1
|
Manjili DA, Babaei FN, Younesirad T, Ghadir S, Askari H, Daraei A. Dysregulated circular RNA and long non-coding RNA-Mediated regulatory competing endogenous RNA networks (ceRNETs) in ovarian and cervical cancers: A non-coding RNA-Mediated mechanism of chemotherapeutic resistance with new emerging clinical capacities. Arch Biochem Biophys 2025; 768:110389. [PMID: 40090441 DOI: 10.1016/j.abb.2025.110389] [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: 11/11/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Cervical cancer (CC) and ovarian cancer (OC) are among the most common gynecological cancers with significant mortality in women, and their incidence is increasing. In addition to the prominent role of the malignant aspect of these cancers in cancer-related women deaths, chemotherapy drug resistance is a major factor that contributes to their mortality and presents a clinical obstacle. Although the exact mechanisms behind the chemoresistance in these cancers has not been revealed, accumulating evidence points to the dysregulation of non-coding RNAs (ncRNAs), particularly long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), as key contributors. These ncRNAs perform the roles of regulators of signaling pathways linked to tumor formation and chemoresistance. Strong data from various recent studies have uncovered that the main mechanism of these ncRNAs in the induction of chemoresistance of CC and OC is done through a dysregulated miRNA sponge activity as competing endogenous RNA (ceRNA) in the competing endogenous RNA networks (ceRNETs), where a miRNA regulating a messenger RNA (mRNA) is trapped, thereby removing its inhibitory effect on the desired mRNA. Understanding these mechanisms is essential to enhancing treatment outcomes and managing the problem of drug resistance. This review provides a comprehensive overview of lncRNA- and circRNA-mediated ceRNETs as the core process of chemoresistance against the commonly used chemotherapeutics, including cisplatin, paclitaxel, oxaliplatin, carboplatin, and docetaxel in CC and OC. Furthermore, we highlight the clinical potential of these ncRNAs serving as diagnostic indicators of chemotherapy responses and therapeutic targets.
Collapse
Affiliation(s)
- Danial Amiri Manjili
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Naghdi Babaei
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tayebeh Younesirad
- Department of Medical Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Sara Ghadir
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hamid Askari
- Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
2
|
Zhang LP, Yang X, Zheng W, Feng KX, Li H. Exploration of chemotherapy-free regimen after multi-line chemotherapy-induced renal impairment in recurrent ovarian cancer: Case report and literature review. Front Oncol 2023; 12:1031045. [PMID: 36741732 PMCID: PMC9892535 DOI: 10.3389/fonc.2022.1031045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023] Open
Abstract
Introduction Platinum-based combination chemotherapy is recommended first choice for relapsed ovarian cancer. However, many of the chemotherapeutic agents are nephrotoxic and can promote kidney dysfunction, which affect the efficacy of cancer treatment and the survival of the patient. There is a need to explore long-term treatments of chemotherapy-free regimen of chronic kidney disease in recurrent ovarian cancer. Case presentation A 41-year-old female patient was presented with stage IIIC well-differentiated ovarian serous papillary adenocarcinoma in 2009. The patient had recurrence of platinum resistance after secondary cytoreductive surgery, and it was difficult to continue chemotherapy after multiple lines of chemotherapy due to myelosuppression, renal impairment and other factors. The patient accepted Niraparib-based treatment regimen after multi-line chemotherapy-induced stage 4 chronic kidney disease. Niraparib combined with anlotinib achieved median PFS of 11 months, disease re-progression, and the patient was switched to niraparib combined with letrozole from October 2021. No evidence of tumor progression was observed till date and the renal toxicity is acceptable. Conclusions In patients with relapsed ovarian cancer, treatment becomes increasingly challenging to subsequent therapies because of renal impairment and emerging drug resistance. Niraparib-based treatment regimen may be a good choice for patients with well-differentiated serous adenocarcinoma of the ovary who are intolerant to chemotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Hu Li
- Department of Gynecology, Guangzhou Panyu Central Hospital, Panyu Cancer Institute, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Yang X, Li Z, Ren H, Peng X, Fu J. New progress of glutamine metabolism in the occurrence, development, and treatment of ovarian cancer from mechanism to clinic. Front Oncol 2022; 12:1018642. [PMID: 36523985 PMCID: PMC9745299 DOI: 10.3389/fonc.2022.1018642] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 10/31/2022] [Indexed: 11/15/2023] Open
Abstract
Glutamine is a non-essential amino acid that can be synthesized by cells. It plays a vital role in the growth and proliferation of mammalian cells cultured in vitro. In the process of tumor cell proliferation, glutamine not only contributes to protein synthesis but also serves as the primary nitrogen donor for purine and pyrimidine synthesis. Studies have shown that glutamine-addicted tumor cells depend on glutamine for survival and reprogram glutamine utilization through the Krebs cycle. Potential therapeutic approaches for ovarian cancer including blocking the entry of glutamine into the tricarboxylic acid cycle in highly aggressive ovarian cancer cells or inhibiting glutamine synthesis in less aggressive ovarian cancer cells. Glutamine metabolism is associated with poor prognosis of ovarian cancer. Combining platinum-based chemotherapy with inhibition of glutamine metabolic pathways may be a new strategy for treating ovarian cancer, especially drug-resistant ovarian cancer. This article reviews the role of glutamine metabolism in the biological behaviors of ovarian cancer cells, such as proliferation, invasion, and drug resistance. Its potential use as a new target or biomarker for ovarian cancer diagnosis, treatment, and the prognosis is investigated.
Collapse
Affiliation(s)
- Xiaojing Yang
- Department of Radiation Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Li
- Department of Radiation Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanru Ren
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China
| | - Xue Peng
- Department of Breast Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Fu
- Department of Radiation Oncology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Ma'rifah AR, Afiyanti Y, Huda MH, Chipojola R, Putri YR, Nasution MAT. Effectiveness of psychoeducation intervention among women with gynecological cancer: a systematic review and meta-analysis of randomized controlled trials. Support Care Cancer 2022; 30:8271-8285. [PMID: 35821448 DOI: 10.1007/s00520-022-07277-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/03/2022] [Indexed: 12/24/2022]
Abstract
Psychoeducation interventions have been suggested to improve sexual functioning and quality of life. The aim of this study was to examine the effects of the psychoeducation intervention on quality of life, sexual function, and psychological outcomes; and to identify the critical components of psychoeducation intervention for women with gynecological cancer. This study was a systematic review and meta-analysis. Psychoeducation interventions were searched using six English databases, eligible studies were extracted, and the risk of bias was evaluated by two authors independently. A random effects model was used to examine the intervention effects. We conducted subgroup analysis and meta-regression to assess the variables underlying the heterogeneity. In total, eight trials were identified, and 1128 participants were included. Women who received the psychoeducation intervention had better quality of life (standardized mean difference (SMD) = 0.59, 95% confidence interval (CI): 0.22, 0.97), sexual functioning (SMD = 0.63, 95% CI: 0.27, 0.99), and psychological outcomes (i.e., anxiety and depression (SMD = - 0.27, 95% CI: - 0.09, - 0.44)). Interventions that combined lecture-based, skill practical, discussion, and counseling were conducted using an online format and were delivered by a team of health professionals that had higher scores of qualities of life. Additionally, involving significant others in the intervention improved sexual functioning. Psychoeducation interventions effectively improve quality of life, sexual functioning, anxiety, and depression. Incorporating lecture-based, practical skills, discussion, and counseling through an online format and involving significant others are beneficial for improving the quality of life and sexual functioning among women with gynecological cancer.
Collapse
Affiliation(s)
- Atun Raudotul Ma'rifah
- Faculty of Nursing, Universitas Indonesia, FIK UI Campus, Jl. Prof. Dr. Bahder DjohanJawa Barat, Depok, Indonesia, 16424.,Harapan Bangsa University Purwokerto, Jl. Raden Patah no 100 Ledug Kembaran Banyumas, Central Java, Indonesia
| | - Yati Afiyanti
- Faculty of Nursing, Universitas Indonesia, FIK UI Campus, Jl. Prof. Dr. Bahder DjohanJawa Barat, Depok, Indonesia, 16424.
| | - Mega Hasanul Huda
- Institute Kesehatan Prima Nusantara Bukit Tinggi, Hermina Hospital Group, Gn. Sahari Sel, Jl. Selangit, RW.10, Kec. Kemayoran, Central Jakarta, Capital Region of Jakarta, Indonesia, 10620
| | | | - Yelmi Reni Putri
- Faculty of Nursing, Universitas Indonesia, FIK UI Campus, Jl. Prof. Dr. Bahder DjohanJawa Barat, Depok, Indonesia, 16424.,Fort De Kock University, Jl. Soekarno Hatta No. 11, Manggis Ganting, Mandingin, Kota Selayan-Kota Bukit Tinggi West Sumatra, Indonesia
| | - M A Tantawi Nasution
- National Library of the Republik of Indonesia, Salemba Raya, 28 A. Kenari, Senen, Central Jakarta, Capital Region of Jakarta, Indonesia
| |
Collapse
|
5
|
Zhang R, Roque DM, Reader J, Lin J. Combined inhibition of IL‑6 and IL‑8 pathways suppresses ovarian cancer cell viability and migration and tumor growth. Int J Oncol 2022; 60:50. [PMID: 35315502 PMCID: PMC8973967 DOI: 10.3892/ijo.2022.5340] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 10/13/2021] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological cancer type in the United States. The success of current chemotherapies is limited by chemoresistance and side effects. Targeted therapy is a promising future direction for cancer therapy. In the present study, the efficacy of co‑targeting IL‑6 and IL‑8 in human ovarian cancer cells by bazedoxifene (Baze) + SCH527123 (SCH) treatment was examined. ELISA, cell viability, cell proliferation, cell migration, cell invasion, western blotting and peritoneal ovarian tumor mouse model analyses were performed to analyze the expression levels of IL‑6 and IL‑8, tumor growth, tumor migration and invasion, and the possible pathways of human ovarian cancer cell lines (SKOV3, CAOV3 and OVCAR3) and patient‑derived OV75 ovarian cancer cells. Each cell line was treated by monotherapy or combination therapy. The results demonstrated that IL‑6 and IL‑8 were secreted by human ovarian cancer cell lines. Compared with the DMSO control, the combination of IL‑6/glycoprotein 130 inhibitor Baze and IL‑8 inhibitor SCH synergistically inhibited cell viability in ovarian cancer cells. Baze + SCH also inhibited cell migration and invasion, suppressed ovarian tumor growth and inhibited STAT3 and AKT phosphorylation, as well as survivin expression. Therefore, co‑targeting the IL‑6 and IL‑8 signaling pathways may be an effective approach for ovarian cancer treatment.
