1
|
Vaicekauskaitė I, Kazlauskaitė P, Gineikaitė R, Čiurlienė R, Lazutka JR, Sabaliauskaitė R. Integrative Analysis of Gene Expression and Promoter Methylation to Differentiate High-Grade Serous Ovarian Cancer from Benign Tumors. Biomedicines 2025; 13:441. [PMID: 40002854 PMCID: PMC11853219 DOI: 10.3390/biomedicines13020441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Ovarian cancer (OC) is the third most common and second most lethal onco-gynecological disease in the world, with high-grade serous ovarian cancer (HGSOC) making up the majority of OC cases worldwide. The current serological biomarkers used for OC diagnosis are lacking sensitivity and specificity, thus new biomarkers are greatly needed. Recently, the chromatin remodeling complex gene ARID1A, Notch and Wnt pathway gene expression, as well as HOX-related gene promoter methylation have been linked with promoting OC. Methods: In this pilot study, 10 gene expression biomarkers and 4 promoter methylation biomarkers were examined as potential diagnostic and prognostic indicators of OC in 65 fresh-frozen gynecologic tumor tissues. Results: Out of 10 genes analyzed, the expression of eight biomarkers was significantly reduced in OC cases compared to benign, and HOX-related gene promoter methylation significantly increased in OC tumors. Out of 14 biomarkers, CTNNB1 showed the best single biomarker separation of HGSOC from benign cases (AUC = 0.97), while a combination of the seven Notch pathway-related gene expressions (NOTCH1, NOTCH2, NOTCH3, NOTCH4, DLL1, JAG2, and HES1) demonstrated the best separation of HGSOC from the benign cases (AUC = 1). Conclusions: The combination of multiple gene expression or gene promoter methylation biomarkers shows great promise for the development of an effective biomarker-based diagnostic approach for OC.
Collapse
Affiliation(s)
- Ieva Vaicekauskaitė
- Institute of Biosciences, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
- National Cancer Institute, 08406 Vilnius, Lithuania
| | - Paulina Kazlauskaitė
- National Cancer Institute, 08406 Vilnius, Lithuania
- Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08661 Vilnius, Lithuania
| | - Rugilė Gineikaitė
- Institute of Biosciences, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
| | - Rūta Čiurlienė
- National Cancer Center, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
| | - Juozas Rimantas Lazutka
- Institute of Biosciences, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
| | - Rasa Sabaliauskaitė
- Institute of Biosciences, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
- National Cancer Institute, 08406 Vilnius, Lithuania
| |
Collapse
|
2
|
Fu M, Deng F, Chen J, Fu L, Lei J, Xu T, Chen Y, Zhou J, Gao Q, Ding H. Current data and future perspectives on DNA methylation in ovarian cancer (Review). Int J Oncol 2024; 64:62. [PMID: 38757340 PMCID: PMC11095605 DOI: 10.3892/ijo.2024.5650] [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: 11/23/2023] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Ovarian cancer (OC) represents the most prevalent malignancy of the female reproductive system. Its distinguishing features include a high aggressiveness, substantial morbidity and mortality, and a lack of apparent symptoms, which collectively pose significant challenges for early detection. Given that aberrant DNA methylation events leading to altered gene expression are characteristic of numerous tumor types, there has been extensive research into epigenetic mechanisms, particularly DNA methylation, in human cancers. In the context of OC, DNA methylation is often associated with the regulation of critical genes, such as BRCA1/2 and Ras‑association domain family 1A. Methylation modifications within the promoter regions of these genes not only contribute to the pathogenesis of OC, but also induce medication resistance and influence the prognosis of patients with OC. As such, a more in‑depth understanding of DNA methylation underpinning carcinogenesis could potentially facilitate the development of more effective therapeutic approaches for this intricate disease. The present review focuses on classical tumor suppressor genes, oncogenes, signaling pathways and associated microRNAs in an aim to elucidate the influence of DNA methylation on the development and progression of OC. The advantages and limitations of employing DNA methylation in the diagnosis, treatment and prevention of OC are also discussed. On the whole, the present literature review indicates that the DNA methylation of specific genes could potentially serve as a prognostic biomarker for OC and a therapeutic target for personalized treatment strategies. Further investigations in this field may yield more efficacious diagnostic and therapeutic alternatives for patients with OC.
