1
|
Gu Q, Zhao H, Zhu T, Lu Y, Lin Y, Yuan H, Lan M. Oriented assembly of hydrophilic nanochains modified by porous zirconium-based coordination polymers for glycopeptides analysis. Talanta 2024; 267:125165. [PMID: 37688895 DOI: 10.1016/j.talanta.2023.125165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Abstract
Mass spectrometry (MS)-based glycoproteomics research requires additional sample pretreatment to improve the effective identification of low-abundance glycopeptides without interference from non-glycoproteins. Herein, an attractive strategy using resorcinol-formaldehyde (RF) resin and zirconium-based coordination polymer (Zr-BCP) was established to prepare one-dimensional porous coordination polymer composites for glycopeptide enrichment before MS analysis. The obtained Fe3O4@RF@Zr-BCP nanochains feature excellent magnetic response (42.26 emu/g), high hydrophilicity (16.0°), and large specific surface area (140.84 m2/g), which provides abundant affinity sites for specific capture of glycopeptides. The materials exhibit outstanding performance in the enrichment of glycopeptides in terms of sensitivity (15 fmol/μL IgG), selectivity (1:200, molar ratio of IgG/BSA), loading capacity (200 mg/g) and recovery (106.4 ± 3.5%). In addition, the developed method based on Fe3O4@RF@Zr-BCP has been successfully applied to capture glycopeptides in tryptic digest of mouse teratoma cell extracts. It is worth emphasizing that compared with dispersed nanoparticles, the one-dimensional chain structure brings extraordinary reusability to Fe3O4@RF@Zr-BCP nanochains, which is conducive to the rapid cyclic enrichment of glycopeptides. This present work provides a potential enrichment platform for comprehensive glycoprotein analysis, and opens a new avenue for the application of oriented-assembly nanochains.
Collapse
Affiliation(s)
- Qinying Gu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Hongli Zhao
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China.
| | - Tianyi Zhu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Yichen Lu
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Yunfan Lin
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Huihui Yuan
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China; Research Center of Analysis and Test, East China University of Science and Technology, Shanghai, 200237, People's Republic of China.
| |
Collapse
|
2
|
Huang W, Chen L, Sun P. ERRα expression in ovarian cancer and promotes ovarian cancer cells migration in vitro. Arch Gynecol Obstet 2022; 305:1525-1534. [PMID: 34797420 DOI: 10.1007/s00404-021-06323-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Ovarian cancer is the leading cause of death from a gynaecological malignancy in the developed world, and is characterized by invasion and metastasis and thus causes a high fatality rate. Estrogen-related receptor alpha (ERRα) has been demonstrated to play a widespread and pathophysiological relevant role in tumourigenesis and development. The aim of this study was to investigate the effect of ERRα expression on the progression of ovarian cancer. METHODS The correlation between ERRα expression level and clinical pathological parameters in ovarian cancer tissues were analysed via cancer public database CPTAC. The expression level of ERRα in ovarian cancer cells were confirmed by RT-qPCR and Western blot methods. The cellular ERRα expression was up-regulated by lentivirus transfection and down-regulated by specific antagonist. The invasion and metastasis capabilities of ovarian cancer cells were characterized by wound healing assay and trans-well chamber assay. RESULTS The CPTAC database showed that the ERRα expression levels were higher in the late-stage and high-grade ovarian cancer tissues than in early-stage and low-grade tissues. Ovarian cancer cells with higher-expression ERRα exhibited stronger invasion and metastasis capabilities in vitro. After up-regulating the ERRα expression level, the invasion and metastasis capabilities of ovarian cancer cells were enhanced, while down-regulation weakened. Moreover, the wound sealing rate was positively correlated with the expression of ERRα mRNA expression level (r = 0.921, P < 0.01), and the cell invasiveness was also positively correlated with the cellular ERRα mRNA expression level (r = 0.926, P < 0.01). CONCLUSIONS Our results suggest that ERRα may promote the progression of ovarian cancer, and may serve as a promising predictive biomarker.
Collapse
Affiliation(s)
- Weiyi Huang
- Department of Gynecology and Obstetrics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital South Branch, Fuzhou, 350028, Fujian, People's Republic of China
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, 350001, Fujian, People's Republic of China
| | - Lili Chen
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, 350001, Fujian, People's Republic of China
- Department of Gynecology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, 18 Daoshan Road, Fuzhou, 350001, Fujian, People's Republic of China.
- Department of Gynecology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China.
| |
Collapse
|
3
|
miR-874-3p mitigates cisplatin resistance through modulating NF-κB/inhibitor of apoptosis protein signaling pathway in epithelial ovarian cancer cells. Mol Cell Biochem 2021; 477:307-317. [PMID: 34716858 DOI: 10.1007/s11010-021-04271-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/29/2021] [Indexed: 10/19/2022]
Abstract
The resistance to cisplatin, the most common platinum chemotherapy drug, may confine the efficacy of treatment in epithelial ovarian cancer patients. Aberrant expression of inhibitor of apoptosis proteins set the stage for resistance to cisplatin in EOC; besides, chemosensitivity in EOC can be chalked up to dysregulation of specific miRNAs. Herein, we investigated whether there is a potential correlation between miR-874-3p and the X-chromosome-linked inhibitor of apoptosis, a member of the IAP protein family in cisplatin-resistant EOC cells. The lower expression of miR-874-3p was found in SKOV3-DDP cells; it was also in association with cisplatin-resistance in EOC cells. XIAP was found to contribute to developing platinum resistance and is an authentic target for miR-874-3p in SKOV3-DDP cells. Consistently, restoration of miR-874-3p expression reversed cisplatin resistance in such cells by modulating XIAP and NF-κB/Survivin signaling pathway. Besides, siRNA knock down of XIAP in SKOV3-DDP cells had an anti-migratory effect like those with miR-874 overexpression. Importantly, the enforced expression of XIAP rescued SKOV3-DDP cells from the cytotoxic effects of miR-874-3p. Finally, miR-874-3p sensitized EOC cells to cisplatin-induced apoptosis, at least in part, through targeting XIAP. The cytotoxic effects of miR-874-3p can be attributed to the targeting XIAP in cisplatin-resistant EOC cells. We believe that the combination of cisplatin with miR-874-3p may make a potential strategy to reverse cisplatin resistance.
Collapse
|
4
|
Grandi G, Perrone AM, Toss A, Vitagliano A, Friso S, Facchinetti F, Cortesi L, Cascinu S, De Iaco P. The generally low sensitivity of CA 125 for FIGO stage I ovarian cancer diagnosis increases for endometrioid histotype. Minerva Med 2020; 111:133-140. [PMID: 32338842 DOI: 10.23736/s0026-4806.20.06474-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The serum marker CA 125 is still the most widely used biomarker for ovarian cancer (OC) diagnosis in gynecological and oncological setting, but its predictive role in early-stage OC is still debated. The aim of this study was to explore the value of CA 125 in distinguishing between early-stage OC and borderline ovarian tumor (BOT) and to evaluate the accuracy of CA 125 in the detection of early stage OC. METHODS A retrospective cohort study was performed at the University Hospital of Bologna (Italy) on 1296 consecutive women suffering from OC or BOT (diagnosed at histology) between 1988-2017. Patients for whom CA 125 level was determined preoperatively were included. The positive cut-off level used was >35 U/mL. RESULTS Of 910 patients, 192 (21.1%) were diagnosed with BOT and 718 (78.9%) with OC. The sensitivity of CA 125 for stage I OC was 54.4 (95% CI: 45.3-63.3) (51.5 for IA, 54.6 for IB, 58.3 for IC), but it increased to 78.0 (95% CI: 63.7-88.0) for stage II. Interestingly, in stage I OC, CA 125 presented a significantly higher sensitivity for the endometrioid histotype [72.4 (95% CI: 52.5-86.5) vs. 49.0 (95% CI: 38.6-59.4), P=0.026]. The positive likelihood ratio of CA 125 for early-stage OC compared to BOT was 1.29 (95% CI: 1.06-1.58). CONCLUSIONS Despite its limited sensitivity for early-stage OCs, CA 125 still represents a useful serum marker to early differentiate between OCs and BOTs. Its sensitivity for stage I OC increases in endometrioid histotype.
