1
|
Kim Y, Park WH, Suh DH, Kim K, No JH, Kim YB. Anticancer Effects of BRD4 Inhibitor in Epithelial Ovarian Cancer. Cancers (Basel) 2024; 16:959. [PMID: 38473320 DOI: 10.3390/cancers16050959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Efforts have been made to develop bromodomain inhibitors as cancer treatments. Sub-pathways, particularly in ovarian cancer, affected by bromodomain-containing protein (BRD) remain unclear. This study verified the antitumor effects of a new drug that can overcome OPT-0139-chemoresistance to treat ovarian cancer. A mouse xenograft model of human ovarian cancer cells, SKOV3 and OVCAR3, was used in this study. Cell viability and proliferation were assessed using MTT and ATP assays. Cell cycle arrest and apoptosis were determined using flow cytometry. BRD4 and c-Myc expression and apoptosis-related molecules were detected using RT-PCR and real-time PCR and Western blot. We confirmed the OPT-0139 effect and mechanism of action in epithelial ovarian cancer. OPT-0139 significantly reduced cell viability and proliferation and induced apoptosis and cell cycle arrest. In the mouse xenograft model, significant changes in tumor growth, volume, weight, and BRD4-related gene expression were observed, suggesting the antitumor effects of BRD4 inhibitors. Combination therapy with cisplatin promoted apoptosis and suppressed tumor growth in vitro and in vivo. Our results suggest OPT-0139, a BRD4 inhibitor, as a promising anticancer drug for the treatment of ovarian cancer by inhibiting cell proliferation, decreasing cell viability, arresting cell cycle, and inducing apoptosis.
Collapse
Affiliation(s)
- Yeorae Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
| | - Wook-Ha Park
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
| | - Dong-Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Jae-Hong No
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Yong-Beom Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
2
|
Poursheikhani A, Yousefi H, Tavakoli-Bazzaz J, Seyed H G. EGFR Blockade Reverses Cisplatin Resistance in Human Epithelial Ovarian Cancer Cells. IRANIAN BIOMEDICAL JOURNAL 2020; 24:370-8. [PMID: 32660222 PMCID: PMC7601546 DOI: 10.29252/ibj.24.6.365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: EOC is one of the most lethal gynecological malignancy worldwide. Although the majority of EOC patients achieve clinical remission after induction therapy, over 80% relapse and succumb to the chemoresistant disease. Previous investigations have demonstrated the association of EGFR with resistance to cytotoxic chemotherapies, hormone therapy, and radiotherapy in the cancers. These studies have highlighted the role of EGFR as an attractive therapeutic target in cisplatin-resistant EOC cells. Methods: The human ovarian cell lines (SKOV3 and OVCAR3) were cultured according to ATCC recommendations. The MTT assay was used to determine the chemosensitivity of the cell lines in exposure to cisplatin and erlotinib. The qRT-PCR was applied to analyze the mRNA expression of the desired genes. Results: Erlotinib in combination with cisplatin reduced the cell proliferation in the chemoresistant EOC cells in comparison to monotherapy of the drugs (p < 0.05). Moreover, erlotinib/cisplatin combination synergistically decreased the expression of anti-apoptotic and also increased pro-apoptotic genes expression (p < 0.05). Cisplatin alone could increase the expression of MDR genes. The data suggested that EGFR and cisplatin drive chemoresistance in the EOC cells through MEKK signal transduction as well as through EGFR/MEKK pathways in the cells, respectively. Conclusion: Our findings propose that EGFR is an attractive therapeutic target in chemoresistant EOC to be exploited in translational oncology, and erlotinib/cisplatin combination treatment is a potential anti-cancer approach to overcome chemoresistance and inhibit the proliferation of the EOC cells.