Collapse
Affiliation(s)
- Mengyu Fu
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Fengying Deng
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jie Chen
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Li Fu
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jiahui Lei
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Ting Xu
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Department of Gynecology and Obstetrics, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215100, P.R. China
| | - Youguo Chen
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Jinhua Zhou
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qinqin Gao
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongmei Ding
- Institute for Fetology, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
3
|
Chen C, Zhu Y, Zhang H, Xiao L. Prognostic Effects of RASSF1A, BRCA1, APC, and p16 Promoter Methylation in Ovarian Cancer: A Meta-Analysis. Gynecol Obstet Invest 2024; 89:363-375. [PMID: 38615670 DOI: 10.1159/000538673] [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: 01/12/2024] [Accepted: 03/25/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION DNA methylation plays an important role in the carcinogenesis, progression, and prognosis of various human cancers. RASSF1A, BRCA1, APC, and p16 are the frequently methylated genes among patients with ovarian cancer. Therefore, our study aimed to better determine the prognostic and cancer characteristics effects of RASSF1A, BRCA1, APC, and p16 promoter methylation in ovarian cancer patients. METHODS Databases such as PubMed, Web of Science, EMBASE, CNKI, and WanFang were searched for published studies up to March 4, 2024. The outcomes are shown as OR and HR with their 95% CIs. Then, the random or fixed-effect model was performed to evaluate the effect sizes. RESULTS Finally, 27 articles were included in this meta-analysis. No significant relationships were observed between RASSF1A, BRCA1, and APC promoter methylation and the clinical prognostic (including overall survival and progression-free survival) and cancer characteristics (including ascites, lymph node metastasis, and pelvic peritoneal metastasis) in ovarian cancer. p16 promoter methylation was significantly related to poor progression-free survival (PFS) (HR = 1.52, 95% CI = 1.14-2.04) and overall survival (OS) (HR = 1.39, 95% CI = 1.06, to 1.83) in univariate and poor PFS in multivariate Cox regression models (HR = 1.42, 95% CI = 1.05-1.92). Besides, our results indicated that the clinical stage was associated with inferior OS while there was no significant association between tumor grade and OS. CONCLUSION RASSF1A, BRCA1, and APC promoter methylation were not significantly associated with clinical prognostic and cancer characteristics. p16 may be a useful biomarker for predicting PFS in ovarian cancer. Furthermore, the clinical stage was significantly associated with OS. In further research, more prospective and multicenter validation studies remain needed.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ying Zhu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haibo Zhang
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lan Xiao
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
4
|
El Gazzar WB, Albakri KA, Hasan H, Badr AM, Farag AA, Saleh OM. Poly(ADP-ribose) polymerase inhibitors in the treatment landscape of triple-negative breast cancer (TNBC). J Oncol Pharm Pract 2023; 29:1467-1479. [PMID: 37559370 DOI: 10.1177/10781552231188903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
OBJECTIVE Chemotherapy is the mainstay for triple-negative breast cancer (TNBC) patients. Over the years, the use of chemotherapy for these patients has demonstrated many adversities, including toxicity and resistance, which suggested the need to develop novel alternative therapeutic options, such as poly(ADP-ribose) polymerase inhibitors (PARPi). Herein, we provide an overview on PARPi, mechanisms of action and the role of biomarkers in PARPi sensitivity trials, clinical advances in PARPi therapy for TNBC patients based on the most recent studies and findings of clinical trials, and challenges that prevent PARP inhibitors from achieving high efficacy such as resistance and overlapping toxicities with other chemotherapies. DATA SOURCES Searching for relevant articles was done using PubMed and Cochrane Library databases by using the keywords including TNBC; chemotherapy; PARPi; BRCA; homologous recombination repair (HRR). Studies had to be published in full-text in English in order to be considered. DATA SUMMARY Although PARPi have been used in the treatment of local/metastatic breast malignancies that are HER2 negative and has a germline BRCA mutation, several questions are still to be answered in order to maximize the clinical benefit of PARP inhibitors in TNBC treatment, such as questions related to the optimal use in the neoadjuvant and metastatic settings as well as the best combinations with various chemotherapies. CONCLUSIONS PARPi are emerging treatment options for patients with gBRCA1/2 mutations. Determining patients that are most likely to benefit from PARPi and identifying the optimal treatment combinations with high efficacy and fewer side effects are currently ongoing.
Collapse
Affiliation(s)
- Walaa Bayoumie El Gazzar
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha City, Egypt
| | | | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Amira M Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amina A Farag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha City, Egypt
| | | |
Collapse
|
5
|
Hunia J, Gawalski K, Szredzka A, Suskiewicz MJ, Nowis D. The potential of PARP inhibitors in targeted cancer therapy and immunotherapy. Front Mol Biosci 2022; 9:1073797. [PMID: 36533080 PMCID: PMC9751342 DOI: 10.3389/fmolb.2022.1073797] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/15/2022] [Indexed: 07/29/2023] Open
Abstract
DNA damage response (DDR) deficiencies result in genome instability, which is one of the hallmarks of cancer. Poly (ADP-ribose) polymerase (PARP) enzymes take part in various DDR pathways, determining cell fate in the wake of DNA damage. PARPs are readily druggable and PARP inhibitors (PARPi) against the main DDR-associated PARPs, PARP1 and PARP2, are currently approved for the treatment of a range of tumor types. Inhibition of efficient PARP1/2-dependent DDR is fatal for tumor cells with homologous recombination deficiencies (HRD), especially defects in breast cancer type 1 susceptibility protein 1 or 2 (BRCA1/2)-dependent pathway, while allowing healthy cells to survive. Moreover, PARPi indirectly influence the tumor microenvironment by increasing genomic instability, immune pathway activation and PD-L1 expression on cancer cells. For this reason, PARPi might enhance sensitivity to immune checkpoint inhibitors (ICIs), such as anti-PD-(L)1 or anti-CTLA4, providing a rationale for PARPi-ICI combination therapies. In this review, we discuss the complex background of the different roles of PARP1/2 in the cell and summarize the basics of how PARPi work from bench to bedside. Furthermore, we detail the early data of ongoing clinical trials indicating the synergistic effect of PARPi and ICIs. We also introduce the diagnostic tools for therapy development and discuss the future perspectives and limitations of this approach.
