1
|
Wu X, Liu J, An R, Yin R, Zhang Y, Zhou H, He A, Wang L, Zhang J, Liu Z, Duan W, Zhu J, Lou G, Chen G, Cheng Y, Xue F, Nick S, Wang H, Li D. First-line bevacizumab plus chemotherapy in Chinese patients with stage III/IV epithelial ovarian cancer, fallopian tube cancer or primary peritoneal cancer: a phase III randomized controlled trial. J Gynecol Oncol 2024; 35:e99. [PMID: 38872480 PMCID: PMC11390243 DOI: 10.3802/jgo.2024.35.e99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/22/2023] [Accepted: 03/31/2024] [Indexed: 06/15/2024] Open
Abstract
OBJECTIVE First-line bevacizumab plus carboplatin and paclitaxel (CP) is approved for stage III/IV ovarian cancer treatment following initial surgical resection, based on global phase III GOG-0218 and ICON7 trials. This study evaluated the efficacy and safety of bevacizumab + CP as first-line ovarian cancer therapy in Chinese patients. METHODS Patients with newly diagnosed, International Federation of Gynecology and Obstetrics (FIGO) stage III/IV epithelial ovarian, fallopian tube, or primary peritoneal cancer post-primary surgery were randomized 1:1 to receive 6 cycles of CP with bevacizumab/placebo, followed by bevacizumab/placebo maintenance until unacceptable toxicity or disease progression. Primary endpoint was investigator-assessed progression-free survival (PFS). Stratification factors were FIGO stage and debulking status (stage III optimally debulked vs stage III suboptimally debulked vs stage IV) and Eastern Cooperative Oncology Group performance status (0 vs 1 or 2). RESULTS Of randomized patients, 51 received bevacizumab + CP and 49 received placebo + CP. Median PFS was 22.6 months with bevacizumab + CP (95% confidence interval [CI]=18.6, not estimable) and 12.3 months (95% CI=9.5, 15.0) with placebo + CP (stratified hazard ratio=0.30; 95% CI=0.17, 0.53). Treatment-related grade 3/4 adverse events occurred in 46 of 49 (94%) patients receiving bevacizumab + CP, and 34 of 50 (68%) receiving placebo + CP. CONCLUSION Bevacizumab + CP showed clinically meaningful improvement in PFS vs placebo + CP, consistent with GOG-0218 results. Safety data were aligned with the known bevacizumab safety profile. These results support first-line bevacizumab + CP therapy in Chinese patients with ovarian cancer. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03635489.
Collapse
Affiliation(s)
- Xiaohua Wu
- Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Jihong Liu
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ruifang An
- First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rutie Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yu Zhang
- Xiangya Hospital Central South University, Changsha, China
| | | | - Aiqin He
- Nantong Tumor Hospital, Nantong, China
| | - Li Wang
- Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jieqing Zhang
- Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Ziling Liu
- The First Hospital of Jilin University, Changchun, China
| | - Wei Duan
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Jianqing Zhu
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Ge Lou
- Harbin Medical University Cancer Hospital, Harbin, China
| | | | | | - Fengxia Xue
- Tianjin Medical University General Hospital, Tianjin, China
| | - Sonja Nick
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | |
Collapse
|
2
|
Sun X, Guo Y. Chemerin Enhances Migration and Invasion of OC Cells via CMKLR1/RhoA/ROCK-Mediated EMT. Int J Endocrinol 2024; 2024:7957018. [PMID: 39104601 PMCID: PMC11300085 DOI: 10.1155/2024/7957018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/26/2024] [Accepted: 03/17/2024] [Indexed: 08/07/2024] Open
Abstract
Chemerin is a newly described adipokine with significant effects on obesity, metabolic disorders, and immune trafficking. Recently, chemerin has gained prominence for its potential roles in cancer and tumorigenesis with pro- or antitumor effects. To date, most referenced multifunctions of chemerin are attributed to the chemokine-like receptor 1 (CMKLR1), distributing broadly in many tissues. This study investigates the in vitro roles of chemerin treatment on migration and invasion of ovarian carcinoma cells (OVCAR-3 and SK-OV-3) and potential underlying mechanisms. Herein, exogenous chemerin treatment promotes growth and invasion of SK-OV-3 cells but has no significant effects on OVCAR-3 cells. SK-OV-3 cells undergo morphological elongation characterized by epithelial-to-mesenchymal transition (EMT) and Ras homologous genome members A (RhoA)/Rho protein-related curl spiral kinase-1 (ROCK1) activation. Furthermore, chemerin-enhanced invasion and EMT of SK-OV-3 cells are effectively blocked by C3 transferase (C3T) and Y27632 and RhoA and ROCK1 inhibitor, respectively. More importantly, RhoA/ROCK1-EMT-mediated SK-OV-3 cell invasion is orchestrated by CMKLR1 upregulation after chemerin treatment (50 ng/mL). The silencing of CMKLR1 significantly (P < 0.0001) reverses the chemerin-enhanced invasion, EMT, and RhoA/ROCK1 activation of SK-OV-3 cells. Our study indicates that chemerin promotes invasion of OC cells via CMKLR1-RhoA/ROCK1-mediated EMT, offering a novel potential target for metastasis of OC.
Collapse
Affiliation(s)
- Xiaojing Sun
- First Hospital of China Medical UniversityDepartment of Obstetrics and Gynecology, Shenyang 110001, Liaoning, China
| | - Yi Guo
- First Hospital of China Medical UniversityDepartment of Obstetrics and Gynecology, Shenyang 110001, Liaoning, China
| |
Collapse
|
3
|
Recht HS, Shampain KL, Flory MN, Nougaret S, Barber EL, Jha P, Maturen KE, Sadowski EA, Shinagare AB, Venkatesan AM, Horowitz JM. Gynecologic oncology tumor board: the central role of the radiologist. Abdom Radiol (NY) 2023; 48:3265-3279. [PMID: 37386301 DOI: 10.1007/s00261-023-03978-y] [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: 03/04/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023]
Abstract
This manuscript is a collaborative, multi-institutional effort by members of the Society of Abdominal Radiology Uterine and Ovarian Cancer Disease Focus Panel and the European Society of Urogenital Radiology Women Pelvic Imaging working group. The manuscript reviews the key role radiologists play at tumor board and highlights key imaging findings that guide management decisions in patients with the most common gynecologic malignancies including ovarian cancer, cervical cancer, and endometrial cancer.
Collapse
Affiliation(s)
- Hannah S Recht
- Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N. St. Clair Street, Suite 800, Chicago, IL, 60611, USA.
| | - Kimberly L Shampain
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Marta N Flory
- Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Stephanie Nougaret
- Montpellier Cancer Institute, University of Montpellier, Monpellier, France
- IRCM, U1198, University of Montpellier, Monpellier, France
| | - Emma L Barber
- Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Priyanka Jha
- Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Katherine E Maturen
- Departments of Radiology and Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Elizabeth A Sadowski
- Departments of Radiology and Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Atul B Shinagare
- Department of Radiology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Aradhana M Venkatesan
- Department of Abdominal Imaging, Division of Diagnostic Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Jeanne M Horowitz
- Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N. St. Clair Street, Suite 800, Chicago, IL, 60611, USA
| |
Collapse
|
4
|
Safety Analysis of Bevacizumab in Ovarian Cancer Patients. J Clin Med 2023; 12:jcm12052065. [PMID: 36902852 PMCID: PMC10003828 DOI: 10.3390/jcm12052065] [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: 01/15/2023] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Bevacizumab (BEV) is beneficial for ovarian cancer patients, but the real world's patient settings differ from those in clinical trials. This study tries to illustrate adverse events in the Taiwanese population. Patients with epithelial ovarian cancer treated with BEV at Kaohsiung Chang Gung Memorial Hospital between 2009 and 2019 were retrospectively reviewed. The receiver operating characteristic curve was adopted to identify the cutoff dose and the presence of BEV-related toxicities. A total of 79 patients receiving BEV in neoadjuvant, frontline, or salvage settings were enrolled. The median follow-up time was 36.2 months. Twenty patients (25.3%) had "De novo" hypertension or the worsening of a preexisting one. Twelve patients (15.2%) had "De novo" proteinuria. Five patients (6.3%) had thromboembolic events/hemorrhage. Four patients (5.1%) had gastrointestinal perforation (GIP), and one patient (1.3%) had wound-healing complications. Patients with BEV-related GIP had at least two risk factors for developing GIP, most of which were conservatively managed. This study revealed a compatible but distinct safety profile from those reported in clinical trials. The presence of BEV-related changes in blood pressure showed a dose-dependent trend. Most of the BEV-related toxicities were managed individually. Patients with potential risks for developing BEV-related GIP should use BEV with caution.
Collapse
|
5
|
Immune Tumor Microenvironment in Ovarian Cancer Ascites. Int J Mol Sci 2022; 23:ijms231810692. [PMID: 36142615 PMCID: PMC9504085 DOI: 10.3390/ijms231810692] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/26/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer (OC) has a specific type of metastasis, via transcoelomic, and most of the patients are diagnosed at advanced stages with multiple tumors spread within the peritoneal cavity. The role of Malignant Ascites (MA) is to serve as a transporter of tumor cells from the primary location to the peritoneal wall or to the surface of the peritoneal organs. MA comprise cellular components with tumor and non-tumor cells and acellular components, creating a unique microenvironment capable of modifying the tumor behavior. These microenvironment factors influence tumor cell proliferation, progression, chemoresistance, and immune evasion, suggesting that MA play an active role in OC progression. Tumor cells induce a complex immune suppression that neutralizes antitumor immunity, leading to disease progression and treatment failure, provoking a tumor-promoting environment. In this review, we will focus on the High-Grade Serous Carcinoma (HGSC) microenvironment with special attention to the tumor microenvironment immunology.
Collapse
|
6
|
Integration of Transcriptome and Epigenome to Identify and Develop Prognostic Markers for Ovarian Cancer. JOURNAL OF ONCOLOGY 2022; 2022:3744466. [PMID: 36081667 PMCID: PMC9448543 DOI: 10.1155/2022/3744466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/04/2022] [Accepted: 06/29/2022] [Indexed: 11/21/2022]
Abstract
DNA methylation is a widely researched epigenetic modification. It is associated with the occurrence and development of cancer and has helped evaluate patients' prognoses. However, most existing DNA methylation prognosis models have not simultaneously considered the changes of the downstream transcriptome. Methods. The RNA-Sequencing data and DNA methylation omics data of ovarian cancer patients were downloaded from The Cancer Genome Atlas (TCGA) database. The Consensus Cluster Plus algorithm was used to construct the methylated molecular subtypes of the ovary. Lasso regression was employed to build a multi-gene signature. An independent data set was applied to verify the prognostic value of the signature. The Gene Set Variation Analysis (GSVA) was used to carry out the enrichment analysis of the pathways linked to the gene signature. The IMvigor 210 cohort was used to explore the predictive efficacy of the gene signature for immunotherapy response. Results. We distinguished ovarian cancer samples into two subtypes with different prognosis, based on the omics data of DNA methylation. Differentially expressed genes and enrichment analysis among subtypes indicated that DNA methylation was related to fatty acid metabolism and the extracellular matrix (ECM)-receptor. Furthermore, we constructed an 8-gene signature, which proved to be efficient and stable in predicting prognostics in ovarian cancer patients with different data sets and distinctive pathological characteristics. Finally, the 8-gene signature could predict patients' responses to immunotherapy. The polymerase chain reaction experiment was further used to verify the expression of 8 genes. Conclusion. We analyzed the prognostic value of the related genes of methylation in ovarian cancer. The 8-gene signature predicted the prognosis and immunotherapy response of ovarian cancer patients well and is expected to be valuable in clinical application.
