1
|
Trifanescu OG, Mitrica RI, Gales LN, Marinescu SA, Motas N, Trifanescu RA, Rebegea L, Gherghe M, Georgescu DE, Serbanescu GL, Bashar HH, Dragosloveanu S, Cristian DA, Anghel RM. Validation of a New Prognostic Score in Patients with Ovarian Adenocarcinoma. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020229. [PMID: 36837431 PMCID: PMC9967083 DOI: 10.3390/medicina59020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/08/2023] [Accepted: 01/19/2023] [Indexed: 01/28/2023]
Abstract
Background and Objectives: This study aimed to assess the impact of clinical prognostic factors and propose a prognostic score that aids the clinician's decision in estimating the risk for patients in clinical practice. Materials and Methods: The study included 195 patients diagnosed with ovarian adenocarcinoma. The therapeutic strategy involved multidisciplinary decisions: surgery followed by adjuvant chemotherapy (80%), neoadjuvant chemotherapy followed by surgery (16.4%), and only chemotherapy in selected cases (3.6%). Results: After a median follow-up of 68 months, in terms of progression-free survival (PFS) and overall survival (OS), Eastern Cooperative Oncology Group (ECOG) performance status of 1 and 2 vs. 0 (hazard ratio-HR = 2.71, 95% confidence interval-CI, 1.96-3.73, p < 0.001 for PFS and HR = 3.19, 95%CI, 2.20-4.64, p < 0.001 for OS), menopausal vs. premenopausal status (HR = 2.02, 95%CI, 1.35-3,0 p < 0.001 and HR = 2.25, 95%CI = 1.41-3.59, p < 0.001), ascites (HR = 1.95, 95%CI 1.35-2.80, p = 0.03, HR = 2.31, 95%CI = 1.52-3.5, p < 0.007), residual disease (HR = 5.12, 95%CI 3.43-7.65, p < 0.0001 and HR = 4.07, 95%CI = 2.59-6.39, p < 0.0001), and thrombocytosis (HR = 2.48 95%CI = 1.72-3.58, p < 0.0001, HR = 3.33, 95%CI = 2.16-5.13, p < 0.0001) were associated with a poor prognosis. An original prognostic score including these characteristics was validated using receiver operating characteristic (ROC) curves (area under the curve-AUC = 0.799 for PFS and AUC = 0.726 for OS, p < 0.001). The median PFS for patients with none, one, two, three, or four (or more) prognostic factors was not reached, 70, 36, 20, and 12 months, respectively. The corresponding median overall survival (OS) was not reached, 108, 77, 60, and 34 months, respectively. Conclusions: Several negative prognostic factors were identified: ECOG performance status ≥ 1, the presence of ascites and residual disease after surgery, thrombocytosis, and menopausal status. These led to the development of an original prognostic score that can be helpful in clinical practice.
Collapse
Affiliation(s)
- Oana Gabriela Trifanescu
- Discipline of Oncology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Radu Iulian Mitrica
- Discipline of Oncology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Laurentia Nicoleta Gales
- Discipline of Oncology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
- Department of Oncology, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
- Correspondence: (L.N.G.); (D.E.G.)
| | - Serban Andrei Marinescu
- Department of Surgery, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Natalia Motas
- Department of Thoracic Surgery, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
- Discipline of Thoracic Surgery, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
| | - Raluca Alexandra Trifanescu
- Discipline of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 011863 Bucharest, Romania
- “C.I. Parhon” Institute of Endocrinology, 011863 Bucharest, Romania
| | - Laura Rebegea
- Discipline of Oncology, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800010 Galati, Romania
| | - Mirela Gherghe
- Department of Nuclear Medicine, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Dragos Eugen Georgescu
- “Dr. Ion Cantacuzino” Department of Surgery, “Carol Davila” University of Medicine and Pharmacy, 030167 Bucharest, Romania
- Correspondence: (L.N.G.); (D.E.G.)
| | - Georgia Luiza Serbanescu
- Discipline of Oncology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Haj Hamoud Bashar
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Hospital, 66421 Homburg, Germany
| | - Serban Dragosloveanu
- Discipline of Orthopedics, “Foisor” Orthopedics Hospital, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
| | - Daniel Alin Cristian
- Discipline of Surgery, Coltea Clinical Hospital, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- Discipline of Oncology, “Carol Davila” University of Medicine and Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| |
Collapse
|
2
|
Trifanescu OG, Gales LN, Tanase BC, Marinescu SA, Trifanescu RA, Gruia IM, Paun MA, Rebegea L, Mitrica R, Serbanescu L, Anghel RM. Prognostic Role of Vascular Endothelial Growth Factor and Correlation with Oxidative Stress Markers in Locally Advanced and Metastatic Ovarian Cancer Patients. Diagnostics (Basel) 2023; 13:diagnostics13010166. [PMID: 36611458 PMCID: PMC9818969 DOI: 10.3390/diagnostics13010166] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) plays an important role in tumor progression in ovarian cancer, but the complex mechanism and interaction with oxidative stress are not fully understood. METHODS A prospective study included 52 patients with ovarian adenocarcinoma stage IIIA-IV. Serum VEGF and reactive oxygen species (ROS) such as malondialdehyde and ceruloplasmin were measured. RESULTS VEGF levels were elevated (mean 1014.7 ± 165 pg/mL), especially in patients with macroscopic residual disease (1058 vs. 810 pg/mL, p = 0.0001). Median progression-free survival (PFS) and overall survival (OS) were 6 and 40 months in patients with a very high VEGF (over 1200 pg/mL), 11 and 48 months in patients with VEGF between 1000-1200 pg/mL, 18 and 84 months in patients with VEGF between 800-1000 pg/mL, and not reached in patients with normal VEGF. Increased VEGF values were associated with a 2.6-fold increased risk of disease progression (HR = 2.60, 95% CI 1.69-3.99), and a 1.4-fold increased risk of death (HR = 1.4, 95% CI 1.15-1.91, p = 0.002). Receiver operator characteristic (ROC) curves were used to validate VEGF as a prognostic factor and the area under the curve (AUC) was 0.814, p = 0.036 for PFS and 0.729, p = 0.043, for OS. There was a positive correlation between VEGF and malondialdehyde, Pearson coefficient of 0.35, p = 0.0001. CONCLUSIONS VEGF and malondialdehyde are important prognostic markers in ovarian cancer, especially in macroscopic residual disease, and there is a positive correlation between angiogenesis and oxidative stress.
Collapse
Affiliation(s)
- Oana Gabriela Trifanescu
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Laurentia Nicoleta Gales
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Medical Oncology II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
- Correspondence: (L.N.G.); (R.A.T.)
| | - Bogdan Cosmin Tanase
- Thoracic Surgery, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Serban Andrei Marinescu
- Oncologic Surgery I, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Raluca Alexandra Trifanescu
- “C. I. Parhon” Institute of Endocrinology, 011863 Bucharest, Romania
- Department of Endocrinology C. I. Parhon, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Correspondence: (L.N.G.); (R.A.T.)
| | - Iuliana Maria Gruia
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Mihai Andrei Paun
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Laura Rebegea
- Medical Clinical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800008 Galați, Romania
| | - Radu Mitrica
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Luiza Serbanescu
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- Department of Oncology, University of Medicine and Pharmacy “Carol Davila”, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology Bucharest, 022328 Bucharest, Romania
| |
Collapse
|
3
|
Liu C, Li Y, Zhu Y, Lu M. The Value of IOTA Simple Rules Combined With CEUS Scoring System in the Diagnosis of Benign and Malignant Ovarian Masses and Its Correlation With MVD and VEGF: A Preliminary Study. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2022; 41:2983-2992. [PMID: 35481545 DOI: 10.1002/jum.15999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/23/2022] [Accepted: 04/17/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVES To investigate the diagnostic value of International Ovarian Tumor Analysis (IOTA) simple rules combined with contrast-enhanced ultrasound (CEUS) scoring system in the differential diagnosis of ovarian tumors, and the correlations of the scoring system with microvessel density (MVD) and vascular endothelial growth factor (VEGF). METHODS One hundred eighty-nine patients with ovarian tumors were examined by routine ultrasound and CEUS. The enhanced characteristics of CEUS were observed, and the masses were classified by IOTA simple rules. To compare the diagnostic value of IOTA simple rules combined with CEUS scoring system and IOTA simple rules in the diagnosis of ovarian tumors. Immunohistochemistry was used to detect the expression of MVD and VEGF in postoperative tissue samples. The correlations between the new scoring system with MVD and VEGF were analyzed. RESULTS The sensitivity (93.98%), specificity (94.34%), positive predictive value (92.86%), negative predictive value (95.24%), and accuracy (94.18%) of IOTA simple rules combined with CEUS scoring system in the diagnosis of ovarian tumors were higher than those of IOTA simple rules alone (all P < .05). The score system was significantly positively correlated with MVD and VEGF, and the r values were 0.77 and 0.63, respectively (P < .001). CONCLUSIONS IOTA simple rules combined with CEUS scoring system was helpful to improve the accuracy of ultrasound diagnosis of ovarian tumors, which was significantly correlated with MVD and VEGF. It could provide important reference information for treatment scheme formulation and prognosis evaluation.
Collapse
Affiliation(s)
- Chun Liu
- Department of Ultrasound, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuan Li
- Department of Ultrasound, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Zhu
- Department of Ultrasound, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Man Lu
- Department of Ultrasound, Sichuan Cancer Hospital Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Guo L, Qin X, Xue L, Yang JY, Zhang Y, Zhu S, Ye G, Tang R, Yang W. A novel form of docetaxel polymeric micelles demonstrates anti-tumor and ascites-inhibitory activities in animal models as monotherapy or in combination with anti-angiogenic agents. Front Pharmacol 2022; 13:964076. [PMID: 36091776 PMCID: PMC9449419 DOI: 10.3389/fphar.2022.964076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
Malignant ascites (MA) is caused by intraperitoneal spread of solid tumor cells and results in a poor quality of life. Chemotherapy is a common first-line treatment for patients with MA. Taxotere ® (DTX) is widely used in solid tumor therapies. However, the low water solubility and side effects caused by additives in the formulation restrict the clinical application of docetaxel. HT001 is a clinical stage docetaxel micelle developed to overcome the solubility issue with improved safety profiles. To support clinical development and expand clinical application of HT001, this study used in vitro and in vivo approaches to investigate the anti-tumor effects of HT001 when applied as monotherapy or in combination with anti-angiogenic agents. HT001 demonstrated comparable anti-proliferative activities as docetaxel in a broad range of cancer cell lines in vitro. Furthermore, HT001 suppressed tumor growth in a dose-dependent manner in A549, MCF-7, and SKOV-3 xenograft tumor mouse models in vivo. In a hepatocellular carcinoma H22 malignant ascites-bearing mouse model, HT001 presented a dose-dependent inhibition of ascites production, prolonged animal survival, and reduced VEGF levels. When dosed at 20 mg/kg, the HT001-treated group exhibited curative results, with no ascites formation in 80% of mice at the end of the study while all the mice in the vehicle control group succumbed. Similar results were obtained in HT001 treatment of mice bearing malignant ascites produced by human ovarian cancer ES-2 cells. Notably, the combination of HT001 with Endostar not only significantly reduced ascites production but also prolonged survival of H22 ascites-bearing mice. HT001 showed similar PK and tissue distribution profiles as DTX in non-rodent hosts. Collectively, these results demonstrate potent anti-tumor activity of HT001 in multiple solid tumor models or malignant ascites models, and reveal synergistic effects with anti-angiogenic agents, supporting the clinical development and clinical expansion plans for HT001.
Collapse
Affiliation(s)
- Leilei Guo
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Xiaokang Qin
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Liting Xue
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Janine Y. Yang
- Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Yumei Zhang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shunwei Zhu
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Gang Ye
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
| | - Renhong Tang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
- *Correspondence: WenQing Yang, ; Renhong Tang,
| | - WenQing Yang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co. Ltd, Nanjing, China
- *Correspondence: WenQing Yang, ; Renhong Tang,
| |
Collapse
|
5
|
Yung MMH, Siu MKY, Ngan HYS, Chan DW, Chan KKL. Orchestrated Action of AMPK Activation and Combined VEGF/PD-1 Blockade with Lipid Metabolic Tunning as Multi-Target Therapeutics against Ovarian Cancers. Int J Mol Sci 2022; 23:ijms23126857. [PMID: 35743298 PMCID: PMC9224484 DOI: 10.3390/ijms23126857] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/06/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is one of the most lethal gynecological malignancies worldwide, and chemoresistance is a critical obstacle in the clinical management of the disease. Recent studies have suggested that exploiting cancer cell metabolism by applying AMP-activated protein kinase (AMPK)-activating agents and distinctive adjuvant targeted therapies can be a plausible alternative approach in cancer treatment. Therefore, the perspectives about the combination of AMPK activators together with VEGF/PD-1 blockade as a dual-targeted therapy against ovarian cancer were discussed herein. Additionally, ferroptosis, a non-apoptotic regulated cell death triggered by the availability of redox-active iron, have been proposed to be governed by multiple layers of metabolic signalings and can be synergized with immunotherapies. To this end, ferroptosis initiating therapies (FITs) and metabolic rewiring and immunotherapeutic approaches may have substantial clinical potential in combating ovarian cancer development and progression. It is hoped that the viewpoints deliberated in this review would accelerate the translation of remedial concepts into clinical trials and improve the effectiveness of ovarian cancer treatment.