Collapse
Affiliation(s)
- Giovanni Grandi
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University Polyclinic of Modena, University of Modena and Reggio Emilia, Modena, Italy -
| | - Anna M Perrone
- Unit of Gynecologic Oncology, S. Orsola-Malpighi Polyclinic, University of Bologna, Bologna, Italy
| | - Angela Toss
- Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Amerigo Vitagliano
- Unit of Gynecology and Obstetrics, Department of Women and Children's Health, University of Padua, Padua, Italy
| | - Stefano Friso
- Unit of Gynecologic Oncology, S. Orsola-Malpighi Polyclinic, University of Bologna, Bologna, Italy
| | - Fabio Facchinetti
- Department of Medical and Surgical Sciences for Mother, Child and Adult, University Polyclinic of Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Cortesi
- Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Stefano Cascinu
- Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Pierandrea De Iaco
- Unit of Gynecologic Oncology, S. Orsola-Malpighi Polyclinic, University of Bologna, Bologna, Italy
| |
Collapse
|
5
|
Pandey D, Wasinghon P, Huang KG. Laparoscopic Management of Peritonitis Due to a Ruptured Ovarian Endometrioma with Extremely High Levels of Cancer Antigen–125 and Cancer Antigen–19-9. J Gynecol Surg 2019. [DOI: 10.1089/gyn.2018.0089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Deeksha Pandey
- Department of Obstetrics and Gynecology, Kasturba Medical College, MAHE (Manipal Academy of Higher Education), Manipal, India
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou Medical Center, Kweishan, Taoyuan, Taiwan
| | - Phornsawan Wasinghon
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou Medical Center, Kweishan, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, Buddhachinnaraj Hospital and Naresuan University, Phitsanulok, Thailand
| | - Kuan-Gen Huang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou Medical Center, Kweishan, Taoyuan, Taiwan
- Chang Gung University College of Medicine, Kweishan, Taoyuan, Taiwan
| |
Collapse
|
6
|
Li Y, Tan C, Liu L, Han L. Significance of blood and salivary IEX-1 expression in diagnosis of epithelial ovarian carcinoma. J Obstet Gynaecol Res 2018; 44:764-771. [PMID: 29431239 PMCID: PMC5900728 DOI: 10.1111/jog.13576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/08/2017] [Indexed: 01/23/2023]
Abstract
AIM This study assesses a clinical potential of immediate early responsive gene X-1 (IEX-1), also named IER3, in the diagnosis of epithelial ovarian carcinoma using blood and salivary specimens. METHODS Immediate early responsive gene X-1 was quantified in blood and saliva by real-time quantitative reverse transcription polymerase chain reaction in 26 cases of epithelial ovarian carcinoma, 37 cases of benign ovarian tumor and 55 cases of healthy women. The receiver operating characteristic (ROC) curve was used to evaluate the diagnostic efficacy of IEX-1. RESULTS Immediate early responsive gene X-1 was expressed in blood and saliva of the benign ovarian tumor group and the healthy women group, both at a level significantly higher than that of the ovarian carcinoma group (P < 0.017). There were no significant differences in IEX-1 expression in blood and saliva (P = 0.376 or 0.621, respectively) between the benign ovarian tumor and the healthy women group. Comparison of IEX-1 expression in blood between the ovarian carcinoma group and the benign ovarian tumor group or the healthy women group demonstrated the ROC-area under curves (AUC) of 0.947 or 0.929, respectively. In discriminating the ovarian carcinoma group from the benign ovarian tumor group, IEX-1 expression in blood demonstrated a sensitivity and specificity of 84.6% and 94.6%, respectively. Similarly, blood IEX-1 expression conferred a sensitivity of 84.6% and specificity of 90.9% in distinguishing the ovarian carcinoma group from the healthy women group. Moreover, salivary IEX-1 expression had ROC-AUC of 0.851 when compared between the ovarian carcinoma group and the benign ovarian tumor group or 0.896 when compared between the ovarian cancer group and the healthy women group. IEX-1 expression was able to discriminate the ovarian carcinoma group from the benign ovarian tumor group with a sensitivity and specificity of 65.4% and 94.6%, respectively, or the ovarian carcinoma from the healthy women with 92.3% sensitivity and 72.5% specificity. CONCLUSION These results suggest the clinical potential of IEX-1 expression in blood and saliva as a sensitive and specific diagnosis for epithelial ovarian carcinoma.
Collapse
Affiliation(s)
- Yuan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chaoyue Tan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liya Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liping Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Denel-Bobrowska M, Łukawska M, Bukowska B, Gajek A, Oszczapowicz I, Marczak A. Molecular mechanism of action of oxazolinoanthracyclines in cells derived from human solid tumors. Part 2. Toxicol In Vitro 2018; 46:323-334. [DOI: 10.1016/j.tiv.2017.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 12/15/2022]
|
8
|
Li Y, Xiao M, Guo F. The role of Sox6 and Netrin-1 in ovarian cancer cell growth, invasiveness, and angiogenesis. Tumour Biol 2017; 39:1010428317705508. [PMID: 28475012 DOI: 10.1177/1010428317705508] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
SOX6 plays important roles in cell proliferation, differentiation, and cell fate determination. It has been confirmed that SOX6 is a tumor suppressor and downregulated in various cancers, including esophageal squamous cell carcinoma, hepatocellular carcinoma, and chronic myeloid leukemia. Netrin-1 is highly expressed in various human cancers and acts as an anti-apoptotic and proangiogenic factor to drive tumorigenesis. The role of SOX6 and netrin-1 in regulating the growth of ovarian tumor cells still remains unclear. Real-time polymerase chain reaction and western blot were used to determine the SOX6 messenger RNA and protein levels, respectively, in ovarian cancer cells and tumor tissues. Stable transfection of SOX6 was conducted to overexpress SOX6 in PA-1 and SW626 cells. Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Invasion of ovarian cancer cells and migration of human umbilical vein endothelial cells were confirmed by Transwell assays. To overexpress netrin-1, ovarian cancer cells with SOX6 restoration was transduced with netrin-1 lentiviral particles. PA-1 xenografts in a nude mice model were used to conduct in vivo evaluation of the role of SOX6 and its relationship with netrin-1 in tumor growth and angiogenesis. In this study, we found significantly reduced SOX6 levels in PA-1, SW626, SK-OV-3, and CaoV-3 ovarian cancer cell lines and human tumor tissues in comparison with normal human ovarian epithelial cells or matched non-tumor tissues. SOX6 overexpression by stable transfection dramatically inhibited proliferation and invasion of PA-1 and SW626 cells. Also, conditioned medium from PA-1 and SW626 cells with SOX6 restoration exhibited reduced ability to induce human umbilical vein endothelial cells migration and tube formation compared with conditioned medium from the cells with transfection control. Furthermore, an inverse relationship between SOX6 and netrin-1 expression was observed in PA-1 and SW626 cells. Overexpression of netrin-1 in ovarian cancer cells with forced SOX6 expression remarkably abrogated the inhibitory effect of SOX6 on proliferation, invasion of the cells, and tumor xenograft growth and vascularity in vivo. Human umbilical vein endothelial cell migration and tube formation were enhanced in the conditioned medium from the ovarian cancer cells transduced with netrin-1 lentivirus particles. Our observations revealed that SOX6 is a tumor suppressor in ovarian cancer cells, and SOX6 exerts an inhibitory effect on the proliferation, invasion, and tumor cell-induced angiogenesis of ovarian cancer cells, whereas nerin-1 plays an opposite role and its expression is inversely correlated with SOX6. Moreover, our findings suggest a new role of SOX6 and netrin-1 for understanding the progression of ovarian cancer and have the potential for the development of new diagnosis and treatment strategies for ovarian cancer.
