines and tissues of gastric adenocarcinoma. Different expression levels of EphA2 and EphrinA-1 were found in two cell lines. The expression of EphA2 and EphrinA-1 was significantly higher in gastric adenocarcinoma tissues than in normal tissues. Statistical analysis showed a significant correlation of EphA2 expression with the depth of tumor invasion, tumor-node-metastasis (TNM) stages, and lymph node metastasis. EphrinA-1 over-expression was significantly correlated with TNM stages and lymph node metastasis, while EphA2 expression was found to be an independent prognostic factor of postoperative gastric adenocarcinoma. In conclusion, the increased expression of EphA2 and EphrinA-1 plays an important role in the progression of human gastric adenocarcinoma, in which elevated EphA2 expression is an independent factor that indicates poor prognosis in postoperative gastric adenocarcinoma.
Collapse
Affiliation(s)
- Wei-Jie Yuan
- Department of General Surgery, Xiangya Hospital, Central South University, Xiangya Road, Changsha, 410008 Hunan Province, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Lee JW, Han HD, Shahzad MMK, Kim SW, Mangala LS, Nick AM, Lu C, Langley RR, Schmandt R, Kim HS, Mao S, Gooya J, Fazenbaker C, Jackson D, Tice DA, Landen CN, Coleman RL, Sood AK. EphA2 immunoconjugate as molecularly targeted chemotherapy for ovarian carcinoma. J Natl Cancer Inst 2009; 101:1193-205. [PMID: 19641174 PMCID: PMC2736292 DOI: 10.1093/jnci/djp231] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 06/03/2009] [Accepted: 06/23/2009] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND EphA2 is overexpressed in many types of human cancer but is absent or expressed at low levels in normal epithelial tissues. We investigated whether a novel immunoconjugate containing an anti-EphA2 monoclonal antibody (1C1) linked to a chemotherapeutic agent (monomethyl auristatin phenylalanine [MMAF]) through a noncleavable linker maleimidocaproyl (mc) had antitumor activity against ovarian cancer cell lines and tumor models. METHODS Specificity of 1C1-mcMMAF was examined in EphA2-positive HeyA8 and EphA2-negative SKMel28 ovarian cancer cells by antibody binding and internalization assays. Controls were phosphate-buffered saline (PBS), 1C1, or control IgG-mcMMAF. Viability and apoptosis were investigated in ovarian cancer cell lines and tumor models (10 mice per group). Antitumor activities were tested in the HeyA8-luc and SKOV3ip1 orthotopic mouse models of ovarian cancer. Endothelial cells were identified by use of immunohistochemistry and anti-CD31 antibodies. All statistical tests were two-sided. RESULTS The 1C1-mcMMAF immunoconjugate specifically bound to EphA2-positive HeyA8 cells but not to EphA2-negative cells and was internalized by HeyA8 cells. Treatment with 1C1-mcMMAF decreased the viability of HeyA8-luc cells in an EphA2-specific manner. In orthotopic mouse models, treatment with 1C1-mcMMAF inhibited tumor growth by 85%-98% compared with that in control mice (eg, for weight of HeyA8 tumors, 1C1-mcMMAF = 0.05 g and control = 1.03 g; difference = 0.98 g, 95% confidence interval [CI] = 0.40 to 1.58 g; P = .001). Even in bulkier disease models with HeyA8-luc cells, 1C1-mcMMAF treatment, compared with control treatment, caused regression of established tumors and increased survival of the mice (eg, 1C1-mcMMAF vs control, mean = 60.6 days vs 29.4 days; difference = 31.2 days, 95% CI = 27.6 to 31.2 days; P = .001). The antitumor effects of 1C1-mcMMAF therapy, in SKOV3ip1 tumors, for example, were statistically significantly related to decreased proliferation (eg, 1C1-mcMMAF vs control, mean = 44.1% vs 55.8% proliferating cells; difference = 11.7%, 95% CI = 2.45% to 20.9%; P = .01) and increased apoptosis of tumor cells (eg, 1C1-mcMMAF vs control, mean = 8.6% vs 0.9% apoptotic cells; difference = 7.7%, 95% CI = 3.8% to 11.7%; P < .001) and of mouse endothelial cells (eg, 1C1-mcMMAF vs control, mean 2.8% vs 0.4% apoptotic endothelial cells; difference = 2.4%, 95% CI = 1.4% to 4.6%; P = .034). CONCLUSION The 1C1-mcMMAF immunoconjugate had antitumor activity in preclinical models of ovarian carcinoma.