Collapse
Affiliation(s)
- Arash Poursheikhani
- Medical Genetics Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Hematology/Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Hematology/Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Louisiana State University, School of Medicine, New Orleans, USA
| | - Javad Tavakoli-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghaffari Seyed H
- Hematology/Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Jiang X. Silencing of heart and neural crest derivatives expressed transcript 2 attenuates transforming growth factor-β1-enhanced apoptosis of human bronchial epithelial cells. Oncol Lett 2018; 16:4997-5005. [PMID: 30250565 PMCID: PMC6144912 DOI: 10.3892/ol.2018.9299] [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: 09/14/2017] [Accepted: 01/22/2018] [Indexed: 01/10/2023] Open
Abstract
Human bronchial epithelial (HBE) cells form the first protective barrier of the airway to protect patients from pulmonary diseases. The present study was performed to illustrate the mechanism underlying the effect of silencing heart and neural crest derivatives expressed transcript 2 (HAND2) on attenuating the transforming growth factor (TGF)-β1-enhanced apoptosis of HBE cells. TGF-β1 (10 µg/ml) was applied to HBE cells, and the HBE cells were transfected with small interfering RNA targeting HAND2 or were transfected with non-specific sequence. Subsequently, cell proliferation was measured using a Cell Counting kit-8 assay, whereas cell cycle and apoptosis status were measured using a flow cytometer. Reverse transcription-quantitative polymerase chain reaction and western blot analyses were performed to detect the expression levels of cell cycle- and apoptosis-related factors. Western blot analysis was also used to detect the phosphorylation levels of extracellular signal-regulated kinase (ERK), P38 and c-Jun-N-terminal kinase (JNK) of mitogen-activated protein kinase (MAPK) pathways. The results showed that TGF-β1 decreased HBE cell proliferation ability, arrested cell cycle at the G2 phase and promoted cell apoptosis with statistical significance. The expression levels of P21 and Cyclin D1 were inhibited, and those of caspase-3, caspase-8 and caspase-9 were promoted by TGF-β1. The phosphorylation levels of ERK, P38 and JNK were increased by TGF-β1. HAND2-silencing significantly alleviated the above functions of TGF-β1 on the HBE cells. In conclusion, the silencing of HAND2 attenuated the TGF-β1-stimulated apoptosis of HBE cells through regulating cell cycle, apoptosis-related factors and ERK/P38/JNK MAPK pathways. This may provide a novel treatment strategy for pulmonary disease, with HAND2 as the novel gene target.
Collapse
Affiliation(s)
- Xiaohui Jiang
- Intensive Care Unit, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
4
|
Abstract
Ovarian cancer is the most common gynecological malignancy in the United States, and prognosis is generally poor because the disease is often diagnosed at an advanced stage. Cyclin-dependent kinases (CDKs) are a family of serine/threonine kinases whose activity is regulated by CDK inhibitors (CKIs) and cyclins. Generally, cyclins and CKIs promote and inhibit CDK activation, respectively. Since cancer commonly involves dysregulation of cell cycle, cyclins and CDKs have been targeted in a variety of tumors using small molecules, peptides, immunotherapy, and CKIs. In this review we discuss the significance of cell cycle dysregulation in ovarian cancer as well as recent advances targeting CDKs in ovarian cancer and potential future directions. Although many of the studies assessing CDK-targeting therapies in ovarian cancer are at an early preclinical stage, there is significant evidence that targeting CDKs, particularly in combination with traditional platinum-based drugs, could have significant efficacy in ovarian cancer. Nevertheless, before these agents can be investigated in humans, additional preclinical development is needed, including using in vivo tumor models and additional studies into their mechanism of action.