Collapse
Affiliation(s)
- Yi Li
- Department of Gynecology, The Second People’s Hospital of Liaocheng City, Linqing, P.R. China
| | - Ming Xiao
- Department of Cardiology, The Second People’s Hospital of Liaocheng City, Linqing, P.R. China
| | - Fangchun Guo
- Department of Medical Ultrasound, The People’s Hospital of Linzi District, Zibo, P.R. China
| |
Collapse
|
9
|
Büyüktiryaki S, Say R, Denizli A, Ersöz A. Phosphoserine imprinted nanosensor for detection of Cancer Antigen 125. Talanta 2017; 167:172-180. [DOI: 10.1016/j.talanta.2017.01.093] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/29/2017] [Accepted: 01/31/2017] [Indexed: 11/15/2022]
|
10
|
Jia H, Zhang Q, Liu F, Zhou D. Prognostic value of MMP-2 for patients with ovarian epithelial carcinoma: a systematic review and meta-analysis. Arch Gynecol Obstet 2017; 295:689-696. [PMID: 27995372 DOI: 10.1007/s00404-016-4257-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/25/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND The reported roles of matrix metalloproteinase 2 (MMP-2) on the prognosis of patients with epithelial ovarian cancers (EOCs) are inconsistent. OBJECTIVE This meta-analysis was performed to evaluate the prognostic significance of MMP-2 for patients with EOCs by analyzing 11 studies. METHODS We systematically searched articles in the Cochrane Library, Pubmed, Embase, China National Knowledge Infrastructure (CNKI), Wanfang Database, and Chinese Biological Medical (CBM) Database, updated to February 1st 2015, with the following search terms: ovarian neoplasm OR ovarian tumor OR ovarian carcinoma OR ovarian malignance OR ovarian cancer AND matrix metalloproteinase-2 OR MMP-2. RESULTS A total of 11 studies involving 1058 patients with EOCs were in accordance with the inclusion criteria. The pooled HR was 1.09 (95% CI 0.32-1.86, p = 0.006) in patients with overexpression of stromal MMP-2 with significant heterogeneity (I 2 = 53.1%, p = 0.074) between studies. For patients with MMP-2 overexpression in tumor cells, the pooled HR was 1.42 (95% CI 1.14-1.70, p = 0.000) with no significant heterogeneity (I 2 = 43.4%, p = 0.078) between studies. Sensitivity analyses were stable. CONCLUSIONS MMP-2 overexpression in tumor cells rather than stroma was significantly associated with poor prognosis in patient with endothelial ovarian cancer; however, the result remains to be confirmed with additional high-quality studies.
Collapse
Affiliation(s)
- Honglei Jia
- Shandong Provincial Hospital Affiliated to Shandong University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021, Shandong, China
| | - Qingyu Zhang
- Qilu Hospital Affiliated to Shandong University, Jinan, 250012, Shandong, China
| | - Fanxiao Liu
- Shandong Provincial Hospital Affiliated to Shandong University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021, Shandong, China
| | - Dongsheng Zhou
- Shandong Provincial Hospital Affiliated to Shandong University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021, Shandong, China.
| |
Collapse
|
11
|
Identification of the key pathway of oxazolinoanthracyclines mechanism of action in cells derived from human solid tumors. Toxicol Appl Pharmacol 2016; 313:159-169. [DOI: 10.1016/j.taap.2016.10.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/08/2016] [Accepted: 10/19/2016] [Indexed: 01/23/2023]
|
12
|
Nordin N, Fadaeinasab M, Mohan S, Mohd Hashim N, Othman R, Karimian H, Iman V, Ramli N, Mohd Ali H, Abdul Majid N. Pulchrin A, a New Natural Coumarin Derivative of Enicosanthellum pulchrum, Induces Apoptosis in Ovarian Cancer Cells via Intrinsic Pathway. PLoS One 2016; 11:e0154023. [PMID: 27136097 PMCID: PMC4852948 DOI: 10.1371/journal.pone.0154023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 04/07/2016] [Indexed: 12/27/2022] Open
Abstract
Drug resistance presents a challenge in chemotherapy and has attracted research interest worldwide and particular attention has been given to natural compounds to overcome this difficulty. Pulchrin A, a new compound isolated from natural products has demonstrated novel potential for development as a drug. The identification of pulchrin A was conducted using several spectroscopic techniques such as nuclear magnetic resonance, liquid chromatography mass spectrometer, infrared and ultraviolet spectrometry. The cytotoxicity effects on CAOV-3 cells indicates that pulchrin A is more active than cisplatin, which has an IC50 of 22.3 μM. Significant changes in cell morphology were present, such as cell membrane blebbing and formation of apoptotic bodies. The involvement of phosphatidylserine (PS) in apoptosis was confirmed by Annexin V-FITC after a 24 h treatment. Apoptosis was activated through the intrinsic pathway by activation of procaspases 3 and 9 as well as cleaved caspases 3 and 9 and ended at the executioner pathway, with the occurrence of DNA laddering. Apoptosis was further confirmed via gene and protein expression levels, in which Bcl-2 protein was down-regulated and Bax protein was up-regulated. Furthermore, the CAOV-3 cell cycle was disrupted at the G0/G1 phase, leading to apoptosis. Molecular modeling of Bcl-2 proteins demonstrated a high- binding affinity, which inhibited the function of Bcl-2 proteins and led to cell death. Results of the current study can shed light on the development of new therapeutic agents, particularly, human ovarian cancer treatments.
Collapse
Affiliation(s)
- Noraziah Nordin
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mehran Fadaeinasab
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Syam Mohan
- Medical Research Center, Jazan University, Jazan, Kingdom of Saudi Arabia
| | - Najihah Mohd Hashim
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Rozana Othman
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hamed Karimian
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Venus Iman
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Noorlela Ramli
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hapipah Mohd Ali
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Nazia Abdul Majid
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
- * E-mail:
| |
Collapse
|
13
|
Yung MMH, Ngan HYS, Chan DW. Targeting AMPK signaling in combating ovarian cancers: opportunities and challenges. Acta Biochim Biophys Sin (Shanghai) 2016; 48:301-17. [PMID: 26764240 PMCID: PMC4886241 DOI: 10.1093/abbs/gmv128] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/29/2015] [Indexed: 12/25/2022] Open
Abstract
The development and strategic application of effective anticancer therapies have turned out to be one of the most critical approaches of managing human cancers. Nevertheless, drug resistance is the major obstacle for clinical management of these diseases especially ovarian cancer. In the past years, substantial studies have been carried out with the aim of exploring alternative therapeutic approaches to enhance efficacy of current chemotherapeutic regimes and reduce the side effects caused in order to produce significant advantages in overall survival and to improve patients' quality of life. Targeting cancer cell metabolism by the application of AMP-activated protein kinase (AMPK)-activating agents is believed to be one of the most plausible attempts. AMPK activators such as 5-aminoimidazole-4-carboxamide 1-β-d-ribofuranoside, A23187, metformin, and bitter melon extract not only prevent cancer progression and metastasis but can also be applied as a supplement to enhance the efficacy of cisplatin-based chemotherapy in human cancers such as ovarian cancer. However, because of the undesirable outcomes along with the frequent toxic side effects of most pharmaceutical AMPK activators that have been utilized in clinical trials, attentions of current studies have been aimed at the identification of replaceable reagents from nutraceuticals or traditional medicines. However, the underlying molecular mechanisms of many nutraceuticals in anticancer still remain obscure. Therefore, better understanding of the functional characterization and regulatory mechanism of natural AMPK activators would help pharmaceutical development in opening an area to intervene ovarian cancer and other human cancers.