Collapse
Affiliation(s)
- Ruijie Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Dana M Roque
- Division of Gynecologic Oncology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jocelyn Reader
- Division of Gynecologic Oncology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
6
|
Yang LQ, Hu HY, Han Y, Tang ZY, Gao J, Zhou QY, Liu YX, Chen HS, Xu TN, Ao L, Xu Y, Che X, Jiang YB, Xu CW, Zhang XC, Jiang YX, Heger M, Wang XM, Cheng SQ, Pan WW. CpG-binding protein CFP1 promotes ovarian cancer cell proliferation by regulating BST2 transcription. Cancer Gene Ther 2022; 29:1895-1907. [PMID: 35864225 PMCID: PMC9750859 DOI: 10.1038/s41417-022-00503-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/28/2022] [Accepted: 06/28/2022] [Indexed: 02/07/2023]
Abstract
Epigenetic alterations have been functionally linked to ovarian cancer development and occurrence. The CXXC zinc finger protein 1 (CFP1) is an epigenetic regulator involved in DNA methylation and histone modification in mammalian cells. However, its role in ovarian cancer cells is unknown. Here, we show that CFP1 protein is highly expressed in human ovarian cancer tissues. Loss of CFP1 inhibited the growth of human ovarian cancer cells, promoted apoptosis, and increased senescence. CFP1 knockdown resulted in reduced levels of SETD1 (a CFP1 partner) and histone H3 trimethylation at the fourth lysine residue (H3K4me3). RNA-sequencing revealed that deletion of CFP1 resulted in mRNA reduction of bone marrow stromal cell antigen 2 (BST2). Bioinformatics analysis and chromatin immunoprecipitation showed that CFP1 binds to the promoter of BST2 and regulates its transcription directly. Overexpression of BST2 rescued the growth inhibitory effect of CFP1 loss. Furthermore, depletion of cullin-RING ubiquitin ligases 4 (CRL4) components ROC1 or CUL4A had significantly inhibited the expression of CFP1 and BST2 similar to MLN4924 treatment that blocked cullin neddylation and inactivated CRL4s. In conclusion, CFP1 promotes ovarian cancer cell proliferation and apoptosis by regulating the transcription of BST2, and the expression of CFP1 was affected by CRL4 ubiquitin ligase complex.
Collapse
Affiliation(s)
- Liu-Qing Yang
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Han-Yin Hu
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Yao Han
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Ze-Yi Tang
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Jie Gao
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Qi-Yin Zhou
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Yi-Xuan Liu
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Hao-Sa Chen
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Tu-Nan Xu
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Lei Ao
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Ying Xu
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Xuan Che
- grid.411870.b0000 0001 0063 8301Department of Anesthesiology, Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children Hospital, Jiaxing University, Jiaxing, 314001 Zhejiang Province China
| | - Ya-Bo Jiang
- grid.73113.370000 0004 0369 1660Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China
| | - Chun-Wei Xu
- grid.256112.30000 0004 1797 9307Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, 350014 Fuzhou, Fujian China
| | - Xian-Chao Zhang
- grid.411870.b0000 0001 0063 8301Institute of Information Network and Artificial Intelligence, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Yu-Xin Jiang
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Michal Heger
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China ,grid.5477.10000000120346234Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands ,grid.5645.2000000040459992XLaboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, the Netherlands
| | - Xiao-Min Wang
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Shu-Qun Cheng
- grid.73113.370000 0004 0369 1660Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai, 200438 China ,grid.411870.b0000 0001 0063 8301G60 STI Valley Industry & Innovation Institute, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| | - Wei-Wei Pan
- grid.411870.b0000 0001 0063 8301Department of Cell Biology, College of Medicine, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China ,grid.411870.b0000 0001 0063 8301G60 STI Valley Industry & Innovation Institute, Jiaxing University, 118 Jiahang Road, Jiaxing, 314001 China
| |
Collapse
|
7
|
Li XF, Sun HY, Hua T, Zhang HB, Tian YJ, Li Y, Kang S. Promoter Methylation of the MGRN1 Gene Predicts Prognosis and Response to Chemotherapy of High-Grade Serous Ovarian Cancer Patients. Front Oncol 2021; 11:659254. [PMID: 34268111 PMCID: PMC8277380 DOI: 10.3389/fonc.2021.659254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/02/2021] [Indexed: 01/24/2023] Open
Abstract
Aberrant DNA methylation is considered to play a critical role in the chemoresistance of epithelial ovarian cancer (EOC). In this study, we explored the relationship between hypermethylation of the Mahogunin Ring Finger 1 (MGRN1) gene promoter and primary chemoresistance and clinical outcomes in high-grade serous ovarian cancer (HGSOC) patients. The MALDI-TOF mass spectrometry assays revealed a strong association between hypermethylation of the MGRN1 upstream region and platinum resistance in HGSOC patients. Spearman’s correlation analysis revealed a significantly negative connection between the methylation level of MGRN1 and its expression in HGSOC. In vitro analysis demonstrated that knockdown of MGRN1 reduced the sensitivity of cells to cisplatin and that expression of EGR1 was significantly decreased in SKOV3 cells with low levels of MGRN1 expression. Similarly, EGR1 mRNA expression was lower in platinum-resistant HGSOC patients and was positively correlated with MGRN1 mRNA expression. Kaplan-Meier analyses showed that high methylation of the MGRN1 promoter region and low expression of MGRN1 were associated with worse survival of HGSOC patients. In multivariable models, low MGRN1 expression was an independent factor predicting poor outcome. Furthermore, low expression of EGR1 was also been confirmed to be significantly related to the poor prognosis of HGSOC patients by Kaplan-Meier. The hypermethylation of the MGRN1 promoter region and low expression of MGRN1 were associated with platinum resistance and poor outcomes in HGSOC patients, probably by altering EGR1 expression.
Collapse
Affiliation(s)
- Xiao-Fei Li
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Hai-Yan Sun
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Tian Hua
- Department of Obstetrics and Gynaecology, Affiliated Xing Tai People Hospital of Hebei Medial University, Xingtai, China
| | - Hai-Bo Zhang
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Yun-Jie Tian
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Yan Li
- Department of Molecular Biology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Shan Kang
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| |
Collapse
|
8
|
Li XF, Hua T, Li Y, Tian YJ, Huo Y, Kang S. The HSP70 gene predicts prognosis and response to chemotherapy in epithelial ovarian cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:806. [PMID: 34268419 PMCID: PMC8246186 DOI: 10.21037/atm-21-2087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/08/2021] [Indexed: 12/21/2022]
Abstract
Background Chemotherapy resistance is an intractable problem in treating patients with epithelial ovarian cancer (EOC). Heat shock proteins (HSPs) act as apoptosis inhibitors and are highly conserved genetically. Most HSPs have strong cytoprotective effects, and their overexpression inhibits apoptosis. This has been demonstrated for HSP70. Heat shock protein 70 (HSP70) expression is abnormally upregulated in malignant cells. Furthermore, HSP70 can inhibit cell death and promote chemotherapeutic resistance. In our study, the relationship between the HSP70 gene and primary chemotherapy resistance and clinical outcome in patients with EOC was explored. Methods Quantitative real-time polymerase chain (qRT-PCR) was applied to determine HSP70 messenger RNA (mRNA) levels, and immunohistochemistry assay was conducted to determine HSP70 protein level. HSP70 overexpression was assessed to clarify its role on chemotherapy resistance to cisplatin in SKOV3 cell lines. Results RT-qPCR assay indicated a strong relationship between HSP70 expression and chemotherapy resistance in patients with EOC. In cultured SKOV3 cells, overexpression of HSP70 inhibited cell sensitivity to cisplatin. Kaplan-Meier analysis demonstrated high HSP70 expression was associated with poor outcome of EOC patients. In multivariate models, high HSP70 expression independently predicted this poor outcome. Conclusions HSP70 predicts the prognosis and response to chemotherapy in EOC patients.
Collapse
Affiliation(s)
- Xiao-Fei Li
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tian Hua
- Department of Obstetrics and Gynecology, Affiliated Xing Tai People Hospital of Hebei Medial University, Xingtai, China
| | - Yan Li
- Department of Molecular Biology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yun-Jie Tian
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Huo
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shan Kang
- Department of Obstetrics and Gynecology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
9
|
Hartnett EG, Knight J, Radolec M, Buckanovich RJ, Edwards RP, Vlad AM. Immunotherapy Advances for Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12123733. [PMID: 33322601 PMCID: PMC7764119 DOI: 10.3390/cancers12123733] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022] Open
Abstract
Simple Summary The overall five-year survival rate in epithelial ovarian cancer is 44% and has only marginally improved in the past two decades. Despite an initial response to standard treatment consisting of chemotherapy and surgical removal of tumor, the lesions invariably recur, and patients ultimately die of chemotherapy resistant disease. New treatment modalities are needed in order to improve the prognosis of women diagnosed with ovarian cancer. One such modality is immunotherapy, which aims to boost the capacity of the patient’s immune system to recognize and attack the tumor cells. We performed a retrospective study to identify some of the most promising immune therapies for epithelial ovarian cancer. Special emphasis was given to immuno-oncology clinical trials. Abstract New treatment modalities are needed in order to improve the prognosis of women diagnosed with epithelial ovarian cancer (EOC), the most aggressive gynecologic cancer type. Most ovarian tumors are infiltrated by immune effector cells, providing the rationale for targeted approaches that boost the existing or trigger new anti-tumor immune mechanisms. The field of immuno-oncology has experienced remarkable progress in recent years, although the results seen with single agent immunotherapies in several categories of solid tumors have yet to extend to ovarian cancer. The challenge remains to determine what treatment combinations are most suitable for this disease and which patients are likely to benefit and to identify how immunotherapy should be incorporated into EOC standard of care. We review here some of the most promising immune therapies for EOC and focus on those currently tested in clinical trials.
Collapse
Affiliation(s)
- Erin G. Hartnett
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Julia Knight
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Mackenzy Radolec
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Ronald J. Buckanovich
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Robert P. Edwards
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
| | - Anda M. Vlad
- Department of Obstetrics and Gynecology and Reproductive Sciences, Magee-Womens Research Institute and Foundation and Magee-Womens Hospital of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (E.G.H.); (M.R.); (R.J.B.); (R.P.E.)
- Correspondence:
| |
Collapse
|
10
|
Xia B, Zhao Z, Wu Y, Wang Y, Zhao Y, Wang J. Circular RNA circTNPO3 Regulates Paclitaxel Resistance of Ovarian Cancer Cells by miR-1299/NEK2 Signaling Pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 21:780-791. [PMID: 32791450 PMCID: PMC7419276 DOI: 10.1016/j.omtn.2020.06.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
Abstract
Circular RNAs (circRNAs) were recently reported to be involved in the pathogenesis of ovarian cancer (OC); however, the molecular mechanisms of circRNAs in tumor progression and paclitaxel (PTX) resistance of OC remain largely undetermined. Here, we focused on circTNPO3 (hsa_circ_0001741), which is located on chromosome 7 (chr7): 128655032–128658211 and derived from TNPO3 gene, and thus we termed as circTNPO3. By microarray and qRT-PCR we identified circTNPO3 to be dramatically high expressed in OC samples and correlated with PTX resistance. Functionally, knockdown of circTNPO3 enhanced cell sensitivity to PTX via promoting PTX-induced apoptosis in vitro and in vivo. In mechanism, circTNPO3 acted as a sponge for microRNA-1299 (miR-1299), and NEK2 (NIMA-related kinase 2) was revealed to be target gene of miR-1299. Subsequently, functional assays illustrated that the oncogenic effects of circTNPO3 were attributed to the regulation of miR-1299/NEK2 axis. In conclusion, circTNPO3 contributed to PTX resistance of OC cells at least partly through upregulating NEK2 expression by sponging miR-1299. circTNPO3/miR-1299/NEK2 signaling pathway might play vital roles in the tumorigenesis and chemoresistance of OC.