Collapse
Affiliation(s)
- Mingo M. H. Yung
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
| | - Michelle K. Y. Siu
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
| | - Hextan Y. S. Ngan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
| | - David W. Chan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Correspondence: or (D.W.C.); (K.K.L.C.); Tel.: +852-3917-9367 or +852-3943-6053 (D.W.C.); +852-2255-4260 (K.K.L.C.); Fax: +852-2816-1947 or +852-2603-5123 (D.W.C.); +852-2255-0947 (K.K.L.C.)
| | - Karen K. L. Chan
- Department of Obstetrics & Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (M.M.H.Y.); (M.K.Y.S.); (H.Y.S.N.)
- Correspondence: or (D.W.C.); (K.K.L.C.); Tel.: +852-3917-9367 or +852-3943-6053 (D.W.C.); +852-2255-4260 (K.K.L.C.); Fax: +852-2816-1947 or +852-2603-5123 (D.W.C.); +852-2255-0947 (K.K.L.C.)
| |
Collapse
|
6
|
The Value of Preoperative Plasma VEGF Levels in Urothelial Carcinoma of the Bladder Treated with Radical Cystectomy. Eur Urol Focus 2021; 8:972-979. [PMID: 34454852 DOI: 10.1016/j.euf.2021.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/15/2021] [Accepted: 08/12/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Elevated preoperative plasma levels of the angiogenesis-related marker VEGF have been associated with worse oncological outcomes in various malignancies. OBJECTIVE To investigate the predictive/prognostic role of VEGF in patients with urothelial carcinoma of the bladder (UCB) treated with radical cystectomy (RC). DESIGN, SETTING, AND PARTICIPANTS VEGF plasma levels were measured preoperatively in 1036 patients with UCB who underwent RC. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The correlation between plasma VEGF levels and pathological and survival outcomes was assessed using logistic regression and Cox regression analyses. Discrimination was assessed using the concordance index (C index). The clinical net benefit was evaluated using decision curve analysis (DCA). RESULTS AND LIMITATIONS Patients with higher pretreatment plasma VEGF levels had poorer recurrence-free survival (RFS), cancer-specific survival (CSS), and overall survival (OS) according to log-rank tests (all p < 0.001). Higher VEGF levels were not independently associated with higher risk of lymph node metastasis, ≥pT3 disease, or non-organ-confined disease (all p > 0.05). Preoperative plasma VEGF levels were independently associated with RFS, CSS, and OS in preoperative and postoperative multivariable models. However, in all cases the C index increased by <0.02 and there was no improvement in net benefit on DCA. A limitation is that none of the patients received current elements of standard of care such as neoadjuvant chemotherapy. CONCLUSIONS Elevated plasma VEGF levels were associated with features of biologically and clinically aggressive disease such as worse survival outcomes among patients with UCB treated with RC. However, VEGF appears to have relatively limited incremental additive value in clinical use. Further study of VEGF for UCB prognostication is warranted before routine use in clinical algorithms. PATIENT SUMMARY Currently available models for predicting outcomes in bladder cancer are less than optimal. A protein called vascular endothelial growth factor (VEGF), which is a marker of the formation of blood vessels (angiogenesis), may have a role in predicting survival outcomes in bladder cancer. TAKE HOME MESSAGE Elevated plasma VEGF levels are associated with worse survival outcomes for patients with urothelial carcinoma of the bladder (UCB) treated with radical cystectomy. VEGF could be used as a part of a biomarker panel to enhance tools currently used for risk stratification for patients with UCB.
Collapse
|
7
|
O GS, R DD, V AT, I AI, P GT. The Plasticity of Circulating Tumor Cells in Ovarian Cancer During Platinum-containing Chemotherapy. Curr Cancer Drug Targets 2021; 21:965-974. [PMID: 34288839 DOI: 10.2174/1568009621666210720141229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Circulating tumor cells (CTCs) are a potential source of metastases and relapses. The data on the ovarian cancer (OC) CTCs molecular characteristics are limited. OBJECTIVE To assess the TGFβ, CXCL2, VEGFA and ERCC1 expression in two OC CTC subpopulations before and during chemotherapy (CT), and its relation to clinical characteristics. METHODS Two CTCs subpopulations (EpCAM+CK18+E-cadherin+; EpCAM+CK18+Vimentin+) were enriched using immunomagnetic separation before treatment and after 3 cycles of platinum-containing CT. Expression of mRNA was assessed using RT-qPCR. RESULTS The study included 31 I-IV stage OC patients. During CT, TGFβ levels increased in both fractions (p=0.054) compared with the initial levels. ERCC1 expression in E-cadherin+ CTCs was higher during neoadjuvant than adjuvant CT (p=0.004). CXCL2 level in E-cadherin+ CTCs increased (p=0.038) during neoadjuvant CT compared with the initial. TGF-β expression in vimentin+ CTCs during CT was negatively correlated to disease stage (p=0.003). Principal component analysis before CT revealed a component combining VEGFA, TGFβ, CXCL2, and a component with ERCC1 and VEGFA; during CT, component 1 contained ERCC1 and VEGFA, component 2 - TGFβ and CXCL2 in both fractions. Increased ERCC1 expression in E-cadherin+ CTCs during CT was associated with decreased progression-free survival (PFS) (HR 1.11 (95% CI 1.03-1.21, p=0.009) in multivariate analysis. CONCLUSION EpCAM+ OC CTCs are phenotypically heterogeneous, which may reflect variability in their metastatic potential. CT changes the molecular characteristics of CTCs. Expression of TGFβ in EpCAM+ CTCs increases during CT. High ERCC1 expression in EpCAM+CK18+E-cadherin+ CTCs during CT is associated with decreased PFS in OC.
Collapse
Affiliation(s)
- Gening Snezhanna O
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Ulyanovsk State University, Ulyanovsk, Russian Federation
| | - Dolgova Dinara R
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Ulyanovsk State University, Ulyanovsk, Russian Federation
| | - Abakumova Tatyana V
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Ulyanovsk State University, Ulyanovsk, Russian Federation
| | - Antoneeva Inna I
- Department of Gynecology, Regional Clinical Oncology Center, Ulyanovsk, Russian Federation
| | - Gening Tatyana P
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Ulyanovsk State University, Ulyanovsk, Russian Federation
| |
Collapse
|
8
|
Maleki P, Sheida SV, Mowla SJ, Soleimani V, Taheri M, Raheb J. LINK-A long non-coding RNA and VEGF RNA expression in epithelial ovarian cancer patients. Hum Antibodies 2020; 28:227-232. [PMID: 32333582 DOI: 10.3233/hab-200411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
LINK-A (long intergenic non-coding RNA for kinase activation) is a newly identified long non-coding RNA with oncogenic function, which leads to the hyperactivation of AKT and HIF1α. thereby, fosters cell proliferation, mobility and metastasis. VEGF (vascular endothelial growth factor), a well-known cytokine has an important role in angiogenesis. In this study, we quantified RNA expression of LINK-A and VEGF on 45 tumor specimens obtained from Iranian patients diagnosed with Epithelial Ovarian Cancer (EOC). Our goal was to evaluate expression of LINK-A lncRNA and VEGF mRNA in ovarian cancer tissues and find the probable correlation of LINK-A with VEGF as a major transcription target for HIF1α. LINK-A and VEGF were remarkably overexpressed in EOC tissues compared to normal tissues (P value: 0.004, 0.0001, respectively), but we did not find correlation between LINK-A and VEGF RNA expressions in this study. LINK-A was significantly overexpressed in higher stages of cancer and tumor grades. VEGF was only significantly elevated in higher stages. This study confirms importance of novel lncRNA of LINK-A in Iranian EOC patients.
Collapse
Affiliation(s)
- Parichehr Maleki
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Sadaf Valeh Sheida
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vahid Soleimani
- Pathology Department, Cancer Institute of Iran, Tehran University of Medical Science, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamshid Raheb
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
9
|
Mercer C, Jones A, Rusling JF, Leech D. Multiplexed Electrochemical Cancer Diagnostics With Automated Microfluidics. ELECTROANAL 2019; 31:208-211. [PMID: 32390709 PMCID: PMC7207070 DOI: 10.1002/elan.201800632] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 10/26/2018] [Indexed: 01/08/2023]
Abstract
Microfluidic platforms can lead to miniaturisation, increased throughput and reduced reagent consumption, particularly when the processes are automated. Here, a programmable microcontroller is used for automation of a microfluidic platform configured to electrochemically determine the levels of 8 proteins simultaneously in complex liquid samples. The platform system is composed of a programmable Arduino microcontroller that controls inexpensive valve actuators, pump, magnetic stirrer and electronic display. The programmable microcontroller results in repeatable timing for each step in a complex assay protocol, such as sandwich immunoassays. Application of the platform is demonstrated using a multiplexed electrochemical immunoassay based on capture at the electrode surface of magnetic particles labelled with horseradish peroxidase and detection antibody. The multiplexed assay protocol is completed in less than 30 mins and results in detection of eight proteins associated with prostate cancer. The approach presented can be used to automate and simplify high-throughput screening campaigns, such as detection of multiple biomarkers in patient samples.
Collapse
Affiliation(s)
- Conan Mercer
- School of Chemistry, and Ryan Institute National University of Ireland Galway University Road, Galway
| | - Abby Jones
- Department of Chemistry University of Connecticut Storrs, CT 06269, USA
| | - James F. Rusling
- School of Chemistry, and Ryan Institute National University of Ireland Galway University Road, Galway
- Department of Chemistry University of Connecticut Storrs, CT 06269, USA
- Institute of Materials Science University of Connecticut Storrs, CT 06269, USA
- Department of Surgery, and Neag Cancer Center UConn Health Farmington, CT 06032, USA
| | - Dónal Leech
- School of Chemistry, and Ryan Institute National University of Ireland Galway University Road, Galway
| |
Collapse
|
10
|
Lin Z, Li D, Cheng W, Wu J, Wang K, Hu Y. MicroRNA-181 Functions as an Antioncogene and Mediates NF-κB Pathway by Targeting RTKN2 in Ovarian Cancers. Reprod Sci 2018; 26:1071-1081. [PMID: 30309296 DOI: 10.1177/1933719118805865] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
MicroRNA (miR)-181 has been reported to participate in carcinogenesis and tumor progression in several malignant cancers, but its expression and biological functions in ovarian cancer have remained largely unclarified. Here, we first measured miR-181 expression in clinical ovarian cancers and found the expression levels of miR-181 were significantly lower in ovarian cancer tissues than that in adjacent tissues. Next, we screened and identified a direct miR-181 target, Rhotekin2 (RTKN2). A correlation between miR-181 and RTKN2 expression was also confirmed in clinical samples of ovarian cancers. Upregulation of miR-181 would specifically and markedly suppress RTKN2 expression. The miR-181-overexpressing subclones showed significant cell growth inhibition by cell apoptosis induction and significant impairment of cell invasiveness in SKOV3 and HO8910 ovarian cancer cells. To identify the mechanisms, we investigated the NF-κB pathway and found that nuclear factor-kappa B (NF-κB), B-cell lymphoma-2 (Bcl-2), and vascular endothelial growth factor (VEGF) were suppressed, whereas IκBα was promoted in miR-181-overexpressing cells. These findings indicate that miR-181 functions as a tumor suppressor and plays a substantial role in inhibiting the tumorigenesis and reversing the metastasis of ovarian cancer through RTKN2-NF-κB signaling pathway in vitro. Taken together, we believe that miR-181 may be a promising therapeutic target for treating malignant ovarian cancers.