Collapse
|
7
|
Tang H, Shan J, Liu J, Wang X, Wang F, Han S, Zhao X, Wang J. Molecular subtypes, clinical significance, and tumor immune landscape of angiogenesis-related genes in ovarian cancer. Front Oncol 2022; 12:995929. [PMID: 36106103 PMCID: PMC9464911 DOI: 10.3389/fonc.2022.995929] [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: 07/16/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis is a physiological process, where new blood vessels are formed from pre-existing vessels through the mechanism called sprouting. It plays a significant role in supporting tumor growth and is expected to provide novel therapeutic ideas for treating tumors that are resistant to conventional therapies. We investigated the expression pattern of angiogenesis-related genes (ARGs) in ovarian cancer (OV) from public databases, in which the patients could be classified into two differential ARG clusters. It was observed that patients in ARGcluster B would have a better prognosis but lower immune cell infiltration levels in the tumor microenvironment. Then ARG score was computed based on differentially expressed genes via cox analysis, which exhibited a strong correlation to copy number variation, immunophenoscore, tumor mutation load, and chemosensitivity. In addition, according to the median risk score, patients were separated into two risk subgroups, of which the low-risk group had a better prognosis, increased immunogenicity, and stronger immunotherapy efficacy. Furthermore, we constructed a prognostic nomogram and demonstrated its predictive value. These findings help us better understand the role of ARGs in OV and offer new perspectives for clinical prognosis and personalized treatment.
Collapse
Affiliation(s)
- Haixia Tang
- Department of Gynecology, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Jingsong Shan
- Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan, China
| | - Juan Liu
- Department of Obstetrics and Gynecology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xuehai Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Fengxu Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Suping Han
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Suping Han, ; Xinyuan Zhao, ; Jinxiu Wang,
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
- *Correspondence: Suping Han, ; Xinyuan Zhao, ; Jinxiu Wang,
| | - Jinxiu Wang
- Department of Gynecology, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
- *Correspondence: Suping Han, ; Xinyuan Zhao, ; Jinxiu Wang,
| |
Collapse
|
8
|
Mitra S, Lami MS, Ghosh A, Das R, Tallei TE, Fatimawali, Islam F, Dhama K, Begum MY, Aldahish A, Chidambaram K, Emran TB. Hormonal Therapy for Gynecological Cancers: How Far Has Science Progressed toward Clinical Applications? Cancers (Basel) 2022; 14:759. [PMID: 35159024 PMCID: PMC8833573 DOI: 10.3390/cancers14030759] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 02/01/2023] Open
Abstract
In recent years, hormone therapy has been shown to be a remarkable treatment option for cancer. Hormone treatment for gynecological cancers involves the use of medications that reduce the level of hormones or inhibit their biological activity, thereby stopping or slowing cancer growth. Hormone treatment works by preventing hormones from causing cancer cells to multiply. Aromatase inhibitors, anti-estrogens, progestin, estrogen receptor (ER) antagonists, GnRH agonists, and progestogen are effectively used as therapeutics for vulvar cancer, cervical cancer, vaginal cancer, uterine cancer, and ovarian cancer. Hormone replacement therapy has a high success rate. In particular, progestogen and estrogen replacement are associated with a decreased incidence of gynecological cancers in women infected with human papillomavirus (HPV). The activation of estrogen via the transcriptional functionality of ERα may either be promoted or decreased by gene products of HPV. Hormonal treatment is frequently administered to patients with hormone-sensitive recurring or metastatic gynecologic malignancies, although response rates and therapeutic outcomes are inconsistent. Therefore, this review outlines the use of hormonal therapy for gynecological cancers and identifies the current knowledge gaps.
Collapse
Affiliation(s)
- Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (M.S.L.); (A.G.); (R.D.)
| | - Mashia Subha Lami
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (M.S.L.); (A.G.); (R.D.)
| | - Avoy Ghosh
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (M.S.L.); (A.G.); (R.D.)
| | - Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (M.S.L.); (A.G.); (R.D.)
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, Indonesia;
- The University Center of Excellence for Biotechnology and Conservation of Wallacea, Institute for Research and Community Services, Sam Ratulangi University, Manado 95115, Indonesia;
| | - Fatimawali
- The University Center of Excellence for Biotechnology and Conservation of Wallacea, Institute for Research and Community Services, Sam Ratulangi University, Manado 95115, Indonesia;
- Pharmacy Study Program, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, Indonesia
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health of Sciences, Daffodil International University, Dhaka 1207, Bangladesh;
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India;
| | - M. Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia;
| | - Afaf Aldahish
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia; (A.A.); (K.C.)
| | - Kumarappan Chidambaram
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia; (A.A.); (K.C.)
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| |
Collapse
|
9
|
Xia X, Li Z, Li Y, Ye F, Zhou X. LncRNA XIST promotes carboplatin resistance of ovarian cancer through activating autophagy via targeting miR-506-3p/FOXP1 axis. J Gynecol Oncol 2022; 33:e81. [PMID: 36335987 PMCID: PMC9634093 DOI: 10.3802/jgo.2022.33.e81] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/05/2022] [Accepted: 08/07/2022] [Indexed: 11/07/2022] Open
Abstract
Objective Resistance to chemotherapy drugs makes ovarian cancer (OC) difficult to treat and ultimately kills patients. Long non-coding RNAs are closely related to carboplatin resistance in OC. In present study, we explored the role of lncRNA X-inactive specific transcript (XIST) on carboplatin resistance in OC. Methods Cell viability, proliferation, and apoptosis were assessed through 2,5-diphenyl-2H-tetrazolium bromide, colony formation, and flow cytometry assays, respectively. Microtubule-associated protein 1A/1B-light chain 3 expression was evaluated by immunofluorescence assay to analyze the cell autophagy. The interaction of XIST/miR-506-3p or miR-506-3p/forkhead box protein P1 (FOXP1) was analyzed using RNA immunoprecipitation (RIP) and dual-luciferases reporter assays. The function of XIST/miR-506-3p/FOXP1 axis in vivo was further confirmed by tumor xenograft study and immunohistochemistry. Results The expression of XIST and FOXP1 increased while miR-506-3p decreased in OC and carboplatin resistance cells. XIST silencing repressed the proliferative and autophagic capacities of carboplatin resistance cells while promoted the apoptosis. XIST overexpression led to the opposite results. XIST targeted miR-506-3p and downregulated its expression. MiR-506-3p inhibition facilitated the proliferative and autophagic capacities while suppressed the apoptosis of cells, XIST knockdown reversed these effects. MiR-506-3p bound to FOXP1. XIST knockdown or miR-506-3p overexpression reversed the increase of cell proliferative and autophagic abilities and the decrease of apoptosis rate induced by FOXP1 overexpression. XIST affected autophagy and carboplatin resistance in vivo via regulating the miR-506-3p/FOXP1 axis. Conclusion XIST knockdown inhibited autophagy and carboplatin resistance of OC through FOXP1/protein kinase B (AKT)/mammalian target of rapamycin pathway by targeting miR-506-3p. Knockdown of XIST inhibited autophagy induced by carboplatin and resistance to carboplatin in ovarian cells. XIST targeted miR-506-3p and reduced its expression. FOXP1 could be a target gene of miR-506-3p. XIST facilitated the autophagy and carboplatin resistance through miR-506-3p/FOXP1 axis in ovarian cancer.
Collapse
Affiliation(s)
- Xiaoyan Xia
- Scientific Research Department, Changsha Health Vocational College, Changsha, P.R. China
| | - Zikui Li
- Department of Obstetrics and Gynecology, The Fist Hospital of Hunan University of Chinese Medicine, Changsha, P.R. China
| | - Yaojun Li
- Nursing College of Changsha Health Vocational College, Changsha, P.R. China
| | - Feng Ye
- Nursing College of Changsha Health Vocational College, Changsha, P.R. China
| | - Xiaoming Zhou
- Department of Cardiology, The Fist Hospital of Hunan University of Chinese Medicine, Changsha, P.R. China
| |
Collapse
|
10
|
Creeden JF, Nanavaty NS, Einloth KR, Gillman CE, Stanbery L, Hamouda DM, Dworkin L, Nemunaitis J. Homologous recombination proficiency in ovarian and breast cancer patients. BMC Cancer 2021; 21:1154. [PMID: 34711195 PMCID: PMC8555001 DOI: 10.1186/s12885-021-08863-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023] Open
Abstract
Homologous recombination and DNA repair are important for genome maintenance. Genetic variations in essential homologous recombination genes, including BRCA1 and BRCA2 results in homologous recombination deficiency (HRD) and can be a target for therapeutic strategies including poly (ADP-ribose) polymerase inhibitors (PARPi). However, response is limited in patients who are not HRD, highlighting the need for reliable and robust HRD testing. This manuscript will review BRCA1/2 function and homologous recombination proficiency in respect to breast and ovarian cancer. The current standard testing methods for HRD will be discussed as well as trials leading to approval of PARPi's. Finally, standard of care treatment and synthetic lethality will be reviewed.
Collapse
Affiliation(s)
- Justin Fortune Creeden
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
- Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Nisha S Nanavaty
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Katelyn R Einloth
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Cassidy E Gillman
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | | | - Danae M Hamouda
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Lance Dworkin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | | |
Collapse
|
11
|
Kim L, Park SA, Yang Y, Kim Y, Heo TH, Kim H. LncRNA SRA mediates cell migration, invasion, and progression of ovarian cancer via NOTCH signaling and epithelial-mesenchymal transition. Biosci Rep 2021; 41:BSR20210565. [PMID: 34402503 PMCID: PMC8421593 DOI: 10.1042/bsr20210565] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/07/2023] Open
Abstract
Long non-coding RNA (lncRNA) is a newly identified regulator of tumor formation and tumor progression. The function and expression of lncRNAs remain to be fully elucidated, but recent studies have begun to address their importance in human health and disease. The lncRNA, SRA, known as steroid receptor activator, acts as an important modulator of gynecological cancer, and its expression may affect biological functions including proliferation, apoptosis, steroid formation, and muscle development. However, it is still not well known whether SRA is involved in the regulation of ovarian cancer. The present study investigated the molecular function and association between SRA expression and clinicopathological factors. In ovarian cancer cell lines, SRA knockdown and overexpression regulated cell migration, proliferation, and invasion. Both in vivo and in vitro experiments using knockdown and overexpression showed that SRA potently regulated epithelial-mesenchymal transition (EMT) and NOTCH pathway components. Further, clinical data confirmed that SRA was a significant predictor of overall survival (OS) and progression-free survival and patients with ovarian cancer exhibiting high expression of SRA exhibited higher recurrence rates than patients with low SRA expression. In conclusion, the present study indicates that SRA has clinical significance as its expression can predict the prognosis of ovarian cancer patients. High expression of the lncRNA SRA is strongly correlated with recurrence-free survival of ovarian cancer patients.
Collapse
Affiliation(s)
- Lee Kyung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, South Korea
| | - Sun-Ae Park
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, South Korea
| | - Yoolhee Yang
- Department of Plastic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Young Tae Kim
- Institute of Women's Life Medical Science, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Tae-Hwe Heo
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, South Korea
| | - Hee Jung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences and BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, South Korea
| |
Collapse
|
12
|
Moody R, Wilson K, Kampan NC, McNally OM, Jobling TW, Jaworowski A, Stephens AN, Plebanski M. Mapping Epitopes Recognised by Autoantibodies Shows Potential for the Diagnosis of High-Grade Serous Ovarian Cancer and Monitoring Response to Therapy for This Malignancy. Cancers (Basel) 2021; 13:cancers13164201. [PMID: 34439354 PMCID: PMC8392293 DOI: 10.3390/cancers13164201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Most women are diagnosed with high-grade serous ovarian cancer (HGSOC) at stage III, when the cancer has already spread, contributing to poor survival outcomes. However, while earlier diagnosis increases survival rates, there is a lack of early diagnosis biomarkers. Previously, autoantibodies specific for phosphorylated heat shock factor 1 (HSF1-PO4) were suggested as a potential diagnostic biomarker for early-stage HGSOC. In the present study, specific regions within HSF1 were identified, tested and confirmed as useful biomarkers, with comparable diagnostic potential to the full protein, across two separate clinical cohorts. Additionally, antibody responses to HSF1-PO4 and the corresponding peptides were found to increase following a round of standard first-line chemotherapy. Together, our data suggest that the identified short peptide sequences could be used as practical alternatives to support early diagnosis or monitor responses to chemotherapy. Abstract Autoantibodies recognising phosphorylated heat shock factor 1 (HSF1-PO4) protein are suggested as potential new diagnostic biomarkers for early-stage high-grade serous ovarian cancer (HGSOC). We predicted in silico B-cell epitopes in human and murine HSF1. Three epitope regions were synthesised as peptides. Circulating immunoglobulin A (cIgA) against the predicted peptide epitopes or HSF1-PO4 was measured using ELISA, across two small human clinical trials of HGSOC patients at diagnosis. To determine whether chemotherapy would promote changes in reactivity to either HSF1-PO4 or the HSF-1 peptide epitopes, IgA responses were further assessed in a sample of patients after a full cycle of chemotherapy. Anti-HSF1-PO4 responses correlated with antibody responses to the three selected epitope regions, regardless of phosphorylation, with substantial cross-recognition of the corresponding human and murine peptide epitope variants. Assessing reactivity to individual peptide epitopes, compared to HSF1-PO4, improved assay sensitivity. IgA responses to HSF1-PO4 further increased significantly post treatment, indicating that HSF1-PO4 is a target for immunity in response to chemotherapy. Although performed in a small cohort, these results offer potential insights into the interplay between autoimmunity and ovarian cancer and offer new peptide biomarkers for early-stage HGSOC diagnosis, to monitor responses to chemotherapy, and widely for pre-clinical HGSOC research.