Collapse
Affiliation(s)
- Bing Xia
- Department of Gynecologic Oncology, Hunan Cancer Hospital and the Affiliated Tumor Hospital of Xiang-Ya School of Medicine, Central South University, Changsha 410078, China
| | - Zitong Zhao
- Department of Gynecologic Oncology, Hunan Cancer Hospital and the Affiliated Tumor Hospital of Xiang-Ya School of Medicine, Central South University, Changsha 410078, China
| | - Yinnayuan Wu
- Department of Gynecologic Oncology, Hunan Cancer Hospital and the Affiliated Tumor Hospital of Xiang-Ya School of Medicine, Central South University, Changsha 410078, China
| | - Ying Wang
- Department of Gynecologic Oncology, Hunan Cancer Hospital and the Affiliated Tumor Hospital of Xiang-Ya School of Medicine, Central South University, Changsha 410078, China
| | - Yan Zhao
- Department of Gynecology and Obstetrics, The Maternal and Child Health Hospital of Hunan, Changsha 410008, China.
| | - Jing Wang
- Department of Gynecologic Oncology, Hunan Cancer Hospital and the Affiliated Tumor Hospital of Xiang-Ya School of Medicine, Central South University, Changsha 410078, China.
| |
Collapse
|
11
|
Braga LDC, Gonçales NG, Furtado RDS, de Andrade WP, Silva LM, da Silva Filho AL. Apoptosis-related gene expression can predict the response of ovarian cancer cell lines to treatment with recombinant human TRAIL alone or combined with cisplatin. Clinics (Sao Paulo) 2020; 75:e1492. [PMID: 32187278 PMCID: PMC7061320 DOI: 10.6061/clinics/2020/e1492] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES The objectives of this study were to determine the sensitivity of ovarian cancer (OC) cell lines (TOV-21G and SKOV-3) to cisplatin and to the recombinant human TRAIL (rhTRAIL), and to evaluate the expression profile of TNFRSF10B, TNFRSF10C, TP53TG5, MDM2, BAX, BCL-2 and CASPASE-8 genes and their participation in the resistance/susceptibility mechanism of these tumor cell lines. METHODS To determine the IC50 values associated with Cisplatin and rhTRAIL, inhibition of cell growth was observed using MTT assays in two human OC cell lines (SKOV-3 and TOV-21G). The analysis of gene expression was performed using quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS Both cell lines had different susceptibility profiles to the tested drugs. In the SKOV-3 cell line, the IC50 values for cisplatin and for rhTRAIL were 270.83 ug/mL and 196.5 ng/mL, respectively. The same concentrations were used for TOV-21G. Different gene expression profiles were observed in each tested cell line. CASPASE-8 and TNFRSF10B expression levels could predict the response of both the cell lines to rhTRAIL alone or the response to a combination of rhTRAIL and cisplatin. In addition, we observed a relationship between BCL-2 and BAX expression that may be helpful in estimating the proliferation rate of the OC cell lines. CONCLUSION SKOV-3 and TOV-21G respond differently to cisplatin and rhTRAIL exposure, and expression of CASPASE-8 and TNFRSF10B are good predictors of responses to these treatments.
Collapse
Affiliation(s)
- Letícia da Conceição Braga
- Servico de Biologia Celular da Diretoria de Pesquisa e Desenvolvimento, Fundacao Ezequiel Dias-Funed, Belo Horizonte, MG, BR
- Departamento de Ginecologia e Obstetricia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, BR
| | - Nikole Gontijo Gonçales
- Servico de Biologia Celular da Diretoria de Pesquisa e Desenvolvimento, Fundacao Ezequiel Dias-Funed, Belo Horizonte, MG, BR
| | - Rafaela de Souza Furtado
- Departamento de Ginecologia e Obstetricia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, BR
| | - Warne Pedro de Andrade
- Departamento de Ginecologia e Obstetricia, Faculdade de Ciencias Medicas e Biologicas de Botucatu, Universidade Estadual Paulista, Botucatu, SP, BR
| | - Luciana Maria Silva
- Servico de Biologia Celular da Diretoria de Pesquisa e Desenvolvimento, Fundacao Ezequiel Dias-Funed, Belo Horizonte, MG, BR
| | - Agnaldo Lopes da Silva Filho
- Departamento de Ginecologia e Obstetricia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, BR
- Departamento de Ginecologia e Obstetricia, Faculdade de Ciencias Medicas e Biologicas de Botucatu, Universidade Estadual Paulista, Botucatu, SP, BR
- *Corresponding author. E-mail:
| |
Collapse
|
12
|
Alotaibi MR, As Sobeai HM, Alaqil FA, Almutairi M, Alhazzani K, Sulaiman AA, Isab AA, Hadal Alotaibi N. A newly synthesized platinum-based compound (PBC-II) increases chemosensitivity of HeLa ovarian cancer cells via inhibition of autophagy. Saudi Pharm J 2019; 27:1203-1209. [PMID: 31885480 PMCID: PMC6921179 DOI: 10.1016/j.jsps.2019.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/02/2019] [Indexed: 11/11/2022] Open
Abstract
There are many mechanisms of resistance, chemoresistance of HeLa cells to anti-cancer agents seems to be autophagy-mediated. While using very effective anti-cancers such as Doxorubicin and cisplatin, cells overcome the cytotoxicity of these drugs through promotion of what so-called cytoprotective autophagy. Here in this study, we sought to introduce a novel platinum-based compound PBC-II that possesses anti-cancer activity. Our data showed that PBC-II is able to induce apoptosis at relatively low concentrations, with no detectable reactive oxygen species (ROS). However, further experiments demonstrated that exposure of HeLa cells to PBC-II did not promote autophagy; rather, it resulted in accumulation of p62 and decrease in LC3-II levels. Autophagy was then promoted in HeLa cells pharmacologically by Doxorubicin and genetically by siRNA IL-10. In order to confirm promotion of autophagy in our model, we performed acridine orange staining to assess for autophagy under microscope as well as via flow cytometry. We then measured protein level of autophagy markers p62 and LC3 by western blot. Our data indicated that PBC-II interferes with therapy-induced autophagy. We also determined PI3K activity while co-incubation of PBC-II with autophagy inducers. It was clear that PI3K activation decreased when PBC-II was co-administered with autophagy inducers. Collectively, PBC-II exerts unique anti-proliferative effects associated with inhibition of autophagy, which indicates that PBC-II is potentially a promising agent to be used in resistant ovarian tumors.
Collapse
Affiliation(s)
- Moureq Rashed Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Homood Moqbel As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | | | - Mashal Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Adam A.A. Sulaiman
- Lab Technical Support Office (LTSO), King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Anvarhusein A. Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Nasser Hadal Alotaibi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakakah 72341, Saudi Arabia
| |
Collapse
|
13
|
Niu Q, Liu Z, Gao J, Wang Q. MiR-338-3p Enhances Ovarian Cancer Cell Sensitivity to Cisplatin by Downregulating WNT2B. Yonsei Med J 2019; 60:1146-1156. [PMID: 31769245 PMCID: PMC6881712 DOI: 10.3349/ymj.2019.60.12.1146] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/26/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Chemoresistance is a concern in ovarian cancer patients, in whom survival remains. MicroRNA, a novel class of small RNAs, have frequently been found to be dysregulated in human malignancies and to act as negative regulators of gene expression. This study aimed to explore the function of miR-338-3p in cisplatin resistance in ovarian cancer and potential molecular mechanisms thereof. MATERIALS AND METHODS The expression levels of miR-338-3p and WNT2B in ovarian cancer tissues and cells were estimated by real-time quantitative polymerase chain reaction (RT-qPCR). In addition, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazol-3-ium bromide (MTT), transwell, and flow cytometry assays were used to assess biological role of miR-338-3p in vitro. Western blot assay was conducted to measure protein expression of WNT2B, epithelial-mesenchymal transition (EMT)-related proteins, and apoptosis-related proteins. The relationship between miR-338-3p and WNT2B was confirmed by dual-luciferase reporter. Finally, a xenograft tumor model was developed to explore the effects of overexpression of miR-338-3p on tumor growth in ovarian cancer in vivo. RESULTS MiR-338-3p was downregulated in cisplatin resistant ovarian cancer tissues and cells. Mechanistically, high expression of miR-338-3p enhanced cell sensitivity to cisplatin by inhibiting proliferation, motility, and EMT and by promoting apoptosis via targeting WNT2B expression in vitro. Furthermore, overexpression of miR-338-3p increased cisplatin sensitivity among ovarian cancer in an in vivo xenograft tumor model. CONCLUSION MiR-338-3p enhances the sensitivity of ovarian cancer cells to cisplatin by downregulating WNT2B.
Collapse
Affiliation(s)
- Qin Niu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Zhenghong Liu
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China.
| | - Jia Gao
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Qiao Wang
- Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
14
|
The Histone Methyltransferase DOT1L Is a Functional Component of Estrogen Receptor Alpha Signaling in Ovarian Cancer Cells. Cancers (Basel) 2019; 11:cancers11111720. [PMID: 31689915 PMCID: PMC6895927 DOI: 10.3390/cancers11111720] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/28/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
Although a large fraction of high-grade serous epithelial ovarian cancers (OCs) expresses Estrogen Receptor alpha (ERα), anti-estrogen-based therapies are still not widely used against these tumors due to a lack of sufficient evidence. The histone methyltransferase Disruptor of telomeric silencing-1-like (DOT1L), which is a modulator of ERα transcriptional activity in breast cancer, controls chromatin functions involved in tumor initiation and progression and has been proposed as a prognostic OC biomarker. As molecular and clinico-pathological data from TCGA suggest a correlation between ERα and DOT1L expression and OC prognosis, the presence and significance of ERα/DOT1L association was investigated in chemotherapy-sensitive and chemotherapy-resistant ER+ OC cells. RNA sequencing before and after inhibition of these factors showed that their activity is implicated in OC cell proliferation and that they functionally cooperate with each other to control the transcription of genes involved in key cancer cell features, such as the cell cycle, epithelial-mesenchymal transition (EMT), drug metabolism, and cell-to-cell signaling, as well as expression of the ERα gene itself. Together with evidence from loss-of-function genetic screens showing that ERα and DOT1L behave as core fitness factors in OC cells, these results suggest that combined inhibition of their activity might be effective against ERα-expressing, chemotherapy-resistant ovarian tumors.