Collapse
Affiliation(s)
- Qi Zhou
- a Department of Obstetrics and Gynecology , The Affiliate Hospital of Guizhou Medical University , Guizhou , China
| |
Collapse
|
5
|
Abstract
PURPOSE P27(kip1) is a negative cell cycle regulator that plays an important role in tumor suppression. Deregulation of p27(kip1) is commonly observed in many human cancers. Numerous studies about p27(kip1) are reported in clinical patients despite variable data for the prognostic of p27(kip1) expression. Here we report a meta-analysis of the association of p27(kip1) expression with the survival of ovarian cancer. METHODS PubMed and Web of science were searched for studies evaluating expression of p27(kip1) and prognostic in ovarian cancer. Published data were extracted and computed into odds ratios (ORs) for death at 3 and 5 years. Data were pooled using the random-effect model. All statistical tests were two-sided. RESULTS Analysis included 9 studies: six studies were reported in European, three studies were reported in American, and one study was reported in Asian. Loss of p27(kip1) was associated with worse overall survival (OS) at both 3 years [OR = 2.61, 95 % confidence interval (CI) 1.95-3.49, p < 0.05] and 5 years (OR = 3.01, 95 % CI 2.17-4.17, p < 0.05). Among studies with different ethnicity (European, American and Asian), the results showed a more significant association in European, including Italy, Germany, and Greece [for both 3-year OS (OR = 3.53, 95 % CI 2.37-5.26) and 5-year OS (OR = 3.66, 95 % CI 2.30-5.83)]. CONCLUSIONS Loss of p27(kip1) is associated with worse survival in ovarian cancer. The development of strategies target p27(kip1) could be a reasonable therapeutic approach.
Collapse
|
6
|
Felix AS, Sherman ME, Hewitt SM, Gunja MZ, Yang HP, Cora RL, Boudreau V, Ylaya K, Lissowska J, Brinton LA, Wentzensen N. Cell-cycle protein expression in a population-based study of ovarian and endometrial cancers. Front Oncol 2015; 5:25. [PMID: 25709969 PMCID: PMC4321403 DOI: 10.3389/fonc.2015.00025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/23/2015] [Indexed: 01/21/2023] Open
Abstract
Aberrant expression of cyclin-dependent kinase (CDK) inhibitors is implicated in the carcinogenesis of many cancers, including ovarian and endometrial cancers. We examined associations between CDK inhibitor expression, cancer risk factors, tumor characteristics, and survival outcomes among ovarian and endometrial cancer patients enrolled in a population-based case-control study. Expression (negative vs. positive) of three CDK inhibitors (p16, p21, and p27) and ki67 was examined with immunohistochemical staining of tissue microarrays. Logistic regression was used to estimate adjusted odds ratios (ORs) and 95% confidence intervals (CIs) for associations between biomarkers, risk factors, and tumor characteristics. Survival outcomes were only available for ovarian cancer patients and examined using Kaplan-Meier plots and Cox proportional hazards regression. Among ovarian cancer patients (n = 175), positive p21 expression was associated with endometrioid tumors (OR = 12.22, 95% CI = 1.45-102.78) and higher overall survival (log-rank p = 0.002). In Cox models adjusted for stage, grade, and histology, the association between p21 expression and overall survival was borderline significant (hazard ratio = 0.65, 95% CI = 0.42-1.05). Among endometrial cancer patients (n = 289), positive p21 expression was inversely associated with age (OR ≥ 65 years of age = 0.25, 95% CI = 0.07-0.84) and current smoking status (OR: 0.33, 95% CI 0.15, 0.72) compared to negative expression. Our study showed heterogeneity in expression of cell-cycle proteins associated with risk factors and tumor characteristics of gynecologic cancers. Future studies to assess these markers of etiological classification and behavior may be warranted.
Collapse
Affiliation(s)
- Ashley S Felix
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA ; Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Mark E Sherman
- Breast and Gynecologic Cancer Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Munira Z Gunja
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Hannah P Yang
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Renata L Cora
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Vicky Boudreau
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Kris Ylaya
- Tissue Array Research Program, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Jolanta Lissowska
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology , Warsaw , Poland
| | - Louise A Brinton
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Nicolas Wentzensen
- Hormonal and Reproductive Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
7
|
Saldanha SN, Tollefsbol TO. Pathway modulations and epigenetic alterations in ovarian tumorbiogenesis. J Cell Physiol 2014; 229:393-406. [PMID: 24105793 DOI: 10.1002/jcp.24466] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 09/06/2013] [Indexed: 12/23/2022]
Abstract
Cellular pathways are numerous and are highly integrated in function in the control of cellular systems. They collectively regulate cell division, proliferation, survival and apoptosis of cells and mutagenesis of key genes that control these pathways can initiate neoplastic transformations. Understanding these pathways is crucial to future therapeutic and preventive strategies of the disease. Ovarian cancers are of three major types; epithelial, germ-cell, and stromal. However, ovarian cancers of epithelial origin, arising from the mesothelium, are the predominant form. Of the subtypes of ovarian cancer, the high-grade serous tumors are fatal, with low survival rate due to late detection and poor response to treatments. Close examination of preserved ovarian tissues and in vitro studies have provided insights into the mechanistic changes occurring in cells mediated by a few key genes. This review will focus on pathways and key genes of the pathways that are mutated or have aberrant functions in the pathology of ovarian cancer. Non-genetic mechanisms that are gaining prominence in the pathology of ovarian cancer, miRNAs and epigenetics, will also be discussed in the review.