Collapse
Affiliation(s)
- Rhiane Moody
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (R.M.); (K.W.); (A.J.)
| | - Kirsty Wilson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (R.M.); (K.W.); (A.J.)
| | - Nirmala Chandralega Kampan
- Department of Obstetrics and Gynaecology, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur 56000, Malaysia;
| | - Orla M. McNally
- Gynaeoncology Unit, Royal Women’s Hospital, Parkville, VIC 3052, Australia;
| | - Thomas W. Jobling
- Department of Gynaecological Oncology, Monash Medical Centre, Bentleigh East, VIC 3165, Australia;
| | - Anthony Jaworowski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (R.M.); (K.W.); (A.J.)
| | - Andrew N. Stephens
- Hudson Institute of Medical Research, Clayton, VIC 3168, Australia;
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; (R.M.); (K.W.); (A.J.)
- Correspondence:
| |
Collapse
|
13
|
Yang M, Chen G, Gao K, Wang Y. Tumor Immunometabolism Characterization in Ovarian Cancer With Prognostic and Therapeutic Implications. Front Oncol 2021; 11:622752. [PMID: 33796460 PMCID: PMC8008085 DOI: 10.3389/fonc.2021.622752] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/01/2021] [Indexed: 12/24/2022] Open
Abstract
Metabolic dysregulation in the tumor microenvironment has significant impact on immune infiltration and immune responses. However, interaction between immunity and metabolism in the ovarian microenvironment requires further exploration. We constructed an immunometabolism gene set and ovarian cancer cohort from The Cancer Genome Atlas (TCGA) and classified these into three immunometabolism subtypes. We explored the relationships between immune infiltration and metabolic reprogramming. Additionally, we built risk score and nomogram as prognostic signatures. Three distinctive immunometabolism subtypes were identified with therapeutic and prognostic implications. Subtype 1, the "immune suppressive-glycan metabolism subtype," featured high levels of immunosuppressive cell infiltration and glycan metabolism activation; Subtype 2, the "immune inflamed-amino acid metabolism subtype," showed abundant adaptive immune cell infiltration and amino acid metabolism activation; Subtype 3, the "immune desert-endocrine subtype," was characterized by low immune cell infiltration and upregulation of hormone biosynthesis. Furthermore, we found that epinephrine biosynthesis displayed a significantly negative correlation with MHC molecules, which may result in defective antigen presentation. We proposed immunometabolism subtypes with prognostic implications and provided new perspectives for the ovarian cancer microenvironment.
Collapse
Affiliation(s)
- Miner Yang
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gaowen Chen
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kunjie Gao
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yifeng Wang
- Department of Gynecology, Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Oliveira DVNP, Prahm KP, Christensen IJ, Hansen A, Høgdall CK, Høgdall EV. Gene expression profile association with poor prognosis in epithelial ovarian cancer patients. Sci Rep 2021; 11:5438. [PMID: 33686173 PMCID: PMC7940404 DOI: 10.1038/s41598-021-84953-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 01/22/2021] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is the eighth most common type of cancer for women worldwide. The current diagnostic and prognostic routine available for OC management either lack specificity or are very costly. Gene expression profiling has shown to be a very effective tool in exploring new molecular markers for patients with OC, although association of such markers with patient survival and clinical outcome is still elusive. Here, we performed gene expression profiling of different subtypes of OC to evaluate its association with patient overall survival (OS) and aggressive forms of the disease. By global mRNA microarray profiling in a total of 196 epithelial OC patients (161 serous, 15 endometrioid, 11 mucinous, and 9 clear cell carcinomas), we found four candidates-HSPA1A, CD99, RAB3A and POM121L9P, which associated with OS and poor clinicopathological features. The overexpression of all combined was correlated with shorter OS and progression-free survival (PFS). Furthermore, the combination of at least two markers were further associated with advanced grade, chemotherapy resistance, and progressive disease. These results indicate that a panel comprised of a few predictors that associates with a more aggressive form of OC may be clinically relevant, presenting a better performance than one marker alone.
Collapse
Affiliation(s)
| | - Kira P Prahm
- Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Ib J Christensen
- Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | | | - Claus K Høgdall
- Department of Gynaecology, Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Estrid V Høgdall
- Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev, Denmark.
| |
Collapse
|
15
|
Yin L, Zhang N, Yang Q. DNA methylation subtypes for ovarian cancer prognosis. FEBS Open Bio 2021; 11:851-865. [PMID: 33278864 PMCID: PMC7931230 DOI: 10.1002/2211-5463.13056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/28/2020] [Accepted: 12/03/2020] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer is one of three major malignancies of the female reproductive system. DNA methylation (MET) is closely related to ovarian cancer occurrence and development, and as such, elucidation of effective MET subtype markers may guide individualized treatment and improve ovarian cancer prognosis. To identify potential markers, we downloaded a total of 571 ovarian cancer MET samples from The Cancer Genome Atlas (TCGA), and established a Cox proportional hazards model using the MET spectrum and clinical pathological parameters. A total of 250 prognosis-related MET loci were obtained by Cox regression, and six molecular subtypes were screened by consensus clustering of CpG loci with a significant difference in both univariate and multivariate analyses. There was a remarkable MET difference between most subtypes. Cluster 2 had the highest MET level and demonstrated the best prognosis, while Clusters 4 and 5 had MET levels significantly lower than those of the other subtypes and demonstrated very poor prognosis. All Cluster 5 samples were at a high grade, while the percentage of stage IV samples in Cluster 4 was greater than in the other subtypes. We obtained five CpG loci using a coexpression network: cg27625732, cg00431050, cg22197830, cg03152385, and cg22809047. Our cluster analysis showed that prognosis in patients with hypomethylation was significantly worse than in patients with hypermethylation. These MET molecular subtypes can be used not only to evaluate ovarian cancer prognosis, but also to fully distinguish the tumor stage and histological grade in patients with ovarian cancer.
Collapse
Affiliation(s)
- Lili Yin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ningning Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
Zhang Y, Wang X, Chen X. Identification of core genes for early diagnosis and the EMT modulation of ovarian serous cancer by bioinformatics perspective. Aging (Albany NY) 2021; 13:3112-3145. [PMID: 33493131 PMCID: PMC7880353 DOI: 10.18632/aging.202524] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/30/2020] [Indexed: 02/05/2023]
Abstract
Ovarian serous carcinoma (OSC), as a common malignant tumor, poses a serious threat to women's health in that epithelial-mesenchymal transformation (EMT)-related modulation becomes heavily implicated in the invasion and progression of OSC. In this study, two core genes (BUB1B and NDC80) among the 16 hub genes have been identified to be involved in the molecular regulation of EMT and associated with the poor early survival of OSC at stages I+II. Through the Gene Regulatory Networks (GRN) analysis of 15 EMT regulators and core genes, it was revealed that TFAP2A and hsa-miR-655 could elaborately modulate EMT development of OSC. Next genetic variation analysis indicated that EMT regulator ELF3 would also serve as a crucial part in the occurrence and progression of OSC. Eventually, survival investigation suggested that TFAP2A, ELF3 and hsa-miR-655 were significantly associated with the overall survival of progressive OSC patients. Thus, combined with diversified bioinformatic analyses, BUB1B, NDC80, TFAP2A, ELF3 and hsa-miR-655 may act as the key biomarkers for early clinical diagnosis and prognosis evaluation of OSC patients as well as potential therapeutic target-points.
Collapse
Affiliation(s)
- Yanna Zhang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, High Technological Development Zone, Chengdu 610041, Sichuan, People's Republic of China
| | - Xun Wang
- State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, High Technological Development Zone, Chengdu 610041, Sichuan, People's Republic of China
| | - Xiancheng Chen
- State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, High Technological Development Zone, Chengdu 610041, Sichuan, People's Republic of China
| |
Collapse
|
17
|
Yao S, Shang W, Huang L, Xu R, Wu M, Wang F. The oncogenic and prognostic role of PDK1 in the progression and metastasis of ovarian cancer. J Cancer 2021; 12:630-643. [PMID: 33403023 PMCID: PMC7778543 DOI: 10.7150/jca.47278] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal of gynecological tumors in women. Tumor metabolism has become a new opportunity in the treatment of tumors. Pyruvate dehydrogenase kinase 1 (PDK1), as a key regulatory enzyme implicated in metabolic reprogramming of tumors, abnormally high expressed in various tumors and involved in the regulation of tumor cell biological behavior. However, the role of PDK1 in the occurrence and development of ovarian cancer remains unclear. Our team identified the expression of PDK1 in ovarian cancer cell lines and tissues through RT-PCR and immunohistochemical staining and evaluated the correlation of PDK1 expression with clinicopathologic features of patients and survival analyses. We used a variety of in vitro experiments to explore the influence of PDK1 on proliferation, invasion, migration, colony formation, apoptosis and the cell cycle of ovarian cancer cell lines CAOV3 and SKOV3. PDK1 was highly expressed in ovarian cancer cell lines and OC tissues. High expression of PDK1 was closely correlated to tumor size, FIGO stage, extraovarian metastases status and distribution. Univariate and multivariate Cox regression analysis identified that PDK1 was an independent prognostic factor for overall survival. Moreover, PDK1 was a superior predictor in prognosis of ovarian cancer and the incorporation of CA125 into PDK1 generated a predictive combination that displayed better predictive accuracy for overall survival. Downregulation of PDK1 suppressed the biological behavior of ovarian cancer cells due to S phase arrest and cellular apoptosis. PDK1 may serve as a novel prognostic biomarker, even a promising antineoplastic target of ovarian cancer.
Collapse
Affiliation(s)
- Shasha Yao
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China.,Department of Laboratory Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, 211100, Nanjing, China
| | - Wenwen Shang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Lei Huang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Rui Xu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Ming Wu
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| | - Fang Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.,National Key Clinical Department of Laboratory Medicine, 210029, Nanjing, China
| |
Collapse
|
18
|
Adaptation of metabolism to multicellular aggregation, hypoxia and obese stromal cell incorporation as potential measure of survival of ovarian metastases. Exp Cell Res 2020; 399:112397. [PMID: 33338477 DOI: 10.1016/j.yexcr.2020.112397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 12/18/2022]
Abstract
Ovarian metastases exfoliate from the primary tumor and it is thought that aggregation supports their survival in the peritoneal cavity during dissemination but the underlying mechanisms are not clearly identified. We have previously shown that ovarian cancer cells acquire an increasingly glycolytic and metabolic flexible phenotype during progression. In the present study, we investigated how hypoxia, aggregation, and the incorporation of the obese stromal vascular fraction (SVF) affect cellular metabolism and the response to common anti-cancer and anti-diabetic drugs. Our results show a reduction of glucose uptake, lactate secretion, cellular respiration and ATP synthesis in response to hypoxia and aggregation, suggesting that the observed reduced proliferation of cells aggregated into spheroids is the result of a down-regulation of respiration. Recruitment of SVF to spheroids increased the spheroids invasive capacity but reduced respiration only in the most aggressive cells. Further, aggregation and hypoxia reduced the response to the metabolic drugs AICAR and metformin, and the chemotherapeutic agents cisplatin and paclitaxel. Our results suggest that the adaptation of cellular metabolism may contribute to enhanced survival under non-permissive conditions, and that these metabolic alterations may provide targets for future interventions that aim to enhance the survival of women with metastatic ovarian cancer.