Collapse
|
15
|
Gong J, Jiang H, Shu C, Hu MQ, Huang Y, Liu Q, Li RF. Prognostic value of lymphocyte-to-monocyte ratio in ovarian cancer: a meta-analysis. J Ovarian Res 2019; 12:51. [PMID: 31151469 PMCID: PMC6544921 DOI: 10.1186/s13048-019-0527-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Prognostic biomarkers are highly needed to properly manage patients with cancer and improve their clinical courses. The relationship between lymphocyte-to-monocyte ratio (LMR) at diagnosis and ovarian cancer prognosis has been extensively studied, but little consensus has been reached regarding its utility as a biomarker of poor outcome. Thus, this study aimed to investigate the potential prognostic value of pretreatment LMR in such patients to shed light on this issue. METHODS We searched the scientific databases of MEDLINE, Embase, Cochrane Library, and WangFang for relevant studies about the inflammatory prognostic factor LMR in ovarian cancer, based on specific inclusion and exclusion criteria. The following parameters were analyzed among others: LMR values and respective cut-offs, patient's overall survival (OS) and progression-free survival (PFS), and clinicopathological features. RESULTS Eight studies, including 2259 patients, were eligible for inclusion in this meta-analysis. We found that low LMR was associated with both poor OS [Hazard ratio (HR): 1.92; 95% confidence interval (CI): 1.58-2.34; p < 0.001] and PFS (HR: 1.70; 95% CI: 1.54-1.88; p < 0.001). Moreover, our findings revealed that low LMR was correlated with high G2/G3 histological grade (OR: 1.67; 95% CI: 1.26-2.20; p < 0.001) and late III-IV FIGO stage tumors (OR: 3.55; 95% CI: 2.68-4.70; p < 0.001), high serum CA-125 level (OR: 2.18; 95% CI: 1.71-2.77; p < 0.001), and presence of malignant ascites (OR: 1.87; 95% CI: 1.11-3.14; p = 0.02) and lymph node metastases (OR: 1.70; 95% CI: 1.13-2.54; p = 0.01). CONCLUSION Pretreatment LMR is a potential prognostic marker of poor outcome in ovarian cancer patients and may thus be important in clinical care and disease control.
Collapse
Affiliation(s)
- Jun Gong
- Department of Abdominal and Pelvic Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Hubei Province, Huangshi, People's Republic of China
| | - Hui Jiang
- Department of Urology, Huangshi Central Hospital of Edong Healthcare Group, Hubei Polytechnic University, Huangshi, Hubei, China
| | - Chang Shu
- Department of Abdominal and Pelvic Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Hubei Province, Huangshi, People's Republic of China
| | - Mei-Qin Hu
- Department of Abdominal and Pelvic Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Hubei Province, Huangshi, People's Republic of China
| | - Yan Huang
- Department of Clinical Laboratory, Huangshi Central Hospital of Edong Healthcare Group, Hubei Polytechnic University, Huangshi, Hubei, China
| | - Qin Liu
- Department of Breast surgery, Thyroid surgery, Huangshi Central Hospital of Edong Healthcare Group, Hubei Polytechnic University, Huangshi, Hubei, China
| | - Rong-Feng Li
- Department of Abdominal and Pelvic Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Hubei Province, Huangshi, People's Republic of China. .,Department of Abdominal and Pelvic Medical Oncology, Huangshi Central Hospital, Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, China.
| |
Collapse
|
16
|
Mensah LB, Morton SW, Li J, Xiao H, Quadir MA, Elias KM, Penn E, Richson AK, Ghoroghchian PP, Liu J, Hammond PT. Layer-by-layer nanoparticles for novel delivery of cisplatin and PARP inhibitors for platinum-based drug resistance therapy in ovarian cancer. Bioeng Transl Med 2019; 4:e10131. [PMID: 31249881 PMCID: PMC6584097 DOI: 10.1002/btm2.10131] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022] Open
Abstract
Advanced staged high-grade serous ovarian cancer (HGSOC) is the leading cause of gynecological cancer death in the developed world, with 5-year survival rates of only 25-30% due to late-stage diagnosis and the shortcomings of platinum-based therapies. A Phase I clinical trial of a combination of free cisplatin and poly(ADP-ribose) polymerase inhibitors (PARPis) showed therapeutic benefit for HGSOC. In this study, we address the challenge of resistance to platinum-based therapy by developing a targeted delivery approach. Novel electrostatic layer-by-layer (LbL) liposomal nanoparticles (NPs) with a terminal hyaluronic acid layer that facilitates CD44 receptor targeting are designed for selective targeting of HGSOC cells; the liposomes can be formulated to contain both cisplatin and the PARPi drug within the liposomal core and bilayer. The therapeutic effectiveness of LbL NP-encapsulated cisplatin and PARPi alone and in combination was compared with the corresponding free drugs in luciferase and CD44-expressing OVCAR8 orthotopic xenografts in female nude mice. The NPs exhibited prolonged blood circulation half-life, mechanistic staged drug release and targeted codelivery of the therapeutic agents to HGSOC cells. Moreover, compared to the free drugs, the NPs resulted in significantly reduced tumor metastasis, extended survival, and moderated systemic toxicity.
Collapse
Affiliation(s)
- Lawrence B. Mensah
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
| | - Stephen W. Morton
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
| | - Jiahe Li
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
| | - Haihua Xiao
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
- Institute of Chemistry, Changchun Institute of Applied ChemistryChinese Academy of Sciences, JilinChangchunP.R. China
| | - Mohiuddin A. Quadir
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
- Department of Coatings and Polymeric MaterialsNorth Dakota State UniversityFargoND, 58108
| | - Kevin M. Elias
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, and Reproductive BiologyBrigham and Women's HospitalBostonMA, 02115
| | - Emily Penn
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
| | - Aysen K. Richson
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
| | - Paiman Peter Ghoroghchian
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Dana‐Farber Cancer InstituteBostonMA, 02115
| | - Joyce Liu
- Dana‐Farber Cancer InstituteBostonMA, 02115
| | - Paula T. Hammond
- The Koch Institute for Integrative Cancer ResearchMassachusetts Institute of Technology (MIT)CambridgeMA, 02142
- Department of Chemical EngineeringMassachusetts Institute of Technology (MIT)CambridgeMA, 02139
| |
Collapse
|
17
|
Sensitization of Drug Resistant Cancer Cells: A Matter of Combination Therapy. Cancers (Basel) 2018; 10:cancers10120483. [PMID: 30518036 PMCID: PMC6315347 DOI: 10.3390/cancers10120483] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/12/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer drug resistance is an enormous problem. It is responsible for most relapses in cancer patients following apparent remission after successful therapy. Understanding cancer relapse requires an understanding of the processes underlying cancer drug resistance. This article discusses the causes of cancer drug resistance, the current combination therapies, and the problems with the combination therapies. The rational design of combination therapy is warranted to improve the efficacy. These processes must be addressed by finding ways to sensitize the drug-resistant cancers cells to chemotherapy, and to prevent formation of drug resistant cancer cells. It is also necessary to prevent the formation of cancer progenitor cells by epigenetic mechanisms, as cancer progenitor cells are insensitive to standard therapies. In this article, we emphasize the role for the rational development of combination therapy, including epigenetic drugs, in achieving these goals.
Collapse
|
18
|
Wang H, Luo Y, Qiao T, Wu Z, Huang Z. Luteolin sensitizes the antitumor effect of cisplatin in drug-resistant ovarian cancer via induction of apoptosis and inhibition of cell migration and invasion. J Ovarian Res 2018; 11:93. [PMID: 30454003 PMCID: PMC6241043 DOI: 10.1186/s13048-018-0468-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 11/04/2018] [Indexed: 12/11/2022] Open
Abstract
Luteolin, a polyphenolic flavone, has been demonstrated to exert anti-tumor activity in various cancer types. Cisplatin drug resistance is a major obstacle in the management of ovarian cancer. In the present study, we investigated the chemo-sensitizing effect of luteolin in both cisplatin-resistant ovarian cancer cell line and a mice xenotransplant model. In vitro, CCK-8 assay showed that luteolin inhibited cell proliferation in a dose-dependent manner, and luteolin enhanced anti-proliferation effect of cisplatin on cisplatin-resistant ovarian cancer CAOV3/DDP cells. Flow cytometry revealed that luteolin enhanced cell apoptosis in combination with cisplatin. Western blotting and qRT-PCR assay revealed that luteolin increased cisplatin-induced downregulation of Bcl-2 expression. In addition, wound-healing assay and Matrigel invasion assay showed that luteolin and cisplatin synergistically inhibited migration and invasion of CAOV3/DDP cells. Moreover, in vivo, luteolin enhanced cisplatin-induced reduction of tumor growth as well as induction of apoptosis. We suggest that luteolin in combination with cisplatin could potentially be used as a new regimen for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Haixia Wang
- Department of Obstetrics and Gynecology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Youjun Luo
- Department of Oncology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Tiankui Qiao
- Department of Oncology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhaoxia Wu
- Department of Traditional Medicine, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Zhonghua Huang
- Department of Obstetrics and Gynecology, Jinshan branch of Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
19
|
Sureechatchaiyan P, Hamacher A, Brockmann N, Stork B, Kassack MU. Adenosine enhances cisplatin sensitivity in human ovarian cancer cells. Purinergic Signal 2018; 14:395-408. [PMID: 30078088 DOI: 10.1007/s11302-018-9622-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 07/20/2018] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is the deadliest gynecologic cancer due to lack of early effective diagnosis and development of resistance to platinum-based chemotherapy. Several studies reported that adenosine concentrations are higher in tumor microenvironment than in non-tumor tissue. This finding inspired us to study the role of adenosine in ovarian cancer cells and to investigate if adenosine pathways offer new treatment options urgently needed to prevent or overcome chemoresistance. The ovarian cancer cell lines HEY, A2780, and its cisplatin-resistant subline A2780CisR were used in this study. Expression and functional activity of adenosine receptors were investigated by RT-PCR, Western blotting, and cAMP assay. A1 and A2B adenosine receptors were expressed and functionally active in all three cell lines. Adenosine showed moderate cytotoxicity (MTT-IC50 values were between 700 and 900 μM) and induced apoptosis in a concentration-dependent manner by increasing levels of sub-G1 and cleaved PARP. Apoptosis was diminished by QVD-OPh, confirming caspase-dependent induction of apoptosis. Forty-eight hours pre-incubation of adenosine prior to cisplatin significantly enhanced cisplatin-induced cytotoxicity in a synergistic manner and increased apoptosis. SLV320 or PSB603, selective A1 and A2B antagonists, was not able to inhibit adenosine-induced increase in cisplatin cytotoxicity or apoptosis whereas dipyridamole, a nucleoside transporter inhibitor, completely abrogated both effects. Mechanistically, adenosine increased pAMPK and reduced pS6K which was prevented by dipyridamole. In conclusion, application of adenosine prior to cisplatin could be a new therapeutic option to increase the potency of cisplatin in a synergistic manner and thus overcome platinum resistance in ovarian cancer.