Collapse
Affiliation(s)
- Jeong-Won Lee
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Hu X, Macdonald DM, Huettner PC, Feng Z, El Naqa IM, Schwarz JK, Mutch DG, Grigsby PW, Powell SN, Wang X. A miR-200 microRNA cluster as prognostic marker in advanced ovarian cancer. Gynecol Oncol 2009; 114:457-64. [PMID: 19501389 DOI: 10.1016/j.ygyno.2009.05.022] [Citation(s) in RCA: 227] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Revised: 05/05/2009] [Accepted: 05/09/2009] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Ovarian cancer is one of the most deadly human cancers, resulting in over 15,000 deaths in the US each year. A reliable method that could predict disease outcome would improve care of patients with this disease. The main aim of this study is to identify novel prognostic biomarkers for advanced ovarian cancer. METHODS We hypothesized that microRNAs (miRNAs) may predict outcome and have examined the prognostic value of these small RNA molecules on disease outcome prediction. miRNAs are a newly identified family of non-coding RNA genes, and recent studies have shown that miRNAs are extensively involved in the tumor development process. We have profiled the expression of miRNAs in advanced ovarian cancer using a novel PCR-based platform and correlated miRNA expression profiles with disease outcome. RESULTS By performing miRNA expression profiling analysis of 55 advanced ovarian tumors, we have shown that three miR-200 miRNAs (miR-200a, miR-200b and miR-429) in the miR-200b-429 cluster are significantly associated with cancer recurrence and overall survival. Further target analysis indicates that these miR-200 miRNAs target multiple genes that are involved in cancer development. In addition, we have also shown that overexpression of this miR-200 cluster inhibits ovarian cancer cell migration. CONCLUSIONS miR-200b-429 may be used as a prognostic marker for ovarian cancer outcome, and low-level expression of miR-200 miRNAs in this cluster predicts poor survival. In addition, our study suggests that miR-200 miRNAs could play an important regulatory role in ovarian cancer.
Collapse
Affiliation(s)
- Xiaoxia Hu
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Kamat AA, Coffey D, Merritt WM, Nugent E, Urbauer D, Lin YG, Edwards C, Broaddus R, Coleman RL, Sood AK. EphA2 overexpression is associated with lack of hormone receptor expression and poor outcome in endometrial cancer. Cancer 2009; 115:2684-92. [PMID: 19396818 PMCID: PMC3139331 DOI: 10.1002/cncr.24335] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND EphA2 is a tyrosine kinase receptor in the ephrin family that is implicated in oncogenesis and angiogenesis. The objective of the current investigation was to study the role of EphA2 in endometrial cancer and its relation to steroid hormone receptor expression. METHODS EphA2, estrogen receptor (ER), progesterone receptor (PR), and Ki-67 expression levels were evaluated using immunohistochemistry in 139 endometrioid endometrial carcinoma (EEC) samples and in 10 benign endometrial samples. Samples were scored by 2 investigators who were blinded to clinical outcome. The results were correlated with clinicopathologic characteristics using univariate and multivariate analysis. A P value <.05 was considered statistically significant. RESULTS High expression of EphA2 was detected in 48% of EEC samples versus 10% of benign samples. EphA2 overexpression was associated significantly with high disease stage (P = .04), high tumor grade (P = .003), increased depth of myometrial invasion (P = .05), low ER expression (P = .01), low PR expression (P = .006), and high Ki-67 expression (P = .04). Low ER and PR expression levels were associated with high tumor grade, positive lymph nodes, high Ki-67 expression, and high EphA2 expression. On univariate analysis of all patients, high EphA2 expression was associated significantly with shorter disease-specific survival (DSS) (P < .001). On multivariate analysis, age (P < .001), high disease stage (P = .002), and high EphA2 expression (P = .04) were independent predictors of poor DSS. CONCLUSIONS EphA2 overexpression was associated with aggressive phenotypic features in EEC and was associated inversely with ER and PR expression. Thus, EphA2 may be an important therapeutic target, especially in patients with hormone receptor-negative endometrial carcinoma.