Collapse
Affiliation(s)
- Mingo M H Yung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hextan Y S Ngan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - David W Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
14
|
Vici P, Pizzuti L, Di Lauro L, Conti L, Mandoj C, Antenucci A, Digiesi G, Sergi D, Amodio A, Marchetti P, Sperati F, Valle M, Garofalo A, Vizza E, Corrado G, Vincenzoni C, Tomao F, Kayal R, Marsella A, Carosi M, Antoniani B, Giordano A, Maugeri-Saccà M, Barba M. Metabolic Determinants and Anthropometric Indicators Impact Clinical-pathological Features in Epithelial Ovarian Cancer Patients. J Cancer 2016; 7:516-22. [PMID: 26958087 PMCID: PMC4780127 DOI: 10.7150/jca.13578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/08/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Over the last twenty years, the efforts of the scientific community devoted to the comprehension and treatment of ovarian cancer have remained poorly remunerative, with the case-fatality ratio of this disease remaining disappointedly high. Limited knowledge of the basic principles regulating ovarian carcinogenesis and factors impacting the course of disease may significantly impair our ability to intervene in early stages and lessen our expectations in terms of treatment outcomes. In the present study, we sought to assess whether metabolic factors and anthropometric indicators, i.e., pre-treatment fasting glucose and body mass index, are associated with renown cancer related prognostic factors such as tumour stage and grade at diagnosis. MATERIALS AND METHODS Study participants were 147 women diagnosed with epithelial ovarian cancer and treated with platinum based regimens and/or surgery at the Regina Elena National Cancer Institute of Rome, Italy. Glucose levels were assessed at the institutional laboratories on venous blood collected in overnight fasting conditions and prior to any therapeutic procedure. Stage was coded according to the FIGO staging system based on the results of the diagnostic workup, while tumour grade was locally assessed by an expert pathologist. Participants' characteristics were descriptively analyzed for the overall study population and in a subgroup of 70 patients for whom data on body mass index (BMI) were available. FIGO stage and grade were compared by categories of pre-treatment fasting glucose defined upon the median value, i.e., 89 mg/dl. The association of interest was tested in regression models including BMI. RESULTS For the overall study population, patients in the lowest category of fasting glucose were significantly more likely to exhibit a FIGO stage III-IV at diagnosis compared with their counterpart in the highest glucose category (81.3 vs 66.7%, p: 0.021). Subgroup analysis in 70 patients with BMI data confirmed this association (81.5 vs 55.8, p: 0.049), which remained significant when tested in regression models including BMI (OR: 0.28 95% CI 0.086-0.89, p: 0.031). No relevant evidence emerged when testing the association between fasting glucose and tumour grade. CONCLUSIONS In patients diagnosed with epithelial ovarian cancer, pre-treatment glucose levels appear to be inversely associated with FIGO stage. Further studies are warranted to eventually confirm and correctly interpret the implications of this novel finding.
Collapse
Affiliation(s)
- Patrizia Vici
- 1. Division of Medical Oncology 2, Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Pizzuti
- 1. Division of Medical Oncology 2, Regina Elena National Cancer Institute, Rome, Italy
| | - Luigi Di Lauro
- 1. Division of Medical Oncology 2, Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Conti
- 2. Division of Clinical Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Chiara Mandoj
- 2. Division of Clinical Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Antenucci
- 2. Division of Clinical Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanna Digiesi
- 2. Division of Clinical Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Domenico Sergi
- 1. Division of Medical Oncology 2, Regina Elena National Cancer Institute, Rome, Italy
| | - Antonella Amodio
- 1. Division of Medical Oncology 2, Regina Elena National Cancer Institute, Rome, Italy
| | - Paolo Marchetti
- 3. Oncology Unit, Sant'Andrea Hospital, La Sapienza University of Rome, Italy
| | - Francesca Sperati
- 4. Biostatistics Unit-Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy
| | - Mario Valle
- 5. General Surgery, Regina Elena National Institute, Rome, Italy
| | - Alfredo Garofalo
- 5. General Surgery, Regina Elena National Institute, Rome, Italy
| | - Enrico Vizza
- 6. Gynecological Oncology, Regina Elena National Cancer Institute, Rome, Italy
| | - Giacomo Corrado
- 6. Gynecological Oncology, Regina Elena National Cancer Institute, Rome, Italy
| | - Cristina Vincenzoni
- 6. Gynecological Oncology, Regina Elena National Cancer Institute, Rome, Italy
| | - Federica Tomao
- 7. Department of Gynecologic Oncology, University “Sapienza”, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ramy Kayal
- 8. Department of Radiology, Regina Elena National Cancer Institute, Rome, Italy
| | - Annalise Marsella
- 8. Department of Radiology, Regina Elena National Cancer Institute, Rome, Italy
| | - Mariantonia Carosi
- 9. Department of Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Barbara Antoniani
- 9. Department of Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Antonio Giordano
- 10. Sbarro Institute for Cancer Research and Molecular Medicine e del Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
- 11. Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Marcello Maugeri-Saccà
- 1. Division of Medical Oncology 2, Regina Elena National Cancer Institute, Rome, Italy
- 12. Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy
| | - Maddalena Barba
- 1. Division of Medical Oncology 2, Regina Elena National Cancer Institute, Rome, Italy
- 12. Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
15
|
Abstract
Ovarian cancer is the most fatal of all gynecologic malignancies. Despite the lack of a recommended screening test for ovarian cancer, NPs can identify risk factors, ensure patients are aware of subtle symptoms, and provide adequate testing and analysis of results.
Collapse
|
16
|
Cao L, Zhang Y, Fu Z, Dong L, Yang S, Meng W, Li Y, Zhang W, Zhang J, Zheng C, Zhu H, Fan L. Diagnostic value of plasma lysophosphatidic acid levels in ovarian cancer patients: A case-control study and updated meta-analysis. J Obstet Gynaecol Res 2015; 41:1951-8. [PMID: 26472266 DOI: 10.1111/jog.12806] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 06/01/2015] [Indexed: 01/01/2023]
Abstract
AIM We investigated whether plasma levels of lysophosphatidic acid (LPA) could serve as a diagnostic indicator for assessing disease progression in ovarian cancer (OC) patients. MATERIAL AND METHODS In this study, we enrolled 98 patients with OC, 70 patients with benign ovarian tumors and 75 healthy controls. Plasma levels of LPA and cancer antigen 125 (CA-125) were measured in all study subjects. The diagnostic values of LPA and CA-125 plasma levels were evaluated and an updated meta-analysis was performed to examine the association between LPA plasma levels and OC progression. Statistical analyses were performed with SPSS 18.0 and R 3.1.0 software. RESULTS In our case-control study, OC patients showed significantly higher plasma LPA levels compared to patients with benign tumors and healthy controls (all P < 0.05). Plasma LPA levels exhibited higher diagnostic sensitivity (P = 0.008) and specificity (P = 0.042) in detecting OC, compared to an established marker, CA-125. Notably, the sensitivity for early stage OC was significantly higher for plasma LPA levels in comparison to CA-125 (P < 0.05). Consistent with this, the area under the receiver-operator curve was greater for LPA (0.899) in comparison to that for CA-125 (0.751). Further, meta-analysis showed that plasma LPA levels were significantly higher in OC patients compared to patients with benign tumors or healthy controls (all P < 0.05). CONCLUSION Plasma LPA levels are elevated in OC patients and correlate with disease progression. Further, LPA shows higher sensitivity and specificity in OC diagnosis, compared to CA-125, especially in early stage OC.