Collapse
|
19
|
Zheng M, Mullikin H, Hester A, Czogalla B, Heidegger H, Vilsmaier T, Vattai A, Chelariu-Raicu A, Jeschke U, Trillsch F, Mahner S, Kaltofen T. Development and Validation of a Novel 11-Gene Prognostic Model for Serous Ovarian Carcinomas Based on Lipid Metabolism Expression Profile. Int J Mol Sci 2020; 21:E9169. [PMID: 33271935 PMCID: PMC7731240 DOI: 10.3390/ijms21239169] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/06/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Biomarkers might play a significant role in predicting the clinical outcomes of patients with ovarian cancer. By analyzing lipid metabolism genes, future perspectives may be uncovered; (2) Methods: RNA-seq data for serous ovarian cancer were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus databases. The non-negative matrix factorization package in programming language R was used to classify molecular subtypes of lipid metabolism genes and the limma package in R was performed for functional enrichment analysis. Through lasso regression, we constructed a multi-gene prognosis model; (3) Results: Two molecular subtypes were obtained and an 11-gene signature was constructed (PI3, RGS, ADORA3, CH25H, CCDC80, PTGER3, MATK, KLRB1, CCL19, CXCL9 and CXCL10). Our prognostic model shows a good independent prognostic ability in ovarian cancer. In a nomogram, the predictive efficiency was notably superior to that of traditional clinical features. Related to known models in ovarian cancer with a comparable amount of genes, ours has the highest concordance index; (4) Conclusions: We propose an 11-gene signature prognosis prediction model based on lipid metabolism genes in serous ovarian cancer.
Collapse
Affiliation(s)
- Mingjun Zheng
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Heather Mullikin
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Anna Hester
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Bastian Czogalla
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Helene Heidegger
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Theresa Vilsmaier
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Aurelia Vattai
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| | - Till Kaltofen
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Maistrasse 11, 80337 Munich, Germany; (M.Z.); (H.M.); (A.H.); (B.C.); (H.H.); (T.V.); (A.V.); (A.C.-R.); (U.J.); (F.T.); (S.M.)
| |
Collapse
|
20
|
Song Z, Guo Q, Wang H, Gao L, Wang S, Liu D, Liu J, Qi Y, Lin B. miR-5193, regulated by FUT1, suppresses proliferation and migration of ovarian cancer cells by targeting TRIM11. Pathol Res Pract 2020; 216:153148. [PMID: 32823233 DOI: 10.1016/j.prp.2020.153148] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/14/2020] [Accepted: 07/24/2020] [Indexed: 12/24/2022]
Abstract
Ovarian cancer is the most lethal gynecological malignancy worldwide. A better understanding of the pathogenesis of ovarian cancer may help to improve the overall survival. Our previous studies have demonstrated that alpha-(1,2)-fucosyltransferase 1 (FUT1) is an oncogenic glycogene in ovarian cancer. However, the underlying mechanism is not fully clarified. In this study, we identified a microRNA as an important downstream regulator for the carcinogenic effect of FUT1 in ovarian cancer. miR-5193 was found down-regulated in ovarian cancer cells, FUT1-overexpression ovarian cancer cells and ovarian tumor samples. MTT, flow cytometry and Transwell assays demonstrated that miR-5193 inhibited the proliferation and migration, and induced the cell cycle arrest and apoptosis of ovarian cancer cells. Real-time PCR and western blot assays showed that miR-5193 downregulated the expression of TRIM11 and upregulated the expression of p53 and p21. Dual luciferase reporter assay indicated that TRIM11 was a direct target of miR‑5193. Rescue experiments confirmed that miR-5193 functioned in ovarian cancer cells by directly targeting TRIM11. Moreover, transfection with miR-5193 mimic in FUT1-overexpression ovarian cancer cells reversed the carcinogenic effect of FUT1. Taken together, our results suggest that miR-5193 is an essential suppressor of human ovarian cancer development, and is an important downstream regulator regarding the carcinogenesis of FUT1 in ovarian cancer.
Collapse
Affiliation(s)
- Zuofei Song
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China; Department of Obstetrics and Gynecology, General Hospital of Northern Theater Command, Shenyang, 110016, People's Republic of China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi 117004, People's Republic of China
| | - Qian Guo
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi 117004, People's Republic of China
| | - Huimin Wang
- Department of Obstetrics and Gynecology, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Lingling Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi 117004, People's Republic of China
| | - Shuang Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi 117004, People's Republic of China
| | - Dawo Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi 117004, People's Republic of China
| | - Juanjuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi 117004, People's Republic of China
| | - Yue Qi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi 117004, People's Republic of China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Benxi 117004, People's Republic of China.
| |
Collapse
|
21
|
Noncoding RNA (ncRNA) Profile Association with Patient Outcome in Epithelial Ovarian Cancer Cases. Reprod Sci 2020; 28:757-765. [PMID: 33125686 PMCID: PMC7862201 DOI: 10.1007/s43032-020-00372-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/21/2020] [Indexed: 12/24/2022]
Abstract
Ovarian cancer (OC) is the second most frequent type of gynecological cancers worldwide. In the past decades, the development of novel diagnostic and prognostic biomarkers available for OC has been limited, reflecting by the lack of specificity of such markers or very costly management. Microarray expression profiling has shown very effective results in exploring new molecular markers for patients with OC. Nonetheless, most screenings are focused on mutations or expression of molecules that are translated into proteins, corresponding to only 2% of the total human genome. In order to account for the vast majority of transcripts, in the present exploratory study, we assessed the expression levels of a comprehensive panel of noncoding RNA in different subtypes of OC. We further evaluated their association with patient overall survival (OS) and aggressive forms of the disease, such as tumor type, stage, and chemotherapy resistance. By microarray profiling in a total of 197 epithelial OC patients (162 serous carcinomas, 15 endometrioid carcinomas, 11 mucinous carcinomas, and 9 clear cell carcinomas), we found two candidates, SNORA68 and SNORD74, which associated with OS and poor clinicopathological features. The overexpression of those two targets combined was correlated with shorter OS and progression-free survival. That association was further observed to correlate with a more aggressive form of the disease. Overall, the results indicate that a panel comprised of SNORA68 and SNORD74 may be clinically relevant, where patients could be offered a more individualized, targeted follow-up, given its further validation on future prospective clinical studies.
Collapse
|
22
|
Ray-Coquard I, Mirza MR, Pignata S, Walther A, Romero I, du Bois A. Therapeutic options following second-line platinum-based chemotherapy in patients with recurrent ovarian cancer: Comparison of active surveillance and maintenance treatment. Cancer Treat Rev 2020; 90:102107. [PMID: 33099187 DOI: 10.1016/j.ctrv.2020.102107] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/30/2022]
Abstract
Most women with advanced ovarian cancer respond to initial treatment, consisting of surgical resection and ≈6 cycles of platinum-based chemotherapy. However, disease recurrence occurs in most patients, and subsequent therapies become necessary. Historically, close monitoring following treatment (active surveillance) was the only available option, as continued maintenance chemotherapy treatment led to increased toxicity without providing any meaningful clinical benefit. Recently, targeted therapy with the angiogenesis inhibitor bevacizumab and the poly(ADP-ribose) polymerase (PARP) inhibitors olaparib, niraparib, and rucaparib have demonstrated significant clinical benefits as maintenance treatment for recurrent disease. Despite consensus guidelines recommending their use, maintenance treatments are currently underutilized. Here, we review evidence from pivotal clinical trials of approved second-line maintenance treatments demonstrating efficacy in terms of progression-free survival and postprogression efficacy outcomes for patients with recurrent ovarian cancer. Adverse events frequently associated with bevacizumab include hypertension, proteinuria, and non-central nervous system bleeding, whereas PARP inhibitors are associated with nausea, vomiting, fatigue, and anemia. Patient-centered outcomes analyses show that PARP inhibitors provide significant benefits to patient health status, even when accounting for the toxicities associated with treatment. Many factors influence the selection of second-line maintenance treatment for patients with recurrent ovarian cancer, including the maintenance treatment received in the first-line setting. Overall, targeted maintenance treatment represents a new standard of care for patients with ovarian cancer, and we recommend that maintenance treatment should be offered to all eligible patients with recurrent ovarian cancer.
Collapse
Affiliation(s)
- Isabelle Ray-Coquard
- Department of Medical Oncology, Centre Léon Bérard and Université Claude Bernard and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon, France.
| | - Mansoor Raza Mirza
- Department of Oncology, Rigshospitalet Copenhagen University Hospital, and Nordic Society of Gynecological Oncology (NGSO), Copenhagen, Denmark.
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Naples, Italy.
| | - Axel Walther
- Bristol Cancer Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK.
| | - Ignacio Romero
- Medical Oncology Department, Instituto Valenciano de Oncologia, Valencia, Spain.
| | - Andreas du Bois
- Department of Gynecology and Gynecological Oncology, Evangelische Kliniken Essen-Mitte (KEM), Essen, Germany.
| |
Collapse
|
23
|
Zheng M, Hu Y, Gou R, Wang J, Nie X, Li X, Liu Q, Liu J, Lin B. Integrated multi-omics analysis of genomics, epigenomics, and transcriptomics in ovarian carcinoma. Aging (Albany NY) 2020; 11:4198-4215. [PMID: 31257224 PMCID: PMC6629004 DOI: 10.18632/aging.102047] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022]
Abstract
In this study, we identified prognostic biomarkers in ovarian carcinoma by integrating multi-omics DNA copy number variation (CNV) and methylation variation (MET) data. CNV, MET, and messenger RNA (mRNA) expression were examined in 351 ovarian carcinoma patients. Genes for which expression was correlated with DNA copy-number or DNA methylation were identified; three ovarian carcinoma gene subtypes were defined based on these correlations. Overall survival and B cell scores were lower, while the macrophage cell score was higher, in the DNA imprinting centre 1 (iC1) subtype than in the iC2 and iC3 subtypes. Comparison of CNV, MET, and mRNA expression among the subtypes identified two genes, ubiquitin B (UBB) and interleukin 18 binding protein (IL18BP), that were associated with prognosis. Mutation spectrum results based on subtype indicated that UBB and IL18BP expression may be influenced by mutation loci. Mutation levels were higher in iC1 samples than in iC2 or iC3 samples, indicating that the iC1 subtype is associated with disease progression. This integrated multi-omics analysis of genomics, epigenomics, and transcriptomics provides new insight into the molecular mechanisms of ovarian carcinoma and may help identify biomolecular markers for early disease diagnosis.
Collapse
Affiliation(s)
- Mingjun Zheng
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Yuexin Hu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Rui Gou
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Xin Nie
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Xiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Qing Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Juanjuan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning 110004, China.,Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| |
Collapse
|
24
|
Ovarian Cancer, Cancer Stem Cells and Current Treatment Strategies: A Potential Role of Magmas in the Current Treatment Methods. Cells 2020; 9:cells9030719. [PMID: 32183385 PMCID: PMC7140629 DOI: 10.3390/cells9030719] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/01/2020] [Accepted: 03/06/2020] [Indexed: 12/18/2022] Open
Abstract
Epithelial ovarian cancer (EOC) constitutes 90% of ovarian cancers (OC) and is the eighth most common cause of cancer-related death in women. The cancer histologically and genetically is very complex having a high degree of tumour heterogeneity. The pathogenic variability in OC causes significant impediments in effectively treating patients, resulting in a dismal prognosis. Disease progression is predominantly influenced by the peritoneal tumour microenvironment rather than properties of the tumor and is the major contributor to prognosis. Standard treatment of OC patients consists of debulking surgery, followed by chemotherapy, which in most cases end in recurrent chemoresistant disease. This review discusses the different origins of high-grade serous ovarian cancer (HGSOC), the major sub-type of EOC. Tumour heterogeneity, genetic/epigenetic changes, and cancer stem cells (CSC) in facilitating HGSOC progression and their contribution in the circumvention of therapy treatments are included. Several new treatment strategies are discussed including our preliminary proof of concept study describing the role of mitochondria-associated granulocyte macrophage colony-stimulating factor signaling protein (Magmas) in HGSOC and its unique potential role in chemotherapy-resistant disease.