Collapse
Affiliation(s)
- Aparna A. Kamat
- Department of Gynecologic Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | | | - William M. Merritt
- Department of Gynecologic Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Elizabeth Nugent
- Department of Gynecologic Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Diana Urbauer
- Department of Biostatistics, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Yvonne G. Lin
- Department of Gynecologic Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | | | - Russell Broaddus
- Department of Pathology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Robert L. Coleman
- Department of Gynecologic Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Anil K. Sood
- Department of Gynecologic Oncology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Department of Cancer Biology, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
80
|
Abstract
Over the past two decades, the 5-year survival for ovarian cancer patients has substantially improved owing to more effective surgery and treatment with empirically optimized combinations of cytotoxic drugs, but the overall cure rate remains approximately 30%. Many investigators think that further empirical trials using combinations of conventional agents are likely to produce only modest incremental improvements in outcome. Given the heterogeneity of this disease, increases in long-term survival might be achieved by translating recent insights at the molecular and cellular levels to personalize individual strategies for treatment and to optimize early detection.
Collapse
Affiliation(s)
- Robert C Bast
- Departments of Experimental Therapeutics and Systems Biology, University of Texas M. D. Anderson Cancer Center, 1515 Holcolmbe Boulevard, Houston, TX 77030, USA.
| | | | | |
Collapse
|
81
|
Blackburn WH, Dickerson EB, Smith MH, McDonald JF, Lyon LA. Peptide-functionalized nanogels for targeted siRNA delivery. Bioconjug Chem 2009; 20:960-8. [PMID: 19341276 PMCID: PMC2765502 DOI: 10.1021/bc800547c] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A major bottleneck in the development of siRNA therapies is their delivery to the desired cell type or tissue, followed by effective passage across the cell membrane with subsequent silencing of the targeted mRNA. To address this problem, we describe the synthesis of core/shell hydrogel nanoparticles (nanogels) with surface-localized peptides that specifically target ovarian carcinoma cell lines possessing high expression levels of the Eph2A receptor. These nanogels are also demonstrated to be highly effective in the noncovalent encapsulation of siRNA and enable cell-specific delivery of the oligonucleotides in serum-containing medium. Cell toxicity and viability assays reveal that the nanogel construct is nontoxic under the conditions studied, as no toxicity or decrease in cell proliferation is observed following delivery. Importantly, a preliminary investigation of gene silencing illustrates that nanogel-mediated delivery of siRNA targeted to the EGF receptor results in knockdown of that receptor. Excellent protection of siRNA during endosomal uptake and endosomal escape of the nanogels is suggested by these results since siRNA activity in the cytosol is required for gene silencing.
Collapse
|
82
|
Abstract
Ovarian cancer is a leading cause of gynecologic cancer death among women. Tumors diagnosed early (in stage I) have a cure rate approaching 90%. However, because specific symptoms and screening tools are lacking, most ovarian cancers are very advanced when finally diagnosed. CA125 expression and pelvic ultrasonography are of limited efficacy in screening, and the search for new, complementary ovarian cancer biomarkers continues. New technology and research techniques have allowed the identification of over 100 possible tumor markers, many of which are still being evaluated for clinical relevance and several of which have entered clinical trials. Here, we review the methods of biomarker discovery, address the significance and functions of newly identified ovarian cancer tumor markers, and provide further insight into the future of ovarian cancer biomarkers.