Collapse
Affiliation(s)
- Parichat Sureechatchaiyan
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine University, Building 26.23.02, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Alexandra Hamacher
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine University, Building 26.23.02, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Nicole Brockmann
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine University, Building 26.23.02, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Bjoern Stork
- Institute of Molecular Medicine, Heinrich-Heine University, Universitaetsstrasse 1, 40225, Duesseldorf, Germany
| | - Matthias U Kassack
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich-Heine University, Building 26.23.02, Universitaetsstrasse 1, 40225, Duesseldorf, Germany.
| |
Collapse
|
20
|
Kleemann M, Schneider H, Unger K, Sander P, Schneider EM, Fischer-Posovszky P, Handrick R, Otte K. MiR-744-5p inducing cell death by directly targeting HNRNPC and NFIX in ovarian cancer cells. Sci Rep 2018; 8:9020. [PMID: 29899543 PMCID: PMC5998049 DOI: 10.1038/s41598-018-27438-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 06/04/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) play an important role in the regulation of gene expression. The binding to target messenger RNAs (mRNAs) results in mRNA cleavage or inhibition of the translational machinery leading to decreased protein levels. Various signalling pathways, including apoptosis are modulated by miRNAs. Here, we investigated the role of miR-744-5p in apoptosis signalling in ovarian cancer cell lines. MiR-744-5p expression was reduced in the cancer cell lines independent of the host gene MAP2K4. Overexpression of miR-744-5p activated the intrinsic apoptotic pathway in SKOV3, OVCAR3 and Cisplatin resistant (A2780-cis) and non-resistant A2780 cells leading to cell death. Notably, miR-744-5p overexpression together with Carboplatin treatment led to at least additive pro-apoptotic effects. Investigation of the apoptotic signalling pathways mediated by miR-744-5p revealed that its elevated expression directly downregulated mRNA and protein expression of nuclear factor I X (NFIX) and heterogeneous nuclear ribonucleoprotein C (HNRNPC). HNRNPC caused diminished miR-21 expression and AKT phosphorylation, while NFIX decreased Bcl2 levels, leading to the detected pro-apoptotic effects. Finally, Kaplan-Meier-Plots showed a prolonged median disease-free survival in ovarian serous cystadenocarcinoma patients with high miR-744 expression.
Collapse
Affiliation(s)
- Michael Kleemann
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Str. 35, 88400, Biberach, Germany. .,University of Ulm, Faculty of Medicine, Albert-Einstein-Allee 11, 89079, Ulm, Germany.
| | - Helga Schneider
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Str. 35, 88400, Biberach, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Philip Sander
- University Medical Center Ulm, Division of Experimental Anesthesiology, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - E Marion Schneider
- University Medical Center Ulm, Division of Experimental Anesthesiology, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Pamela Fischer-Posovszky
- University Medical Center Ulm, Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Eythstr. 24, 89075, Ulm, Germany
| | - René Handrick
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Str. 35, 88400, Biberach, Germany
| | - Kerstin Otte
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, Hubertus-Liebrecht-Str. 35, 88400, Biberach, Germany
| |
Collapse
|
21
|
Hedemann N, Rogmans C, Sebens S, Wesch D, Reichert M, Schmidt-Arras D, Oberg HH, Pecks U, van Mackelenbergh M, Weimer J, Arnold N, Maass N, Bauerschlag DO. ADAM17 inhibition enhances platinum efficiency in ovarian cancer. Oncotarget 2018; 9:16043-16058. [PMID: 29662625 PMCID: PMC5882316 DOI: 10.18632/oncotarget.24682] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 02/28/2018] [Indexed: 12/18/2022] Open
Abstract
Chemotherapeutic resistance evolves in about 70 % of ovarian cancer patients and is a major cause of death in this tumor entity. Novel approaches to overcome these therapeutic limitations are therefore highly warranted. A disintegrin and metalloprotease 17 (ADAM17) is highly expressed in ovarian cancer and required for releasing epidermal growth factor receptor (EGFR) ligands like amphiregulin (AREG). This factor has recently been detected in ascites of advanced stage ovarian cancer patients. However, it is not well understood, whether and how ADAM17 might contribute to chemo resistance of ovarian cancer. In this study, we identified ADAM17 as an essential upstream regulator of AREG release under chemotherapeutic treatment in ovarian cancer cell lines and patient derived cells. In the majority of ovarian cancer cells cisplatin treatment resulted in enhanced ADAM17 activity, as shown by an increased shedding of AREG. Moreover, both mRNA and the protein content of AREG were dose-dependently increased by cisplatin exposure. Consequently, cisplatin strongly induced phosphorylation of ADAM17-downstream mediators, the EGFR and extracellular signal-regulated kinases (ERK). Phorbol 12-myristate 13-acetate (PMA), similarly to cisplatin, mediated AREG shedding and membrane fading of surface ADAM17. Inhibition of ADAM17 with either GW280264X or the anti-ADAM17 antibody D1 (A12) as well as silencing of ADAM17 by siRNA selectively reduced AREG release. Thus, ADAM17 inhibition sensitized cancer cells to cisplatin-induced apoptosis, and significantly reduced cell viability. Based on these findings, we propose that targeting of ADAM17 in parallel to chemotherapeutic treatment suppresses survival pathways and potentially diminish evolving secondary chemo resistance mechanisms.
Collapse
Affiliation(s)
- Nina Hedemann
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Christoph Rogmans
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Daniela Wesch
- Institute of Immunology, Christian-Albrechts-University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Manuel Reichert
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Hans-Heinrich Oberg
- Institute of Immunology, Christian-Albrechts-University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Ulrich Pecks
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Marion van Mackelenbergh
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jörg Weimer
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Norbert Arnold
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nicolai Maass
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
22
|
Yang SD, Ahn SH, Kim JI. 3-Oxoacid CoA transferase 1 as a therapeutic target gene for cisplatin-resistant ovarian cancer. Oncol Lett 2017; 15:2611-2618. [PMID: 29434981 DOI: 10.3892/ol.2017.7560] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/03/2017] [Indexed: 01/28/2023] Open
Abstract
Ovarian cancer (OC) is the second leading cause of mortality from gynecological malignancies and has the highest mortality rate worldwide. As it is commonly asymptomatic during the early stages of the disease, >70% of patients with OC are diagnosed at advanced stages with metastasis. Despite treatment methods, including optimal debulking surgery and chemotherapy with the platinum-based drug cisplatin, OC recurrence is often inevitable, with an overall 5-year survival rate of 45%, mostly due to the steady development of cisplatin resistance. To identify genes involved in cisplatin resistance, the present study determined the half-maximal inhibitory concentrations of eight different OC cell lines and classified them into two groups (sensitive and resistant). mRNA expression was analyzed with GeneChip Human Gene 1.0 ST Arrays, and DNA methylation profiles were evaluated with the HumanMethylation450 BeadChip. Using an integrated approach of analyzing gene expression levels and DNA methylation profiles simultaneously, 26 genes were selected that were differentially expressed and methylated between the resistant and sensitive groups. Among these 26 genes, 3-oxoacid CoA transferase 1 (OXCT1), which was demonstrated to be downregulated and hypermethylated at promoter CpGs in the cisplatin-resistant group compared with the cisplatin-sensitive group, was selected for further investigation. Treatment with a DNA methyltransferase inhibitor restored hypermethylation-mediated gene silencing of OXCT1 in the cisplatin-resistant group, but not in the cisplatin-sensitive group. Furthermore, overexpression of OXCT1 conferred sensitivity to cisplatin in OC cells. The results of the present study suggest that OXCT1 serves an important role in conferring cisplatin sensitivity, and may provide a potential therapeutic target for cisplatin chemotherapy in patients with recurrent OC.
Collapse
Affiliation(s)
- San-Duk Yang
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - So Hee Ahn
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 110-799, Republic of Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea.,Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 110-799, Republic of Korea
| |
Collapse
|
23
|
Huang YH, Peng W, Furuuchi N, DuHadaway JB, Jimbo M, Pirritano A, Dunton CJ, Daum GS, Leiby BE, Brody JR, Sawicki JA. Insights from HuR biology point to potential improvement for second-line ovarian cancer therapy. Oncotarget 2017; 7:21812-24. [PMID: 26943573 PMCID: PMC5008325 DOI: 10.18632/oncotarget.7840] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/21/2016] [Indexed: 12/28/2022] Open
Abstract
This retrospective study aimed to investigate the role that an RNA-binding protein, HuR, plays in the response of high-grade serous ovarian tumors to chemotherapeutics. We immunohistochemically stained sections of 31 surgically-debulked chemo-naïve ovarian tumors for HuR and scored the degree of HuR cytoplasmic staining. We found no correlation between HuR intracellular localization in tumor sections and progression free survival (PFS) of these patients, 29 of whom underwent second-line gemcitabine/platin combination therapy for recurrent disease. Ribonucleoprotein immunoprecipitation (RNP-IP) analysis of ovarian cancer cells in culture showed that cytoplasmic HuR increases deoxycytidine kinase (dCK), a metabolic enzyme that activates gemcitabine. The effects of carboplatin treatment on HuR and WEE1 (a mitotic inhibitor) expression, and on cell cycle kinetics, were also examined. Treatment of ovarian cancer cells with carboplatin results in increased HuR cytoplasmic expression and elevated WEE1 expression, arresting cell cycle G2/M transition. This may explain why HuR cytoplasmic localization in chemo-naïve tumors is not predictive of therapeutic response and PFS following second-line gemcitabine/platin combination therapy. These results suggest treatment of recurrent ovarian tumors with a combination of gemcitabine, carboplatin, and a WEE1 inhibitor may be potentially advantageous as compared to current clinical practices.