Collapse
|
25
|
Zheng M, Hu Y, Gou R, Nie X, Li X, Liu J, Lin B. Identification three LncRNA prognostic signature of ovarian cancer based on genome-wide copy number variation. Biomed Pharmacother 2020; 124:109810. [PMID: 32000042 DOI: 10.1016/j.biopha.2019.109810] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/10/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Ovarian cancer is one of the most common malignant tumors of the female reproductive system, which seriously threatens the health of patients. It is of great significance to identify biomarkers to improve the clinical status of ovarian cancer patients. METHODS Methylation, RNA- sequencing, Copy number variation (CNV), mutation and clinical characteristics of ovarian cancer and control samples were downloaded from The Cancer Genome Atlas database (TCGA). The "iClusterPlus". R package was used to cluster the molecular subtypes. The copy number variation of the entire lncRNA genome was analyzed using GISTIC. The prognosis-associated lncRNA related to CNV was screened as potential targets for ovarian cancer. RESULTS Six molecular subtypes were identified based on multi-omics analysis and DElncRNAs are significantly enriched in specific molecular subtypes. The deletion or amplification of lncRNA copy number affects the occurrence and development of ovarian cancer to some extent. Three prognostic-associated lncRNA including LOC101927151, LINC00861 and LEMD1-AS1 were selected. These lncRNAs can be used as biomarkers to predict survival in patients with ovarian cancer. The accuracy of results were verified using the Gene Expression Omnibus (GEO) dataset. CONCLUSION Based on genome-wide copy number variation, prognostic-associated lncRNAs were identified as new biomolecular markers for ovarian cancer.
Collapse
Affiliation(s)
- Mingjun Zheng
- Department of Gynaecology and Obstetrics, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China; Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Yuexin Hu
- Department of Gynaecology and Obstetrics, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Rui Gou
- Department of Gynaecology and Obstetrics, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Xin Nie
- Department of Gynaecology and Obstetrics, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Xiao Li
- Department of Gynaecology and Obstetrics, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Juanjuan Liu
- Department of Gynaecology and Obstetrics, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China
| | - Bei Lin
- Department of Gynaecology and Obstetrics, Shengjing Hospital Affiliated to China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, China.
| |
Collapse
|
26
|
Ovarian cancer: An update on imaging in the era of radiomics. Diagn Interv Imaging 2019; 100:647-655. [DOI: 10.1016/j.diii.2018.11.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 12/13/2022]
|
27
|
Role of Survivin and p53 Expression in Response of Primary Culture of Ovarian Cancer Cells to Treatment With Chemotherapeutic Agents. Int J Gynecol Cancer 2019; 28:1239-1246. [PMID: 29727353 DOI: 10.1097/igc.0000000000001281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Ovarian cancer is associated with a high relapse rate and is the fifth leading cause of cancer deaths in women. The genetic profile of a tumor is responsible for deciding response to chemotherapeutic agents. In this study, we investigate the relation between survivin and p53 expression and response to chemotherapeutic agents of primary cultures of ovarian cancer cells established from ascitic fluid. MATERIALS AND METHOD Ascitic fluid and Dulbecco's modified Eagle medium was mixed in equal proportion in culture flasks and incubated to establish primary culture. The cells were treated with different combinations of carboplatin and paclitaxel with and without survivin small interfering RNA transfection. Cell survival was estimated by MTT assay. Survivin and p53 expression was quantified by real-time polymerase chain reaction. RESULTS Out of 19 ascitic fluid samples, 13 primary cultures of ovarian cancer cells were established. The half maximal inhibitory concentration doses of carboplatin (≥70 μg/mL) and paclitaxel (≥18 μg/mL) were high for 10/13 and 5/13 patients, respectively. Survivin messenger RNA expression was significantly downregulated on treatment with carboplatin (100 μg/mL), paclitaxel (12.5 μg/mL), and a combination of carboplatin (50 μg/mL) and paclitaxel (6.25 μg/mL). Only paclitaxel-treated ovarian cancer cells showed decrease in expression of p53. Survivin small interfering RNA increased sensitivity of the primary cultures to chemotherapeutic agents. CONCLUSIONS The present study highlights the fact that establishing primary cultures from ascitic fluid may help to develop personalized treatment regime for individual patients based on their molecular profile. Our study also shows that supplementing taxols drugs with survivin inhibitors may prove to be beneficial in the treatment of ovarian cancer patients.
Collapse
|
28
|
Sait KHW, Alam Q, Anfinan N, Al-Ghamdi O, Malik A, Noor R, Jahan F, Tarique M. Structure-based virtual screening and molecular docking for the identification of potential novel EGFRkinase inhibitors against ovarian cancer. Bioinformation 2019; 15:287-294. [PMID: 31285646 PMCID: PMC6599442 DOI: 10.6026/97320630015287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 03/31/2019] [Indexed: 12/24/2022] Open
Abstract
Epidermal Growth Factor Receptor (EGFR) is, for the most part, deregulated and over-communicated in ovarian disease, which is legitimately connected with STAT3 enactment that prompts the collection of hostile to apoptotic occasions and along these lines, docetaxel medicate obstruction happens. As to, expanding of docetaxel medicate affectability by focusing on EGFR receptor alongside docetaxel drugs is one of the real techniques in ovarian disease treatment. In this specific circumstance, utilizing atomic recreation considers, the present examination depicted the auxiliary and pragmatic properties of IBS Database mixes as a potential inhibitor of EGFR tyrosine kinase, and furthermore ADMET had researched its Pharmacokinetic profile. As indicated by the outcomes, STOCK1N-98911, STOCK1N- 98869, and STOCK1N-98896 have appeared tremendous restricting vitality by associating with critical build ups in the dynamic site. Natural movement range forecast of these mixes indicated potential anticancer properties by demonstrating important collaboration with EGFR tyrosine kinase. Besides, the investigation is likewise valuable for further clinical based examinations and furthermore for the approval of toxicological and pharmacokinetic contemplate.
Collapse
Affiliation(s)
- Khalid Hussain Wali Sait
- Department of Obstetrics and Gynecology, Gynecology Oncology Unite, Faculty of Medicine, King Abdulaziz University, Jeddah, SaudiArabia
| | - Qamre Alam
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah. Saudi Arabia
| | - Nisrin Anfinan
- Department of Obstetrics andGynecology, Gynecology Oncology Unite, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Othman Al-Ghamdi
- Department of Biological Sciences, Faculty of Science, University of Jeddah, Kingdom of Saudi Arabia
| | - Arshi Malik
- Department of Clinical Biochemistry, College ofMedicine, King Khalid University, Abha, Saudi Arabia
| | - Rana Noor
- Department of Biochemistry, Faculty of Dentistry, Jamia Millia Islamia, JamiaNagar, New Delhi-110025, India
| | - Farheen Jahan
- Department of Biosciences Jamia Millia Islamia, Jamia Nagar, New Delhi-110025, India
| | - Mohammed Tarique
- 8Center forInterdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi-110025, India
| |
Collapse
|
29
|
Activity Behaviors and Physiological Characteristics of Women With Advanced-Stage Ovarian Cancer: A Preliminary Cross-sectional Investigation. Int J Gynecol Cancer 2019; 28:604-613. [PMID: 29369120 DOI: 10.1097/igc.0000000000001197] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Ovarian cancer (OC) survivors experience many disease and treatment adverse effects. However, the impact of OC and its treatment on objective activity behaviors and physiological status have not been examined. The purpose of this study was to compare objectively measured activity behaviors and physiological characteristics of advanced-stage OC survivors to age-matched controls. METHODS Twenty stage III-IV OC survivors and 20 controls completed assessments of activity behaviors (7-day accelerometry), physical function (400-meter walk as indicator of cardiorespiratory fitness, repeated chair rise, 6-meter walking tests), muscle strength (1-repetition maximum and handgrip), body composition (dual-energy x-ray absorptiometry), and musculoskeletal morphology (peripheral quantitative computed tomography). RESULTS Compared with controls, OC survivors spent more time/day in prolonged sedentary bouts (P = 0.039, r = 0.32), had lower cardiorespiratory fitness (P = 0.041, r = 0.33) and upper body strength (P = 0.023, r = 0.37), had higher areal bone mineral content (P = 0.047, r = 0.33) and volumetric trabecular density (P = 0.048, r = 0.31), but were not different in other measures of body composition nor in muscle morphology (P > 0.050). Only 20% (n = 4) of OC survivors accrued 150 minutes/week or greater moderate and vigorous physical activity (MVPA) time in 10-minute bouts or greater. Moderate and vigorous physical activity time/day in 10-minute bouts or greater was strongly associated with cardiorespiratory fitness (P = 0.001, ρ = -0.702) and lower extremity function (P = 0.019, ρ = -0.519) and moderately associated with muscle cross-sectional area (P = 0.035, ρ = 0.473). CONCLUSIONS Posttreatment OC survivors spent more time in prolonged sedentary bouts and had lower cardiorespiratory fitness and upper body strength compared with controls. Moderate and vigorous physical activity was associated with physical function and muscle cross-sectional area. Future studies should test the efficacy of exercise interventions to increase MVPA, reduce sedentary behavior, and increase cardiorespiratory fitness and muscle strength in OC survivors.
Collapse
|
30
|
Ramalho S, Andrade LADA, Filho CC, Natal RDA, Pavanello M, Ferracini AC, Sallum LF, Sarian LO, Derchain S. Role of discoidin domain receptor 2 (DDR2) and microRNA-182 in survival of women with high-grade serous ovarian cancer. Tumour Biol 2019; 41:1010428318823988. [PMID: 30810094 DOI: 10.1177/1010428318823988] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The objective of this study is to evaluate the relationship between discoidin domain receptor 2 (DDR2) and miR-182 expression with response to platinum-based chemotherapy and survival in women with high-grade serous ovarian cancer (HGSOC). We evaluated 78 women with HGSOC stages I-IV, diagnosed between 1996 and 2013, and followed up until 2016. DDR2 expression was assessed using immunohistochemistry on tissue microarray slides. The microRNAs were evaluated by qRT-PCR. DDR2 expression was high in 11 (14.1%) women. PFS was significantly lower in women with FIGO stage I/II - versus III/IV, post-surgery residual disease and high expression of DDR2. Women with postsurgery residual disease, FIGO stage I/II - versus III/IV and DDR2 expression had worse OS, but only post-surgery residual disease remained an independent prognostic factor for worse OS in multivariable analysis. miR-182 expression levels were significantly lower in patients harboring tumors with higher expression of DDR2 (p < 0.001). In this relatively large cohort of women with HSGOC, higher DDR2 expression was associated with lower miR-182 levels and worse PFS, suggesting that these molecules may be associated with mechanisms of HGSOC progression.
Collapse
Affiliation(s)
- Susana Ramalho
- 1 Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Cássio Cardoso Filho
- 1 Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Rodrigo de Andrade Natal
- 3 Laboratory of Investigative and Molecular Pathology (LAPE), Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Marina Pavanello
- 4 Postgraduate Program in Medical Sciences, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Amanda Canato Ferracini
- 4 Postgraduate Program in Medical Sciences, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Luis Felipe Sallum
- 1 Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Luis Otávio Sarian
- 1 Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Sophie Derchain
- 1 Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| |
Collapse
|
31
|
Rodriguez-Aguayo C, Bayraktar E, Ivan C, Aslan B, Mai J, He G, Mangala LS, Jiang D, Nagaraja AS, Ozpolat B, Chavez-Reyes A, Ferrari M, Mitra R, Siddik ZH, Shen H, Yang X, Sood AK, Lopez-Berestein G. PTGER3 induces ovary tumorigenesis and confers resistance to cisplatin therapy through up-regulation Ras-MAPK/Erk-ETS1-ELK1/CFTR1 axis. EBioMedicine 2019; 40:290-304. [PMID: 30655206 PMCID: PMC6411965 DOI: 10.1016/j.ebiom.2018.11.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/07/2018] [Accepted: 11/21/2018] [Indexed: 02/04/2023] Open
Abstract
Background Inflammatory mediator prostaglandin E2–prostaglandin E2 receptor EP3 (PTGER3) signaling is critical for tumor-associated angiogenesis, tumor growth, and chemoresistance. However, the mechanism underlying these effects in ovarian cancer is not known. Methods An association between higher tumoral expression of PTGER3 and shorter patient survival in the ovarian cancer dataset of The Cancer Genome Atlas prompted investigation of the antitumor effects of PTGER3 downmodulation. PTGER3 mRNA and protein levels were higher in cisplatin-resistant ovarian cancer cells than in their cisplatin-sensitive counterparts. Findings Silencing of PTGER3 via siRNA in cancer cells was associated with decreased cell growth and less invasiveness, as well as cell-cycle arrest and increased apoptosis, mediated through the Ras-MAPK/Erk-ETS1-ELK1/CFTR1 axis. Furthermore, sustained PTGER3 silencing with multistage vector and liposomal 2’-F-phosphorodithioate-siRNA–mediated silencing of PTGER3 combined with cisplatin resulted in robust antitumor effects in cisplatin-resistant ovarian cancer models. Interpretation These findings identify PTGER3 as a potential therapeutic target in chemoresistant ovarian cancers expressing high levels of this oncogenic protein. Fund National Institutes of Health/National Cancer Institute, USA.