Collapse
Affiliation(s)
- Sabita N Saldanha
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama; Department of Biological Sciences, Alabama State University, Montgomery, Alabama
| | | |
Collapse
|
8
|
Rescigno P, Cerillo I, Ruocco R, Condello C, De Placido S, Pensabene M. New hypothesis on pathogenesis of ovarian cancer lead to future tailored approaches. BIOMED RESEARCH INTERNATIONAL 2013; 2013:852839. [PMID: 24063014 PMCID: PMC3766984 DOI: 10.1155/2013/852839] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 05/14/2013] [Indexed: 11/17/2022]
Abstract
In the last decades, management of epithelial ovarian cancer (EOC) has been based on the staging system of the International Federation of Gynecology and Obstetrics (FIGO), and different classifications have been proposed for EOC that take account of grade of differentiation, histological subtype, and clinical features. However, despite taxonomic efforts, EOC appears to be not a unique disease; its subtypes differ for epidemiological and genetic risk factors, precursor lesions, patterns of spread, response to chemotherapy, and prognosis. Nevertheless, carboplatin plus paclitaxel combination represents the only standard treatment in adjuvant and advanced settings. This paper summarizes theories about the classification and origin of EOC and classical and new prognostic factors. It presents data about standard treatment and novel agents. We speculate about the possibility to create tailored therapy based on specific mutations in ovarian cancer and to personalize prevention.
Collapse
Affiliation(s)
- P. Rescigno
- Department of Clinical Medicine and Surgery, University Hospital and University Federico II, via Pansini 5, 80131 Naples, Italy
| | - I. Cerillo
- Department of Clinical Medicine and Surgery, University Hospital and University Federico II, via Pansini 5, 80131 Naples, Italy
| | - R. Ruocco
- Department of Clinical Medicine and Surgery, University Hospital and University Federico II, via Pansini 5, 80131 Naples, Italy
| | - C. Condello
- Department of Clinical Medicine and Surgery, University Hospital and University Federico II, via Pansini 5, 80131 Naples, Italy
| | - S. De Placido
- Department of Clinical Medicine and Surgery, University Hospital and University Federico II, via Pansini 5, 80131 Naples, Italy
| | - M. Pensabene
- Department of Clinical Medicine and Surgery, University Hospital and University Federico II, via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
9
|
Heeran MC, Høgdall CK, Kjaer SK, Christensen L, Blaakaer J, Christensen IJ, Hogdall EVS. Limited prognostic value of tissue protein expression levels of cyclin E in Danish ovarian cancer patients: from the Danish 'MALOVA' ovarian cancer study. APMIS 2012; 120:846-54. [PMID: 22958293 DOI: 10.1111/j.1600-0463.2012.02913.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 04/02/2012] [Indexed: 11/29/2022]
Abstract
The primary objective of this study was to assess the expression of cyclin E in tumour tissues from 661 patients with epithelial ovarian tumours. The second was to evaluate whether cyclin E tissue expression levels correlate with clinico-pathological parameters and prognosis of the disease. Using tissue arrays (TA), we analysed the cyclin E expression levels in tissues from 168 women with borderline ovarian tumours (BOT) (147 stage I, 4 stage II, 17 stage III) and 493 Ovarian cancer (OC) patients (127 stage I, 45 stage II, 276 stage III, 45 stage IV). Using a 10% cut-off level for cyclin E overexpression, 20% of the BOTs were positive with a higher proportion of serous than mucinous tumours. Sixty-two per cent of the OCs were positive for cyclin E expression with the highest percentage found in clear cell carcinomas. Results based on univariate and multivariate survival analyses with a 10% cut-off value showed that cyclin E had no independent prognostic value. In conclusion, we found cyclin E expression in tumour tissue to be of limited prognostic value to Danish OC patients.