Collapse
Affiliation(s)
- Jaromir Hunia
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Karol Gawalski
- Doctoral School, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | | | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
6
|
Gull N, Jones MR, Peng PC, Coetzee SG, Silva TC, Plummer JT, Reyes ALP, Davis BD, Chen SS, Lawrenson K, Lester J, Walsh C, Rimel BJ, Li AJ, Cass I, Berg Y, Govindavari JPB, Rutgers JKL, Berman BP, Karlan BY, Gayther SA. DNA methylation and transcriptomic features are preserved throughout disease recurrence and chemoresistance in high grade serous ovarian cancers. J Exp Clin Cancer Res 2022; 41:232. [PMID: 35883104 PMCID: PMC9327231 DOI: 10.1186/s13046-022-02440-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 07/13/2022] [Indexed: 11/18/2022] Open
Abstract
Background Little is known about the role of global DNA methylation in recurrence and chemoresistance of high grade serous ovarian cancer (HGSOC). Methods We performed whole genome bisulfite sequencing and transcriptome sequencing in 62 primary and recurrent tumors from 28 patients with stage III/IV HGSOC, of which 11 patients carried germline, pathogenic BRCA1 and/or BRCA2 mutations. Results Landscapes of genome-wide methylation (on average 24.2 million CpGs per tumor) and transcriptomes in primary and recurrent tumors showed extensive heterogeneity between patients but were highly preserved in tumors from the same patient. We identified significant differences in the burden of differentially methylated regions (DMRs) in tumors from BRCA1/2 compared to non-BRCA1/2 carriers (mean 659 DMRs and 388 DMRs in paired comparisons respectively). We identified overexpression of immune pathways in BRCA1/2 carriers compared to non-carriers, implicating an increased immune response in improved survival (P = 0.006) in these BRCA1/2 carriers. Conclusion These findings indicate methylome and gene expression programs established in the primary tumor are conserved throughout disease progression, even after extensive chemotherapy treatment, and that changes in methylation and gene expression are unlikely to serve as drivers for chemoresistance in HGSOC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02440-z.
Collapse
|
7
|
Li XF, Zhang HB, Huo Y. High HOXA9 gene expression predicts response to chemotherapy and prognosis of high-grade serous ovarian cancer patients. J Int Med Res 2022; 50:3000605221135864. [PMID: 36366735 PMCID: PMC9659939 DOI: 10.1177/03000605221135864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/22/2022] [Indexed: 02/28/2025] Open
Abstract
OBJECTIVE High-grade serous ovarian cancer (HGSOC) is a deadly malignancy. Homeobox protein A9 (HOXA9) is linked with serous papillary histotype differentiation, and inappropriate HOXA9 expression is a step in ovarian cancer that induces aberrant differentiation. This study aimed to reveal the significance of HOXA9 in HGSOC. METHODS HOXA9 mRNA and protein expression were examined by quantitative PCR and immunohistochemistry, respectively. The chi-square test was used to evaluate associations between HOXA9 expression and clinical characteristics. The prognostic value of HOXA9 was calculated by the Kaplan-Meier method. The Kaplan-Meier Plotter database was used to assess the prognostic value of HOXA9. RESULTS The mRNA and protein expression of HOXA9 were significantly upregulated in chemotherapy-resistant HGSOC compared with chemotherapy-sensitive HGSOC. The chi-square test showed that high HOXA9 expression was significantly related with grade, clinical stage, and residual disease. High HOXA9 expression was significantly associated with poor prognosis. The Kaplan-Meier Plotter database further confirmed these results. Cox hazard regression showed that high HOXA9 expression was an independent prognostic factor for survival in HGSOC patients. CONCLUSION This study showed that HOXA9 expression was associated with chemotherapy resistance and poor outcomes in HGSOC patients. High HOXA9 expression might be a prognostic indicator for HGSOC.
Collapse
Affiliation(s)
- Xiao-fei Li
- Department of Obstetrics and Gynecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Hai-Bo Zhang
- Department of Obstetrics and Gynecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Yan Huo
- Department of Intensive Care Unit, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| |
Collapse
|
8
|
Faaborg L, Andersen RF, Waldstrøm M, Henriksen JR, Adimi P, Jakobsen A, Steffensen KD. Prognostic Impact of Circulating Methylated Homeobox A9 DNA in Patients Undergoing Treatment for Recurrent Ovarian Cancer. Cancers (Basel) 2022; 14:1766. [PMID: 35406538 PMCID: PMC8997085 DOI: 10.3390/cancers14071766] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 01/23/2023] Open
Abstract
Methylated Homeobox A9 circulating tumor DNA (meth-HOXA9) has been suggested as a blood-based biomarker in epithelial ovarian cancer (EOC), although its prognostic significance remains unproven. The aim of the present study was to investigate the prognostic impact of meth-HOXA9 in patients with recurrent EOC. DNA was purified from 4 mL plasma and, following bilsulfite conversion, meth-HOXA9 was analyzed using a methylation-specific droplet digital PCR. Detection of meth-HOXA9 was reported as a percentage of total DNA and as a binary variable (detectable and undetectable). Meth-HOXA9 status and its dynamics during palliative treatment were correlated with overall survival (OS) as the primary endpoint. At baseline, meth-HOXA9 was detected in 65.9% (83/126) of the patients. The median OS was 8.9 and 17.9 months in patients with detectable and undetectable meth-HOXA9 at baseline (hazard ratio: 2.04, p = 0.002), which remained significant in the multivariate analysis. Median OS in patients with an increase in meth-HOXA9 after one treatment cycle was 5.3 months compared to 33 months in patients with undetectable meth-HOXA9 (p < 0.001). Meth-HOXA9 was significantly related to poor survival and may serve as a prognostic marker in patients with recurrent EOC. The longitudinal monitoring of meth-HOXA9 is clinically feasible with the perspective of aiding clinical decision making.