Collapse
Affiliation(s)
- Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emine Bayraktar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Medical Biology, Faculty of Medicine, University of Gaziantep, Gaziantep 27310, Turkey
| | - Cristina Ivan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Burcu Aslan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Guangan He
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lingegowda S Mangala
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dahai Jiang
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Archana S Nagaraja
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Arturo Chavez-Reyes
- Centro de Investigación y Estudios Avanzados del IPN, Unidad Monterrey, Apodaca, NL, CP. 66600, Mexico
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Rahul Mitra
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zahid H Siddik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Xianbin Yang
- AM Biotechnologies LLC, 12521 Gulf Freeway, Houston, TX 77034, USA
| | - Anil K Sood
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Zheng MJ, Li X, Hu YX, Dong H, Gou R, Nie X, Liu Q, Ying-Ying H, Liu JJ, Lin B. Identification of molecular marker associated with ovarian cancer prognosis using bioinformatics analysis and experiments. J Cell Physiol 2019; 234:11023-11036. [PMID: 30633343 DOI: 10.1002/jcp.27926] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/25/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Ovarian cancer is one of the three major malignant tumors of the female reproductive system, and the mortality associated with ovarian cancer ranks first among gynecologic malignant tumors. The pathogenesis of ovarian cancer is not yet clearly defined but elucidating this process would be of great significance for clinical diagnosis, prevention, and treatment. For this study, we used bioinformatics to identify the key pathogenic genes and reveal the potential molecular mechanisms of ovarian cancer; we used immunohistochemistry to validate them. METHODS We analyzed and integrated four gene expression profiles (GSE14407, GSE18520, GSE26712, and GSE54388), which were downloaded from the Gene Expression Omnibus (GEO) database, with the aim of obtaining a common differentially expressed gene (DEG). Then, we performed Gene Ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway analysis using the Database for Annotation, Visualization, and Integrated Discovery (DAVID). We then established a protein-protein interaction (PPI) network of the DEGs through the Search Tool for the Retrieval of Interacting Genes (STRING) database and selected hub genes. Finally, survival analysis of the hub genes was performed using a Kmplotter online tool. RESULTS A total of 226 DEGs were detected after the analysis of the four gene expression profiles; of these, 87 were upregulated genes and 139 were downregulated. GO analysis results showed that DEGs were significantly enriched in biological processes including the G2/M transition of the mitotic cell cycle, the apoptotic process, cell proliferation, blood coagulation, and positive regulation of the canonical Wnt signaling pathway. KEGG analysis results showed that DEGs were particularly enriched in the cell cycle, the p53 signaling pathway, the Wnt signaling pathway, the Ras signaling pathway, the Rap1 signaling pathway, and tyrosine metabolism. We selected 50 hub genes from the PPI network, which had 147 nodes and 655 edges, and 30 of them were associated with the prognosis of ovarian cancer. We performed immunohistochemistry on phosphoserine aminotransferase 1 (PSAT1). PSAT1 was highly expressed in cancer tissues, and its expression level was related to clinical stage and tissue differentiation in ovarian cancer. A Cox proportional risk model suggested that high expression of PSAT1 and late clinical stage were independent risk factors for survival and prognosis of ovarian cancer patients. CONCLUSION The detection of DEGs using bioinformatics analysis might be crucial to understanding the pathogenesis of ovarian cancer, especially the molecular mechanisms of its development. The association between PSAT1 expression and the occurrence, development, and prognosis of ovarian cancer was further verified by immunohistochemistry. The PSAT1 expression can be used as a prognostic marker to provide a potential target for the diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ming-Jun Zheng
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Xiao Li
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Yue-Xin Hu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Hui Dong
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Rui Gou
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Xin Nie
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Qing Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Hao Ying-Ying
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Juan-Juan Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital Affiliated to China Medical University, Shenyang, China
| |
Collapse
|
33
|
A Physiological Profile of Ovarian Cancer Survivors to Inform Tailored Exercise Interventions and the Development of Exercise Oncology Guidelines. Int J Gynecol Cancer 2018; 27:1560-1567. [PMID: 29036032 DOI: 10.1097/igc.0000000000001044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Physical activity has become increasingly important in supportive cancer care. However, physical activity and exercise guidelines for ovarian cancer survivors remain generic. The aim of this narrative review is to summarize existing data regarding the physiological characteristics (treatment-related adverse effects, concurrent comorbidities, body weight and composition, physical fitness and function, and physical activity behavior) of ovarian cancer survivors to further understanding of their cancer-specific physical activity and exercise needs. We also highlight gaps in the current knowledge base. METHODS We undertook a narrative review of current literature on the physiological status of ovarian cancer survivors. We defined physiological status as treatment-related adverse effects, concurrent comorbidities, body weight and composition, physical fitness and function, and physical activity behavior. RESULTS In addition to disease- and treatment-related symptoms and adverse effects, the majority of ovarian cancer survivors have comorbidities, which may adversely affect treatment effectiveness and safety, as well as survival. Despite high overweight and obesity rates, a large percentage of women are malnourished at diagnosis, with potentially compromised muscle mass and muscle density. Low muscle density at diagnosis and loss of muscle mass during treatment may be associated with worse survival outcomes. A small number of studies have observed impaired physical function and cardiorespiratory fitness in ovarian cancer survivors. The majority of ovarian cancer survivors are insufficiently active or sedentary. CONCLUSIONS Our review suggests that ovarian cancer survivors could benefit from physical activity and exercise oncology interventions aimed at addressing detrimental changes to physiological status due to disease and treatment. However, current knowledge gaps regarding the physiological characteristics of ovarian cancer survivors throughout the entire survivorship spectrum challenge the development of tailored exercise intervention studies and exercise oncology guidelines.
Collapse
|
34
|
Establishment of Primary Cell Culture From Ascitic Fluid and Solid Tumor Obtained From Epithelial Ovarian Carcinoma Patients. Int J Gynecol Cancer 2018; 27:2000-2005. [PMID: 28816710 DOI: 10.1097/igc.0000000000001087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Ovarian cancer is the seventh leading cause of cancer death worldwide. This is mainly due to late diagnosis and high rate of relapse and resistance following chemotherapy. In the present study, we describe simple and cost-effective method to establish primary culture from ascitic fluid and solid tumor obtained from epithelial ovarian carcinoma patient, which may provide a better tool for in vitro testing of drug sensitivity and designing individualized treatment protocol. METHODS Complete Dulbecco modified Eagle medium (DMEM) was prepared by supplementing DMEM with 10% fetal bovine serum and antibiotics (ciprofloxacin and amphotericin B). Establishment of primary culture of ovarian cancer cells from ascites fluid and solid tumor was done by using complete DMEM media. RESULTS Primary cultures of ovarian cancer cells were established from ascitic fluid and solid tumor tissue. Of the 7 ascitic fluid samples, we were able to establish 5 primary cultures of ovarian cancer cells. All the 7 samples were diagnosed as serous papillary adenocarcinoma. Some fibroblasts were also attached to culture flask on day 4; they were removed by exposing them to trypsin for a brief period. On day 7, grape-like clusters were visualized under inverted microscope. The cells became confluent on the 10th and 11th day and showed cobblestone appearance, which is a hallmark of ovarian cancer cells. Senescent irregularly shaped cells that have ceased dividing were seen after 8 to 10 passages. CONCLUSION This study highlights the fact that establishing primary cultures from ascitic fluid or solid tumor tissue may help us to understand the molecular profile of the cancer cells, which allow us to select the best chemotherapeutic agent for ovarian cancer patients and thus take a step toward patient-tailored therapy so that patients are not exposed to drugs to which they are not likely to respond.
Collapse
|
35
|
Health-related quality of life and pelvic floor dysfunction in advanced-stage ovarian cancer survivors: associations with objective activity behaviors and physiological characteristics. Support Care Cancer 2018; 26:2239-2246. [DOI: 10.1007/s00520-018-4069-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/22/2018] [Indexed: 12/23/2022]
|
36
|
Intestinal Permeability and Toxicity of Second-Line Therapeutic Agents in Ovarian Cancer. TUMORI JOURNAL 2018; 94:624-6. [DOI: 10.1177/030089160809400436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
37
|
Palliative Radiation Therapy for Recurrent Ovarian Cancer: Efficacy and Predictors of Clinical Response. Int J Gynecol Cancer 2018; 28:43-50. [DOI: 10.1097/igc.0000000000001139] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
ObjectiveThis study aimed to report response rates and predictors of response to palliative radiotherapy (RT) for recurrent ovarian cancer.Methods/MaterialsDatabase review identified 64 patients with symptomatic ovarian cancer recurrence who received a total of 76 courses of RT for 103 indications from March 2003 to August 2014. Radiotherapy indications were pain (44%), bleeding (32%), obstruction (15%), and other (9%). Responses were categorized as complete, partial, or none; all response (AR) was the sum of complete and partial responses. Response rates were compared using a χ2 test. Multivariate analysis was performed using logistic regression. Patients were followed up for symptom recurrence and death.ResultsResponse rates were significantly higher for pain (AR, 87%) and bleeding (93%) than for obstruction (62%) and other (60%; P < 0.01). Patients treated for pain at nonbony sites had higher response rates (AR 96%) compared with those treated at bony sites (75%; P = 0.04). Patients with clear cell histology had the lowest response rates (AR, 60%) compared with those with serous (82%), endometrioid (95%), or other Müllerian histology (85%; P = 0.01). Platinum status at diagnosis or the time of RT was not associated with response, nor was tumor size or number of prior chemotherapy regimens. On multivariate analysis, histology, RT indication, and RT dose were independent predictors of response (all P < 0.01).ConclusionsPalliative RT provides relief of pain and bleeding in most patients with ovarian cancer recurrence. Patients with symptomatic obstruction, bony involvement, and clear cell histology may experience lower clinical response rates.
Collapse
|
38
|
Loss of PRP4K drives anoikis resistance in part by dysregulation of epidermal growth factor receptor endosomal trafficking. Oncogene 2017; 37:174-184. [PMID: 28892043 PMCID: PMC5770602 DOI: 10.1038/onc.2017.318] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 07/06/2017] [Accepted: 08/04/2017] [Indexed: 12/16/2022]
Abstract
Anoikis acts as a critical barrier to metastasis by inducing cell death upon cancer cell detachment from the extracellular matrix (ECM), thereby preventing tumor cell dissemination to secondary sites. The induction of anoikis requires the lysosomal-mediated downregulation of epidermal growth factor receptors (EGFRs) leading to termination of pro-survival signaling. In this study, we demonstrate that depletion of pre-mRNA splicing factor 4 kinase (PRP4K; also known as PRPF4B) causes dysregulation of EGFR trafficking and anoikis resistance. We also report a novel cytoplasmic localization of PRP4K at the late endosome, and demonstrate both nuclear and cytoplasmic localization in breast, lung and ovarian cancer tissue. Mechanistically, depletion of PRP4K leads to reduced EGFR degradation following cell detachment from the ECM and correlates with increased TrkB, vimentin and Zeb1 expression. As a result, PRP4K loss promotes sustained growth factor signaling and increased cellular resistance to anoikis in vitro and in a novel zebrafish xenotransplantation model of anoikis sensitivity, as well as increased metastasis in a mouse model of ovarian cancer. Thus, PRP4K may serve as a potential biomarker of anoikis sensitivity in ovarian and other epithelial cancers.