Collapse
Affiliation(s)
- Yu-Hung Huang
- Lankenau Institute for Medical Research, Wynnewood, PA 19086, USA.,Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Weidan Peng
- Lankenau Institute for Medical Research, Wynnewood, PA 19086, USA
| | - Narumi Furuuchi
- Lankenau Institute for Medical Research, Wynnewood, PA 19086, USA
| | | | - Masaya Jimbo
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrea Pirritano
- Main Line Gynecologic Oncology, Lankenau Medical Center, Wynnewood, PA 19096, USA
| | - Charles J Dunton
- Lankenau Institute for Medical Research, Wynnewood, PA 19086, USA.,Main Line Gynecologic Oncology, Lankenau Medical Center, Wynnewood, PA 19096, USA
| | - Gary S Daum
- Main Line Health Laboratories, Lankenau Medical Center, Wynnewood, PA 19096, USA
| | - Benjamin E Leiby
- Division of Biostatistics, Thomas Jefferson University, Philadelphia, PA 19107, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jonathan R Brody
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Janet A Sawicki
- Lankenau Institute for Medical Research, Wynnewood, PA 19086, USA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
24
|
Wehbe M, Lo C, Leung AWY, Dragowska WH, Ryan GM, Bally MB. Copper (II) complexes of bidentate ligands exhibit potent anti-cancer activity regardless of platinum sensitivity status. Invest New Drugs 2017; 35:682-690. [PMID: 28733701 PMCID: PMC5694505 DOI: 10.1007/s10637-017-0488-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/28/2017] [Indexed: 01/06/2023]
Abstract
Insensitivity to platinum, either through inherent or acquired resistance, is a major clinical problem in the treatment of many solid tumors. Here, we explored the therapeutic potential of diethyldithiocarbamate (DDC), pyrithione (Pyr), plumbagin (Plum), 8-hydroxyquinoline (8-HQ), clioquinol (CQ) copper complexes in a panel of cancer cell lines that differ in their sensitivity to platins (cisplatin/carboplatin) using a high-content imaging system. Our data suggest that the copper complexes were effective against both platinum sensitive (IC50 ~ 1 μM platinum) and insensitive (IC50 > 5 μM platinum) cell lines. Furthermore, copper complexes of DDC, Pyr and 8-HQ had greater therapeutic activity compared to the copper-free ligands in all cell lines; whereas the copper-dependent activities of Plum and CQ were cell-line specific. Four of the copper complexes (Cu(DDC)2, Cu(Pyr)2, Cu(Plum)2 and Cu(8-HQ)2) showed IC50 values less than that of cisplatin in all tested cell lines. The complex copper DDC (Cu(DDC)2) was selected for in vivo evaluation due to its low nano-molar range activity in vitro and the availability of an injectable liposomal formulation. Liposomal (Cu(DDC)2) was tested in a fast-growing platinum-resistant A2780-CP ovarian xenograft model and was found to achieve a statistically significant reduction (50%; p < 0.05) in tumour size. This work supports the potential use of copper-based therapeutics to treat cancers that are insensitive to platinum drugs.
Collapse
Affiliation(s)
- Mohamed Wehbe
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Cody Lo
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Ada W Y Leung
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Wieslawa H Dragowska
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Gemma M Ryan
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Marcel B Bally
- Experimental Therapeutics, British Columbia Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2146 East Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Center for Drug Research and Development, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
25
|
An J, Lv W, Zhang Y. LncRNA NEAT1 contributes to paclitaxel resistance of ovarian cancer cells by regulating ZEB1 expression via miR-194. Onco Targets Ther 2017; 10:5377-5390. [PMID: 29180871 PMCID: PMC5691924 DOI: 10.2147/ott.s147586] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Chemoresistance is one of the major obstacles for cancer therapy in the clinic. Nuclear paraspeckle assembly transcript 1 (NEAT1) has been reported as an oncogene in most malignancies such as lung cancer, esophageal cancer, and gastric cancer. This study is designed to investigate the function of NEAT1 in paclitaxel (PTX) resistance of ovarian cancer and its potential molecular mechanism. Patients and methods The expressions of NEAT1 and miR-194 in ovarian cancer tissues and cells were estimated by quantitative real-time polymerase chain reaction (qRT-PCR). MTT, flow cytometry, and Western blot assays were used to assess the effect of NEAT1 on PTX resistance in PTX-resistant ovarian cancer cells. Luciferase reporter assay was applied to examine the association between NEAT1, zinc finger E-box-binding homeobox 1 (ZEB1) and miR-194. Xenograft tumor model was established to confirm the biological role of NEAT1 in PTX resistance of ovarian cancer in vivo. Results NEAT1 was upregulated, and miR-194 was downregulated in PTX-resistant ovarian cancer tissues and cells. Functionally, NEAT1 knockdown enhanced cell sensitivity to PTX via promoting PTX-induced apoptosis in vitro. NEAT1 was identified as a molecular sponge of miR-194 to upregulate ZEB1 expression. Mechanistically, NEAT1-knockdown-induced PTX sensitivity was mediated by miR-194/ZEB1 axis. Moreover, NEAT1 knockdown improved PTX sensitivity of ovarian cancer in vivo. Conclusion NEAT1 contributed to PTX resistance of ovarian cancer cells at least partly through upregulating ZEB1 expression by sponging miR-194, elucidating a novel regulatory pathway of chemoresistance in PTX-resistant ovarian cancer cells and providing a possible long noncoding RNA (lncRNA)-targeted therapy for ovarian cancer.
Collapse
Affiliation(s)
- Jihong An
- Department of Clinical Pharmacy, Huaihe Hospital of Henan University, Kaifeng, People's Republic of China
| | - Weiling Lv
- Department of Clinical Pharmacy, Huaihe Hospital of Henan University, Kaifeng, People's Republic of China
| | - Yongzhou Zhang
- Department of Clinical Pharmacy, Huaihe Hospital of Henan University, Kaifeng, People's Republic of China
| |
Collapse
|
26
|
Kleemann M, Bereuther J, Fischer S, Marquart K, Hänle S, Unger K, Jendrossek V, Riedel CU, Handrick R, Otte K. Investigation on tissue specific effects of pro-apoptotic micro RNAs revealed miR-147b as a potential biomarker in ovarian cancer prognosis. Oncotarget 2017; 8:18773-18791. [PMID: 27821806 PMCID: PMC5386646 DOI: 10.18632/oncotarget.13095] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 10/27/2016] [Indexed: 11/25/2022] Open
Abstract
The development and progression of cancer can be ascribed to imbalances in gene regulation leading to aberrant cellular behavior. The loss of micro RNAs (miRNAs) exhibiting tumor-suppressive function has been demonstrated to be often causative for uncontrolled cell proliferation, migration or tissue infiltration. The installation of de novo tumor suppressive function by using pro-apoptotic miRNAs might be a promising therapeutic approach. In addition, there is a great demand for novel biomarkers for the prognosis of cancer, which prompted us to transfer a high content miRNA screening initially performed to identify bioprocess relevant miRNAs in Chinese hamster ovary (CHO) cells to human cancer cell lines . Analysis of screened miRNAs exhibiting strongest pro-apoptotic effects discovered globally and cross-species active candidates. The recovery rate of apoptosis inducing miRNAs was highest in the human ovarian carcinoma cell line SKOV3. Focusing on ovarian cell lines miR-1912, miR-147b and miR-3073a showed significant apoptosis induction in cell lines with different genetic background (SKOV3p53null, OVCAR3p53R248Q, TOV21G, TOV112Dp53R175H, A2780, A2780-cisp53K351N) alone and additive effects in combination with carboplatin. While expression analysis revealed a low endogenous expression of miR-1912 and miR-147b in SKOV3, miRNA expression was highly upregulated upon apoptosis induction using chemotherapeutics. Ectopic introduction of these miRNAs lead to enhanced activation of caspase-dependent death signaling and an induction of the pro-apoptotic proteins Bak1 and Bax and a reduced expression of Bcl2 and Bcl-xL. Finally, analysis of The Cancer Genome Atlas data revealed the expression of hsa-miR-147b-5p to show a positive influence on the median survival of ovarian cancer patients.
Collapse
Affiliation(s)
- Michael Kleemann
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400 Biberach, Germany.,University of Ulm, Faculty of Medicine, 89079 Ulm, Germany
| | - Jeremias Bereuther
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400 Biberach, Germany
| | - Simon Fischer
- Boehringer Ingelheim Pharma GmbH and Co.KG, BP Process Development Germany, 88400 Biberach, Germany
| | - Kim Marquart
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400 Biberach, Germany
| | - Simon Hänle
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400 Biberach, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical School, 45122 Essen, Germany
| | | | - René Handrick
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400 Biberach, Germany
| | - Kerstin Otte
- Institute of Applied Biotechnology, University of Applied Sciences Biberach, 88400 Biberach, Germany
| |
Collapse
|
27
|
Minor DR, Moores SP, Chan JK. Prolonged survival after intraperitoneal interleukin-2 immunotherapy for recurrent ovarian cancer. Gynecol Oncol Rep 2017; 22:43-44. [PMID: 29034306 PMCID: PMC5633818 DOI: 10.1016/j.gore.2017.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 09/19/2017] [Accepted: 09/23/2017] [Indexed: 01/09/2023] Open
Abstract
A case report of a 14 year remission of recurrent ovarian cancer with intraperitoneal aldesleukin (IL-2) is presented. Intraperitoneal IL-2 was given with little toxicity. Immunotherapy may have the potential for durable remissions in ovarian cancer.
Collapse
Affiliation(s)
- David R Minor
- California Pacific Medical Center Research Institute, 2200 Webster St. Suite 511, San Francisco, CA 94115, United States
| | - Samantha P Moores
- California Pacific Medical Center Research Institute, 2200 Webster St., San Francisco, CA 94115, United States
| | - John K Chan
- California Pacific Medical Center Research Institute/Palo Alto Medical Foundation, 3838 California St., San Francisco, CA 94118, United States
| |
Collapse
|
28
|
Bober SL, Recklitis CJ, Michaud AL, Wright AA. Improvement in sexual function after ovarian cancer: Effects of sexual therapy and rehabilitation after treatment for ovarian cancer. Cancer 2017; 124:176-182. [PMID: 28881456 DOI: 10.1002/cncr.30976] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/24/2017] [Accepted: 08/09/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Sexual dysfunction is a distressing long-term effect after gynecological cancer and affects the majority of survivors for years after the completion of therapy. Despite its prevalence, treatment-related sexual dysfunction is underrecognized and undertreated for survivors. Thus, the aim of this study was to develop and test a brief psychoeducational intervention for managing sexual dysfunction for women who have undergone treatment for ovarian cancer (OC). METHODS Forty-six OC survivors with documented, treatment-related sexual dysfunction received a single half-day group intervention that included sexual health education and rehabilitation training, relaxation and cognitive behavioral therapy skills to address sexual symptoms, and a single tailored booster telephone call 4 weeks after the group. Assessment measures were completed at the baseline (baseline 1), after an 8-week no-treatment run-in period (baseline 2), and then again 2 and 6 months after the intervention. The Female Sexual Function Index (FSFI) was used to assess sexual functioning, and the Brief Symptom Inventory 18 (BSI-18) was used to capture psychological distress. RESULTS Between baseline 1 and baseline 2, there were no significant changes in the study measures, and this indicated no natural improvement during the run-in period. In contrast, the total FSFI scores improved significantly from baseline 1 to the 2- (n = 45; P < .0005) and 6-month time points (n = 42; P < .05). The BSI-18 scores were also significantly improved at the 2- (P < .005) and 6-month time points (P < .01) in comparison with baseline 1. CONCLUSIONS This brief behavioral intervention led to significant improvements in overall sexual functioning and psychological distress that were maintained at the 6-month follow-up. The results demonstrate the feasibility of this brief, low-intensity behavioral intervention and support the development of a larger randomized controlled trial. Cancer 2018;124:176-82. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Sharon L Bober
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Christopher J Recklitis
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | | | - Alexi A Wright
- Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Le T, Kennedy EB, Dodge J, Elit L. Follow-up of patients who are clinically disease-free after primary treatment for fallopian tube, primary peritoneal, or epithelial ovarian cancer: a Program in Evidence-Based Care guideline adaptation. ACTA ACUST UNITED AC 2016; 23:343-350. [PMID: 27803599 DOI: 10.3747/co.23.3042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND A need for follow-up recommendations for survivors of fallopian tube, primary peritoneal, or epithelial ovarian cancer after completion of primary treatment was identified by Cancer Care Ontario's Program in Evidence-Based Care. METHODS We searched for existing guidelines, conducted a systematic review (medline, embase, and cdsr, January 2010 to March 2015), created draft recommendations, and completed a comprehensive review process. Outcomes included overall survival, quality of life, and patient preferences. RESULTS The Cancer Australia guidance document Follow Up of Women with Epithelial Ovarian Cancer was adapted for the Ontario context. A key randomized controlled trial found that the overall survival rate did not differ between asymptomatic women who received early treatment based on elevated serum cancer antigen 125 (ca125) alone and women who waited for the appearance of clinical symptoms before initiating treatment (hazard ratio: 0.98; 95% confidence interval: 0.80 to 1.20; p = 0.85); in addition, patients in the delayed treatment group reported good global health scores for longer. No randomized studies were found for other types of follow-up. We recommend that survivors be made aware of the potential harms and benefits of surveillance, including a discussion of the limitations of ca125 testing. Women could be offered the option of no formal follow-up or a follow-up schedule that is agreed upon by the woman and her health care provider. Education about the most common symptoms of recurrence should be provided. Alternative models of care such as nurse-led or telephone-based follow-up (or both) could be emerging options. CONCLUSIONS The recommendations provided in this guidance document have a limited evidence base. Recommendations should be updated as further information becomes available.