Collapse
Affiliation(s)
- Louise Faaborg
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (P.A.); (A.J.); (K.D.S.)
- Department of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 5000 Odense C, Denmark
| | - Rikke Fredslund Andersen
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark;
| | - Marianne Waldstrøm
- Department of Pathology, Lillebaelt Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark;
| | - Jon Røikjær Henriksen
- Department of Oncology, Odense University Hospital, J.B. Winsløws Vej 4, 5000 Odense C, Denmark;
| | - Parvin Adimi
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (P.A.); (A.J.); (K.D.S.)
| | - Anders Jakobsen
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (P.A.); (A.J.); (K.D.S.)
- Department of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 5000 Odense C, Denmark
| | - Karina Dahl Steffensen
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; (P.A.); (A.J.); (K.D.S.)
- Department of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 5000 Odense C, Denmark
| |
Collapse
|
9
|
Liu W, Jiang Q, Sun C, Liu S, Zhao Z, Wu D. Developing a 5-gene prognostic signature for cervical cancer by integrating mRNA and copy number variations. BMC Cancer 2022; 22:192. [PMID: 35184747 PMCID: PMC8859909 DOI: 10.1186/s12885-022-09291-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 02/04/2022] [Indexed: 11/10/2022] Open
Abstract
Background Cervical cancer is frequently detected gynecological cancer all over the world. This study was designed to develop a prognostic signature for an effective prediction of cervical cancer prognosis. Methods Differentially expressed genes (DEGs) were identified based on copy number variation (CNV) data and expression profiles from different databases. A prognostic model was constructed and further optimized by stepwise Akaike information criterion (stepAIC). The model was then evaluated in three groups (training group, test group and validation group). Functional analysis and immune analysis were used to assess the difference between high-risk and low-risk groups. Results The study developed a 5-gene prognostic model that could accurately classify cervical cancer samples into high-risk and low-risk groups with distinctly different prognosis. Low-risk group exhibited more favorable prognosis and higher immune infiltration than high-risk group. Both univariate and multivariate Cox regression analysis showed that the risk score was an independent risk factor for cervical cancer. Conclusions The 5-gene prognostic signature could serve as a predictor for identifying high-risk cervical cancer patients, and provided potential direction for studying the mechanism or drug targets of cervical cancer. The integrated analysis of CNV and mRNA expanded a new perspective for exploring prognostic signatures in cervical cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09291-z.
Collapse
|
10
|
Yari H, Shabani S, Nafissi N, Majidzadeh T, Mahjoubi F. Investigation of promoter methylation patterns association with genes expression profile of ISL1, MGMT and DMNT3b in tissue of breast cancer patients. Mol Biol Rep 2022; 49:847-857. [PMID: 34997427 DOI: 10.1007/s11033-021-06546-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVES Cancer initiation and progression could influenced by both genetic and epigenetic events revealing of the overlap between epigenetic and genetic alteration can give important insights into cancer biology. METHODS AND RESULTS In this experiment ISL1, MGMT, DMNT3b genes were candidate to investigate both methylation status and expression profile by using methylation-specific PCR and real time PCR in 40 breast cancer patients, respectively, also we have assessed relation of the promoter methylation status and expression variation of the target genes. The mean level of methylation of ISL1 and MGMT in tumor tissues were significantly greater than normal tissues. In Contrast, DMNT3b gene was showed lower mean level of methylation in tumor tissue compared to normal tissues, however, this was not statistically significant. Relative expression analysis was displayed a significant reduction in expression level of ISL1 and MGMT in tumor tissues. Furthermore, there was a meaningful association between down expression of ISL1 with histological grade, Her2 and ER status. Moreover, MGMT down expression was significantly associated with tumor sizes. Any remarkable relation was not observed between DMNT3b expression level and clinic pathological features. At the end, significant relation between methylation status and expression level has been revealed. CONCLUSIONS In this study all observed results were exactly in line with the results were obtained from articles which were based on the methylation research and illustrate that the real-time PCR and methylation methods are in correlated with each other, furthermore, selected genes are capable to use as a potential biomarkers, however, more research on extended cases are needed.