Collapse
Affiliation(s)
- Mel C Heeran
- Department of Pathology, Herlev Hospital, University of Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
10
|
Wang R, Wang Z, Yang J, Liu X, Wang L, Guo X, Zeng F, Wu M, Li G. LRRC4 inhibits the proliferation of human glioma cells by modulating the expression of STMN1 and microtubule polymerization. J Cell Biochem 2012; 112:3621-9. [PMID: 21809374 DOI: 10.1002/jcb.23293] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
LRRC4 is a tumor suppressor of glioma, and it is epigenetically inactivated commonly in glioma. Our previous study has shown that induction of LRRC4 expression inhibits the proliferation of glioma cells. However, little is known about the mechanisms underlying the action of LRRC4 in glioma cells. We employed two-dimensional fluorescence differential gel electrophoresis (2-D DIGE) and MALDI -TOF/TOF-MS/MS to identify 11 differentially expressed proteins, including the significantly down-regulated STMN1 expression in the LRRC4-expressing U251 glioma cells. The levels of STMN1 expression appeared to be positively associated with the pathogenic degrees of human glioma. Furthermore, induction of LRRC4 over-expression inhibited the STMN1 expression and U251 cell proliferation in vitro, and the glioma growth in vivo. In addition, induction of LRRC4 or knockdown of STMN1 expression induced cell cycle arrest in U251 cells, which was associated with modulating the p21, cyclin D1, and cyclin B expression, and the ERK phosphorylation, and inhibiting the CDK5 and cdc2 kinase activities, but increasing the microtubulin polymerization in U251 cells. LRRC4, at least partially by down-regulating the STMN1expression, acts as a major glioma suppressor, induces cell cycle arrest and modulates the dynamic process of microtubulin, leading to the inhibition of glioma cell proliferation and growth. Potentially, modulation of LRRC4 or STMN1 expression may be useful for design of new therapies for the intervention of glioma.
Collapse
Affiliation(s)
- Rong Wang
- Cancer Research Institute, Central South University, Changsha, Hunan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Gyorffy B, Lánczky A, Szállási Z. Implementing an online tool for genome-wide validation of survival-associated biomarkers in ovarian-cancer using microarray data from 1287 patients. Endocr Relat Cancer 2012; 19:197-208. [PMID: 22277193 DOI: 10.1530/erc-11-0329] [Citation(s) in RCA: 675] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The validation of prognostic biomarkers in large independent patient cohorts is a major bottleneck in ovarian cancer research. We implemented an online tool to assess the prognostic value of the expression levels of all microarray-quantified genes in ovarian cancer patients. First, a database was set up using gene expression data and survival information of 1287 ovarian cancer patients downloaded from Gene Expression Omnibus and The Cancer Genome Atlas (Affymetrix HG-U133A, HG-U133A 2.0, and HG-U133 Plus 2.0 microarrays). After quality control and normalization, only probes present on all three Affymetrix platforms were retained (n=22,277). To analyze the prognostic value of the selected gene, we divided the patients into two groups according to various quantile expressions of the gene. These groups were then compared using progression-free survival (n=1090) or overall survival (n=1287). A Kaplan-Meier survival plot was generated and significance was computed. The tool can be accessed online at www.kmplot.com/ovar. We used this integrative data analysis tool to validate the prognostic power of 37 biomarkers identified in the literature. Of these, CA125 (MUC16; P=3.7×10(-5), hazard ratio (HR)=1.4), CDKN1B (P=5.4×10(-5), HR=1.4), KLK6 (P=0.002, HR=0.79), IFNG (P=0.004, HR=0.81), P16 (P=0.02, HR=0.66), and BIRC5 (P=0.00017, HR=0.75) were associated with survival. The combination of several probe sets can further increase prediction efficiency. In summary, we developed a global online biomarker validation platform that mines all available microarray data to assess the prognostic power of 22,277 genes in 1287 ovarian cancer patients. We specifically used this tool to evaluate the effect of 37 previously published biomarkers on ovarian cancer prognosis.