Collapse
Affiliation(s)
- T Le
- The Ottawa Hospital, Ottawa
| | - E B Kennedy
- Program in Evidence-Based Care, Cancer Care Ontario and McMaster University, Hamilton
| | - J Dodge
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Hamilton, ON
| | - L Elit
- Juravinski Cancer Centre, Hamilton, ON
| |
Collapse
|
30
|
Bonito NA, Borley J, Wilhelm-Benartzi CS, Ghaem-Maghami S, Brown R. Epigenetic Regulation of the Homeobox Gene MSX1 Associates with Platinum-Resistant Disease in High-Grade Serous Epithelial Ovarian Cancer. Clin Cancer Res 2016; 22:3097-3104. [PMID: 26763252 PMCID: PMC4849558 DOI: 10.1158/1078-0432.ccr-15-1669] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 12/24/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Although high-grade serous ovarian cancer (HGSOC) is frequently chemoresponsive, a proportion of patients do not respond to platinum-based chemotherapy at presentation or have progression-free survival (PFS) of less than 6 months. Validated predictive biomarkers of lack of response would enable alternative treatment stratification for these patients and identify novel mechanisms of intrinsic resistance. Our aim was to identify DNA methylation biomarkers of poor response to chemotherapy and demonstrate involvement of the associated gene in platinum drug cell sensitivity. EXPERIMENTAL DESIGN DNA methylation was investigated in independent tumor cohorts using Illumina HumanMethylation arrays and gene expression by Affymetrix arrays and qRT-PCR. The role of Msh homeobox 1 (MSX1) in drug sensitivity was investigated by gene reintroduction and siRNA knockdown of ovarian cancer cell lines. RESULTS CpG sites at contiguous genomic locations within the MSX1 gene have significantly lower levels of methylation in independent cohorts of HGSOC patients, which recur by 6 months compared with after 12 months (P < 0.05, q < 0.05, n = 78), have poor RECIST response (P < 0.05, q < 0.05, n = 61), and are associated with PFS in an independent cohort (n = 146). A decrease in methylation at these CpG sites correlates with decreased MSX1 gene expression. MSX1 expression is associated with PFS (HR, 0.92; 95% CI, 0.85-0.99; P = 0.029; n = 309). Cisplatin-resistant ovarian cancer cell lines have reduced MSX1 expression, and MSX1 overexpression leads to cisplatin sensitization, increased apoptosis, and increased cisplatin-induced p21 expression. CONCLUSIONS Hypomethylation of CpG sites within the MSX1 gene is associated with resistant HGSOC disease at presentation and identifies expression of MSX1 as conferring platinum drug sensitivity. Clin Cancer Res; 22(12); 3097-104. ©2016 AACR.
Collapse
Affiliation(s)
- Nair A. Bonito
- Department of Surgery & Cancer, Imperial College London, London W12 0NN, UK
| | - Jane Borley
- Department of Surgery & Cancer, Imperial College London, London W12 0NN, UK
| | | | | | - Robert Brown
- Department of Surgery & Cancer, Imperial College London, London W12 0NN, UK
- Institute of Cancer Research, Sutton, London SM2 5NG, UK
| |
Collapse
|
31
|
Pathak HB, Zhou Y, Sethi G, Hirst J, Schilder RJ, Golemis EA, Godwin AK. A Synthetic Lethality Screen Using a Focused siRNA Library to Identify Sensitizers to Dasatinib Therapy for the Treatment of Epithelial Ovarian Cancer. PLoS One 2015; 10:e0144126. [PMID: 26637171 PMCID: PMC4670180 DOI: 10.1371/journal.pone.0144126] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/15/2015] [Indexed: 02/07/2023] Open
Abstract
Molecular targeted therapies have been the focus of recent clinical trials for the treatment of patients with recurrent epithelial ovarian cancer (EOC). The majority have not fared well as monotherapies for improving survival of these patients. Poor bioavailability, lack of predictive biomarkers, and the presence of multiple survival pathways can all diminish the success of a targeted agent. Dasatinib is a tyrosine kinase inhibitor of the Src-family kinases (SFK) and in preclinical studies shown to have substantial activity in EOC. However, when evaluated in a phase 2 clinical trial for patients with recurrent or persistent EOC, it was found to have minimal activity. We hypothesized that synthetic lethality screens performed using a cogently designed siRNA library would identify second-site molecular targets that could synergize with SFK inhibition and improve dasatinib efficacy. Using a systematic approach, we performed primary siRNA screening using a library focused on 638 genes corresponding to a network centered on EGFR, HER2, and the SFK-scaffolding proteins BCAR1, NEDD9, and EFS to screen EOC cells in combination with dasatinib. We followed up with validation studies including deconvolution screening, quantitative PCR to confirm effective gene silencing, correlation of gene expression with dasatinib sensitivity, and assessment of the clinical relevance of hits using TCGA ovarian cancer data. A refined list of five candidates (CSNK2A1, DAG1, GRB2, PRKCE, and VAV1) was identified as showing the greatest potential for improving sensitivity to dasatinib in EOC. Of these, CSNK2A1, which codes for the catalytic alpha subunit of protein kinase CK2, was selected for additional evaluation. Synergistic activity of the clinically relevant inhibitor of CK2, CX-4945, with dasatinib in reducing cell proliferation and increasing apoptosis was observed across multiple EOC cell lines. This overall approach to improving drug efficacy can be applied to other targeted agents that have similarly shown poor clinical activity.
Collapse
Affiliation(s)
- Harsh B. Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
- * E-mail:
| | - Yan Zhou
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Geetika Sethi
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jeff Hirst
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Russell J. Schilder
- Department of Gynecologic Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Erica A. Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Andrew K. Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- University of Kansas Cancer Center, Kansas City, Kansas, United States of America
| |
Collapse
|
32
|
Yu AS, Zhao L. Effects of the GSK-3β inhibitor (2Z,3E)-6-bromoindirubin-3'-oxime upon ovarian cancer cells. Tumour Biol 2015; 37:4857-64. [PMID: 26526575 DOI: 10.1007/s13277-015-4344-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 10/28/2015] [Indexed: 02/08/2023] Open
Abstract
Ovarian cancer (OC) is a deadly disease, and despite improvements in treatment, overall 5-year survival is low. Glycogen synthase kinase (GSK)-3β is a multifunctional serine/threonine kinase. We wished to ascertain if the GSK-3β inhibitor (2Z,3E)-6-bromoindirubin-3'-oxime, known as "BIO," can suppress OC development. The OC cell lines A2780 and OVCAR3 were exposed to BIO. At different time points, cell proliferation, apoptosis, cell cycle, and cell invasion/cell migration assays were carried out. Phalloidin staining was undertaken to observe lamellipodia formation. Real-time reverse transcription-polymerase chain reaction and western blotting were used to assess expression of messenger RNA (mRNA) and protein of GSK-3β, cyclin D1, matrix metalloproteinase (MMP)-9, and p21. BIO suppressed the proliferation, invasion, and migration of OC cells; reduced lamellipodia formation; and induced G1 arrest of the cell cycle. BIO exposure led to a significant downregulation of mRNA and protein expression of cyclin D1 and MMP9 in comparison with untreated control cells. In contrast, BIO exposure upregulated mRNA and protein expression of p21 in comparison with untreated control cells. Besides, GSK-3β small interfering RNA (siRNA) transfection in ovarian cancer cells also downregulated GSK-3β, cyclin D1, and MMP9 protein expression while upregulated p21 expression. These data suggest that BIO, as an inhibitor of GSK-3β, can suppress OC development. Therefore, BIO could be a candidate drug for the treatment of OC.
Collapse
Affiliation(s)
- Ai-Song Yu
- Department of Preventive Medicine, School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Lin Zhao
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, Shenyang, 110042, People's Republic of China. .,Clinical Oncology College, Dalian Medical University, Shenyang, 110042, People's Republic of China.
| |
Collapse
|
33
|
An apoptosis-enhancing drug overcomes platinum resistance in a tumour-initiating subpopulation of ovarian cancer. Nat Commun 2015; 6:7956. [PMID: 26234182 PMCID: PMC4532886 DOI: 10.1038/ncomms8956] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/29/2015] [Indexed: 02/07/2023] Open
Abstract
High-grade serous ovarian cancers (HGSCs) are deadly malignancies that relapse despite carboplatin chemotherapy. Here we show that 16 independent primary HGSC samples contain a CA125-negative population enriched for carboplatin-resistant cancer initiating cells. Transcriptome analysis reveals upregulation of homologous recombination DNA repair and anti-apoptotic signals in this population. While treatment with carboplatin enriches for CA125-negative cells, co-treatment with carboplatin and birinapant eliminates these cells in HGSCs expressing high levels of the inhibitor of apoptosis protein cIAP in the CA125-negative population. Birinapant sensitizes CA125-negative cells to carboplatin by mediating degradation of cIAP causing cleavage of caspase 8 and restoration of apoptosis. This co-therapy significantly improves disease-free survival in vivo compared with either therapy alone in tumour-bearing mice. These findings suggest that therapeutic strategies that target CA125-negative cells may be useful in the treatment of HGSC. Despite normalization of the CA125 serum biomarker at the completion of carboplatin therapy the vast majority of patients with high grade serous ovarian cancers relapse. Here, Janzen et al., identify a sub-population of tumor cells that are CA125 negative, cancer initiating and platinum resistant but readily eliminated with the addition of apoptosis enhancing drugs to carboplatin.