Collapse
Affiliation(s)
- Zilin Lin
- 1 Medical Department, General Hospital of the People's Liberation Army, Beijing, China
| | - Dehao Li
- 1 Medical Department, General Hospital of the People's Liberation Army, Beijing, China
| | - Wenjia Cheng
- 2 Surgery of Nanlou Department, General Hospital of the People's Liberation Army, Beijing, China
| | - Jiajia Wu
- 1 Medical Department, General Hospital of the People's Liberation Army, Beijing, China
| | - Kun Wang
- 1 Medical Department, General Hospital of the People's Liberation Army, Beijing, China
| | - Yi Hu
- 3 Department of Oncology, General Hospital of the People's Liberation Army, Beijing, China
| |
Collapse
|
11
|
Pazos MC, Sequeira G, Bocchicchio S, May M, Abramovich D, Parborell F, Tesone M, Irusta G. PDGFB as a vascular normalization agent in an ovarian cancer model treated with a gamma-secretase inhibitor. J Cell Physiol 2018; 233:5949-5961. [PMID: 29266203 DOI: 10.1002/jcp.26404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/12/2017] [Indexed: 12/21/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related deaths in women. In the past 20 years, the canonical types of drugs used to treat ovarian cancer have not been replaced and the survival rates have not changed. These facts show the clear need to find new therapeutic strategies for this illness. Thus, the aim of the present study was to investigate the effect of a gamma-secretase inhibitor (DAPT) in combination with the Platelet-derived growth factor B (PDGFB) on an ovarian cancer xenograft model. To achieve this goal, we analyzed the effect of the administration of DAPT alone and the co-administration of DAPT and recombinant PDGFB on parameters associated with tumor growth and angiogenesis in an orthotopic experimental model of ovarian cancer. We observed that the dose of DAPT used was ineffective to reduce ovarian tumor growth, but showed anticancer activity when co-administered with recombinant PDGFB. The administration of PDGFB alone normalized tumor vasculature by increasing periendothelial coverage and vascular functionality. Interestingly, this effect exerted by PDGFB was also observed in the presence of DAPT. Our findings suggest that PDGFB is able to improve tumor vascularity and allows the anticancer action of DAPT in the tumor. We propose that this therapeutic strategy could be a new tool for ovarian cancer treatment and deserves further studies.
Collapse
Affiliation(s)
- Maria C Pazos
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado, Buenos Aires, Argentina
| | - Gonzalo Sequeira
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado, Buenos Aires, Argentina
| | - Sebastian Bocchicchio
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado, Buenos Aires, Argentina
| | - Maria May
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado, Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado, Buenos Aires, Argentina
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado, Buenos Aires, Argentina
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental (IByME-CONICET), Vuelta de Obligado, Buenos Aires, Argentina
| |
Collapse
|
12
|
Ruscito I, Cacsire Castillo-Tong D, Vergote I, Ignat I, Stanske M, Vanderstichele A, Glajzer J, Kulbe H, Trillsch F, Mustea A, Kreuzinger C, Benedetti Panici P, Gourley C, Gabra H, Nuti M, Taube ET, Kessler M, Sehouli J, Darb-Esfahani S, Braicu EI. Characterisation of tumour microvessel density during progression of high-grade serous ovarian cancer: clinico-pathological impact (an OCTIPS Consortium study). Br J Cancer 2018; 119:330-338. [PMID: 29955134 PMCID: PMC6070919 DOI: 10.1038/s41416-018-0157-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 05/22/2018] [Accepted: 06/01/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND High-grade serous ovarian cancer (HGSOC) intratumoural vasculature evolution remains unknown. The study investigated changes in tumour microvessel density (MVD) in a large cohort of paired primary and recurrent HGSOC tissue samples and its impact on patients' clinico-pathological outcome. METHODS A total of 222 primary (pOC) and recurrent (rOC) intra-patient paired HGSOC were assessed for immunohistochemical expression of angiogenesis-associated biomarkers (CD31, to evaluate MVD, and VEGF-A). Expression profiles were compared between pOCs and rOCs and correlated with patients' data. RESULTS High intratumoural MVD and VEGF-A expression were observed in 75.7% (84/111) and 20.7% (23/111) pOCs, respectively. MVDhigh and VEGF(+) samples were detected in 51.4% (57/111) and 20.7% (23/111) rOCs, respectively. MVDhigh/VEGF(+) co-expression was found in 19.8% (22/111) and 8.1% (9/111) of pOCs and rOCs, respectively (p = 0.02). Pairwise analysis showed no significant change in MVD (p = 0.935) and VEGF-A (p = 0.121) levels from pOCs to rOCs. MVDhigh pOCs were associated with higher CD3(+) (p = 0.029) and CD8(+) (p = 0.013) intratumoural effector TILs, while VEGF(+) samples were most frequently encountered among BRCA-mutated tumours (p = 0.019). Multivariate analysis showed VEGF and MVD were not independent prognostic factors for OS. CONCLUSIONS HGSOC intratumoural vasculature did not undergo significant changes during disease progression. High concentration of CD31(+) vessels seems to promote recruitment of effector TILs. The study also provides preliminary evidence of the correlation between VEGF-positivity and BRCA status.
Collapse
Affiliation(s)
- Ilary Ruscito
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany. .,Cell Therapy Unit and Laboratory of Tumor Immunology, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - Dan Cacsire Castillo-Tong
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Ignace Vergote
- Division of Gynaecological Oncology, Leuven Cancer Institute, Department of Gynaecology and Obstetrics, University Hospital Leuven, Catholic University of Leuven, Herestraat 49, B-3000, Leuven, Belgium
| | - Iulia Ignat
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Mandy Stanske
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Adriaan Vanderstichele
- Division of Gynaecological Oncology, Leuven Cancer Institute, Department of Gynaecology and Obstetrics, University Hospital Leuven, Catholic University of Leuven, Herestraat 49, B-3000, Leuven, Belgium
| | - Jacek Glajzer
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Hagen Kulbe
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistrasse 15, Munich, Germany.,Department of Gynecology and Gynecologic Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 46, Hamburg, Germany
| | - Alexander Mustea
- Department of Gynecology and Obstetrics, University Medicine of Greifswald, Greifswald, Germany
| | - Caroline Kreuzinger
- Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | | | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, MRC IGMM, Western General Hospital, University of Edinburgh Cancer Research, UK Centre, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Hani Gabra
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK.,Clinical Discovery Unit, AstraZeneca, Cambridge, UK
| | - Marianna Nuti
- Cell Therapy Unit and Laboratory of Tumor Immunology, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Eliane T Taube
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Mirjana Kessler
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Silvia Darb-Esfahani
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Elena Ioana Braicu
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
13
|
[Recommendations for biomarker testing in epithelial ovarian cancer. A national consensus statement by the Spanish Society of Pathology and the Spanish Society of Medical Oncology]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2018; 51:84-96. [PMID: 29602379 DOI: 10.1016/j.patol.2017.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 11/24/2022]
Abstract
Advances in the understanding of the histological and molecular characteristics of ovarian cancer now allow 5subtypes to be identified, leading to a more refined therapeutic approach and improved clinical trials. Each of the subtypes has specific histological features and a particular biomarker expression, as well as mutations in different genes, some of which have prognostic and predictive value. CA125 and HE4 are examples of ovarian cancer biomarkers used in diagnosis and follow-up. Currently, somatic or germinal mutations on BRCA1 and BRCA2 genes are the most important biomarkers in epithelial ovarian cancer, having prognostic and predictive value. In this article, a group of experts from the Spanish Society of Medical Oncology and the Spanish Society of Pathology review the histological and molecular characteristics of the 5subtypes of ovarian cancer and describe the most useful biomarkers and mutations for diagnosis, screening and tailored treatment strategy.
Collapse
|
14
|
Nuclear factor 90 promotes angiogenesis by regulating HIF-1α/VEGF-A expression through the PI3K/Akt signaling pathway in human cervical cancer. Cell Death Dis 2018; 9:276. [PMID: 29449553 PMCID: PMC5833414 DOI: 10.1038/s41419-018-0334-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 01/28/2023]
Abstract
Vascular endothelial growth factor A (VEGF-A), a fundamental component of angiogenesis, provides nutrients and oxygen to solid tumors, and enhances tumor cell survival, invasion, and migration. Nuclear factor 90 (NF90), a double-stranded RNA-binding protein, is strongly expressed in several human cancers, promotes tumor growth by reducing apoptosis, and increasing cell cycle process. The mechanisms by which cervical cancer cells inducing VEGF-A expression and angiogenesis upon NF90 upregulation remain to be fully established. We demonstrated that NF90 is upregulated in human cervical cancer specimens and the expression of NF90 is paralleled with that of VEGF-A under hypoxia. The expressions of hypoxia inducible factor-1α (HIF-1α) and VEGF-A are downregulated upon NF90 knockdown, which can be rescued by ectopic expression of NF90. Suppression of NF90 decreases the tube formation and cell migration of HUVECs. Moreover, the PI3K/Akt signaling pathway participates in the regulation. Knockdown of NF90 also reduces the tumor growth and angiogenesis of cervical cancer cell line in the mouse xenograft model. Taken together, suppression of NF90 in cervical cancer cell lines can decrease VEGF-A expression, inhibit angiogenesis, and reduce tumorigenic capacity in vivo.
Collapse
|
15
|
The effect of celecoxib on tumor growth in ovarian cancer cells and a genetically engineered mouse model of serous ovarian cancer. Oncotarget 2018; 7:39582-39594. [PMID: 27074576 PMCID: PMC5129955 DOI: 10.18632/oncotarget.8659] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/28/2016] [Indexed: 12/20/2022] Open
Abstract
Our objective was to evaluate the effect of the COX-2 inhibitor, celecoxib, on (1) proliferation and apoptosis in human ovarian cancer cell lines and primary cultures of ovarian cancer cells, and (2) inhibition of tumor growth in a genetically engineered mouse model of serous ovarian cancer under obese and non-obese conditions. Celecoxib inhibited cell proliferation in three ovarian cancer cell lines and five primary cultures of human ovarian cancer after 72 hours of exposure. Treatment with celecoxib resulted in G1 cell cycle arrest, induction of apoptosis, inhibition of cellular adhesion and invasion and reduction of expression of hTERT mRNA and COX-2 protein in all of the ovarian cancer cell lines. In the KpB mice fed a high fat diet (obese) and treated with celecoxib, tumor weight decreased by 66% when compared with control animals. Among KpB mice fed a low fat diet (non-obese), tumor weight decreased by 46% after treatment with celecoxib. In the ovarian tumors from obese and non-obese KpB mice, treatment with celecoxib as compared to control resulted in decreased proliferation, increased apoptosis and reduced COX-2 and MMP9 protein expression, as assessed by immunohistochemistry. Celecoxib strongly decreased the serum level of VEGF and blood vessel density in the tumors from the KpB ovarian cancer mouse model under obese and non-obese conditions. This work suggests that celecoxib may be a novel chemotherapeutic agent for ovarian cancer prevention and treatment and be potentially beneficial in both obese and non-obese women.
Collapse
|
16
|
Jang K, Kim M, Gilbert CA, Simpkins F, Ince TA, Slingerland JM. VEGFA activates an epigenetic pathway upregulating ovarian cancer-initiating cells. EMBO Mol Med 2017; 9:304-318. [PMID: 28179359 PMCID: PMC5331266 DOI: 10.15252/emmm.201606840] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The angiogenic factor, VEGFA, is a therapeutic target in ovarian cancer (OVCA). VEGFA can also stimulate stem‐like cells in certain cancers, but mechanisms thereof are poorly understood. Here, we show that VEGFA mediates stem cell actions in primary human OVCA culture and OVCA lines via VEGFR2‐dependent Src activation to upregulate Bmi1, tumor spheres, and ALDH1 activity. The VEGFA‐mediated increase in spheres was abrogated by Src inhibition or SRC knockdown. VEGFA stimulated sphere formation only in the ALDH1+ subpopulation and increased OVCA‐initiating cells and tumor formation in vivo through Bmi1. In contrast to its action in hemopoietic malignancies, DNA methyl transferase 3A (DNMT3A) appears to play a pro‐oncogenic role in ovarian cancer. VEGFA‐driven Src increased DNMT3A leading to miR‐128‐2 methylation and upregulation of Bmi1 to increase stem‐like cells. SRC knockdown was rescued by antagomir to miR‐128. DNMT3A knockdown prevented VEGFA‐driven miR‐128‐2 loss, and the increase in Bmi1 and tumor spheres. Analysis of over 1,300 primary human OVCAs revealed an aggressive subset in which high VEGFA is associated with miR‐128‐2 loss. Thus, VEGFA stimulates OVCA stem‐like cells through Src‐DNMT3A‐driven miR‐128‐2 methylation and Bmi1 upregulation.