Collapse
Affiliation(s)
- Hadi Yari
- Human Genetics Dept., Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Pajouhesh Blv, Tehran Karaj High Way, Tehran, Iran
| | - Samira Shabani
- Human Genetics Dept., Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Pajouhesh Blv, Tehran Karaj High Way, Tehran, Iran
| | - Nahid Nafissi
- Surgery Department of General Surgery, Iran University of Medical Science, Tehran, Iran
| | - Tayebeh Majidzadeh
- Human Genetics Dept., Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Pajouhesh Blv, Tehran Karaj High Way, Tehran, Iran
| | - Frouzandeh Mahjoubi
- Human Genetics Dept., Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Pajouhesh Blv, Tehran Karaj High Way, Tehran, Iran.
| |
Collapse
|
11
|
Brown LJ, Achinger-Kawecka J, Portman N, Clark S, Stirzaker C, Lim E. Epigenetic Therapies and Biomarkers in Breast Cancer. Cancers (Basel) 2022; 14:474. [PMID: 35158742 PMCID: PMC8833457 DOI: 10.3390/cancers14030474] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/07/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Epigenetic therapies remain a promising, but still not widely used, approach in the management of patients with cancer. To date, the efficacy and use of epigenetic therapies has been demonstrated primarily in the management of haematological malignancies, with limited supportive data in solid malignancies. The most studied epigenetic therapies in breast cancer are those that target DNA methylation and histone modification; however, none have been approved for routine clinical use. The majority of pre-clinical and clinical studies have focused on triple negative breast cancer (TNBC) and hormone-receptor positive breast cancer. Even though the use of epigenetic therapies alone in the treatment of breast cancer has not shown significant clinical benefit, these therapies show most promise in use in combinations with other treatments. With improving technologies available to study the epigenetic landscape in cancer, novel epigenetic alterations are increasingly being identified as potential biomarkers of response to conventional and epigenetic therapies. In this review, we describe epigenetic targets and potential epigenetic biomarkers in breast cancer, with a focus on clinical trials of epigenetic therapies. We describe alterations to the epigenetic landscape in breast cancer and in treatment resistance, highlighting mechanisms and potential targets for epigenetic therapies. We provide an updated review on epigenetic therapies in the pre-clinical and clinical setting in breast cancer, with a focus on potential real-world applications. Finally, we report on the potential value of epigenetic biomarkers in diagnosis, prognosis and prediction of response to therapy, to guide and inform the clinical management of breast cancer patients.
Collapse
Affiliation(s)
- Lauren Julia Brown
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Joanna Achinger-Kawecka
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Neil Portman
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Susan Clark
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Clare Stirzaker
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Elgene Lim
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| |
Collapse
|
12
|
Tserpeli V, Stergiopoulou D, Londra D, Giannopoulou L, Buderath P, Balgkouranidou I, Xenidis N, Grech C, Obermayr E, Zeillinger R, Pavlakis K, Rampias T, Kakolyris S, Kasimir-Bauer S, Lianidou ES. Prognostic Significance of SLFN11 Methylation in Plasma Cell-Free DNA in Advanced High-Grade Serous Ovarian Cancer. Cancers (Basel) 2021; 14:cancers14010004. [PMID: 35008168 PMCID: PMC8750111 DOI: 10.3390/cancers14010004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Epigenetic alterations in ctDNA are highly promising as a source of novel potential liquid biopsy biomarkers and comprise a very promising liquid biopsy approach in ovarian cancer, for early diagnosis, prognosis and response to treatment. Methods: In the present study, we examined the methylation status of six gene promoters (BRCA1, CST6, MGMT, RASSF10, SLFN11 and USP44) in high-grade serous ovarian cancer (HGSOC). We evaluated the prognostic significance of DNA methylation of these six gene promoters in primary tumors (FFPEs) and plasma cfDNA samples from patients with early, advanced and metastatic HGSOC. Results: We report for the first time that the DNA methylation of SLFN11 in plasma cfDNA was significantly correlated with worse PFS (p = 0.045) in advanced stage HGSOC. Conclusions: Our results strongly indicate that SLFN11 epigenetic inactivation could be a predictor of resistance to platinum drugs in ovarian cancer. Our results should be further validated in studies based on a larger cohort of patients, in order to further explore whether the DNA methylation of SLFN11 promoter could serve as a potential prognostic DNA methylation biomarker and a predictor of resistance to platinum-based chemotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Victoria Tserpeli
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (V.T.); (D.S.); (D.L.); (L.G.)
| | - Dimitra Stergiopoulou
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (V.T.); (D.S.); (D.L.); (L.G.)
| | - Dora Londra
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (V.T.); (D.S.); (D.L.); (L.G.)
| | - Lydia Giannopoulou
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (V.T.); (D.S.); (D.L.); (L.G.)
| | - Paul Buderath
- Department of Gynecology and Obstetrics, University Hospital of Essen, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany; (P.B.); (S.K.-B.)
| | - Ioanna Balgkouranidou
- Department of Oncology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.B.); (N.X.); (S.K.)
| | - Nikolaos Xenidis
- Department of Oncology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.B.); (N.X.); (S.K.)
| | - Christina Grech
- Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (E.O.); (R.Z.)
| | - Eva Obermayr
- Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (E.O.); (R.Z.)
| | - Robert Zeillinger
- Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (E.O.); (R.Z.)
| | - Kitty Pavlakis
- Pathology Department, IASO Women’s Hospital, 15123 Athens, Greece;
| | - Theodoros Rampias
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Stylianos Kakolyris
- Department of Oncology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.B.); (N.X.); (S.K.)