Collapse
|
34
|
Ocak M, Gillman AG, Bresee J, Zhang L, Vlad AM, Müller C, Schibli R, Edwards WB, Anderson CJ, Gach HM. Folate receptor-targeted multimodality imaging of ovarian cancer in a novel syngeneic mouse model. Mol Pharm 2015; 12:542-53. [PMID: 25536192 PMCID: PMC4319688 DOI: 10.1021/mp500628g] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
A new
transplantable ovarian tumor model is presented using a novel
folate receptor (FR) positive, murine ovarian cancer cell line that
emulates the human disease and induces widespread intraperitoneal
(i.p.) tumors in immunocompetent mice within 4–8 weeks of implantation.
Tumor development was monitored using a new positron emission tomography
(PET) FR-targeting reporter with PET/computerized tomography (PET/CT)
and fluorescence molecular tomography (FMT) using a commercial FR-targeting
reporter. Conventional structural magnetic resonance imaging (MRI)
was also performed. Adult female C57BL/6 mice were injected i.p. with
6 × 106 MKP-L FR+ cells. Imaging was performed weekly
beginning 2 weeks after tumor induction. The albumin-binding, FR-targeting
ligand cm09 was radiolabeled with the positron emitter 68Ga and used to image the tumors with a small animal PET/CT. The FR-reporter
FolateRSense 680 (PerkinElmer) was used for FMT and flow cytometry.
Preclinical MRI (7 T) without FR-targeting was compared with the PET
and FMT molecular imaging. Tumors were visible by all three imaging
modalities. PET/CT had the highest imaging sensitivity at 3–3.5
h postadministration (mean %IA/g mean > 6) and visualized tumors
earlier
than the other two modalities with lower kidney uptake (mean %IA/g
mean < 17) than previously reported FR-targeting agents in late
stage disease. FMT showed relatively low FR-targeted agent in the
bladder and kidneys, but yielded the lowest anatomical image resolution.
MRI produced the highest resolution images, but it was difficult to
distinguish tumors from abdominal organs during early progression
since a FR-targeting MRI reporter was not used. Nevertheless, there
was good correlation of imaging biomarkers between the three modalities.
Tumors in the mouse ovarian cancer model could be detected using FR-targeted
imaging as early as 2 weeks post i.p. injection of tumor cells. An
imaging protocol should combine one or more of the modalities, e.g.,
PET/CT or PET/MRI for optimal tumor detection and delineation from
surrounding tissues.
Collapse
Affiliation(s)
- Meltem Ocak
- Department of Radiology, ‡Cancer Institute, §Department of Obstetrics, Gynecology & Reproductive Sciences, ∥Magee Womens Research Institute, ⊥Department of Pharmacology & Chemical Biology, #Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania 15213, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Chen S, Chen X, Xiu YL, Sun KX, Zong ZH, Zhao Y. microRNA 490-3P enhances the drug-resistance of human ovarian cancer cells. J Ovarian Res 2014; 7:84. [PMID: 25297343 PMCID: PMC4158041 DOI: 10.1186/s13048-014-0084-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 08/22/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are non-coding, single-stranded small RNAs that regulate gene expression negatively, which is involved in fundamental cellular processes. In this study, we investigated the role of miR-490-3P in the development of drug resistance in ovarian cancer cells. METHODS The human ovarian carcinoma cell line A2780 and A2780/Taxol were exposed to paclitaxel in the presence or absence of microRNA 490-3P transfection, after which cell viability were performed by CCK-8 assay. Reverse transcription polymerase chain reaction (RT-PCR) and western blotting were used to assess the mRNA and protein expression levels of GST-π, MDR1 or P-gp. RESULTS Our results showed higher miR-490-3P mRNA expression level in A2780/Taxol cells than in A2780 cells (p < 0.05). Following miR-490-3P transfection, both A2780 and A2780/Taxol cells showed decreased sensitivity to paclitaxel. The mRNA expression levels of MDR1, GST-π (p < 0.05) and protein expression levels of P-gp, GST-π were up-regulated [corrected] after miR-490-3P transfection in comparison to mock and negative control cancer cells. CONCLUSION Our results demonstrate for the first time that microRNA 490-3P may be involved in the development of drug resistance in ovarian cancer.
Collapse
|
36
|
Xiu YL, Zhao Y, Gou WF, Chen S, Takano Y, Zheng HC. Anacardic acid enhances the proliferation of human ovarian cancer cells. PLoS One 2014; 9:e99361. [PMID: 24921663 PMCID: PMC4055655 DOI: 10.1371/journal.pone.0099361] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 05/14/2014] [Indexed: 12/28/2022] Open
Abstract
Background Anacardic acid (AA) is a mixture of 2-hydroxy-6-alkylbenzoic acid homologs. Certain antitumor activities of AA have been reported in a variety of cancers. However, the function of AA in ovarian cancer, to date, has remained unknown. Methods Ovarian cancer cell lines were exposed to AA, after which cell proliferation, apoptosis, invasion and migration assays were performed. Phalloidin staining was used to observe lamellipodia formation. Reverse transcription polymerase chain reaction (RT-PCR) and western blotting were used to assess the mRNA and protein expression levels of Phosphatidylinositol 3-kinase (PI3K), vascular endothelial growth factor (VEGF) and caspase 3. Results Our results showed that AA promotes ovarian cancer cell proliferation, inhibits late apoptosis, and induces cell migration and invasion, as well as lamellipodia formation. AA exposure significantly up-regulated PI3K and VEGF mRNA and protein expression, while, in contrast, it down-regulated caspase 3 mRNA and protein expression in comparison to untreated control cells. Conclusion Taken together, our results demonstrate for the first time that AA may potentiate the proliferation, invasion, metastasis and lamellipodia formation in ovarian cancer cell lines via PI3K, VEGF and caspase 3 pathways.
Collapse
Affiliation(s)
- Yin-Ling Xiu
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Yang Zhao
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Wen-Feng Gou
- Department of Biochemistry and Molecular Biology, Institute of Pathology and Pathophysiology, College of Basic Medicine, China Medical University, Shenyang, P.R. China
| | - Shuo Chen
- Department of Gynecology, The First Affiliated Hospital of China Medical University, Shenyang, P.R. China
| | - Yasuo Takano
- Clinical Cancer Institute, Kanagawa Cancer Center, Yokohama, Japan
| | - Hua-Chuan Zheng
- Department of Biochemistry and Molecular Biology, Institute of Pathology and Pathophysiology, College of Basic Medicine, China Medical University, Shenyang, P.R. China
- * E-mail:
| |
Collapse
|
37
|
Koti M, Gooding RJ, Nuin P, Haslehurst A, Crane C, Weberpals J, Childs T, Bryson P, Dharsee M, Evans K, Feilotter HE, Park PC, Squire JA. Identification of the IGF1/PI3K/NF κB/ERK gene signalling networks associated with chemotherapy resistance and treatment response in high-grade serous epithelial ovarian cancer. BMC Cancer 2013; 13:549. [PMID: 24237932 PMCID: PMC3840597 DOI: 10.1186/1471-2407-13-549] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/31/2013] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Resistance to platinum-based chemotherapy remains a major impediment in the treatment of serous epithelial ovarian cancer. The objective of this study was to use gene expression profiling to delineate major deregulated pathways and biomarkers associated with the development of intrinsic chemotherapy resistance upon exposure to standard first-line therapy for ovarian cancer. METHODS The study cohort comprised 28 patients divided into two groups based on their varying sensitivity to first-line chemotherapy using progression free survival (PFS) as a surrogate of response. All 28 patients had advanced stage, high-grade serous ovarian cancer, and were treated with standard platinum-based chemotherapy. Twelve patient tumours demonstrating relative resistance to platinum chemotherapy corresponding to shorter PFS (< eight months) were compared to sixteen tumours from platinum-sensitive patients (PFS > eighteen months). Whole transcriptome profiling was performed using an Affymetrix high-resolution microarray platform to permit global comparisons of gene expression profiles between tumours from the resistant group and the sensitive group. RESULTS Microarray data analysis revealed a set of 204 discriminating genes possessing expression levels which could influence differential chemotherapy response between the two groups. Robust statistical testing was then performed which eliminated a dependence on the normalization algorithm employed, producing a restricted list of differentially regulated genes, and which found IGF1 to be the most strongly differentially expressed gene. Pathway analysis, based on the list of 204 genes, revealed enrichment in genes primarily involved in the IGF1/PI3K/NF κB/ERK gene signalling networks. CONCLUSIONS This study has identified pathway specific prognostic biomarkers possibly underlying a differential chemotherapy response in patients undergoing standard platinum-based treatment of serous epithelial ovarian cancer. In addition, our results provide a pathway context for further experimental validations, and the findings are a significant step towards future therapeutic interventions.
Collapse
Affiliation(s)
- Madhuri Koti
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Robert J Gooding
- Department of Physics, Engineering Physics and Astronomy, Queen’s University, Kingston, ON, Canada
| | - Paulo Nuin
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
- Ontario Cancer Biomarker Network, Toronto, ON, Canada
| | - Alexandria Haslehurst
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Colleen Crane
- Department of Pathology, The Ottawa Hospital, Ottawa, ON, Canada
| | - Johanne Weberpals
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Timothy Childs
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Peter Bryson
- Department of Obstetrics and Gynecology, Queen’s University, Kingston, ON, Canada
| | - Moyez Dharsee
- Ontario Cancer Biomarker Network, Toronto, ON, Canada
| | - Kenneth Evans
- Ontario Cancer Biomarker Network, Toronto, ON, Canada
| | - Harriet E Feilotter
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Paul C Park
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Jeremy A Squire
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
- Departments of Genetics and Pathology, Faculdade de Medicina de Ribeirão Preto, University of Sao Paulo, Brazil
| |
Collapse
|
38
|
Investigation of bendamustine HCL in a phase 2 study in women with resistant ovarian cancer. Invest New Drugs 2012; 31:160-6. [PMID: 22580577 DOI: 10.1007/s10637-012-9827-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 04/25/2012] [Indexed: 10/28/2022]
Abstract
We investigated the safety and efficacy of 90 mg/m(2) bendamustine HCL, administered intravenously on days 1 and 2 every 28 days in 10 women with platinum and taxane resistant epithelial ovarian cancer. There were no objective tumor responses observed; 2 patients had stable disease. Plasma samples collected at pre-treatment and end of cycle one were analyzed for changes in circulating total cytokeratin 18 and caspase cleaved cytokeratin 18 as exploratory early biomarkers of bendamustine-induced tumor cell death. All patients had measureable levels of both total and cleaved caspase 3 cytokeratin 18, but no relationship with response was possible due to the lack of clinical benefit in treated patients. Due to the high incidence of adverse events and absence of objective responses, only ten patients were treated as predefined by the Simon Two-Stage Design in the protocol. Overall, the regimen was not well tolerated and was associated with fatigue and a greater number of gastrointestinal side effects as compared to previously reported experiences in different patient populations. However, our study subjects did experience less bone marrow suppression. The lack of tolerability could reflect the degree of tumor burden within the peritoneal cavity as well as the high number of prior regimens (median of 5) received by the patients participating in this study.
Collapse
|