Collapse
|
39
|
Anastasi E, Gigli S, Santulli M, Tartaglione S, Ballesio L, Porpora MG, Granato T, Catalano C, Angeloni A, Manganaro L. Role of Galectin-3 Combined with Multi- Detector Contrast Enhanced Computed Tomography in Predicting Disease Recurrence in Patients with Ovarian Cancer. Asian Pac J Cancer Prev 2017; 18:1277-1282. [PMID: 28610414 PMCID: PMC5555535 DOI: 10.22034/apjcp.2017.18.5.1277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Galectin-3 (Gal-3) is an endogenous β-galactoside-binding lectin, playing an important role in the pathogenesis of multiple malignancies. Aim of the study was to evaluate in a group of patients treated for ovarian cancer (EOC), the role of Gal-3 combined with multi-detector contrast-enhanced computed tomography (MDCT), as predictor of recurrence disease. Seventeen follow-up patients with recurrent ovarian cancer and 13 follow-up patients with stable ovarian disease, who performed MDCT at one-year follow-up after cytoreductive treatment, were enrolled. Serum Gal-3 concentrations were determined by using ELISA method. Twenty healthy controls were included in the analysis. Two radiologist blinded to patients status, reviewed MDCT exams, recording the following signs of disease recurrence: local tumor spread, enlarged lymph-nodes, carcinomatosis implants and metastases. We calculated the respective threshold values of Gal-3 identified by ROC curve analysis for each imaging findings related to disease recurrence: lymphoadenopathies 92.45 ng/ml (AUC: 0.81, Se=91% Spe=73%), carcinomatosis 85.95 ng/ml (AUC: 0.93 Se= 93.7%, Spe=92.8%), local tumor spread 99.05 (AUC: 0.90, Se=100%, Spe=73%) and metastasis 99.05ng/ml (AUC: 0,78, Se=100%, Spe=70%). A significant correlation between high Gal-3 serum levels and presence of local tumor spread (n=11/17, p:0.001), carcinomatosis (n=16/17, p:0.00), lymphoadenopathies (n=15/17, p:0.00) and metastasis (n=11/17, p:0.003) related with recurrence disease was observed. Patients with recurrence of ovarian cancer presents higher Gal-3 values compared to women with stable diseases. Gal-3 combined to CECT should be used to improve the monitoring of EOC patients.
Collapse
Affiliation(s)
- Emanuela Anastasi
- Department of Molecular Medicine, "Sapienza" University of Rome, Policlinico Umberto I, Viale Regina Elena 324, 00161 Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lin M, Gao Y, Diefenbach TJ, Shen JK, Hornicek FJ, Park YI, Xu F, Lu TJ, Amiji M, Duan Z. Facial Layer-by-Layer Engineering of Upconversion Nanoparticles for Gene Delivery: Near-Infrared-Initiated Fluorescence Resonance Energy Transfer Tracking and Overcoming Drug Resistance in Ovarian Cancer. ACS APPLIED MATERIALS & INTERFACES 2017; 9:7941-7949. [PMID: 28177223 DOI: 10.1021/acsami.6b15321] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Development of multidrug resistance (MDR) contributes to the majority of treatment failures in clinical chemotherapy. We report facial layer-by-layer engineered upconversion nanoparticles (UCNPs) for near-infrared (NIR)-initiated tracking and delivery of small interfering RNA (siRNA) to enhance chemotherapy efficacy by silencing the MDR1 gene and resensitizing resistant ovarian cancer cells to drug. Layer-by-layer engineered UCNPs were loaded with MDR1 gene-silencing siRNA (MDR1-siRNA) by electrostatic interaction. The delivery vehicle enhances MDR1-siRNA cellular uptake, protects MDR1-siRNA from nuclease degradation, and promotes endosomal escape for silencing the MDR gene. The intrinsic photon upconversion of UCNPs provides an unprecedented opportunity for monitoring intracellular attachment and release of MDR1-siRNA by NIR-initiated fluorescence resonance energy transfer occurs between donor UCNPs and acceptor fluorescence dye-labeled MDR1-siRNA. Enhanced chemotherapeutic efficacy in vitro was demonstrated by cell viability assay. The developed delivery vehicle holds great potential in delivery and imaging-guided tracking of therapeutic gene targets for effective treatment of drug-resistant cancers.
Collapse
Affiliation(s)
| | | | - Thomas J Diefenbach
- Ragon Institute of MGH, Massachusetts Institute of Technology (MIT) and Harvard University , Boston, Massachusetts 02139, United States
| | | | | | - Yong Il Park
- School of Chemical Engineering, Chonnam National University , Gwangju 61186, Republic of Korea
| | | | | | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University , Boston, Massachusetts 02115, United States
| | | |
Collapse
|
41
|
Current and future potential of metallo drugs: Revisiting DNA-binding of metal containing molecules and their diverse mechanism of action. Inorganica Chim Acta 2016. [DOI: 10.1016/j.ica.2016.01.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
42
|
Dungl DA, Maginn EN, Stronach EA. Preventing Damage Limitation: Targeting DNA-PKcs and DNA Double-Strand Break Repair Pathways for Ovarian Cancer Therapy. Front Oncol 2015. [PMID: 26579492 DOI: 10.3389/fonc.2015.00240] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Platinum-based chemotherapy is the cornerstone of ovarian cancer treatment, and its efficacy is dependent on the generation of DNA damage, with subsequent induction of apoptosis. Inappropriate or aberrant activation of the DNA damage response network is associated with resistance to platinum, and defects in DNA repair pathways play critical roles in determining patient response to chemotherapy. In ovarian cancer, tumor cell defects in homologous recombination - a repair pathway activated in response to double-strand DNA breaks (DSB) - are most commonly associated with platinum-sensitive disease. However, despite initial sensitivity, the emergence of resistance is frequent. Here, we review strategies for directly interfering with DNA repair pathways, with particular focus on direct inhibition of non-homologous end joining (NHEJ), another DSB repair pathway. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a core component of NHEJ and it has shown considerable promise as a chemosensitization target in numerous cancer types, including ovarian cancer where it functions to promote platinum-induced survival signaling, via AKT activation. The development of pharmacological inhibitors of DNA-PKcs is on-going, and clinic-ready agents offer real hope to patients with chemoresistant disease.
Collapse
Affiliation(s)
- Daniela A Dungl
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Elaina N Maginn
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Euan A Stronach
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| |
Collapse
|
43
|
Dungl DA, Maginn EN, Stronach EA. Preventing Damage Limitation: Targeting DNA-PKcs and DNA Double-Strand Break Repair Pathways for Ovarian Cancer Therapy. Front Oncol 2015. [PMID: 26579492 DOI: 10.3389/fonc.2015.00240]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Platinum-based chemotherapy is the cornerstone of ovarian cancer treatment, and its efficacy is dependent on the generation of DNA damage, with subsequent induction of apoptosis. Inappropriate or aberrant activation of the DNA damage response network is associated with resistance to platinum, and defects in DNA repair pathways play critical roles in determining patient response to chemotherapy. In ovarian cancer, tumor cell defects in homologous recombination - a repair pathway activated in response to double-strand DNA breaks (DSB) - are most commonly associated with platinum-sensitive disease. However, despite initial sensitivity, the emergence of resistance is frequent. Here, we review strategies for directly interfering with DNA repair pathways, with particular focus on direct inhibition of non-homologous end joining (NHEJ), another DSB repair pathway. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a core component of NHEJ and it has shown considerable promise as a chemosensitization target in numerous cancer types, including ovarian cancer where it functions to promote platinum-induced survival signaling, via AKT activation. The development of pharmacological inhibitors of DNA-PKcs is on-going, and clinic-ready agents offer real hope to patients with chemoresistant disease.
Collapse
Affiliation(s)
- Daniela A Dungl
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Elaina N Maginn
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Euan A Stronach
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| |
Collapse
|
44
|
Eng KH, Hanlon BM, Bradley WH, Szender JB. Prognostic factors modifying the treatment-free interval in recurrent ovarian cancer. Gynecol Oncol 2015; 139:228-35. [PMID: 26383827 PMCID: PMC4630152 DOI: 10.1016/j.ygyno.2015.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 09/09/2015] [Accepted: 09/12/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVES While primary treatment for high-grade serous ovarian cancer tends to be uniform - maximal debulking and platinum/taxane adjuvant chemotherapy - there is little standardization of treatment in the recurrent setting beyond the exhaustive use of platinum therapies. Using secondary data from multiple centers participating in the Cancer Genome Atlas study (TCGA), we seek to characterize clinical features, timing and serial response data to provide empirical evidence for treatment expectations in the recurrent setting. METHODS We conducted a retrospective survival analysis of TCGA study primary and secondary patient chemotherapy regimens by characterizing the dynamics of 1119 lines of therapy comprising the treatment of 461 high-grade serous ovarian cancer patients. All patients with post-surgical drug therapy information from the TCGA database were included in this study. RESULTS A complete response to adjuvant therapy led to longer overall survival, but did not affect treatment free intervals (TFIs) after relapse of disease. A strong predictor of the TFI on the next treatment regimen was the previous TFI with a decaying effect. The number of previous treatments, of platinum treatments, and the length of time from surgery all have an exponentially decreasing effect on TFI. Re-treatment times appear to cluster at predictable times following surgery. CONCLUSIONS While patients experience a consistent reduction in TFI with increasing re-treatment, the initial adjuvant interval is unrelated to later interval lengths. Platinum re-treatment remained an effective option in patients typically thought to be platinum resistant and the timing of monitoring visits may drive overall re-treatment patterns.
Collapse
MESH Headings
- Age Factors
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carboplatin/administration & dosage
- Chemotherapy, Adjuvant
- Cisplatin/administration & dosage
- Cytoreduction Surgical Procedures
- Deoxycytidine/administration & dosage
- Deoxycytidine/analogs & derivatives
- Disease-Free Survival
- Docetaxel
- Doxorubicin/administration & dosage
- Female
- Humans
- Middle Aged
- Multivariate Analysis
- Neoplasm Grading
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/therapy
- Neoplasm, Residual
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Neoplasms, Cystic, Mucinous, and Serous/therapy
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/therapy
- Paclitaxel/administration & dosage
- Prognosis
- Proportional Hazards Models
- Retrospective Studies
- Survival Analysis
- Taxoids/administration & dosage
- Time Factors
- Topotecan/administration & dosage
- Gemcitabine
Collapse
Affiliation(s)
- Kevin H Eng
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, United States.
| | - Bret M Hanlon
- Department of Statistics, University of Wisconsin-Madison, Madison, WI, United States
| | - William H Bradley
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - J Brian Szender
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY, United States
| |
Collapse
|
45
|
Dungl DA, Maginn EN, Stronach EA. Preventing Damage Limitation: Targeting DNA-PKcs and DNA Double-Strand Break Repair Pathways for Ovarian Cancer Therapy. Front Oncol 2015; 5:240. [PMID: 26579492 PMCID: PMC4620694 DOI: 10.3389/fonc.2015.00240] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/10/2015] [Indexed: 12/22/2022] Open
Abstract
Platinum-based chemotherapy is the cornerstone of ovarian cancer treatment, and its efficacy is dependent on the generation of DNA damage, with subsequent induction of apoptosis. Inappropriate or aberrant activation of the DNA damage response network is associated with resistance to platinum, and defects in DNA repair pathways play critical roles in determining patient response to chemotherapy. In ovarian cancer, tumor cell defects in homologous recombination – a repair pathway activated in response to double-strand DNA breaks (DSB) – are most commonly associated with platinum-sensitive disease. However, despite initial sensitivity, the emergence of resistance is frequent. Here, we review strategies for directly interfering with DNA repair pathways, with particular focus on direct inhibition of non-homologous end joining (NHEJ), another DSB repair pathway. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a core component of NHEJ and it has shown considerable promise as a chemosensitization target in numerous cancer types, including ovarian cancer where it functions to promote platinum-induced survival signaling, via AKT activation. The development of pharmacological inhibitors of DNA-PKcs is on-going, and clinic-ready agents offer real hope to patients with chemoresistant disease.