Collapse
Affiliation(s)
- Kibeom Jang
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Minsoon Kim
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Candace A Gilbert
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fiona Simpkins
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Obstetrics & Gynecology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tan A Ince
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, USA.,Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA .,Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
17
|
Xu CH, He ZH, Xu H. Association of four genetic polymorphisms in the vascular endothelial growth factor-A gene and development of ovarian cancer: a meta-analysis. Oncotarget 2017; 8:73063-73078. [PMID: 29069849 PMCID: PMC5641192 DOI: 10.18632/oncotarget.20379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 08/09/2017] [Indexed: 12/20/2022] Open
Abstract
This study meta-analyzed the literature on possible association of four polymorphisms (+936C/T, −460C/T, −2578C/A and −1154G/A) in the vascular endothelial growth factor (VEGF)-A gene with risk of ovarian cancer. Meta-analysis of 7 case-control studies involving +936C/T, 4 studies involving −460C/T, 4 studies involving −2578C/A and 2 studies involving −1154G/A showed significant association between −460C/T and ovarian cancer risk. This risk was observed in the total population (allelic model, OR 1.80, 95% CI 1.26–2.59, P = 0.001; recessive model, OR 1.84, 95% CI 1.13–2.98, P = 0.01; dominant model, OR 0.51, 95% CI 0.39–0.67, P < 0.001; homozygous model, OR 2.48, 95% CI 1.72–3.56, P < 0.001; heterozygous model, OR 1.67, 95% CI 1.26–2.21, P < 0.001) and in the subgroup of Asian study participants. The CA genotype at −2578C/A was a risk factor in the total population, while the CT genotype at +936C/T was a protective factor in Caucasians. None of the five genetic models suggested a significant association between −1154G/A and ovarian cancer risk in the entire study population, or between +936C/T and risk in Asian or Chinese participants. These findings should be verified in large, well-designed studies.
Collapse
Affiliation(s)
- Chao-Huan Xu
- Department of Gynaecology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhong-Hui He
- Department of Gynaecology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Hong Xu
- Department of Gynaecology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
18
|
Serum Vascular Endothelial Growth Factor-A as a Prognostic Biomarker for Epithelial Ovarian Cancer. Int J Gynecol Cancer 2017; 27:1325-1332. [DOI: 10.1097/igc.0000000000001027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BackgroundBevacizumab, which targets vascular endothelial growth factor (VEGF)-A, has recently been proven to be effective for the treatment of epithelial ovarian cancer (EOC). Thus, interest in VEGF-A has increased. There are few reports on concomitant detection of both ligands and its soluble receptors in serum samples, and the significance of serum VEGF-A in EOC is unclear, unlike the situation with tissue samples. We conducted the present study to explore the levels of serum VEGF family and its receptors and to evaluate their utility as prognostic biomarkers.MethodsA total of 128 patients with EOC, who were consecutively treated at Tottori University Hospital between 2006 and 2012, were included. Blood samples were collected before initial surgery. Serum concentrations of VEGF-A, VEGF-C, VEGFR-1, and VEGFR-2 were analyzed by enzyme-linked immunosorbent assay. We also examined the mRNA and protein expression of VEGF-A in tumor tissue from 30 cases by real-time reverse transcription polymerase chain reaction and immunohistochemistry.ResultsThe levels of VEGF-A in patients with stage III/IV disease were significantly higher than those with stage I/II disease (P = 0.0036). On the other hand, the level of VEGFR-2 in stage III/IV was significantly lower than that in stage I/II (P = 0.0026). With the cutoff value of VEGF/VEGFRs at the median level, the overall survival (OS) for patients with high VEGF-A levels was significantly lower than those with low levels (P = 0.015). Patients with high VEGFR-2 levels showed better prognosis than those with low VEGFR-2 levels (P = 0.023). Multivariate analysis revealed that International Federation of Gynecology and Obstetrics stage and serum VEGF-A were independent prognostic factors for OS [hazard ratio 2.01, 95% confidence interval (1.13–3.63), P = 0.017]. There was no significant correlation between mRNA or protein expression and serum levels of VEGF-A.ConclusionsSerum VEGF-A is an independent prognostic factor for OS in patients with EOC, implying that serum VEGF-A is a prognostic biomarker for EOC. Further study to validate the data is needed.
Collapse
|
19
|
Recommendations for biomarker testing in epithelial ovarian cancer: a National Consensus Statement by the Spanish Society of Pathology and the Spanish Society of Medical Oncology. Clin Transl Oncol 2017; 20:274-285. [PMID: 28815456 DOI: 10.1007/s12094-017-1719-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 07/22/2017] [Indexed: 12/20/2022]
Abstract
Because of advances in the understanding of histological and molecular characteristics in ovarian cancer, it is now possible to recognize the existence of five subtypes, which in turn has allowed a more refined therapeutic approach and better design of clinical trials. Each of these five subtypes has specific histological features and a particular biomarker expression, as well as mutations in different genes, some of which have prognostic and predictive value. CA125 and HE4 are examples of ovarian cancer biomarkers used in the diagnosis and follow-up of these malignancies. Currently, somatic or germinal mutations on BRCA1 and BRCA2 genes are the most important biomarkers in epithelial ovarian cancer having prognostic and predictive value. This article will review the histological and molecular characteristics of the five subtypes of ovarian cancer, describing the most important biomarkers and mutations that can guide in diagnosis, screening and tailored treatment strategy.
Collapse
|
20
|
Phelps DL, Borley JV, Flower KJ, Dina R, Darb-Esfahani S, Braicu I, Sehouli J, Fotopoulou C, Wilhelm-Benartzi CS, Gabra H, Yazbek J, Chatterjee J, Ip J, Khan H, Likos-Corbett MT, Brown R, Ghaem-Maghami S. Methylation of MYLK3 gene promoter region: a biomarker to stratify surgical care in ovarian cancer in a multicentre study. Br J Cancer 2017; 116:1287-1293. [PMID: 28350786 PMCID: PMC5482730 DOI: 10.1038/bjc.2017.83] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 02/22/2017] [Accepted: 03/03/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Survival benefit from surgical debulking of ovarian cancer (OC) is well established, but some women, despite total macroscopic clearance of disease, still have poor prognosis. We aimed to identify biomarkers to predict benefit from conventional surgery. METHODS Clinical data from women debulked for high-stage OC were analysed (Hammersmith Hospital, London, UK; 2001-2014). Infinium's HumanMethylation27 array interrogated tumour DNA for differentially methylated CpG sites, correlated to survival, in patients with the least residual disease (RD; Hammersmith Array). Validation was performed using bisulphite pyrosequencing (Charité Hospital, Berlin, Germany cohort) and The Cancer Genome Atlas' (TCGA) methylation data set. Kaplan-Meier curves and Cox models tested survival. RESULTS Altogether 803 women with serous OC were studied. No RD was associated with significantly improved overall survival (OS; hazard ratio (HR) 1.25, 95% CI 1.06-1.47; P=0.0076) and progression-free survival (PFS; HR 1.23, 95% CI 1.05-1.43; P=0.012; Hammersmith database n=430). Differentially methylated loci within FGF4, FGF21, MYLK2, MYLK3, MYL7, and ITGAE associated with survival. Patients with the least RD had significantly better OS with higher methylation of MYLK3 (Hammersmith (HR 0.51, 95% CI 0.31-0.84; P=0.01), Charité (HR 0.46, 95% CI 0.21-1.01; P=0.05), and TCGA (HR 0.64, 95% CI 0.44-0.93; P=0.02)). CONCLUSIONS MYLK3 methylation is associated with improved OS in patients with the least RD, which could potentially be used to determine response to surgery.
Collapse
Affiliation(s)
- David L Phelps
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Jane V Borley
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Kirsty J Flower
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Roberto Dina
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | | | - Ioana Braicu
- Department of Gynaecology, Virchow Campus, Universitätsmedizin, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynaecology, Virchow Campus, Universitätsmedizin, Berlin, Germany
| | - Christina Fotopoulou
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
- Department of Gynaecology, Virchow Campus, Universitätsmedizin, Berlin, Germany
| | | | - Hani Gabra
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Joseph Yazbek
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Jayanta Chatterjee
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Jacey Ip
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Harun Khan
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | | | - Robert Brown
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Sadaf Ghaem-Maghami
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
21
|
Zonta YR, Martinez M, Camargo ICC, Domeniconi RF, Lupi Júnior LA, Pinheiro PFF, Reiter RJ, Martinez FE, Chuffa LGA. Melatonin Reduces Angiogenesis in Serous Papillary Ovarian Carcinoma of Ethanol-Preferring Rats. Int J Mol Sci 2017; 18:ijms18040763. [PMID: 28398226 PMCID: PMC5412347 DOI: 10.3390/ijms18040763] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/24/2017] [Accepted: 03/30/2017] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is a hallmark of ovarian cancer (OC); the ingrowth of blood vessels promotes rapid cell growth and the associated metastasis. Melatonin is a well-characterized indoleamine that possesses important anti-angiogenic properties in a set of aggressive solid tumors. Herein, we evaluated the role of melatonin therapy on the angiogenic signaling pathway in OC of an ethanol-preferring rat model that mimics the same pathophysiological conditions occurring in women. OC was chemically induced with a single injection of 7,12-dimethylbenz(a)anthracene (DMBA) under the ovarian bursa. After the rats developed serous papillary OC, half of the animals received intraperitoneal injections of melatonin (200 µg/100 g body weight/day) for 60 days. Melatonin-treated animals showed a significant reduction in OC size and microvessel density. Serum levels of melatonin were higher following therapy, and the expression of its receptor MT1 was significantly increased in OC-bearing rats, regardless of ethanol intake. TGFβ1, a transforming growth factor-beta1, was reduced only after melatonin treatment. Importantly, vascular endothelial growth factor (VEGF) was severely reduced after melatonin therapy in animals given or not given ethanol. Conversely, the levels of VEGF receptor 1 (VEGFR1) was diminished after ethanol consumption, regardless of melatonin therapy, and VEGFR2 was only reduced following melatonin. Hypoxia-inducible factor (HIF)-1α was augmented with ethanol consumption, and, notably, melatonin significantly reduced their levels. Collectively, our results suggest that melatonin attenuates angiogenesis in OC in an animal model of ethanol consumption; this provides a possible complementary therapeutic opportunity for concurrent OC chemotherapy.
Collapse
MESH Headings
- Alcohol Drinking/physiopathology
- Animals
- Antioxidants/administration & dosage
- Antioxidants/pharmacology
- Blotting, Western
- Cystadenocarcinoma, Papillary/blood supply
- Cystadenocarcinoma, Papillary/drug therapy
- Cystadenocarcinoma, Papillary/metabolism
- Cystadenocarcinoma, Serous/blood supply
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/metabolism
- Ethanol/administration & dosage
- Female
- Food Preferences
- Immunohistochemistry
- Injections, Intraperitoneal
- Melatonin/administration & dosage
- Melatonin/pharmacology
- Microscopy, Fluorescence
- Neovascularization, Pathologic/prevention & control
- Ovarian Neoplasms/blood supply
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Rats
- Receptor, Melatonin, MT1/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor Receptor-1/metabolism
Collapse
Affiliation(s)
- Yohan Ricci Zonta
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Marcelo Martinez
- Department of Morphology and Pathology, Universidade Federal de São Carlos (UFSCar), São Carlos-SP 13565-905, Brazil.
| | - Isabel Cristina C Camargo
- Department of Biotechnology, School of Sciences, Humanities and Languages, São Paulo State University (UNESP), Assis-SP 19806-900, Brazil.
| | - Raquel F Domeniconi
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Luiz Antonio Lupi Júnior
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Patricia Fernanda F Pinheiro
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, USA.
| | - Francisco Eduardo Martinez
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| | - Luiz Gustavo A Chuffa
- Department of Anatomy, Institute of Biosciences, São Paulo State University (UNESP), Botucatu-SP 18618-970, Brazil.
| |
Collapse
|
22
|
Anti-tumour activity of tivozanib, a pan-inhibitor of VEGF receptors, in therapy-resistant ovarian carcinoma cells. Sci Rep 2017; 7:45954. [PMID: 28383032 PMCID: PMC5382685 DOI: 10.1038/srep45954] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 03/08/2017] [Indexed: 01/18/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most fatal gynaecological malignancy. Despite initial therapeutic response, the majority of advanced-stage patients relapse and succumb to chemoresistant disease. Overcoming drug resistance is the key to successful treatment of EOC. Members of vascular endothelial growth factor (VEGF) family are overexpressed in EOC and play key roles in its malignant progression though their contribution in development of the chemoresistant disease remains elusive. Here we show that expression of the VEGF family is higher in therapy-resistant EOC cells compared to sensitive ones. Overexpression of VEGFR2 correlated with resistance to cisplatin and combination with VEGFR2-inhibitor apatinib synergistically increased cisplatin sensitivity. Tivozanib, a pan-inhibitor of VEGF receptors, reduced proliferation of the chemoresistant EOC cells through induction of G2/M cell cycle arrest and apoptotic cell death. Tivozanib decreased invasive potential of these cells, concomitant with reduction of intercellular adhesion molecule-1 (ICAM-1) and diminishing the enzymatic activity of urokinase-type plasminogen activator (uPA) and matrix metalloproteinase-2 (MMP-2). Moreover, tivozanib synergistically enhanced anti-tumour effects of EGFR-directed therapies including erlotinib. These findings suggest that the VEGF pathway has potential as a therapeutic target in therapy-resistant EOC and VEGFR blockade by tivozanib may yield stronger anti-tumour efficacy and circumvent resistance to EGFR-directed therapies.