Collapse
Affiliation(s)
- Liyan Cao
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Yunjie Zhang
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Zhanzhao Fu
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Lixin Dong
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Sen Yang
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Wei Meng
- Department of Oncology, The First Affiliated Hospital, Hebei North University, Zhangjiakou, China
| | - Yiyang Li
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Wenchao Zhang
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Jialing Zhang
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Changjun Zheng
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - He Zhu
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| | - Limei Fan
- Radiation Oncology Division, The First Hospital of Qin Huang Dao City, Qin Huang Dao, China
| |
Collapse
|
17
|
Ma LS, Yan QI, Huang Y, Zhao W, Zhu YU. Downregulation of human epidermal growth factor receptor 2 by short hairpin RNA increases chemosensitivity of human ovarian cancer cells. Oncol Lett 2015; 9:2211-2217. [PMID: 26137043 DOI: 10.3892/ol.2015.3033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 01/16/2015] [Indexed: 02/07/2023] Open
Abstract
The aim of the current study was to investigate the suppressive effects of pSilencer T7-human epidermal growth factor receptor 2 (HER2)-short hairpin RNA (shRNA) recombinant plasmids on human SKOV3 ovarian cancer cell growth and sensitivity to carboplatin (CBP). Three different pairs of shRNAs (shRNAa, shRNAb and shRNAc), targeting the HER2 gene, were selected and transfected into human SKOV3 cells, respectively. The expression levels of HER2 were then detected by immunohistochemical (IHC), semi-quantitative reverse transcription-polymerase chain reaction and western blot analyses. In addition, cell cycle and cell growth were investigated using cell counting kit-8 (CCK-8). The results of the IHC and western blot analyses revealed that shRNAb significantly inhibited HER2 protein expression in SKOV3 cells. shRNAb exhibited an improved effect on HER2 expression compared with shRNAa (P<0.01), while shRNAc did not affect HER2 expression. Nontransfected and nonspecific shRNA groups were used as the negative controls. Knockdown of HER2 expression by shRNA was initiated at 24 h following transfection, achieving an optimum effect at 48 h and lasting for at least 72 h after the treatment. The CCK-8 cell growth assay indicated that the knockdown of HER2 expression in the SKOV3 cell line resulted in significant growth suppression and cell cycle arrest. In addition, inhibition of HER2 significantly increased SKOV3 cell sensitivity to CBP treatment. In conclusion, pSilencer T7-HER2-shRNA significantly inhibited HER2 expression in human ovarian cancer cells in vitro and induced chemotherapeutic sensitivity to CBP.
Collapse
Affiliation(s)
- Li Shan Ma
- Department of Obstetrics and Gynecology, Yangpu Hospital Affiliated to Tongji University, Shanghai 200090, P.R. China
| | - Q I Yan
- Department of Obstetrics and Gynecology, Jiangwan Hospital, Shanghai 200434, P.R. China
| | - Yongfang Huang
- Department of Obstetrics and Gynecology, Jiangwan Hospital, Shanghai 200434, P.R. China
| | - Wenxia Zhao
- Department of Obstetrics and Gynecology, Jiangwan Hospital, Shanghai 200434, P.R. China
| | - Y U Zhu
- Department of Obstetrics and Gynecology, Jiangwan Hospital, Shanghai 200434, P.R. China
| |
Collapse
|
18
|
The clinical value of dynamic contrast-enhanced MRI in differential diagnosis of malignant and benign ovarian lesions. Tumour Biol 2015; 36:5515-22. [DOI: 10.1007/s13277-015-3219-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/03/2015] [Indexed: 10/23/2022] Open
|
19
|
Leng R, Liao G, Wang H, Kuang J, Tang L. Rac1 expression in epithelial ovarian cancer: effect on cell EMT and clinical outcome. Med Oncol 2015; 32:329. [PMID: 25585684 DOI: 10.1007/s12032-014-0329-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/05/2014] [Indexed: 02/03/2023]
Abstract
Ras-related C3 botulinum toxin substrate 1 (rac1) has been implicated in tumor epithelial-mesenchymal transition (EMT); however, limited information is available regarding the role of rac1 in epithelial ovarian cancer (EOC). This study aimed to evaluate the correlation of rac1 expression with EMT and EOC prognosis. Rac1 protein levels of 150 EOC specimens were evaluated by immunohistochemical staining. Survival analysis was performed to determine the correlation between rac1 expression and survival. Cellular and molecular changes were also examined after rac1 in ovarian cancer cells was silenced in vitro and in vivo. The mechanism of rac1 on EMT was investigated by Western blot analysis. Rac1 was highly expressed in EOC. Rac1 overexpression was closely associated with advanced stage based on International Federation of Gynecology and Obstetrics, poor grade, serum Ca-125, and residual tumor size. Survival analyses demonstrated that patients with high rac1 expression levels were more susceptible to early tumor recurrence with very poor prognosis. This study revealed that rac1 downregulation decreased cell EMT and proliferation capability in vitro and in vivo. Rac1 expression possibly altered cell EMT by interacting with p21-activated kinase 1 and p38 mitogen-activated protein kinase signaling pathways. The present study showed that rac1 overexpression is associated with cell EMT and poor EOC prognosis. Rac1 possibly plays an important role in predicting EOC metastasis.
Collapse
Affiliation(s)
- Ruobing Leng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | | | | | | | | |
Collapse
|
20
|
Kelemen LE, Terry KL, Goodman MT, Webb PM, Bandera EV, McGuire V, Rossing MA, Wang Q, Dicks E, Tyrer JP, Song H, Kupryjanczyk J, Dansonka-Mieszkowska A, Plisiecka-Halasa J, Timorek A, Menon U, Gentry-Maharaj A, Gayther SA, Ramus SJ, Narod SA, Risch HA, McLaughlin JR, Siddiqui N, Glasspool R, Paul J, Carty K, Gronwald J, Lubiński J, Jakubowska A, Cybulski C, Kiemeney LA, Massuger LFAG, van Altena AM, Aben KKH, Olson SH, Orlow I, Cramer DW, Levine DA, Bisogna M, Giles GG, Southey MC, Bruinsma F, Kjær SK, Høgdall E, Jensen A, Høgdall CK, Lundvall L, Engelholm SA, Heitz F, du Bois A, Harter P, Schwaab I, Butzow R, Nevanlinna H, Pelttari LM, Leminen A, Thompson PJ, Lurie G, Wilkens LR, Lambrechts D, Van Nieuwenhuysen E, Lambrechts S, Vergote I, Beesley J, Fasching PA, Beckmann MW, Hein A, Ekici AB, Doherty JA, Wu AH, Pearce CL, Pike MC, Stram D, Chang-Claude J, Rudolph A, Dörk T, Dürst M, Hillemanns P, Runnebaum IB, Bogdanova N, Antonenkova N, Odunsi K, Edwards RP, Kelley JL, Modugno F, Ness RB, Karlan BY, Walsh C, Lester J, Orsulic S, Fridley BL, Vierkant RA, Cunningham JM, Wu X, Lu K, Liang D, Hildebrandt MA, Weber RP, Iversen ES, Tworoger SS, Poole EM, Salvesen HB, Krakstad C, Bjorge L, Tangen IL, Pejovic T, Bean Y, Kellar M, Wentzensen N, Brinton LA, Lissowska J, Garcia-Closas M, Campbell IG, Eccles D, Whittemore AS, Sieh W, Rothstein JH, Anton-Culver H, Ziogas A, Phelan CM, Moysich KB, Goode EL, Schildkraut JM, Berchuck A, Pharoah PD, Sellers TA, Brooks-Wilson A, Cook LS, Le ND. Consortium analysis of gene and gene-folate interactions in purine and pyrimidine metabolism pathways with ovarian carcinoma risk. Mol Nutr Food Res 2014; 58:2023-35. [PMID: 25066213 PMCID: PMC4197821 DOI: 10.1002/mnfr.201400068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 04/29/2014] [Accepted: 06/01/2014] [Indexed: 11/07/2022]
Abstract
SCOPE We reevaluated previously reported associations between variants in pathways of one-carbon (1-C) (folate) transfer genes and ovarian carcinoma (OC) risk, and in related pathways of purine and pyrimidine metabolism, and assessed interactions with folate intake. METHODS AND RESULTS Odds ratios (OR) for 446 genetic variants were estimated among 13,410 OC cases and 22,635 controls, and among 2281 cases and 3444 controls with folate information. Following multiple testing correction, the most significant main effect associations were for dihydropyrimidine dehydrogenase (DPYD) variants rs11587873 (OR = 0.92; p = 6 × 10⁻⁵) and rs828054 (OR = 1.06; p = 1 × 10⁻⁴). Thirteen variants in the pyrimidine metabolism genes, DPYD, DPYS, PPAT, and TYMS, also interacted significantly with folate in a multivariant analysis (corrected p = 9.9 × 10⁻⁶) but collectively explained only 0.2% of OC risk. Although no other associations were significant after multiple testing correction, variants in SHMT1 in 1-C transfer, previously reported with OC, suggested lower risk at higher folate (p(interaction) = 0.03-0.006). CONCLUSION Variation in pyrimidine metabolism genes, particularly DPYD, which was previously reported to be associated with OC, may influence risk; however, stratification by folate intake is unlikely to modify disease risk appreciably in these women. SHMT1 SNP-by-folate interactions are plausible but require further validation. Polymorphisms in selected genes in purine metabolism were not associated with OC.