Collapse
Affiliation(s)
- Balázs Gyorffy
- Research Laboratory of Pediatrics and Nephrology, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | |
Collapse
|
12
|
Hershko DD. Cyclin-dependent kinase inhibitor p27 as a prognostic biomarker and potential cancer therapeutic target. Future Oncol 2010; 6:1837-47. [PMID: 21142858 DOI: 10.2217/fon.10.144] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The prognosis and clinical management of patients with cancer is commonly determined by traditional clinical and pathological factors. Nevertheless, patients may present with significantly different clinical outcomes despite similar clinicopathological features. This has prompted intense research to find biological markers that may closely reflect tumor biology and thereby clinical outcome. This article presents the current knowledge on the prognostic significance of p27 expression in cancer and its potential role as a target for future therapy.
Collapse
Affiliation(s)
- Dan D Hershko
- Department of Surgery & Breast Health Institute, Rambam Health Care Campus & the Technion – Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
13
|
Le Page C, Huntsman DG, Provencher DM, Mes-Masson AM. Predictive and prognostic protein biomarkers in epithelial ovarian cancer: recommendation for future studies. Cancers (Basel) 2010; 2:913-54. [PMID: 24281100 PMCID: PMC3835111 DOI: 10.3390/cancers2020913] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 04/19/2010] [Accepted: 05/13/2010] [Indexed: 12/18/2022] Open
Abstract
Epithelial ovarian cancer is the most lethal gynecological malignancy. Due to its lack of symptoms, this disease is diagnosed at an advanced stage when the cancer has already spread to secondary sites. While initial rates of response to first treatment is >80%, the overall survival rate of patients is extremely low, mainly due to development of drug resistance. To date, there are no reliable clinical factors that can properly stratify patients for suitable chemotherapy strategies. Clinical parameters such as disease stage, tumor grade and residual disease, although helpful in the management of patients after their initial surgery to establish the first line of treatment, are not efficient enough. Accordingly, reliable markers that are independent and complementary to clinical parameters are needed for a better management of these patients. For several years, efforts to identify prognostic factors have focused on molecular markers, with a large number having been investigated. This review aims to present a summary of the recent advances in the identification of molecular biomarkers in ovarian cancer patient tissues, as well as an overview of the need and importance of molecular markers for personalized medicine in ovarian cancer.
Collapse
Affiliation(s)
- Cécile Le Page
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CR/CHUM), Institut du cancer de Montréal, 1560 Sherbrooke Est, Montreal, H2L4M1, QC, Canada; E-Mails: (C.L.P.); (D.M.P.)
| | - David G. Huntsman
- Department of Pathology and Genetic Pathology Evaluation Centre of the Prostate Research Center, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver General Hospital, Vancouver, Canada; E-Mail: (D.G.H.)
- Translational and Applied Genomics, BC Cancer Agency, Room 3427, 600 West 10th Avenue, Vancouver, V5Z 4E6, BC, Canada
| | - Diane M. Provencher
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CR/CHUM), Institut du cancer de Montréal, 1560 Sherbrooke Est, Montreal, H2L4M1, QC, Canada; E-Mails: (C.L.P.); (D.M.P.)
- Département d’Obstétrique et Gynécologie, Clinique de Gynécologie Oncologie, Université de Montréal, 1560 Sherbrooke Est, Montreal, H2L4M1, QC, Canada; E-Mail:
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l’Université de Montréal (CR/CHUM), Institut du cancer de Montréal, 1560 Sherbrooke Est, Montreal, H2L4M1, QC, Canada; E-Mails: (C.L.P.); (D.M.P.)