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital of Essen, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany; (P.B.); (S.K.-B.)
| | - Evi S. Lianidou
- Analysis of Circulating Tumor Cells, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (V.T.); (D.S.); (D.L.); (L.G.)
- Correspondence: ; Tel.: +30-210-7274311
| |
Collapse
|
13
|
Wang J, Li J, Chen R, Yue H, Li W, Wu B, Bai Y, Zhu G, Lu X. DNA methylation-based profiling reveals distinct clusters with survival heterogeneity in high-grade serous ovarian cancer. Clin Epigenetics 2021; 13:190. [PMID: 34645493 PMCID: PMC8515755 DOI: 10.1186/s13148-021-01178-3] [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: 02/21/2021] [Accepted: 09/29/2021] [Indexed: 12/27/2022] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is the most common type of epigenetically heterogeneous ovarian cancer. Methylation typing has previously been used in many tumour types but not in HGSOC. Methylation typing in HGSOC may promote the development of personalized care. The present study used DNA methylation data from The Cancer Genome Atlas database and identified four unique methylation subtypes of HGSOC. With the poorest prognosis and high frequency of residual tumours, cluster 4 featured hypermethylation of a panel of genes, which indicates that demethylation agents may be tested in this group and that neoadjuvant chemotherapy may be used to reduce the possibility of residual lesions. Cluster 1 and cluster 2 were significantly associated with metastasis genes and metabolic disorders, respectively. Two feature CpG sites, cg24673765 and cg25574024, were obtained through Cox proportional hazards model analysis of the CpG sites. Based on the methylation level of the two CpG sites, the samples were classified into high- and low-risk groups to identify the prognostic information. Similar results were obtained in the validation set. Taken together, these results explain the epigenetic heterogeneity of HGSOC and provide guidance to clinicians for the prognosis of HGSOC based on DNA methylation sites.
Collapse
Affiliation(s)
- Jieyu Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Jun Li
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China
| | - Ruifang Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China
| | - Huiran Yue
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China
| | - Wenzhi Li
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Beibei Wu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China
| | - Yang Bai
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China
| | - Guohua Zhu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China
| | - Xin Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, No. 128, Shenyang Road, Yangpu District, Shanghai, 200090, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Faaborg L, Jakobsen A, Waldstrøm M, Petersen CB, Andersen RF, Steffensen KD. HOXA9-methylated DNA as a diagnostic biomarker of ovarian malignancy. Biomark Med 2021; 15:1309-1317. [PMID: 34514844 DOI: 10.2217/bmm-2021-0144] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aim: In ovarian cancer, methylated HOXA9 (meth-HOXA9) has been proposed as a relevant biomarker, however, its role in the carcinogenic development remains unknown. This study aimed at evaluating meth-HOXA9 as a diagnostic biomarker in ovarian cancer. Materials & methods: The meth-HOXA9 status was examined in 138 tissue specimens encompassing normal ovaries, benign- and borderline tumors, and ovarian cancer using droplet digital PCR. Results: Meth-HOXA9 was detected in 93% (82/88) and 88% (14/16) of ovarian cancer and borderline tumors, respectively. In patients with benign ovarian tumors meth-HOXA9 was detected in 17% (3/18). Using receiver operating characteristic (ROC) analysis meth-HOXA9 had a diagnostic accuracy of 98%. Conclusion: Meth-HOXA9 is highly cancer specific and could serve as a general diagnostic marker of ovarian malignancy.
Collapse
Affiliation(s)
- Louise Faaborg
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, 7100, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, 5000, Denmark
| | - Anders Jakobsen
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, 7100, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, 5000, Denmark
| | - Marianne Waldstrøm
- Institute of Regional Health Research, University of Southern Denmark, Odense, 5000, Denmark.,Department of Pathology, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, 7100, Denmark
| | - Christina B Petersen
- Department of Pathology, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, 7100, Denmark
| | - Rikke F Andersen
- Department of Clinical Biochemistry, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, 7100, Denmark
| | - Karina D Steffensen
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, 7100, Denmark.,Institute of Regional Health Research, University of Southern Denmark, Odense, 5000, Denmark.,Center for Shared Decision Making, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, 7100, Denmark
| |
Collapse
|
15
|
Werner B, Yuwono N, Duggan J, Liu D, David C, Srirangan S, Provan P, DeFazio A, Arora V, Farrell R, Lee YC, Warton K, Ford C. Cell-free DNA is abundant in ascites and represents a liquid biopsy of ovarian cancer. Gynecol Oncol 2021; 162:720-727. [PMID: 34454680 DOI: 10.1016/j.ygyno.2021.06.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/15/2021] [Accepted: 06/27/2021] [Indexed: 02/09/2023]
Abstract
OBJECTIVE Malignant ascites is a common clinical feature of ovarian cancer and represents a readily accessible sample of tumour cells and tumour DNA. This study aimed to characterise the cell-free DNA (cfDNA) in ascites in terms of its size profile, stability and cell-free tumour DNA (cftDNA) content. METHODS Cell spheroids, loose cells and cell-free fluid was collected from ascites from 18 patients with ovarian cancer. cfDNA was isolated and assessed for size by electrophoresis, concentration by fluorometry,cftDNA content by methylation specific qPCR of HOXA9 and IFFO1 promoter regions and by targeted sequencing. Stability was assessed after ascites fluid was stored at 4 °C for 24 and 72 h before fractionating. RESULTS The concentration of cfDNA in ascites ranged from 6.6 to 300 ng/mL. cfDNA size distribution resembled blood plasma-derived cfDNA, with major peaks corresponding to mono- and di-nucleosome DNA fragments. High molecular weight cfDNA was observed in 7 of 18 patients and appeared to be associated with extracellular vesicles. IFFO1 and HOXA9 methylation was proportionately higher in cfDNA than spheroid- and loose-cell fractions and was not observed in healthy primary cells. Variant allele frequency was highest in cfDNA compared to single cells and spheroids from ascites. Though cancer cell numbers in ascites declined to near zero in recurrent ascites from one patient undertaking chemotherapy, cftDNA could still be sampled. cfDNA size, concentration and tumour content was stable over 72 h. CONCLUSION cfDNA in ovarian cancer ascites demonstrates inter-patient variability, yet is consistently a rich source of cftDNA, which is a stable substrate. This supports the wider clinical use of ascites in the molecular analysis of ovarian cancer.