Collapse
Affiliation(s)
- Linda E. Kelemen
- Department of Population Health Research, Alberta Health Services-Cancer Care, Calgary, AB, Canada
- Departments of Medical Genetics and Oncology, University of Calgary, Calgary, AB, Canada
| | - Kathryn L. Terry
- Obstetrics and Gynecology Epidemology Center, Brigham and Women's Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Marc T. Goodman
- Cancer Prevention and Control, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Community and Population Health Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Penelope M. Webb
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Elisa V. Bandera
- Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, The State University of New Jersey, New Brunswick, NJ, USA
| | - Valerie McGuire
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Qinggang Wang
- Department of Population Health Research, Alberta Health Services-Cancer Care, Calgary, AB, Canada
| | - Ed Dicks
- Department of Oncology, University of Cambridge, Cambridge, UK
| | | | - Honglin Song
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jolanta Kupryjanczyk
- Department of Pathology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | | | - Joanna Plisiecka-Halasa
- Department of Pathology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Agnieszka Timorek
- Department of Obstetrics, Gynecology and Oncology, IInd Faculty of Medicine, Warsaw Medical University and Brodnowski Hospital, Warsaw, Poland
| | - Usha Menon
- Gynaecological Cancer Research Centre, Department of Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Aleksandra Gentry-Maharaj
- Gynaecological Cancer Research Centre, Department of Women's Cancer, Institute for Women's Health, University College London, London, UK
| | - Simon A. Gayther
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Susan J. Ramus
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Steven A. Narod
- Women's College Research Institute, University of Toronto, Toronto, ON, Canada
| | - Harvey A. Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - John R. McLaughlin
- Prosserman Centrre for Health Research at the Samuel Lunenfeld Research Institute, Toronto, ON, Canada
| | - Nadeem Siddiqui
- Department of Gynaecological Oncology, Glasgow Royal Infirmary, Glasgow, UK
| | | | - James Paul
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Karen Carty
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Jacek Gronwald
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Jan Lubiński
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Anna Jakubowska
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Lambertus A. Kiemeney
- Department for Health Evidence, Radboud university medical center, Nijmegen, Netherlands
- Department of Urology, Radboud university medical center, Nijmegen, Netherlands
- Comprehensive Cancer Center, The Netherlands, Utrecht, Netherlands
| | | | - Anne M. van Altena
- Department of Gynaecology, Radboud university medical center, Nijmegen, Netherlands
| | - Katja K. H. Aben
- Department for Health Evidence, Radboud university medical center, Nijmegen, Netherlands
- Comprehensive Cancer Center, The Netherlands, Utrecht, Netherlands
| | - Sara H. Olson
- Memorial Sloan-Kettering Cancer Center, Department of Epidemiology and Biostatistics, New York, NY, USA
| | - Irene Orlow
- Memorial Sloan-Kettering Cancer Center, Department of Epidemiology and Biostatistics, New York, NY, USA
| | - Daniel W. Cramer
- Obstetrics and Gynecology Epidemology Center, Brigham and Women's Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
| | - Douglas A. Levine
- Gynecology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Maria Bisogna
- Gynecology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Graham G. Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, University of Melbourne, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Melissa C. Southey
- Department of Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Fiona Bruinsma
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Susanne Krüger Kjær
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
- The Juliane Marie Centre, Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Estrid Høgdall
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Allan Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Claus K. Høgdall
- The Juliane Marie Centre, Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lene Lundvall
- The Juliane Marie Centre, Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Svend-Aage Engelholm
- Department of Radiation Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Florian Heitz
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte/ Evang. Huyssens-Stiftung/ Knappschaft GmbH, Essen, Germany
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | - Andreas du Bois
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte/ Evang. Huyssens-Stiftung/ Knappschaft GmbH, Essen, Germany
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | - Philipp Harter
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte/ Evang. Huyssens-Stiftung/ Knappschaft GmbH, Essen, Germany
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | | | - Ralf Butzow
- Department of Pathology, Helsinki University Central Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Liisa M. Pelttari
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Arto Leminen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Pamela J. Thompson
- Cancer Prevention and Control, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Community and Population Health Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Galina Lurie
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA
| | - Lynne R. Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA
| | - Diether Lambrechts
- Vesalius Research Center, VIB, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Oncology, University of Leuven, Belgium
| | - Els Van Nieuwenhuysen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Sandrina Lambrechts
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Ignace Vergote
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology and Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Jonathan Beesley
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - AOCS Study Group/ACS Investigators
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Matthias W. Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Alexander Hein
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Arif B. Ekici
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Jennifer A. Doherty
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Community and Family Medicine, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Anna H. Wu
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Celeste L. Pearce
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Malcolm C. Pike
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Memorial Sloan-Kettering Cancer Center, Department of Epidemiology and Biostatistics, New York, NY, USA
| | - Daniel Stram
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Rudolph
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Matthias Dürst
- Department of Gynecology, Jena University Hospital-Friedrich Schiller University, Jena, Germany
| | - Peter Hillemanns
- Clinics of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany
| | - Ingo B. Runnebaum
- Department of Gynecology, Jena University Hospital-Friedrich Schiller University, Jena, Germany
| | - Natalia Bogdanova
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Natalia Antonenkova
- Byelorussian Institute for Oncology and Medical Radiology Aleksandrov N.N., Minsk, Belarus
| | - Kunle Odunsi
- Department of Gynecological Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Robert P. Edwards
- Department of Obstetrics, Gynecology and Reproductive Sciences and Ovarian Cancer Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph L. Kelley
- Department of Obstetrics, Gynecology and Reproductive Sciences and Ovarian Cancer Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francesmary Modugno
- Department of Obstetrics, Gynecology and Reproductive Sciences and Ovarian Cancer Center of Excellence, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health and Womens Cancer Research Program, Magee-Womens Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Roberta B. Ness
- The University of Texas School of Public Health, Houston, TX, USA
| | - Beth Y. Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christine Walsh
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sandra Orsulic
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Brooke L. Fridley
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Robert A. Vierkant
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Julie M. Cunningham
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, Mayo Clinic, Rochester, MN, USA
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karen Lu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dong Liang
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | | | - Rachel Palmieri Weber
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - Edwin S. Iversen
- Duke Cancer Institute, Durham, NC, USA
- Department of Statistical Science, Duke University, Durham, NC, USA
| | - Shelley S. Tworoger
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth M. Poole
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Helga B. Salvesen
- Department of Gynecology and Obstetrics, Haukeland University Horpital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Camilla Krakstad
- Department of Gynecology and Obstetrics, Haukeland University Horpital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Line Bjorge
- Department of Gynecology and Obstetrics, Haukeland University Horpital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ingvild L. Tangen
- Department of Gynecology and Obstetrics, Haukeland University Horpital, Bergen, Norway
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tanja Pejovic
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Yukie Bean
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Melissa Kellar
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Louise A. Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Montserrat Garcia-Closas