- Département de Medicine, Université de Montréal, 1560 Sherbrooke Est, Montreal, H2L4M1, QC, Canada
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-514-890-8000 ext 25496; Fax: +1-514-412-7703
| |
Collapse
|
14
|
Abstract
Dysregulation of the cell cycle is an important prerequisite for cancer development. p27 has an established role in cell cycle control and hence may be disrupted during carcinogenesis. The influence of p27 expression, including its subcellular location, on tumor behavior in ovarian cancer has been controversial. The purpose of this study was to evaluate the expression of p27 in a large population of patients with ovarian cancer and correlate this to clinicopathologic variables including overall survival. Using a tissue microarray of 339 primary ovarian cancers, the expression of p27 was assessed immunohistochemically. Coupled to a comprehensive database of clinicopathologic variables, its effect on these factors and survival was studied. Cytoplasmic p27 showed a progressively negative impact on overall survival (P=0.004). Tumors displaying nuclear p27 also had poorer prognosis (P=0.014). Factors shown to predict prognosis independently of each other were age, stage, and the absence of macroscopic disease after surgery. Cytoplasmic p27 expression, but not nuclear, was independently predictive of prognosis on multivariate analysis (P=0.042). Both subcellular locations of p27 expression were more frequently observed in serous compared with mucinous subtypes. Cytoplasmic p27 independently predicts poorer prognosis in ovarian carcinoma. These results seem counterintuitive, when considering the antiproliferative role of p27, but may reflect a more complex function of p27 within cell cycle regulation. These data support a novel role for p27 within the cytoplasm, possibly through effects on apoptosis, cellular motility, and drug resistance.
Collapse
|
15
|
Gadducci A, Cosio S, Tana R, Genazzani AR. Serum and tissue biomarkers as predictive and prognostic variables in epithelial ovarian cancer. Crit Rev Oncol Hematol 2008; 69:12-27. [PMID: 18595727 DOI: 10.1016/j.critrevonc.2008.05.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 04/17/2008] [Accepted: 05/08/2008] [Indexed: 11/29/2022] Open
Abstract
Tumour stage, residual disease after initial surgery, histological type and tumour grade are the most important clinical-pathological factors related to the clinical outcome of patients with epithelial ovarian cancer. In the last years, several investigations have assessed different biological variables in sera and in tissue samples from patients with this malignancy in order to detect biomarkers able to reflect either the response to chemotherapy or survival. The present paper reviewed the literature data about the predictive or prognostic relevance of serum CA 125, soluble cytokeratin fragments, serum human kallikreins, serum cytokines, serum vascular endothelial growth factor and plasma d-dimer as well as of tissue expression of cell cycle- and apoptosis-regulatory proteins, human telomerase reverse transcriptase, membrane tyrosine kinase receptors and matrix metalloproteinases. A next future microarray technology will hopefully offer interesting perspectives of translational research for the identification of novel predictive and prognostic biomarkers for epithelial ovarian cancer.
Collapse
Affiliation(s)
- Angiolo Gadducci
- Department of Procreative Medicine, Division of Gynecology and Obstetrics, University of Pisa, Via Roma 56, Pisa 56127, Italy.