Collapse
Affiliation(s)
- Daniela A Dungl
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Elaina N Maginn
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| | - Euan A Stronach
- Molecular Therapy Laboratory, Department of Surgery and Cancer, Ovarian Cancer Action Research Centre, Imperial College London , London , UK
| |
Collapse
|
46
|
Evangelista L, Palma MD, Gregianin M, Nardin M, Roma A, Nicoletto MO, Nardelli GB, Zagonel V. Diagnostic and prognostic evaluation of fluorodeoxyglucose positron emission tomography/computed tomography and its correlation with serum cancer antigen-125 (CA125) in a large cohort of ovarian cancer patients. J Turk Ger Gynecol Assoc 2015; 16:137-44. [PMID: 26401105 DOI: 10.5152/jtgga.2015.15251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/16/2015] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE We evaluated the efficacy of 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) in recurrent disease, response to therapy, and long-term follow-up of ovarian cancer (OC) patients in relation to cancer antigen-125 (CA125) levels and the prognostic meaning of this modality in this subset of subjects. MATERIAL AND METHODS Between 2005 and 2015, we retrospectively evaluated 125 patients affected by OC who underwent FDG PET/CT imaging at our institution. The indications for PET/CT were recurrence of disease in 78 patients, therapy response assessment in 29, and follow-up in 18. The results of FDG PET/CT were compared with those of histopathology and clinical and radiological progression during follow-up for at least 6 months. The median long-term follow-up was 33 months. The diagnostic accuracies for the different clinical settings were evaluated. The relationships among global survival (GS), FDG PET/CT results, and CA125 levels were evaluated by both Kaplan-Meier and Cox regression analysis. RESULTS CA125 results were positive (>35 UI/mL) in 62 patients and negative in 63 (49% vs. 51%). The sensitivity and specificity of CA125 were 72% and 91%, respectively. PET/CT imaging showed a sensitivity of 98.6% and a specificity of 77.8% for the assessment of recurrent disease, and a sensitivity of 72.7% and a specificity of 88.9% for therapy evaluation. Meanwhile, in 18 patients evaluated during follow-up, the specificity was 82.3%. GS was significantly higher in case of negative CA125 values at the time of FDG PET/CT, of a negative PET/CT scan and when no evidence of peritoneum recurrence and distant metastases was determined by PET. Multivariate regression analysis showed that only age and peritoneum recurrence as determined by PET were identified as independent predictors of poor prognosis. CONCLUSION Metabolic imaging with FDG PET/CT proved useful in patients where OC recurrence was suspected, even when the value of tumor marker CA125 was in a normal range. A positive PET/CT scan and the presence of peritoneum recurrence at PET were associated with a poor prognosis after approximately 30 months.
Collapse
Affiliation(s)
- Laura Evangelista
- Radiotherapy and Nuclear Medicine Unit, Istituto Oncologico VenetoIOV - IRCCS, Padova, Italy
| | | | - Michele Gregianin
- Radiotherapy and Nuclear Medicine Unit, Istituto Oncologico VenetoIOV - IRCCS, Padova, Italy
| | - Margherita Nardin
- Oncological Radiology Unit, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Anna Roma
- Medical Oncology Unit, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | | | | | - Vittorina Zagonel
- Medical Oncology Unit, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| |
Collapse
|
47
|
Reyes-González JM, Armaiz-Peña GN, Mangala LS, Valiyeva F, Ivan C, Pradeep S, Echevarría-Vargas IM, Rivera-Reyes A, Sood AK, Vivas-Mejía PE. Targeting c-MYC in Platinum-Resistant Ovarian Cancer. Mol Cancer Ther 2015; 14:2260-9. [PMID: 26227489 DOI: 10.1158/1535-7163.mct-14-0801] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 07/19/2015] [Indexed: 11/16/2022]
Abstract
The purpose of this study was to investigate the molecular and therapeutic effects of siRNA-mediated c-MYC silencing in cisplatin-resistant ovarian cancer. Statistical analysis of patient's data extracted from The Cancer Genome Atlas (TCGA) portal showed that the disease-free (DFS) and the overall (OS) survival were decreased in ovarian cancer patients with high c-MYC mRNA levels. Furthermore, analysis of a panel of ovarian cancer cell lines showed that c-MYC protein levels were higher in cisplatin-resistant cells when compared with their cisplatin-sensitive counterparts. In vitro cell viability, growth, cell-cycle progression, and apoptosis, as well as in vivo therapeutic effectiveness in murine xenograft models, were also assessed following siRNA-mediated c-MYC silencing in cisplatin-resistant ovarian cancer cells. Significant inhibition of cell growth and viability, cell-cycle arrest, and activation of apoptosis were observed upon siRNA-mediated c-MYC depletion. In addition, single weekly doses of c-MYC-siRNA incorporated into 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG-2000)-based nanoliposomes resulted in significant reduction in tumor growth. These findings identify c-MYC as a potential therapeutic target for ovarian cancers expressing high levels of this oncoprotein.
Collapse
Affiliation(s)
- Jeyshka M Reyes-González
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Guillermo N Armaiz-Peña
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lingegowda S Mangala
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fatma Valiyeva
- University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico
| | - Cristina Ivan
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunila Pradeep
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Adrian Rivera-Reyes
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, Puerto Rico
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pablo E Vivas-Mejía
- Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico. University of Puerto Rico Comprehensive Cancer Center, San Juan, Puerto Rico.
| |
Collapse
|
48
|
Yang X, Shen J, Gao Y, Feng Y, Guan Y, Zhang Z, Mankin H, Hornicek FJ, Duan Z. Nsc23925 prevents the development of paclitaxel resistance by inhibiting the introduction of P-glycoprotein and enhancing apoptosis. Int J Cancer 2015; 137:2029-39. [PMID: 25904021 DOI: 10.1002/ijc.29574] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/13/2015] [Accepted: 04/14/2015] [Indexed: 12/12/2022]
Abstract
Strategies to prevent the emergence of drug resistance will increase the effectiveness of chemotherapy treatment and prolong survival of women with ovarian cancer. The aim of our study is to determine the effects of NSC23925 on preventing the development of paclitaxel resistance in ovarian cancer both in cultured cells in vitro and in mouse xenograft models in vivo, and to further elucidate these underlying mechanisms. We first developed a paclitaxel-resistant ovarian cancer cell line, and demonstrated that NSC23925 could prevent the introduction of paclitaxel resistance by specifically inhibiting the overexpression of P-glycoprotein (Pgp) in vitro. The paclitaxel-resistant ovarian cancer cells were then established in a mouse model by continuous paclitaxel treatment in combination with or without NSC23925 administration in the mice. The majority of mice continuously treated with paclitaxel alone eventually developed paclitaxel resistance with overexpression of Pgp and antiapoptotic proteins, whereas mice remained sensitivity to paclitaxel and displayed lower expression levels of Pgp and antiapoptotic proteins after administered continuously with combination of paclitaxel-NSC23925. Paclitaxel-NSC23925-treated mice experienced significantly longer overall survival time than paclitaxel-treated mice. Furthermore, the combination of paclitaxel and NSC23925 therapy did not induce obvious toxicity as measured by mice body weight changes, blood cell counts and histology of internal organs. Collectively, our observations provide evidence that NSC23925 in combination with paclitaxel may prevent the onset of Pgp or antiapoptotic-mediated paclitaxel resistance, and improve the long-term clinical outcome in patients with ovarian cancer.
Collapse
Affiliation(s)
- Xiaoqian Yang
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jacson Shen
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Yan Gao
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Yong Feng
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Yichun Guan
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhan Zhang
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Henry Mankin
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
49
|
OVCAR-3 spheroid-derived cells display distinct metabolic profiles. PLoS One 2015; 10:e0118262. [PMID: 25688563 PMCID: PMC4331360 DOI: 10.1371/journal.pone.0118262] [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: 05/21/2014] [Accepted: 01/07/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction Recently, multicellular spheroids were isolated from a well-established epithelial ovarian cancer cell line, OVCAR-3, and were propagated in vitro. These spheroid-derived cells displayed numerous hallmarks of cancer stem cells, which are chemo- and radioresistant cells thought to be a significant cause of cancer recurrence and resultant mortality. Gene set enrichment analysis of expression data from the OVCAR-3 cells and the spheroid-derived putative cancer stem cells identified several metabolic pathways enriched in differentially expressed genes. Before this, there had been little previous knowledge or investigation of systems-scale metabolic differences between cancer cells and cancer stem cells, and no knowledge of such differences in ovarian cancer stem cells. Methods To determine if there were substantial metabolic changes corresponding with these transcriptional differences, we used two-dimensional gas chromatography coupled to mass spectrometry to measure the metabolite profiles of the two cell lines. Results These two cell lines exhibited significant metabolic differences in both intracellular and extracellular metabolite measurements. Principal components analysis, an unsupervised dimensional reduction technique, showed complete separation between the two cell types based on their metabolite profiles. Pathway analysis of intracellular metabolomics data revealed close overlap with metabolic pathways identified from gene expression data, with four out of six pathways found enriched in gene-level analysis also enriched in metabolite-level analysis. Some of those pathways contained multiple metabolites that were individually statistically significantly different between the two cell lines, with one of the most broadly and consistently different pathways, arginine and proline metabolism, suggesting an interesting hypothesis about cancerous and stem-like metabolic phenotypes in this pair of cell lines. Conclusions Overall, we demonstrate for the first time that metabolism in an ovarian cancer stem cell line is distinct from that of more differentiated isogenic cancer cells, supporting the potential importance of metabolism in the differences between cancer cells and cancer stem cells.
Collapse
|
50
|
Hung SW, Marrache S, Cummins S, Bhutia YD, Mody H, Hooks SB, Dhar S, Govindarajan R. Defective hCNT1 transport contributes to gemcitabine chemoresistance in ovarian cancer subtypes: overcoming transport defects using a nanoparticle approach. Cancer Lett 2015; 359:233-40. [PMID: 25600708 DOI: 10.1016/j.canlet.2015.01.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/14/2015] [Accepted: 01/14/2015] [Indexed: 11/30/2022]
Abstract
Nucleoside analogs are used as chemotherapeutic options for the treatment of platinum-resistant ovarian cancers. Human concentrative nucleoside transporter 1 (hCNT1) is implicated in sensitizing solid tumors to nucleoside analogs although its role in determining drug efficacy in ovarian cancers remains unclear. Here we examined the functional expression of hCNT1 and compared its contributions toward gemcitabine efficacy in histological subtypes of ovarian cancer. Radioactivity analysis identified hCNT1-mediated (3)H-gemcitabine transport in ovarian cancer cells to be significantly reduced compared with that of normal ovarian surface epithelial cells. Biochemical and immunocytochemical analysis identified that unlike normal ovarian cells which expressed high levels of hCNT1 at the apical cell surface, the transporter was either diminished in expression and/or mislocalized in cell lines of various subtypes of ovarian cancer. Retroviral expression of hCNT1 selectively rescued gemcitabine transport in cell lines representing serous, teratocarcinoma, and endometrioid subtypes, but not clear cell carcinoma (CCC). In addition, exogenous hCNT1 predominantly accumulated in intracytoplasmic vesicles in CCC suggesting defective cellular trafficking of hCNT1 as a contributing factor to transport deficiency. Despite diminution of hCNT1 transport in the majority of ovarian cancers and apparent trafficking defects with CCC, the chemotherapeutic efficacy of gemcitabine was broadly enhanced in all subtypes when delivered via engineered nanoparticles (NPs). Additionally, by bypassing the transport requirement, the delivery of a gemcitabine-cisplatin combination in NP formulation increased their synergistic interactions. These findings uncover hCNT1 as a putative determinant for nucleoside analog chemoresistance in ovarian cancer and may help rationalize drug selection and delivery strategies for various histological subtypes of ovarian cancer.
Collapse
Affiliation(s)
- Sau Wai Hung
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Sean Marrache
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Shannon Cummins
- Department of Biological Sciences, University of Georgia, Athens, GA, USA
| | - Yangzom D Bhutia
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Hardik Mody
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Shelley B Hooks
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Shanta Dhar
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Rajgopal Govindarajan
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA.
| |
Collapse
|