Collapse
|
23
|
Stine JE, Guo H, Sheng X, Han X, Schointuch MN, Gilliam TP, Gehrig PA, Zhou C, Bae-Jump VL. The HMG-CoA reductase inhibitor, simvastatin, exhibits anti-metastatic and anti-tumorigenic effects in ovarian cancer. Oncotarget 2016; 7:946-60. [PMID: 26503475 PMCID: PMC4808044 DOI: 10.18632/oncotarget.5834] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/23/2015] [Indexed: 01/07/2023] Open
Abstract
Ovarian cancer is the 5th leading cause of cancer death among women in the United States. The mevalonate pathway is thought to be a potential oncogenic pathway in the pathogenesis of ovarian cancer. Simvastatin, a 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) inhibitor, is a widely used drug for inhibiting the synthesis of cholesterol and may also have anti-tumorigenic activity. Our goal was to evaluate the effects of simvastatin on ovarian cancer cell lines, primary cultures of ovarian cancer cells and in an orthotopic ovarian cancer mouse model. Simvastatin significantly inhibited cellular proliferation, induced cell cycle G1 arrest and apoptosis, and caused cellular stress via reduction in the enzymatic activity of HMGCR and inhibition of the MAPK and mTOR pathways in ovarian cancer cells. Furthermore, simvastatin induced DNA damage and reduced cell adhesion and invasion. Simvastatin also exerted anti-proliferative effects on primary cell cultures of ovarian cancer. Treatment with simvastatin in an orthotopic mouse model reduced ovarian tumor growth, coincident with decreased Ki-67, HMGCR, phosphorylated-Akt and phosphorylated-p42/44 protein expression. Our findings demonstrate that simvastatin may have therapeutic benefit for ovarian cancer treatment and be worthy of further exploration in clinical trials.
Collapse
Affiliation(s)
- Jessica E Stine
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Hui Guo
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Gynecologic Oncology, ShanDong Cancer Hospital & Institute, Jinan University, Jinan, P.R. China
| | - Xiugui Sheng
- Department of Gynecologic Oncology, ShanDong Cancer Hospital & Institute, Jinan University, Jinan, P.R. China
| | - Xiaoyun Han
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,Department of Gynecologic Oncology, ShanDong Cancer Hospital & Institute, Jinan University, Jinan, P.R. China
| | - Monica N Schointuch
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Timothy P Gilliam
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Paola A Gehrig
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
24
|
Pierscianek D, Wolf S, Keyvani K, El Hindy N, Stein KP, Sandalcioglu IE, Sure U, Mueller O, Zhu Y. Study of angiogenic signaling pathways in hemangioblastoma. Neuropathology 2016; 37:3-11. [PMID: 27388534 DOI: 10.1111/neup.12316] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/16/2016] [Accepted: 05/22/2016] [Indexed: 01/30/2023]
Abstract
Hemangioblastoma (HB) is mainly located in the brain and the spinal cord. The tumor is composed of two major components, namely neoplastic stromal cells and abundant microvessels. Thus, hyper-vascularization is the hallmark of this tumor. Despite the identification of germline and/or epigenetic mutations of Von Hippel Lindau (VHL) gene as an important pathogenic mechanism of HB, little is known about the molecular signaling involved in this highly vascularized tumor. The present study investigated the key players of multiple angiogenic signaling pathways including VEGF/VEGFR2, EphB4/EphrinB2, SDF1α/CXCR4 and Notch/Dll4 pathways in surgical specimens of 22 HB. The expression of key angiogenic factors was detected by RT2 -PCR and Western blot. Immunofluorescent staining revealed the cellular localization of these proteins. We demonstrated a massive upregulation of mRNA levels of VEGF and VEGFR2, CXCR4 and SDF1α, EphB4 and EphrinB2, as well as the main components of Dll4-Notch signaling in HB. An increase in the protein expression of VEGF, CXCR4 and the core-components of Dll4-Notch signaling was associated with an activation of Akt and Erk1/2 and accompanied by an elevated expression of PCNA. Immuofluorescent staining revealed the expression of VEGF and CXCR4 in endothelial cells as well as in tumor cells. Dll4 protein was predominantly found in tumor cells, whereas EphB4 immunoreactivity was exclusively detected in endothelial cells. We conclude that multiple key angiogenic pathways were activated in HB, which may synergistically contribute to the abundant vascularization in this tumor. Identification of these aberrant pathways provides potential targets for a possible future application of anti-angiogenic therapy for this tumor, particularly when a total surgical resection becomes difficult due to the localization or multiplicity of the tumor.
Collapse
Affiliation(s)
| | - Stefanie Wolf
- Department of Neurosurgery, Universitatsklinikum Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University Hospital Essen, Essen, Germany
| | | | - Klaus-Peter Stein
- Department of Neurosurgery, KRH Hospital Nordstadt, Hannover, Germany
| | | | - Ulrich Sure
- Department of Neurosurgery, Universitatsklinikum Essen, Germany
| | - Oliver Mueller
- Department of Neurosurgery, Universitatsklinikum Essen, Germany
| | - Yuan Zhu
- Department of Neurosurgery, Universitatsklinikum Essen, Germany
| |
Collapse
|
25
|
Szubert S, Moszynski R, Michalak S, Nowicki M, Sajdak S, Szpurek D. The associations between serum VEGF, bFGF and endoglin levels with microvessel density and expression of proangiogenic factors in malignant and benign ovarian tumors. Microvasc Res 2016; 107:91-6. [PMID: 27312585 DOI: 10.1016/j.mvr.2016.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/27/2016] [Accepted: 06/12/2016] [Indexed: 11/19/2022]
Abstract
AIM OF THE STUDY To investigate whether serum levels of VEGF, bFGF and endoglin correlate with tumor VEGF and bFGF expression or microvessel density (MVD) in ovarian cancer. PATIENTS AND METHODS Forty five patients with epithelial ovarian cancers (EOCs) and 38 patients with benign ovarian tumors (BOTs) were included into the study. Serum levels of VEGF, bFGF and endoglin were assessed using ELISA. The expression of VEGF and bFGF in tumor samples were evaluated using ELISA of supernatants obtained from tumor homogenization. MVD was analyzed using immunohistochemistry with antibodies against CD31, CD34 and CD105. RESULTS Serum VEGF levels were significantly higher in EOCs than in BOTs (436.6pg/ml [19.67-2860] vs 295.5pg/ml [123-539], P=0.025). Serum endoglin levels were lowered in the group EOCs when compared to BOTs (33,720g/ml [12,220-73,940] vs 42,390pg/ml [19,380-56,910], P=0.015). There were no differences in bFGF levels between studied groups. EOCs have significantly higher CD105 MVD (25 vessels/mm2 [0-57] vs 6 vessels/mm2 [0-70], P<0.001) and tumor VEGF (405.9pg/mg protein [0-3000] vs 2.225 [0-634.7], P<0.001) expression than BOTs, while, bFGF expression was higher in BOTs than in EOCs (2076pg/mg protein [668.1-8718] vs 847.3pg/mg protein [188.9-8333], P=0.003). In patients with EOCs we have observed negative correlation between serum VEGF concentration and its tissue expression (r Spearman=-0.571, P=0.0261), and serum VEGF concentration correlated positively with CD34-MVD (r Spearman=0.545, P=0.0289). In a multiple regression analysis we have observed only the negative correlation between serum VEGF and CD105-MVD (r=-0.5288, P=0.0427). CONCLUSIONS Serum VEGF is a useful marker for prediction of ovarian cancer MVD and tumor VEGF expression.
Collapse
Affiliation(s)
- Sebastian Szubert
- Division of Gynecological Surgery, Poznan University of Medical Sciences, Poland.
| | - Rafal Moszynski
- Division of Gynecological Surgery, Poznan University of Medical Sciences, Poland
| | - Slawomir Michalak
- Department of Neurochemistry and Neuropathology, Poznan University of Medical Sciences, Poland; Neuroimmunological Unit Polish Academy of Sciences, Poland
| | - Michal Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poland
| | - Stefan Sajdak
- Division of Gynecological Surgery, Poznan University of Medical Sciences, Poland
| | - Dariusz Szpurek
- Division of Gynecological Surgery, Poznan University of Medical Sciences, Poland; Department of Obstetrics and Gynecology, Leszno Regional Hospital, Leszno, Poland
| |
Collapse
|
26
|
Antonângelo L, Tuma T, Fabro A, Acencio M, Terra R, Parra E, Vargas F, Takagaki T, Capelozzi V. Id-1, Id-2, and Id-3 co-expression correlates with prognosis in stage I and II lung adenocarcinoma patients treated with surgery and adjuvant chemotherapy. Exp Biol Med (Maywood) 2016; 241:1159-68. [PMID: 26869608 DOI: 10.1177/1535370216632623] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/22/2016] [Indexed: 12/25/2022] Open
Abstract
Inhibitors of DNA binding/inhibitors of differentiation (Id) protein family have been shown to be involved in carcinogenesis. However, the roles of Id during lung adenocarcinoma (ADC) progression remain unclear. Eighty-eight ADC samples were evaluated for Id-1,2,3 level and angiogenesis (CD 34 and VEGF microvessel density) by immunohistochemistry and morphometry. The impact of these markers was tested on follow-up until death or recurrence. A significant difference between tumor and normal tissue was found for Id-1,2,3 expression (P < 0.01). In addition, high levels of nuclear Id-1 were associated with higher angiogenesis in the tumor stroma (P < 0.01). Equally significant was the association between patients in T1-stage and low cytoplasmic Id-2, as well as patients in stage-IIb and low Id-3. High cytoplasm Id-3 expression was also directly associated to lymph nodes metastasis (P = 0.05). Patients at stages I to III, with low Id-1 and Id-3 cytoplasm histoscores showed significant long metastasis-free survival time than those with high Id-1 or Id-3 expression (P = 0.04). Furthermore, high MVD-CD34 and MVD-VEGF expression were associated with short recurrence-free survival compared to low MVD-CD34 and MVD-VEGF expressions (P = 0.04). Cox model analyses controlled for age, lymph node metastasis, and adjuvant treatments showed that nuclear Id-1, cytoplasmic Id-3, and MVD-CD34 were significantly associated with survival time. Median score for nuclear Id-1 and cytoplasmic Id-3 divided patients in two groups, being that those with increased Id-1 and Id-3 presented higher risk of death. Ids showed an independent prognostic value in patients with lung ADC, regardless of disease stage. Id-1 and Id-3 should be considered new target candidates in the development of personalized therapy in lung ADC.
Collapse
Affiliation(s)
- Leila Antonângelo
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| | - Taila Tuma
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| | - Alexandre Fabro
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| | - Milena Acencio
- Pulmonary Division, Heart Institute Clinics Hospital, University of Sao Paulo, Sao Paulo 01246903, Brazil
| | - Ricardo Terra
- Pulmonary Division, Heart Institute Clinics Hospital, University of Sao Paulo, Sao Paulo 01246903, Brazil
| | - Edwin Parra
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| | - Francisco Vargas
- Pulmonary Division, Heart Institute Clinics Hospital, University of Sao Paulo, Sao Paulo 01246903, Brazil
| | - Teresa Takagaki
- Pulmonary Division, Heart Institute Clinics Hospital, University of Sao Paulo, Sao Paulo 01246903, Brazil
| | - Vera Capelozzi
- Department of Pathology, University of Sao Paulo, Sao Paulo 01246903,Brazil
| |
Collapse
|
27
|
Coosemans A, Decoene J, Baert T, Laenen A, Kasran A, Verschuere T, Seys S, Vergote I. Immunosuppressive parameters in serum of ovarian cancer patients change during the disease course. Oncoimmunology 2015; 5:e1111505. [PMID: 27141394 DOI: 10.1080/2162402x.2015.1111505] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/15/2015] [Accepted: 10/17/2015] [Indexed: 12/30/2022] Open
Abstract
Neoplastic cells can escape immune control leading to cancer growth. Regulatory T cells (Treg), myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) are crucial in immune escape. TAM are divided based on their immune profile, M1 are immunostimulatory while M2 are immunosuppressive. Research so far has mainly focused on the intratumoral behavior of these cells. This study, on the other hand, explored the systemic changes of the key metabolites [IL-4 (interleukin), IL-13, arginase, IL-10, VEGF-A (vascular endothelial growth factor), CCL-2 (chemokine (C-C) motif ligand 2) and TGF-β (transforming growth factor)] linked to Treg, MDSC and TAM during the course of the disease in ovarian and fallopian tube cancer patients. Serum samples were therefore analyzed at diagnosis, after (interval)-debulking surgery and after chemotherapy (paclitaxel-carboplatin). We also determined galectin-1 (gal-1), involved in angiogenesis and tumor-mediated immune evasion. We found significantly lower levels of IL-10, VEGF-A, TGF-β and arginase and higher levels of gal-1 after chemotherapy compared to diagnosis. After debulking surgery, a decrease in IL-10 was significant. Gal-1 and CCL-2 appeared independent prognostic factors for progression-free and overall survival (OS) (multivariate analysis). These results will help us in the decision making of future therapies in order to further modulate the immune system in a positive way.