| | | | | | | |
Collapse
|
16
|
Chu IM, Hengst L, Slingerland JM. The Cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy. Nat Rev Cancer 2008; 8:253-67. [PMID: 18354415 DOI: 10.1038/nrc2347] [Citation(s) in RCA: 754] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cyclin-dependent kinase (Cdk) inhibitor p27 (also known as KIP1) regulates cell proliferation, cell motility and apoptosis. Interestingly, the protein can exert both positive and negative functions on these processes. Diverse post-translational modifications determine the physiological role of p27. Phosphorylation regulates p27 binding to and inhibition of cyclin-Cdk complexes, its localization and its ubiquitin-mediated proteolysis. In cancers, p27 is inactivated through impaired synthesis, accelerated degradation and by mislocalization. Moreover, studies in several tumour types indicate that p27 expression levels have both prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Isabel M Chu
- Braman Family Breast Cancer Institute, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, Miami, Florida 33136, USA
| | | | | |
Collapse
|
17
|
Nam EJ, Kim YT. Alteration of cell-cycle regulation in epithelial ovarian cancer. Int J Gynecol Cancer 2008; 18:1169-82. [PMID: 18298566 DOI: 10.1111/j.1525-1438.2008.01191.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In spite of the clinical importance of epithelial ovarian cancer (EOC), little is known about the pathobiology of its precursor lesions and progression. Regulatory mechanisms of the cell cycle are mainly composed of cyclins, cyclin-dependent kinases (CDK), and CDK inhibitors. Alteration of these mechanisms results in uncontrolled cell proliferation, which is a distinctive feature of human cancers. This review describes the current state of knowledge about the alterations of cell-cycle regulations in the context of p16-cyclin D1-CDK4/6-pRb pathway, p21-p27-cyclin E-CDK2 pathway, p14-MDM2-p53 pathway, and ATM-Chk2-CDC25 pathway, respectively. Recent evidence suggests that ovarian cancer is a heterogenous group of neoplasms with several different histologic types, each with its own underlying molecular genetic mechanism. Therefore, expression of cell cycle regulatory proteins should be tested separately according to each histologic type. In serous ovarian carcinoma, high expression of p16, p53, and p27 and low expression of p21 and cyclin E were shown. In addition, this review focuses on the prognostic significance of cell cycle-regulating proteins in EOC. However, it is difficult to compare the results from different groups due to diverse methodologies and interpretations. Accordingly, researchers should establish standardized criteria for the interpretation of immunohistochemical results.
Collapse
Affiliation(s)
- E J Nam
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seodaemun-gu, Seoul, Korea
| | | |
Collapse
|
18
|
Minichromosome maintenance proteins 2 and 5 in non-benign epithelial ovarian tumours: relationship with cell cycle regulators and prognostic implications. Br J Cancer 2007; 97:1124-34. [PMID: 17940502 PMCID: PMC2360432 DOI: 10.1038/sj.bjc.6603992] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Minichromosome maintenance proteins (MCM) have recently emerged as novel proliferation markers with prognostic implications in several tumour types. This is the first study investigating MCM-2 and MCM-5 immunohistochemical expression in a series of ovarian adenocarcinomas and low malignant potential (LMP) tumours aiming to determine possible associations with clinicopathological parameters, the conventional proliferation index Ki-67, cell cycle regulators (p53, p27(Kip1), p21(WAF1) and pRb) and patients' outcome. Immunohistochemistry was applied in a series of 43 cases of ovarian LMP tumours and 85 cases of adenocarcinomas. Survival analysis was restricted to adenocarcinomas. The median MCM-2 and MCM-5 labelling indices (LIs) were significantly higher in adenocarcinomas compared to LMP tumours (P<0.0001 for both associations). In adenocarcinomas, the levels of MCM-2 and MCM-5 increased significantly with advancing tumour stage (P=0.0052 and P=0.0180, respectively), whereas both MCM-2 and MCM-5 increased significantly with increasing tumour grade (P=0.0002 and P=0.0006, respectively) and the presence of bulky residual disease (P<0.0001 in both relationships). A strong positive correlation was established between MCM-2 or MCM-5 expression level and Ki-67 LI (P<0.0001) as well as p53 protein (P=0.0038 and P=0.0500, respectively). Moreover, MCM-2 LI was inversely correlated with p27(Kip-1) LI (P=0.0068). Finally, both MCM-2 and MCM-5 were associated significantly with adverse patients' outcome in both univariate (> or =20 vs >20%, P=0.0011 and > or =25 vs <25%, P=0.0100, respectively) and multivariate (P=0.0001 and 0.0090, respectively) analysis. An adequately powered independent group of 45 patients was used in order to validate our results in univariate survival analysis. In this group, MCM-2 and MCM-5 expression retained their prognostic significance (P<0.0001 in both relationships). In conclusion, MCM-2 and MCM-5 proteins appear to be promising as prognostic markers in patients with ovarian adenocarcinomas.
Collapse
|