Collapse
Affiliation(s)
- Celestine S Tung
- University of Texas, MD Anderson Cancer Center, Department of Gynecologic Oncology, 1515 Holcombe Blvd, Unit 1362, Houston, TX 77030, USA.
| | | | | |
Collapse
|
83
|
Margaryan NV, Strizzi L, Abbott DE, Seftor EA, Rao MS, Hendrix MJC, Hess AR. EphA2 as a promoter of melanoma tumorigenicity. Cancer Biol Ther 2009; 8:279-88. [PMID: 19223760 DOI: 10.4161/cbt.8.3.7485] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The greatest health threat from malignant melanoma is death due to metastatic disease. Consequently, the identification of markers predictive of metastatic disease is essential for identifying new therapeutic targets. EphA2, a protein tyrosine kinase receptor commonly expressed in epithelial cells, has been found to be overexpressed and constitutively active in melanoma tumor cells having a metastatic phenotype as characterized by increased invasion, proliferation and vasculogenic mimicry (VM). Based on this observation, we hypothesized that increased expression of EphA2 by melanoma tumor cells could promote these characteristics of a metastatic phenotype in addition to promoting tumorigenicity as a whole. We analyzed a panel of human melanoma tumor cell lines derived from patient tissues classified as primary (either radial growth phase or vertical growth phase) and/or metastatic for the expression of EphA2 and found a correlation between increased EphA2 expression and metastatic potential. Experiments using the most metastatic of the human melanoma cell lines demonstrated that downregulation of EphA2 results in a significant decrease in invasion, proliferation, clonogenicity and VM in vitro, in addition to suppressed tumorigenicity in an orthotopic mouse model. Lastly, utilization of a human phospho-kinase array revealed increased phosphorylation of several different protein kinases involved in mediating various aspects of cellular proliferation. To the best of our knowledge these results provide the first direct in vivo evidence demonstrating a role for EphA2 in promoting melanoma tumorigenicity and suggest EphA2 as a significant molecular target for the therapeutic intervention of malignant melanoma.
Collapse
|
84
|
Wykosky J, Debinski W. The EphA2 receptor and ephrinA1 ligand in solid tumors: function and therapeutic targeting. Mol Cancer Res 2009; 6:1795-806. [PMID: 19074825 DOI: 10.1158/1541-7786.mcr-08-0244] [Citation(s) in RCA: 246] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The Eph receptor tyrosine kinases and ephrin ligands have been studied extensively for their roles in developmental processes. In recent years, Eph receptors and ephrins have been found to be integral players in cancer formation and progression. Among these are EphA2 and ephrinA1, which are involved in the development and maintenance of many different types of solid tumors. The function of EphA2 and ephrinA1 in tumorigenesis and tumor progression is complex and seems to be dependent on cell type and microenvironment. These variables affect the expression of the EphA2 and ephrinA1 proteins, the pathways through which they induce signaling, and the functional consequences of that signaling on the behavior of tumor cells and tumor-associated cells. This review will specifically focus on the roles that EphA2 and ephrinA1 play in the different cell types that contribute to the malignancy of solid tumors, with emphasis on the opportunities for therapeutic targeting.
Collapse
Affiliation(s)
- Jill Wykosky
- Department of Neurosurgery, Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | |
Collapse
|
85
|
Wesa AK, Herrem CJ, Mandic M, Taylor JL, Vasquez C, Kawabe M, Tatsumi T, Leibowitz MS, Finke JH, Bukowski RM, Bruckheimer E, Kinch MS, Storkus WJ. Enhancement in specific CD8+ T cell recognition of EphA2+ tumors in vitro and in vivo after treatment with ligand agonists. THE JOURNAL OF IMMUNOLOGY 2008; 181:7721-7. [PMID: 19017961 DOI: 10.4049/jimmunol.181.11.7721] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The EphA2 receptor tyrosine kinase is an attractive therapeutic target that is commonly overexpressed on solid tumors, with the degree of overexpression associated with disease progression, metastatic potential, and poor prognosis. Agonistic mAbs or ligand (ephrinA1)-Fc fusion protein are capable of inducing EphA2 internalization and degradation, thereby (at least transiently) eliminating the influence of this oncoprotein. We and others have also shown that EphA2 contains multiple peptide epitopes that can be recognized by effector CD4(+) and CD8(+) T cells isolated from tumor-bearing patients. Herein, we show that "agonist" reagents that trigger the proteasome-dependent degradation of tumor cell EphA2 result in the improved presentation of peptides derived from (both the extracellular and intracellular domains of) EphA2 in MHC class I complexes expressed on the tumor cell membrane for at least 48 h, as manifested by increased recognition by EphA2-specific CD8(+) T cells in vitro. We also observed that while delivery of ephrinA1-Fc fusion protein or agonist mAb into EphA2(+) tumor lesions promotes EphA2 degradation in situ, this single administration of agent does not dramatically alter tumor progression in a humanized SCID model. However, when combined with the adoptive transfer of normally nontherapeutic (human) anti-EphA2 CD8(+) CTL, this dual-agent regimen results in complete tumor eradication. These results suggest that strategies targeting the conditional proteasome-mediated destruction of tumor cell EphA2 may enable EphA2-specific CD8(+) T cells (of modest functional avidity) to realize improved therapeutic potential.