Collapse
Affiliation(s)
- An Coosemans
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium; Department of Oncology, Laboratory of Gynecologic Oncology, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Judit Decoene
- Department of Gynecology and Obstetrics, UZ Leuven , Leuven, Belgium
| | - Thaïs Baert
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium; Department of Oncology, Laboratory of Gynecologic Oncology, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Annouschka Laenen
- Biostatistics and Statistical Bioinformatics Center of Leuven, KU Leuven , Leuven, Belgium
| | - Ahmad Kasran
- Department of Microbiology and Immunology, Laboratory of Clinical Immunology, KU Leuven , Leuven, Belgium
| | - Tina Verschuere
- Department of Neuroscience, Laboratory of Experimental Neurosurgery, KU Leuven , Leuven, Belgium
| | - Sven Seys
- Department of Microbiology and Immunology, Laboratory of Clinical Immunology, KU Leuven , Leuven, Belgium
| | - Ignace Vergote
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium; Department of Oncology, Laboratory of Gynecologic Oncology, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| |
Collapse
|
28
|
Desai A, Xu J, Aysola K, Akinbobuyi O, White M, Reddy VE, Okoli J, Clark C, Partridge EE, Childs E, Beech DJ, Rice MV, Reddy E, Rao VN. Molecular Mechanism Linking BRCA1 Dysfunction to High Grade Serous Epithelial Ovarian Cancers with Peritoneal Permeability and Ascites. ACTA ACUST UNITED AC 2015; 1. [PMID: 26665166 DOI: 10.15744/2454-3284.1.103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ovarian cancer constitutes the second most common gynecological cancer with a five-year survival rate of 40%. Among the various histotypes associated with hereditary ovarian cancer, high-grade serous epithelial ovarian carcinoma (HGSEOC) is the most predominant and women with inherited mutations in BRCA1 have a lifetime risk of 40-60%. HGSEOC is a challenge for clinical oncologists, due to late presentation of patient, diagnosis and high rate of relapse. Ovarian tumors have a wide range of clinical presentations including development of ascites as a result of deregulated endothelial function thereby causing increased vascular permeability of peritoneal vessels. The molecular mechanisms remain elusive. Studies have shown that fallopian tube cancers develop in women with BRCA1 gene mutations more often than previously suspected. Recent studies suggest that many primary peritoneal cancers and some high-grade serous epithelial ovarian carcinomas actually start in the fallopian tubes. In this article we have addressed the molecular pathway of a recently identified potential biomarker Ubc9 whose deregulated expression due to BRCA1 dysfunction can result in HGSEOC with peritoneal permeability and formation of ascites. We also discuss the role of downstream targets Caveolin-1 and Vascular Endothelial Growth Factor (VEGF) in the pathogenesis of ascites in ovarian carcinomas. Finally we hypothesize a signaling axis between Ubc9 over expression, loss of Caveolin-1 and induction of VEGF in BRCA1 mutant HGSEOC cells. We suggest that Ubc9-mediated stimulation of VEGF as a novel mechanism underlying ovarian cancer aggressiveness and ascites formation. Agents that target Ubc9 and VEGF signaling may represent a novel therapeutic strategy to impede peritoneal growth and spread of HGSEOC.
Collapse
Affiliation(s)
- A Desai
- Cancer Biology Program, Department of OB/GYN, School of Medicine, Georgia Cancer Center for Excellence, Grady Health System, Atlanta, USA
| | - J Xu
- Department of Internal Medicine, School of Medicine, Georgia Cancer Center for Excellence, Grady Health System, Atlanta, USA
| | - K Aysola
- Department of Surgery, Morehouse, School of Medicine, Georgia Cancer Center for Excellence, Grady Health System, Atlanta, USA
| | - O Akinbobuyi
- Department of Internal Medicine, University of Buffalo, Erie County Medical Center, Buffalo NY
| | - M White
- Philadelphia College of Osteopathic Medicine, Suwanee GA
| | - V E Reddy
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Caron JM, Caron JM. Methyl Sulfone Blocked Multiple Hypoxia- and Non-Hypoxia-Induced Metastatic Targets in Breast Cancer Cells and Melanoma Cells. PLoS One 2015; 10:e0141565. [PMID: 26536104 PMCID: PMC4633041 DOI: 10.1371/journal.pone.0141565] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/10/2015] [Indexed: 12/18/2022] Open
Abstract
Metastatic cancer causes 90% of cancer deaths. Unlike many primary tumors, metastatic tumors cannot be cured by surgery alone. Metastatic cancer requires chemotherapy. However, metastatic cells are not easily killed by chemotherapy. These problems with chemotherapy are caused in part by the metastatic cell niche: hypoxia. Here we show that the molecule, methyl sulfone, normalized metastatic metabolism of hypoxic breast cancer and melanoma cells by altering several metabolic functions of the cells. Under hypoxia, methyl sulfone decreased expression of the master regulator of hypoxia, HIF-1α, and reduced levels of the glycolytic enzymes, PKM2, LDHA, GLUT1, the pro-angiogenic protein, VEGF, and the iron-sulfur metabolism molecules, miR-210 and transferrin, all of which promote metastasis. Conversely, methyl sulfone increased levels of ISCU1/2 and ferroportin, proteins associated with iron-sulfur cluster biogenesis and iron homeostasis in normal cells. These data identify methyl sulfone as a multi-targeting molecule that blocks the survival/proliferative effect of hypoxia on metastatic cells and brings normality back to cellular metabolism.
Collapse
|
30
|
Ranjbar R, Nejatollahi F, Nedaei Ahmadi AS, Hafezi H, Safaie A. Expression of Vascular Endothelial Growth Factor (VEGF) and Epidermal Growth Factor Receptor (EGFR) in Patients With Serous Ovarian Carcinoma and Their Clinical Significance. IRANIAN JOURNAL OF CANCER PREVENTION 2015; 8:e3428. [PMID: 26478789 PMCID: PMC4606373 DOI: 10.17795/ijcp-3428] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/12/2015] [Indexed: 12/19/2022]
Abstract
Background: Vascular endothelial growth factor (VEGF) has an essential role in tumor metastasis by inducing the construction of abnormal blood vessels. Epidermal growth factor receptor (EGFR) is involved in different parts of cancer growth such as tumor initiation, angiogenesis and metastasis. Objectives: The aim of this study was to evaluate the expression of VEGF and EGFR in ovarian cancer in southern Iran and to assess the correlation between expression of these two markers and patients’ age, tumor stage, and grade. Patients and Methods: In this cross-sectional study, 50 paraffin blocks of serous ovarian adenocarcinomas and 50 paraffin-embedded specimens from control individuals operated for reasons other than malignancy were immunohistochemically stained using anti-human VEGF and EGFR antibodies. Results: A significant difference in the frequency of positive expression of VEGF was observed in ovarian cancer patients (25.0%) compared with the control group (8.0%) (P = 0.023). A significant difference between EGFR expression in patients (56.8%) and controls (24.0%) was also obtained (P = 0.001). No significant correlation between VEGF and EGFR expression and patients’ age, tumor grade and stage were detected (P > 0.05). Conclusions: The significant increase in both VEGF and EGFR in the patients with ovarian cancer compared to healthy individuals could have prognostic value. Identifying these markers may be useful for chemopreventive and chemotherapeutic strategies for patients with serous ovarian cancer.
Collapse
Affiliation(s)
- Reza Ranjbar
- Molecular Biology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Foroogh Nejatollahi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, IR Iran ; Shiraz HIV/AIDS Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Ahmad Sina Nedaei Ahmadi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Hossein Hafezi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Akbar Safaie
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, IR Iran
| |
Collapse
|
31
|
Abstract
PURPOSE Although an anti-tumor effect of emodin has been reported before, its effect on human gynecological cancer cells has so far not been studied. Here, we assessed the effect of emodin on cervical cancer-derived (Hela), choriocarcinoma-derived (JAR) and ovarian cancer-derived (HO-8910) cells, and investigated the possible underlying molecular and cellular mechanisms. METHODS AND RESULTS The respective cells were treated with 0, 5, 10 or 15 μM emodin for 72 h. Subsequently, MTT and Transwell in vitro migration assays revealed that emodin significantly decreased the viability and invasive capacity of the gynecological cancer-derived cells tested. We found that emodin induced apoptosis and significantly decreased mitochondrial membrane potential and ATP release in these cells. We also found that emodin may exert its apoptotic effects via regulating the activity of caspase-9 and the expression of cleaved-caspase-3. Moreover, we found that emodin induced a cell cycle arrest at the G0/G1 phase, possibly through down-regulating the key cell cycle regulators Cyclin D and Cyclin E. Interestingly, emodin also led to autophagic cell death, as revealed by increased MAP LC3 expression, a marker of the autophagosome, and decreased expression of the autophagy regulators Beclin-1 and Atg12-Atg5. Finally, we found that the protein levels of both VEGF and VEGFR-2 were significantly decreased in emodin-treated cells, suggesting an anti-angiogenic effect of emodin on gynecological cancer-derived cells. CONCLUSIONS Our results suggest that emodin exhibits an anti-tumor effect on gynecological cancer-derived cells, possibly through multiple mechanisms including the induction of apoptosis and autophagy, the arrest of the cell cycle, and the inhibition of angiogenesis. Our findings may provide a basis for the design of potential emodin-based strategies for the treatment of gynecological tumors.
Collapse
|
32
|
Baker AF, Malm SW, Pandey R, Laughren C, Cui H, Roe D, Chambers SK. Evaluation of a hypoxia regulated gene panel in ovarian cancer. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2015; 8:45-56. [PMID: 25998313 PMCID: PMC4449346 DOI: 10.1007/s12307-015-0166-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/26/2015] [Indexed: 01/02/2023]
Abstract
A panel of nine hypoxia regulated genes, selected from a previously published fifty gene panel, was investigated for its ability to predict hypoxic ovarian cancer phenotypes. All nine genes including vascular endothelial growth factor A, glucose transporter 1, phosphoglycerate mutase 1, lactate dehydrogenase A, prolyl 4-hydroxylase, alpha-polypeptide 1, adrenomedullin, N-myc downstream regulated 1, aldolase A, and carbonic anhydrase 9 were upregulated in the HEY and OVCAR-3 human ovarian cell lines cultured in vitro under hypoxic compared to normoxic conditions as measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The gene panel was also elevated in HEY xenograft tumor tissue compared to HEY cells cultured in normoxia. The HEY xenograft tissue demonstrated heterogeneous positive immunohistochemical staining for the exogenous hypoxia biomarker pimonidazole, and the hypoxia regulated protein carbonic anhydrase IX. A quantitative nuclease protection assay (qNPA) was developed which included the nine hypoxia regulated genes. The qNPA assay provided similar results to those obtained using qRT-PCR for cultured cell lines. The qNPA assay was also evaluated using paraffin embedded fixed tissues including a set of five patient matched primary and metastatic serous cancers and four normal ovaries. In this small sample set the average gene expression was higher in primary and metastatic cancer tissue compared to normal ovaries for the majority of genes investigated. This study supports further evaluation by qNPA of this gene panel as an alternative or complimentary method to existing protein biomarkers to identify ovarian cancers with a hypoxic phenotype.