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, UK and Breakthrough Breast Cancer Research Centre, London, UK
| | - Ian G. Campbell
- Department of Pathology, University of Melbourne, Melbourne, VIC, Australia
- Cancer Genetics Laboratory, Research Division, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, VIC, Australia
| | - Diana Eccles
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - Alice S. Whittemore
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Weiva Sieh
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph H. Rothstein
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California Irvine, Irvine, CA, USA
- Genetic Epidemiology Research Institute, UCI Center for Cancer Genetics Research & Prevention, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Argyrios Ziogas
- Department of Epidemiology, University of California Irvine, Irvine, CA, USA
| | - Catherine M. Phelan
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kirsten B. Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Ellen L. Goode
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Joellen M. Schildkraut
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute, Durham, NC, USA
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Paul D.P. Pharoah
- Department of Oncology, University of Cambridge, Cambridge, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Thomas A. Sellers
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Angela Brooks-Wilson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, BC, Canada and Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC Canada
| | - Linda S. Cook
- Department of Population Health Research, Alberta Health Services-Cancer Care, Calgary, AB, Canada
- Division of Epidemiology and Biostatistics, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Nhu D. Le
- Cancer Control Research, BC Cancer Agency, Vancouver, BC, Canada
| | | |
Collapse
|
21
|
Haslene-Hox H, Madani A, Berg KCG, Woie K, Salvesen HB, Wiig H, Tenstad O. Quantification of the concentration gradient of biomarkers between ovarian carcinoma interstitial fluid and blood. BBA CLINICAL 2014; 2:18-23. [PMID: 26673827 PMCID: PMC4633919 DOI: 10.1016/j.bbacli.2014.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 12/25/2022]
Abstract
Background Tumor interstitial fluid (TIF) rather than plasma should be used in cancer biomarker discovery because of the anticipated higher concentration of locally produced proteins in the tumor microenvironment. Nevertheless, the actual TIF-to-plasma gradient of tumor specific proteins has not been quantified. We present the proof-of-concept for the quantification of the postulated gradient between TIF and plasma. Methods TIF was collected by centrifugation from serous (n = 19), endometrioid (n = 9) and clear cell (n = 3) ovarian carcinomas with early (n = 15) and late stage (n = 16) disease in grades 1 (n = 2), 2 (n = 8) and 3 (n = 17), and ELISA was used for the determination of CA-125, osteopontin and VEGF-A. Results All three markers were significantly up-regulated in TIF compared with plasma (p < 0.0001). The TIF-to-plasma ratio of the ovarian cancer biomarker CA-125 ranged from 1.4 to 24,300 (median = 194) and was inversely correlated to stage (p = 0.0006). The cancer related osteopontin and VEGF-A had TIF-to-plasma ratios ranging from 1 to 62 (median = 15) and 2 to 1040 (median = 59), respectively. The ratios were not affected by tumor stage, indicative of more widespread protein expression. Conclusion We present absolute quantitative data on the TIF-to-plasma gradient of selected proteins in the tumor microenvironment, and demonstrate a substantial and stage dependent gradient for CA-125 between TIF and plasma, suggesting a relation between total tumor burden and tissue-to-plasma gradient. General significance We present novel quantitative data on biomarker concentration in the tumor microenvironment, and a new strategy for biomarker selection, applicable in future biomarker studies. Quantification of CA-125, VEGF and osteopontin in tumor interstitial fluid (TIF) A large TIF-to-plasma gradient was observed for CA-125, the highest in early stage. Lower VEGF and osteopontin gradient indicate more widespread protein expression.
Collapse
Affiliation(s)
- Hanne Haslene-Hox
- Department of Biomedicine, University of Bergen, Norway ; Department of Bioprocess Technology, SINTEF Materials and Chemistry, Trondheim, Norway
| | - Amina Madani
- Department of Biomedicine, University of Bergen, Norway
| | - Kaja C G Berg
- Department of Biomedicine, University of Bergen, Norway
| | - Kathrine Woie
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Helga B Salvesen
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway ; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Norway
| | - Olav Tenstad
- Department of Biomedicine, University of Bergen, Norway
| |
Collapse
|
22
|
Cohen JG, White M, Cruz A, Farias-Eisner R. In 2014, can we do better than CA125 in the early detection of ovarian cancer? World J Biol Chem 2014; 5:286-300. [PMID: 25225597 PMCID: PMC4160523 DOI: 10.4331/wjbc.v5.i3.286] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/12/2014] [Accepted: 05/14/2014] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is a lethal gynecologic malignancy with greater than 70% of women presenting with advanced stage disease. Despite new treatments, long term outcomes have not significantly changed in the past 30 years with the five-year overall survival remaining between 20% and 40% for stage III and IV disease. In contrast patients with stage I disease have a greater than 90% five-year overall survival. Detection of ovarian cancer at an early stage would likely have significant impact on mortality rate. Screening biomarkers discovered at the bench have not translated to success in clinical trials. Existing screening modalities have not demonstrated survival benefit in completed prospective trials. Advances in high throughput screening are making it possible to evaluate the development of ovarian cancer in ways never before imagined. Data in the form of human “-omes” including the proteome, genome, metabolome, and transcriptome are now available in various packaged forms. With the correct pooling of resources including prospective collection of patient specimens, integration of high throughput screening, and use of molecular heterogeneity in biomarker discovery, we are poised to make progress in ovarian cancer screening. This review will summarize current biomarkers, imaging, and multimodality screening strategies in the context of emerging technologies.
Collapse
|
23
|
Mosch CG, Jaschinski T, Eikermann M. Impact of epithelial ovarian cancer screening on patient-relevant outcomes in average-risk postmenopausal women. Hippokratia 2014. [DOI: 10.1002/14651858.cd011210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Christoph G Mosch
- University Witten/Herdecke; Institute for Research in Operative Medicine (IFOM) - Department for Evidence-based Health Services Research; Ostmerheimer Str. 200 (Building 38) Cologne Germany 51109
| | - Thomas Jaschinski
- University Witten/Herdecke; Institute for Research in Operative Medicine (IFOM) - Department for Evidence-based Health Services Research; Ostmerheimer Str. 200 (Building 38) Cologne Germany 51109
| | - Michaela Eikermann
- Medical advisory service of social health insurance (MDS); Department of Evidence-based medicine; Theodor-Althoff-Straße 47 Essen North Rhine Westphalia Germany 51109
| |
Collapse
|
24
|
Trabectedin as a single agent and in combination with pegylated liposomal doxorubicin - activity against ovarian cancer cells. Contemp Oncol (Pozn) 2014; 18:149-52. [PMID: 25520572 PMCID: PMC4268992 DOI: 10.5114/wo.2014.43153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 09/27/2013] [Accepted: 12/17/2013] [Indexed: 02/07/2023] Open
Abstract
Over 225 000 new cases of ovarian cancer are diagnosed each year. Symptoms are often vague, so most cases are detected when the disease is at an advanced stage. There is a need to find new drugs which will be able to treat ovarian cancer effectively. One of the most promising antineoplastic agents is trabectedin (Yondelis), derived from the marine tunicate Ecteinascidia turbinata, approved by the European Union in July 2007 for the treatment of soft-tissue sarcomas. This drug shows a mechanism of action based on the inhibition of the nucleotide excision repair system. Trabectedin shows anti-tumour activity in vitro and in vivo in ovarian, breast, prostate, renal, melanoma and non-small cell lung cancer cell lines. Trabectedin in combination with pegylated liposomal doxorubicin demonstrates synergistic antineoplastic activity.
Collapse
|
25
|
Pro-apoptotic activity of new analog of anthracyclines--WP 631 in advanced ovarian cancer cell line. Toxicol In Vitro 2013; 28:273-81. [PMID: 24287111 DOI: 10.1016/j.tiv.2013.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 09/05/2013] [Accepted: 11/15/2013] [Indexed: 11/22/2022]
Abstract
In this work we investigated the mode of cell death induced by WP 631, a novel anthracycline antibiotic, in the ovarian cancer cell line (OV-90) derived from the malignant ascites of a patient diagnosed with advanced disease. The effects were compared with those of doxorubicin (DOX), a first generation anthracycline. The ability of WP 631 to induce apoptosis and necrosis was examined by double staining with Annexin V and propidium iodide, measurements of the level of intracellular calcium ions and cytochrome c, PARP cleavage. We also investigated the possible involvement of the caspases activation, DNA degradation (comet assay) and intracellular reactive oxygen species (ROS) production in the development of the apoptotic events and their significance for drug efficiency. The results obtained clearly demonstrate that antiproliferative capacity of WP 631 in tested cell line was a few times greater than that of DOX. Furthermore, ovarian cancer cells treated with WP 631 showed a higher mean level of basal DNA damage in comparison to DOX. In conclusion, WP 631 is able to induce caspase - dependent apoptosis in human ovarian cancer cells. Obtained results suggested that WP 631 may be a candidate for further evaluation as chemotherapeutic agents for human cancers.