Collapse
Affiliation(s)
- Bonnita Werner
- Gynaecological Cancer Research Group, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Australia
| | - Nicole Yuwono
- Gynaecological Cancer Research Group, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Australia
| | - Jennifer Duggan
- Gynaecological Oncology Department, Royal Hospital for Women, Sydney, Australia
| | - Dongli Liu
- Gynaecological Cancer Research Group, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Australia
| | - Catherine David
- Gynaecological Oncology Department, Royal Hospital for Women, Sydney, Australia
| | - Sivatharsny Srirangan
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, Australia
| | - Pamela Provan
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | | | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Gynaecological Oncology, Westmead Hospital, Sydney, Australia
| | - Vivek Arora
- Gynaecological Oncology Department, Royal Hospital for Women, Sydney, Australia; Prince of Wales Private Hospital, Sydney, Australia; School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Australia
| | | | - Yeh Chen Lee
- Gynaecological Oncology Department, Royal Hospital for Women, Sydney, Australia; Faculty of Medicine and Health, University of New South Wales, Australia
| | - Kristina Warton
- Gynaecological Cancer Research Group, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Australia
| | - Caroline Ford
- Gynaecological Cancer Research Group, School of Women's and Children's Health, Faculty of Medicine and Health, University of New South Wales, Australia.
| |
Collapse
|
16
|
Faaborg L, Fredslund Andersen R, Waldstrøm M, Høgdall E, Høgdall C, Adimi P, Jakobsen A, Dahl Steffensen K. Analysis of HOXA9 methylated ctDNA in ovarian cancer using sense-antisense measurement. Clin Chim Acta 2021; 522:152-157. [PMID: 34419462 DOI: 10.1016/j.cca.2021.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022]
Abstract
DNA promoter methylation is an early event in tumorigenesis and holds promise as a valuable marker in ovarian cancer (OC). It can be measured using circulating tumor specific DNA (ctDNA) isolated from the bloodstream. Sensitivity, however, is a limiting factor of its diagnostic feasibility in OC. DNA methylation analyses are based on bisulfite conversion, resulting in two DNA strands that are no longer complementary. The current standard strategy would then target only one of the double stranded DNA strands, but the potential to increase the sensitivity by targeting both DNA strands is available. In this study, we aimed at evaluating the diagnostic potential of methylated HOXA9 ctDNA in OC by targeting both the DNA sense and antisense strand. Methylated HOXA9 was detected in the plasma of 47/79 (59.5%) patients with newly diagnosed OC using sense-antisense droplet digital PCR. Simultaneous sense-antisense measurement increased the sensitivity by 14.6% (51.9% to 59.5%) as compared to antisense only. In patients with FIGO stage I-II disease the sensitivity was increased by 25%. In conclusion, simultaneous measurement targeting both DNA strands can increase the sensitivity and the analytical approach appears valuable in the diagnostic setting of OC.
Collapse
Affiliation(s)
- Louise Faaborg
- Department of Oncology, Vejle Hospital, Lillebaelt Hospital - University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 5000 Odense C, Denmark.