Collapse
Affiliation(s)
- Amanda F. Baker
- University of Arizona Cancer Center and College of Medicine, Tucson, Arizona ,University of Arizona Cancer Center, 1515 N. Campbell Ave Room 3977A, Tucson, AZ 85724 Arizona
| | - Scott W. Malm
- University of Arizona Cancer Center and College of Pharmacy, Tucson, Arizona
| | - Ritu Pandey
- University of Arizona Cancer Center, 1515 N. Campbell Ave Room 3977A, Tucson, AZ 85724 Arizona
| | - Cindy Laughren
- University of Arizona Cancer Center, 1515 N. Campbell Ave Room 3977A, Tucson, AZ 85724 Arizona
| | - Haiyan Cui
- University of Arizona Cancer Center, 1515 N. Campbell Ave Room 3977A, Tucson, AZ 85724 Arizona
| | - Denise Roe
- University of Arizona Cancer Center and Mel and Enid Zuckerman College of Public Health, Tucson, Arizona
| | - Setsuko K. Chambers
- University of Arizona Cancer Center and College of Medicine, Tucson, Arizona
| |
Collapse
|
33
|
Ptak A, Gregoraszczuk EL. Effects of bisphenol A and 17β-estradiol on vascular endothelial growth factor A and its receptor expression in the non-cancer and cancer ovarian cell lines. Cell Biol Toxicol 2015; 31:187-97. [DOI: 10.1007/s10565-015-9303-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022]
|
34
|
PAN XINTING, ZHU QINGYUN, SUN YUNBO, LI LIANDI, ZHU YUNPENG, ZHAO ZHIHUI, ZUO JIANXIN, FANG WEI, LI KUN. PLGA/poloxamer nanoparticles loaded with EPAS1 siRNA for the treatment of pancreatic cancer in vitro and in vivo. Int J Mol Med 2015; 35:995-1002. [DOI: 10.3892/ijmm.2015.2096] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 02/03/2015] [Indexed: 11/06/2022] Open
|
35
|
Prognostic significance of pretreatment VEGF, survivin, and Smac/DIABLO serum levels in patients with serous ovarian carcinoma. Tumour Biol 2015; 36:4157-65. [PMID: 25577253 PMCID: PMC4529473 DOI: 10.1007/s13277-015-3050-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/02/2015] [Indexed: 12/15/2022] Open
Abstract
The second mitochondria-derived activator of caspase (Smac/DIABLO), vascular endothelial growth factor (VEGF), and survivin are known to play a significant role in the growth and development of numerous tumors. Serum concentrations of VEGF, survivin, and Smac/DIABLO were analyzed in 92 patients with serous ovarian cancer and 94 healthy controls. Values were correlated with clinicopathological characteristics and outcomes. The median pretreatment serum VEGF and survivin levels in patients with serous ovarian carcinoma were significantly higher, while Smac/DIABLO levels were significantly lower than that in healthy controls. Receiver operating characteristic (ROC) curve analysis showed that the best cutoff point for VEGF was determined to be 345 pg/ml; with 83 % sensitivity and 65 % specificity. For survivin, the cutoff point was 110 pg/ml and for Smac/DIABLO was 75 pg/ml, with 82 and 62 % sensitivity and 43 and 87 % specificity, respectively. In the patients group, higher VEGF and survivin levels and lower Smac/DIABLO levels in sera were significantly associated with poorer overall survival (OS) and disease-free survival (DFS). Preoperative measurement of serum VEGF, survivin, and Smac/DIABLO may be of help in early detection of serous ovarian cancer and may provide important information about the patient's outcome and prognosis.
Collapse
|
36
|
De Francesco EM, Lappano R, Santolla MF, Marsico S, Caruso A, Maggiolini M. HIF-1α/GPER signaling mediates the expression of VEGF induced by hypoxia in breast cancer associated fibroblasts (CAFs). Breast Cancer Res 2014; 15:R64. [PMID: 23947803 PMCID: PMC3978922 DOI: 10.1186/bcr3458] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/17/2013] [Accepted: 08/15/2013] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Carcinoma-associated fibroblasts (CAFs) play a pivotal role in cancer progression by contributing to invasion, metastasis and angiogenesis. Solid tumors possess a unique microenvironment characterized by local hypoxia, which induces gene expression changes and biological features leading to poor outcomes. Hypoxia Inducible Factor 1 (HIF-1) is the main transcription factor that mediates the cell response to hypoxia through different mechanisms that include the regulation of genes strongly associated with cancer aggressiveness. Among the HIF-1 target genes, the G-protein estrogen receptor (GPER) exerts a stimulatory role in diverse types of cancer cells and in CAFs. METHODS We evaluated the regulation and function of the key angiogenic mediator vascular endothelial growth factor (VEGF) in CAFs exposed to hypoxia. Gene expression studies, Western blotting analysis and immunofluorescence experiments were performed in CAFs and breast cancer cells in the presence of cobalt chloride (CoCl₂) or cultured under low oxygen tension (2% O₂), in order to analyze the involvement of the HIF-1α/GPER signaling in the biological responses to hypoxia. We also explored the role of the HIF-1α/GPER transduction pathway in functional assays like tube formation in human umbilical vein endothelial cells (HUVECs) and cell migration in CAFs. RESULTS We first determined that hypoxia induces the expression of HIF-1α and GPER in CAFs, then we ascertained that the HIF-1α/GPER signaling is involved in the regulation of VEGF expression in breast cancer cells and in CAFs exposed to hypoxia. We also assessed by ChIP assay that HIF-1α and GPER are both recruited to the VEGF promoter sequence and required for VEGF promoter stimulation upon hypoxic condition. As a biological counterpart of these findings, conditioned medium from hypoxic CAFs promoted tube formation in HUVECs in a HIF-1α/GPER dependent manner. The functional cooperation between HIF-1α and GPER in CAFs was also evidenced in the hypoxia-induced cell migration, which involved a further target of the HIF-1α/GPER signaling like connective tissue growth factor (CTGF). CONCLUSIONS The present results provide novel insight into the role elicited by the HIF-1α/GPER transduction pathway in CAFs towards the hypoxia-dependent tumor angiogenesis. Our findings further extend the molecular mechanisms through which the tumor microenvironment may contribute to cancer progression.
Collapse
|
37
|
Zhang F, Li C, Liu H, Wang Y, Chen Y, Wu X. The functional proteomics analysis of VEGF-treated human epithelial ovarian cancer cells. Tumour Biol 2014; 35:12379-87. [DOI: 10.1007/s13277-014-2552-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/25/2014] [Indexed: 12/12/2022] Open
|
38
|
Zhang W, Zhao CG, Sun HY, Zheng WE, Chen H. Expression characteristics of KAI1 and vascular endothelial growth factor and their diagnostic value for hepatocellular carcinoma. Gut Liver 2014. [PMID: 25071074 DOI: 10.5009/gn13331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND/AIMS We tried to investigate the expression characteristics of KAI1, a suppressor of wide-spectrum tumor metastasis, and vascular endothelial growth factor (VEGF), the most common angiogenesis factor, and then to analyze their diagnostic value for hepatocellular carcinoma (HCC). METHODS The protein and mRNA expression levels of KAI1 or VEGF in HCC tissues and in self-controlled para-carcinoma tissues were analyzed by Western blot and real-time polymerase chain reaction, respectively. Serum levels of KAI1 and VEGF in the patients with HCC, benign liver disease or in healthy controls were quantitatively detected by enzyme-linked immunosorbent assay. RESULTS The expression level of KAI1 was downregulated, while the expression level of VEGF was upregulated in the tissues or serum of the patients with HCC. The expression level of serum KAI1 in HCC patients was correlated with TNM staging, intrahepatic metastasis, lymph node or peritoneal metastasis, and portal vein thrombus. In addition to the factors that were correlated with KAI1 expression, VEGF expression was also closely related to the α-fetoprotein level of the patients. The area under the receiver operating characteristic curve for the diagnosis of HCC was 0.907 for KAI1 and 0.779 for VEGF. The sensitivity of serum KAI1 levels in the diagnosis of HCC was 86.96%; the accuracy was 83.06%, while the sensitivity, the accuracy and the negative predictive value were improved to 91.86%, 84.68%, and 78.79% according to the combined detection of KAI1 and VEGF, respectively. CONCLUSIONS A combined detection of KAI1 and VEGF may greatly improve the efficiency of diagnosis and form a reliable panel of diagnostic markers for HCC.
Collapse
Affiliation(s)
- Wu Zhang
- Department of Adiotherapy, The Third Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| | | | - Hong Yu Sun
- Department of Adiotherapy, The Third Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| | - Wei E Zheng
- Department of Adiotherapy, The Third Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| | - Hua Chen
- Department of Adiotherapy, The Third Affiliated Hospital, Wenzhou, China
| |
Collapse
|
39
|
Wang W, Ren F, Wu Q, Jiang D, Li H, Shi H. MicroRNA-497 suppresses angiogenesis by targeting vascular endothelial growth factor A through the PI3K/AKT and MAPK/ERK pathways in ovarian cancer. Oncol Rep 2014; 32:2127-33. [PMID: 25176450 DOI: 10.3892/or.2014.3439] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 08/05/2014] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) have been shown to play an important role in diverse biological processes and cancer progression. The objective of the present study was to investigate the role of miR-497 in ovarian cancer angiogenesis. We found that miR-497 expression was downregulated in human ovarian cancer tissues, and the low miR-497 expression was significantly associated with increased angiogenesis. Functionally, exogenous expression of miR-497 suppressed the ability of ovarian cancer cells to promote capillary tube formation of endothelial cells. We further disclosed that miR-497 exerted its function of anti-angiogenesis by suppressing VEGFA expression in ovarian cancer cells and, in turn, impairing the VEGFR2-mediated PI3K/AKT and MAPK/ERK pathways. Our findings suggest that downregulation of miR-497 may contribute to angiogenesis in ovarian cancer. miR-497 may be a promising candidate target for prevention and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Wei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Fang Ren
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qinghua Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Dazhi Jiang
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hongjun Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Huirong Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
40
|
Xie Y, Hicks MJ, Kaminsky SM, Moore MAS, Crystal RG, Rafii A. AAV-mediated persistent bevacizumab therapy suppresses tumor growth of ovarian cancer. Gynecol Oncol 2014; 135:325-32. [PMID: 25108232 DOI: 10.1016/j.ygyno.2014.07.105] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 01/11/2023]
Abstract
RATIONALE Anti-angiogenesis therapies such as bevacizumab, the monoclonal antibody to vascular endothelial growth factor (VEGF), have been used against ovarian cancer, but transient and low peritoneal drug levels are likely a factor in treatment failure. We hypothesized that a single administration of adeno-associated virus (AAV)-mediated intraperitoneal expression of bevacizumab would direct persistent expression and suppress growth and metastasis of ovarian cancer. METHODS AAVrh.10BevMab, a rhesus serotype 10 adeno-associated viral vector coding for bevacizumab, was evaluated for the capacity of a single intraperitoneal administration to persistently suppress peritoneal tumor growth in an intraperitoneal model of ovarian carcinomatosis with human ovarian cancer cells in nude immunodeficient mice. RESULTS The data demonstrates that AAVrh10.BevMab mediates persistent and high levels of bevacizumab in the peritoneal cavity following a single intraperitoneal administration in mice. In AAVrh10.BevMab treated A2780 human ovarian cancer-bearing mice, tumor growth was significantly suppressed (p<0.05) and the area of blood vessels in the tumor was decreased (p<0.04). Survival of mice with A2780 xenografts or SK-OV3 xenografts was greatly prolonged in the presence of AAVrh10.BevMab (p<0.001). Administration of AAVrh10.BevMab 4days after A2780-luciferase cell implantation reduced tumor growth (p<0.01) and increased mouse survival (p<0.0001). Combination of AAVrh10.BevMab with cytotoxic reagents paclitaxel or topotecan proved to be more effective in increasing survival than treatment with cytotoxic reagent alone. CONCLUSION A single administration of AAVrh10.BevMab provides sustained and high local expression of bevacizumab in the peritoneal cavity, and significantly suppresses peritoneal carcinomatosis and increases survival in an ovarian cancer murine model.