Collapse
Affiliation(s)
- Amy K Wesa
- Department of Dermatology, University of Pittsburgh School ofMedicine, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Lu C, Shahzad MM, Wang H, Landen CN, Kim SW, Allen J, Nick AM, Jennings N, Kinch MS, Bar-Eli M, Sood AK. EphA2 overexpression promotes ovarian cancer growth. Cancer Biol Ther 2008; 7:1098-103. [PMID: 18443431 PMCID: PMC2705979 DOI: 10.4161/cbt.7.7.6168] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Silencing EphA2 has been shown to result in anti-tumor efficacy. However, it is not known whether increasing EphA2 expression specifically results in increased tumor growth and progression. We examined the effects of stable EphA2 transfection into poorly invasive ovarian cancer cells with regard to in vitro invasive and in vivo metastatic potential. RESULTS In low cell density, EphA2-overexpressing A2780 cells (A2780-EphA2) displayed less cell-cell contact, increased cell-extracellular matrix (ECM) attachment and anchorage-independent cell growth compared to empty vector controls. There was no significant effect on anchorage-dependent cell proliferation, migration or invasion. Increased expression of EphA2 promoted tumor growth and enhanced the metastatic potential in A2780-EphA2 human ovarian cancer xenografts. The overexpression of EphA2 resulted in enhanced microvessel density (MVD), but had no effect on tumor cell proliferation. METHODS EphA2 gene was introduced into A2780 cells by retroviral infection. The effects of increased EphA2 expression were examined on cellular morphology, and anchorage-dependent and independent cell growth. Furthermore, the effect of EphA2 overexpression on metastatic ability was determined using an orthotopic nude mouse model of ovarian carcinoma. CONCLUSIONS EphA2 promotes tumor growth by enhancing cell-ECM adhesion, increasing anchorage-independent growth and promoting angiogenesis.