Collapse
|
26
|
Jiang W, Huang R, Duan C, Fu L, Xi Y, Yang Y, Yang WM, Yang D, Yang DH, Huang RP. Identification of five serum protein markers for detection of ovarian cancer by antibody arrays. PLoS One 2013; 8:e76795. [PMID: 24116163 PMCID: PMC3792870 DOI: 10.1371/journal.pone.0076795] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 08/28/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Protein and antibody arrays have emerged as a promising technology to study protein expression and protein function in a high-throughput manner. These arrays also represent a new opportunity to profile protein expression levels in cancer patients' samples and to identify useful biosignatures for clinical diagnosis, disease classification, prediction, drug development and patient care. We applied antibody arrays to discover a panel of proteins which may serve as biomarkers to distinguish between patients with ovarian cancer and normal controls. METHODOLOGY/PRINCIPAL FINDINGS Using a case-control study design of 34 ovarian cancer patients and 53 age-matched healthy controls, we profiled the expression levels of 174 proteins using antibody array technology and determined the CA125 level using ELISA. The expression levels of those proteins were analyzed using 3 discriminant methods, including artificial neural network, classification tree and split-point score analysis. A panel of 5 serum protein markers (MSP-alpha, TIMP-4, PDGF-R alpha, and OPG and CA125) was identified, which could effectively detect ovarian cancer with high specificity (95%) and high sensitivity (100%), with AUC =0.98, while CA125 alone had an AUC of 0.87. CONCLUSIONS/SIGNIFICANCE Our pilot study has shown the promising set of 5 serum markers for ovarian cancer detection.
Collapse
Affiliation(s)
- Weidong Jiang
- RayBiotech, Inc, Norcross, Georgia, United States of America
| | - Ruochun Huang
- RayBiotech, Inc, Norcross, Georgia, United States of America
- RayBiotech, Inc, Guangzhou, China
| | - Chaohui Duan
- Department of Clinical Laboratory, the Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liwu Fu
- The Affiliated Cancer Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Yun Xi
- Department of Clinical Laboratory, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuebo Yang
- Department of Obstetrics and Gynecology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei-Min Yang
- Department of Gynecology, Wuxi Maternal and Child Health Hospital, Wuxi, China
| | - Dongzi Yang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dong-Hua Yang
- Biosample Repository, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Ruo-Pan Huang
- RayBiotech, Inc, Norcross, Georgia, United States of America
- RayBiotech, Inc, Guangzhou, China
- South China Biochip Research Center, Guangzhou, China
| |
Collapse
|
27
|
González Martín A, Redondo A, Jurado M, De Juan A, Romero I, Bover I, Del Campo JM, Cervantes A, García Y, López-Guerrero JA, Mendiola C, Palacios J, Rubio MJ, Poveda Velasco A. GEICO (Spanish Group for Investigation on Ovarian Cancer) treatment guidelines in ovarian cancer 2012. Clin Transl Oncol 2013; 15:509-25. [PMID: 23468275 PMCID: PMC3695314 DOI: 10.1007/s12094-012-0995-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 12/21/2012] [Indexed: 01/23/2023]
Abstract
In 2006, under the auspices of The Spanish Research Group for Ovarian Cancer (Spanish initials GEICO), the first "Treatment Guidelines in Ovarian Cancer" were developed and then published in Clinical and Translational Oncology by Poveda Velasco et al. (Clin Transl Oncol 9(5):308-316, 2007). Almost 6 years have elapsed and over this time, we have seen some important developments in the treatment of ovarian cancer. Significant changes were also introduced after the GCIG-sponsored 4th Consensus Conference on Ovarian Cancer by Stuart et al. (Int J Gynecol Cancer 21:750-755, 2011). So we decided to update the treatment guidelines in ovarian cancer and, with this objective, a group of investigators of the GEICO group met in February 2012. This study summarizes the presentations, discussions and evidence that were reviewed during the meeting and during further discussions of the manuscript.
Collapse
Affiliation(s)
- A González Martín
- Medical Oncology Department, MD Anderson Cancer Center, C/Arturo Soria, 270, 28033, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zhou S, Tang L, Wang H, Dai J, Zhang J, Shen L, Ng SW, Berkowitz RS. Overexpression of c-Abl predicts unfavorable outcome in epithelial ovarian cancer. Gynecol Oncol 2013; 131:69-76. [PMID: 23820113 DOI: 10.1016/j.ygyno.2013.06.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/21/2013] [Accepted: 06/23/2013] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Abelson tyrosine kinase (c-Abl) has been shown to promote solid tumor invasion and metastasis. However, little is known regarding whether c-Abl contributes to the development or progression of epithelial ovarian cancer (EOC). The aims of this study are to determine the expression of c-Abl and investigate a possible relationship between c-Abl and prognosis in EOC. METHODS c-Abl protein level was evaluated in 137 EOC specimens by immunohistochemical staining and 32 EOC specimens by Western blot analysis. Expression of c-Abl in ovarian cancer cell lines was measured by Western blot analysis and immunofluorescence. Survival analysis was performed to assess the correlation between c-Abl expression and survival. RESULTS Immunohistochemical staining and Western blot analysis revealed that c-Abl was overexpressed in EOC compared with samples from a non-invasive ovarian tumor and normal ovaries (P<0.05). Furthermore, expression of c-Abl was significantly associated with advanced FIGO stage, poor grade, serum Ca-125 and residual tumor size (P<0.05). By Western blot analysis, c-Abl expression was examined in four ovarian cancer cell lines. Meanwhile, immunofluorescence was performed to show c-Abl expression in SKOV3 and 3AO cell lines. Survival analysis demonstrated that patients with low c-Abl staining had a significantly better survival compared to patients with high c-Abl staining (P<0.05). In multivariate analysis, c-Abl overexpression, poor grade, advanced stage and suboptimal surgical debulking were independent prognostic factors of poor survival. CONCLUSIONS Our present study finds that c-Abl overexpression is associated with an unfavorable outcome. c-Abl may be a crucial predictor for EOC metastasis.
Collapse
Affiliation(s)
- Suiyang Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Accuracy of Positron Emission Tomography/Computed Tomography in the Diagnosis and Restaging for Recurrent Ovarian Cancer. Int J Gynecol Cancer 2013; 23:598-607. [DOI: 10.1097/igc.0b013e31828a183c] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
30
|
Chandler K, Goldman R. Glycoprotein disease markers and single protein-omics. Mol Cell Proteomics 2013; 12:836-45. [PMID: 23399550 PMCID: PMC3617330 DOI: 10.1074/mcp.r112.026930] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/07/2013] [Indexed: 12/14/2022] Open
Abstract
Glycoproteins are well represented among biomarkers for inflammatory and cancer diseases. Secreted and membrane-associated glycoproteins make excellent targets for noninvasive detection. In this review, we discuss clinically applicable markers of cancer diseases and methods for their analysis. High throughput discovery continues to supply marker candidates with unusual glycan structures, altered glycoprotein abundance, or distribution of site-specific glycoforms. Improved analytical methods are needed to unlock the potential of these discoveries in validated clinical assays. A new generation of targeted quantitative assays is expected to advance the use of glycoproteins in early detection of diseases, molecular disease classification, and monitoring of therapeutic interventions.
Collapse
Affiliation(s)
- Kevin Chandler
- From the Departments of ‡Biochemistry and Molecular and Cellular Biology and
| | - Radoslav Goldman
- From the Departments of ‡Biochemistry and Molecular and Cellular Biology and
- ¶Oncology, Georgetown University, Washington, D.C. 20057
| |
Collapse
|