Collapse
Affiliation(s)
- Yi Xie
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Martin J Hicks
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Malcolm A S Moore
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, United States
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, United States.
| | - Arash Rafii
- Department of Genetic Medicine, Weill Cornell Medical College-Qatar, Doha, Qatar
| |
Collapse
|
41
|
Zhang W, Zhao CG, Sun HY, Zheng WE, Chen H. Expression characteristics of KAI1 and vascular endothelial growth factor and their diagnostic value for hepatocellular carcinoma. Gut Liver 2014; 8:536-42. [PMID: 25071074 PMCID: PMC4164248 DOI: 10.5009/gnl13331] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 11/19/2013] [Accepted: 12/30/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/AIMS We tried to investigate the expression characteristics of KAI1, a suppressor of wide-spectrum tumor metastasis, and vascular endothelial growth factor (VEGF), the most common angiogenesis factor, and then to analyze their diagnostic value for hepatocellular carcinoma (HCC). METHODS The protein and mRNA expression levels of KAI1 or VEGF in HCC tissues and in self-controlled para-carcinoma tissues were analyzed by Western blot and real-time polymerase chain reaction, respectively. Serum levels of KAI1 and VEGF in the patients with HCC, benign liver disease or in healthy controls were quantitatively detected by enzyme-linked immunosorbent assay. RESULTS The expression level of KAI1 was downregulated, while the expression level of VEGF was upregulated in the tissues or serum of the patients with HCC. The expression level of serum KAI1 in HCC patients was correlated with TNM staging, intrahepatic metastasis, lymph node or peritoneal metastasis, and portal vein thrombus. In addition to the factors that were correlated with KAI1 expression, VEGF expression was also closely related to the α-fetoprotein level of the patients. The area under the receiver operating characteristic curve for the diagnosis of HCC was 0.907 for KAI1 and 0.779 for VEGF. The sensitivity of serum KAI1 levels in the diagnosis of HCC was 86.96%; the accuracy was 83.06%, while the sensitivity, the accuracy and the negative predictive value were improved to 91.86%, 84.68%, and 78.79% according to the combined detection of KAI1 and VEGF, respectively. CONCLUSIONS A combined detection of KAI1 and VEGF may greatly improve the efficiency of diagnosis and form a reliable panel of diagnostic markers for HCC.
Collapse
Affiliation(s)
- Wu Zhang
- Department of Adiotherapy, The Third Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| | | | - Hong Yu Sun
- Department of Adiotherapy, The Third Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| | - Wei E Zheng
- Department of Adiotherapy, The Third Affiliated Hospital, Wenzhou Medical College, Wenzhou, China
| | - Hua Chen
- Department of Adiotherapy, The Third Affiliated Hospital, Wenzhou, China
| |
Collapse
|
42
|
Conteduca V, Kopf B, Burgio SL, Bianchi E, Amadori D, De Giorgi U. The emerging role of anti-angiogenic therapy in ovarian cancer (review). Int J Oncol 2014; 44:1417-24. [PMID: 24626312 DOI: 10.3892/ijo.2014.2334] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 01/28/2014] [Indexed: 11/06/2022] Open
Abstract
The introduction of new therapeutic agents into clinical practice of ovarian cancer, in addition to the role of surgery and chemotherapy, has been the subject of numerous studies because this tumor remains worldwide the most lethal gynecological cancer. It is now known that angiogenesis plays a vital role for ovarian physiology, but also in ovarian carcinogenesis and so it has become the main target of ovarian cancer treatment. In this review, the most common molecular pathways of angiogenesis have been investigated leading to the identification of novel targets, including monoclonal antibodies and tyrosine kinase inhibitors. The fundamental targets of anti-angiogenic drugs are vascular endothelial growth factor receptor and its ligand, but also platelet-derived growth factor, fibroblast growth factor and angiopoietin. Moreover, improved knowledge of angiogenic process allowed the discovery of other molecules, such as semaphorins, neuropilins, clusterin, some transcriptional factors, and the identification of features, including stemness, epithelial-mesenchymal transition, downregulation of certain microRNAs, the alteration of immune system, that contribute to angiogenesis and possibly to resistance mechanisms. The following patent and literature review aim to highlight recent findings of approved and novel anti-angiogenic drugs that make the treatment of patients with ovarian cancer a rapidly growing field of oncology.
Collapse
Affiliation(s)
- Vincenza Conteduca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.) - IRCCS, Meldola (FC), Italy
| | - Barbara Kopf
- Istituto Oncologico della Svizzera Italiana (IOSI), Ospedale San Giovanni, Bellinzona, Switzerland
| | - Salvatore Luca Burgio
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.) - IRCCS, Meldola (FC), Italy
| | - Emanuela Bianchi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.) - IRCCS, Meldola (FC), Italy
| | - Dino Amadori
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.) - IRCCS, Meldola (FC), Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.) - IRCCS, Meldola (FC), Italy
| |
Collapse
|
43
|
Matias-Guiu X, Davidson B. Prognostic biomarkers in endometrial and ovarian carcinoma. Virchows Arch 2014; 464:315-31. [PMID: 24504546 DOI: 10.1007/s00428-013-1509-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 11/05/2013] [Accepted: 11/07/2013] [Indexed: 02/06/2023]
Abstract
This article reviews the main prognostic and predictive biomarkers of endometrial (EC) and ovarian carcinoma (OC). In EC, prognosis still relies on conventional pathological features such as histological type and grade, as well as myometrial or lymphovascular space invasion. Estrogen receptor, p53, Ki-67, and ploidy analysis are the most promising biomarkers among a long list of molecules that have been proposed. Also, a number of putative predictive biomarkers have been proposed in molecular targeted therapy. In OC, prognosis is predominantly dependent on disease stage at diagnosis and the extent of residual disease at primary operation. Diagnostic markers which aid in establishing histological type in OC are available. However, not a single universally accepted predictive or prognostic marker exists to date. Targeted therapy has been growingly focused at in recent years, in view of the frequent development of chemoresistance at recurrent disease. The present review emphasizes the crucial role of correct pathological classification and stringent selection criteria of the material studied as basis for any evaluation of biological markers. It further emphasizes the promise of targeted therapy in EC and OC, while simultaneously highlighting the difficulties remaining before this can become standard of care.
Collapse
Affiliation(s)
- Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, IRBLLEIDA, University of Lleida, Av. Alcalde Rovira Roure 80, 25198, Lleida, Spain,
| | | |
Collapse
|
44
|
Extracellular matrix metalloproteinase inducer (EMMPRIN) expression correlates positively with active angiogenesis and negatively with basic fibroblast growth factor expression in epithelial ovarian cancer. J Cancer Res Clin Oncol 2013; 140:361-9. [PMID: 24374756 PMCID: PMC3923122 DOI: 10.1007/s00432-013-1569-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/08/2013] [Indexed: 02/07/2023]
Abstract
Purpose The primary aim of this paper was to evaluate the expression of extracellular matrix metalloproteinase inducer (EMMPRIN) and its relationship with proangiogenic factors and microvessel density (MVD) in ovarian cancer. Methods The study group included 58 epithelial ovarian cancers (EOCs), 35 benign ovarian tumors, and 21 normal ovaries. The expression of EMMPRIN, vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (bFGF) was assessed by ELISA of tissue homogenates. Antibodies against CD105, CD31, and CD34 were used to immunohistochemically assess MVD. Results We have found significantly higher EMMPRIN expression in EOC than in benign ovarian tumors and normal ovaries. Similarly, the VEGF expression was higher in EOC than in benign ovarian tumors and normal ovaries. By contrast, bFGF expression was lower in EOC than in benign ovarian tumors and ovary samples. EMMPRIN expression in EOC was directly correlated with VEGF expression and CD105-MVD, but inversely correlated with bFGF expression. Grade 2/3 ovarian cancers had increased expression of EMMPRIN and VEGF, increased CD105-MVD, and lowered expression of bFGF compared to grade 1 ovarian cancers. Moreover, EMMPRIN expression was higher in advanced (FIGO III and IV) ovarian cancer. Conclusions The upregulation of EMMPRIN and VEGF expression is correlated with increased CD105-MVD and silenced bFGF, which suggests early and/or reactivated angiogenesis in ovarian cancer. Aggressive EOC is characterized by the following: high expression of EMMPRIN and VEGF, high CD105-MVD, and low expression of bFGF.
Collapse
|
45
|
Becker MA, Farzan T, Harrington SC, Krempski JW, Weroha SJ, Hou X, Kalli KR, Wong TW, Haluska P. Dual HER/VEGF receptor targeting inhibits in vivo ovarian cancer tumor growth. Mol Cancer Ther 2013; 12:2909-16. [PMID: 24130056 DOI: 10.1158/1535-7163.mct-13-0547] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ovarian cancer mortality ranks highest among all gynecologic cancers with growth factor pathways playing an integral role in tumorigenesis, metastatic dissemination, and therapeutic resistance. The HER and VEGF receptor (VEGFR) are both overexpressed and/or aberrantly activated in subsets of ovarian tumors. While agents targeting either the HER or VEGF pathways alone have been investigated, the impact of these agents have not led to overall survival benefit in ovarian cancer. We tested the hypothesis that cotargeting HER and VEGFR would maximize antitumor efficacy at tolerable doses. To this end, ovarian cancer xenografts grown intraperitoneally in athymic nude mice were tested in response to AC480 (pan-HER inhibitor, "HERi"), cediranib (pan-VEGFR inhibitor "VEGFRi"), or BMS-690514 (combined HER/VEGFR inhibitor "EVRi"). EVRi was superior to both HERi and VEGFRi in terms of tumor growth, final tumor weight, and progression-free survival. Correlative tumor studies employing phosphoproteomic antibody arrays revealed distinct agent-specific alterations, with EVRi inducing the greatest overall effect on growth factor signaling. These data suggest that simultaneous inhibition of HER and VEGFR may benefit select subsets of ovarian cancer tumors. To this end, we derived a novel HER/VEGF signature that correlated with poor overall survival in high-grade, late stage, serous ovarian cancer patient tumors.
Collapse
Affiliation(s)
- Marc A Becker
- Corresponding Author: Paul Haluska, Division of Medical Oncology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN 55905.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
The VEGF pathway in lung cancer. Cancer Chemother Pharmacol 2013; 72:1169-81. [DOI: 10.1007/s00280-013-2298-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 09/13/2013] [Indexed: 12/21/2022]
|
47
|
Tanase CP, Enciu AM, Mihai S, Neagu AI, Calenic B, Cruceru ML. Anti-cancer Therapies in High Grade Gliomas. CURR PROTEOMICS 2013; 10:246-260. [PMID: 24228024 PMCID: PMC3821381 DOI: 10.2174/1570164611310030007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 05/10/2013] [Accepted: 06/11/2013] [Indexed: 12/28/2022]
Abstract
High grade gliomas represent one of the most aggressive and treatment-resistant types of human cancer, with only 1–2 years median survival rate for patients with grade IV glioma. The treatment of glioblastoma is a considerable therapeutic challenge; combination therapy targeting multiple pathways is becoming a fast growing area of research. This review offers an up-to-date perspective of the literature about current molecular therapy targets in high grade glioma, that include angiogenic signals, tyrosine kinase receptors, nodal signaling proteins and cancer stem cells related approaches. Simultaneous identification of proteomic signatures could provide biomarker panels for diagnostic and personalized treatment of different subsets of glioblastoma. Personalized medicine is starting to gain importance in clinical care, already having recorded a series of successes in several types of cancer; nonetheless, in brain tumors it is still at an early stage.
Collapse
Affiliation(s)
- Cristiana Pistol Tanase
- Victor Babes National Institute of Pathology, Department of Biochemistry-Proteomics, no 99-101 Splaiul Inde-pendentei, 050096 sect 5 Bucharest, Romania
| | | | | | | | | | | |
Collapse
|
48
|
VEGF +936C/T and +460C/T gene polymorphisms and oral cancer risk: a meta-analysis. Mol Biol Rep 2013; 40:6637-43. [PMID: 24057253 DOI: 10.1007/s11033-013-2778-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 09/14/2013] [Indexed: 10/26/2022]
Abstract
Many studies have examined the association between the VEGF +936C/T (rs833061) and +460C/T (rs3025039) gene polymorphisms and oral cancer risk in various populations, but their results have been inconsistent. To assess this relationship more precisely, we performed a meta-analysis. The PubMed, Embase, Web of Science, and China National Knowledge Infrastructure databases were searched for case-control studies that were published up to January 2013. Data were extracted and pooled odds ratios (ORs) with 95 % confidence intervals (CIs) were calculated. Ultimately, six studies were included, comprising 1006 oral cancer cases and 1016 controls. Overall, the pooled OR for VEGF +936 T allele carriers (TC + TT) versus the wild-type homozygotes (CC) was 1.28 (95 % CI 1.04-1.58; P = 0.228 for heterogeneity), the pooled OR for TT versus CC was 1.64 (95 % CI 1.34-1.98; P = 0.315 for heterogeneity), and the pooled OR for the T allele versus the C allele was 1.42 (95 % CI 1.22-1.76; P = 0.286 for heterogeneity). In the stratified analysis by ethnicity, significant risks were found among Caucasians but not Asians. However, there were no associations between VEGF +460C/T and oral cancer risk in only two of the included studies. In conclusion, this meta-analysis demonstrates that the VEGF +936 T allele may be associated with an increased risk of oral cancer, especially among Caucasian populations.
Collapse
|