Collapse
Affiliation(s)
- Chunhua Lu
- Department of Gynecologic Oncology, UTMD Anderson Cancer Center; Houston, Texas USA
| | - Mian M.K. Shahzad
- Department of Gynecologic Oncology, UTMD Anderson Cancer Center; Houston, Texas USA
- Department of Obstetrics and Gynecology; Baylor College of Medicine; Houston, Texas USA
| | - Hua Wang
- Department of Cancer Biology; UTMD Anderson Cancer Center; Houston, Texas USA
| | - Charles N. Landen
- Department of Gynecologic Oncology, UTMD Anderson Cancer Center; Houston, Texas USA
| | - Seung W. Kim
- Department of Cancer Biology; UTMD Anderson Cancer Center; Houston, Texas USA
| | - Julie Allen
- Department of Cancer Biology; UTMD Anderson Cancer Center; Houston, Texas USA
| | - Alpa M. Nick
- Department of Gynecologic Oncology, UTMD Anderson Cancer Center; Houston, Texas USA
| | - Nicholas Jennings
- Department of Gynecologic Oncology, UTMD Anderson Cancer Center; Houston, Texas USA
| | | | - Menashe Bar-Eli
- Department of Cancer Biology; UTMD Anderson Cancer Center; Houston, Texas USA
| | - Anil K. Sood
- Department of Gynecologic Oncology, UTMD Anderson Cancer Center; Houston, Texas USA
- Department of Cancer Biology; UTMD Anderson Cancer Center; Houston, Texas USA
| |
Collapse
|
87
|
Holm R, de Putte GV, Suo Z, Lie AK, Kristensen GB. Expressions of EphA2 and EphrinA-1 in early squamous cell cervical carcinomas and their relation to prognosis. Int J Med Sci 2008; 5:121-6. [PMID: 18566674 PMCID: PMC2424178 DOI: 10.7150/ijms.5.121] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 06/04/2008] [Indexed: 11/05/2022] Open
Abstract
By using immunohistochemistry we investigated the expression of EphA2 and EphrinA-1 in 217 early squamous cell cervical carcinomas and examine their prognostic relevance. For EphA2 expression, 21 tumors (10%) showed negative, 108 (50%) weak positive, 69 (32%) moderate positive and 19 (9%) strong positive, whereas for EphrinA-1 expression, 33 tumors (15%) showed negative, 91 (42%) weak positive, 67 (31%) moderate positive and 26 (12%) strong positive. In univariate analysis high expression (strong staining) of EphrinA-1 was associated with poor disease-free (P = 0.033) and disease-specific (P = 0.039) survival. However, in the multivariate analyses neither EphrinA-1 nor EphA2 was significantly associated to survival. The increased levels of EphA2 and EphrinA-1 in a relative high number of early stage squamous cell carcinomas suggested that these two proteins may play an important role in the development of a subset of early cervical cancers. However, EphA2 and EphrinA-1 were not independently associated with clinical outcome.
Collapse
Affiliation(s)
- Ruth Holm
- Division of Pathology, The Norwegian Radium Hospital, Rikshospitalet University Hospital, Oslo, Norway.
| | | | | | | | | |
Collapse
|
88
|
Hess AR, Margaryan NV, Seftor EA, Hendrix MJC. Deciphering the signaling events that promote melanoma tumor cell vasculogenic mimicry and their link to embryonic vasculogenesis: role of the Eph receptors. Dev Dyn 2008; 236:3283-96. [PMID: 17557303 DOI: 10.1002/dvdy.21190] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
During embryogenesis, the primordial microcirculation is formed through a process known as vasculogenesis. The term "vasculogenic mimicry" has been used to describe the manner in which highly aggressive, but not poorly aggressive melanoma tumor cells express endothelial and epithelial markers and form vasculogenic-like networks similar to embryonic vasculogenesis. Vasculogenic mimicry is one example of the remarkable plasticity demonstrated by aggressive melanoma cells and suggests that these cells have acquired an embryonic-like phenotype. Since the initial discovery of tumor cell vasculogenic mimicry by our laboratory, we have been focusing on understanding the molecular mechanisms that regulate this process. This review will highlight recent findings identifying key signal transduction events that regulate melanoma vasculogenic mimicry and their similarity to the signal transduction events responsible for promoting embryonic vasculogenesis and angiogenesis. Specifically, this review will focus on the role of the Eph receptors and ligands in embryonic vasculogenesis, angiogenesis, and vasculogenic mimicry.
Collapse
Affiliation(s)
- Angela R Hess
- Children's Memorial Research Center, Program in Cancer Biology and Epigenomics, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer, Chicago, Illinois 60614-3394, USA.
| | | | | | | |
Collapse
|
89
|
Lazar AJF, Das P, Tuvin D, Korchin B, Zhu Q, Jin Z, Warneke CL, Zhang PS, Hernandez V, Lopez-Terrada D, Pisters PW, Pollock RE, Lev D. Angiogenesis-Promoting Gene Patterns in Alveolar Soft Part Sarcoma. Clin Cancer Res 2007; 13:7314-21. [DOI: 10.1158/1078-0432.ccr-07-0174] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|