| | - Rikke Fredslund Andersen
- Department of Clinical Biochemistry, Vejle Hospital, Lillebaelt Hospital - University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark
| | - Marianne Waldstrøm
- Department of Oncology, Vejle Hospital, Lillebaelt Hospital - University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; Department of Pathology, Vejle Hospital, Lillebaelt Hospital - University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark
| | - Estrid Høgdall
- Department of Pathology, Herlev Hospital, University of Copenhagen, Borgmester Ib Juuls Vej 1, 2730 Herlev, Denmark
| | - Claus Høgdall
- Department of Gynecology, The Juliane Marie Centre, Righospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Parvin Adimi
- Department of Oncology, Vejle Hospital, Lillebaelt Hospital - University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark
| | - Anders Jakobsen
- Department of Oncology, Vejle Hospital, Lillebaelt Hospital - University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 5000 Odense C, Denmark
| | - Karina Dahl Steffensen
- Department of Oncology, Vejle Hospital, Lillebaelt Hospital - University Hospital of Southern Denmark, Beriderbakken 4, 7100 Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, J.B. Winsløws Vej 19, 5000 Odense C, Denmark
| |
Collapse
|
17
|
Baranova I, Kovarikova H, Laco J, Sedlakova I, Vrbacky F, Kovarik D, Hejna P, Palicka V, Chmelarova M. Identification of a four-gene methylation biomarker panel in high-grade serous ovarian carcinoma. Clin Chem Lab Med 2021; 58:1332-1340. [PMID: 32145055 DOI: 10.1515/cclm-2019-1319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/04/2020] [Indexed: 12/31/2022]
Abstract
Background The lack of effective biomarkers for the screening and early detection of ovarian cancer (OC) is one of the most pressing problems in oncogynecology. Because epigenetic alterations occur early in the cancer development, they provide great potential to serve as such biomarkers. In our study, we investigated a potential of a four-gene methylation panel (including CDH13, HNF1B, PCDH17 and GATA4 genes) for the early detection of high-grade serous ovarian carcinoma (HGSOC). Methods For methylation detection we used methylation sensitive high-resolution melting analysis and real-time methylation specific analysis. We also investigated the relation between gene hypermethylation and gene relative expression using the 2-ΔΔCt method. Results The sensitivity of the examined panel reached 88.5%. We were able to detect methylation in 85.7% (12/14) of early stage tumors and in 89.4% (42/47) of late stage tumors. The total efficiency of the panel was 94.4% and negative predictive value reached 90.0%. The specificity and positive predictive value achieved 100% rates. Our results showed lower gene expression in the tumor samples in comparison to control samples. The more pronounced downregulation was measured in the group of samples with detected methylation. Conclusions In our study we designed the four-gene panel for HGSOC detection in ovarian tissue with 100% specificity and sensitivity of 88.5%. The next challenge is translation of the findings to the less invasive source for biomarker examination, such as plasma. Our results indicate that combination of examined genes deserve consideration for further testing in clinical molecular diagnosis of HGSOC.
Collapse
Affiliation(s)
- Ivana Baranova
- Institute of Clinical Biochemistry and Diagnostics, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Helena Kovarikova
- Institute of Clinical Biochemistry and Diagnostics, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Laco
- The Fingerland Department of Pathology, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Iva Sedlakova
- Department of Obstetrics and Gynecology, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Filip Vrbacky
- The 4th Department of Internal Medicine - Hematology, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Dalibor Kovarik
- Department of Forensic Medicine, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Petr Hejna
- Department of Forensic Medicine, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Vladimir Palicka
- Institute of Clinical Biochemistry and Diagnostics, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marcela Chmelarova
- Institute of Clinical Biochemistry and Diagnostics, Charles University Faculty of Medicine and University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
18
|
Kalachand RD, Stordal B, Madden S, Chandler B, Cunningham J, Goode EL, Ruscito I, Braicu EI, Sehouli J, Ignatov A, Yu H, Katsaros D, Mills GB, Lu KH, Carey MS, Timms KM, Kupryjanczyk J, Rzepecka IK, Podgorska A, McAlpine JN, Swisher EM, Bernards SS, O'Riain C, O'Toole S, O'Leary JJ, Bowtell DD, Thomas DM, Prieske K, Joosse SA, Woelber L, Chaudhry P, Häfner N, Runnebaum IB, Hennessy BT. BRCA1 Promoter Methylation and Clinical Outcomes in Ovarian Cancer: An Individual Patient Data Meta-Analysis. J Natl Cancer Inst 2021; 112:1190-1203. [PMID: 32413141 DOI: 10.1093/jnci/djaa070] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/23/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND BRCA1 methylation has been associated with homologous recombination deficiency, a biomarker of platinum sensitivity. Studies evaluating BRCA1-methylated tubal and ovarian cancer (OC) do not consistently support improved survival following platinum chemotherapy. We examine the characteristics of BRCA1-methylated OC in a meta-analysis of individual participant data. METHODS Data of 2636 participants across 15 studies were analyzed. BRCA1-methylated tumors were defined according to their original study. Associations between BRCA1 methylation and clinicopathological characteristics were evaluated. The effects of methylation on overall survival (OS) and progression-free survival (PFS) were examined using mixed-effects models. All statistical tests were 2-sided. RESULTS 430 (16.3%) tumors were BRCA1-methylated. BRCA1 methylation was associated with younger age and advanced-stage, high-grade serous OC. There were no survival differences between BRCA1-methylated and non-BRCA1-methylated OC (median PFS = 20.0 vs 18.5 months, hazard ratio [HR] = 1.01, 95% CI = 0.87 to 1.16; P = .98; median OS = 46.6 vs 48.0 months, HR = 1.02, 95% CI = 0.87 to 1.18; P = .96). Where BRCA1/2 mutations were evaluated (n = 1248), BRCA1 methylation displayed no survival advantage over BRCA1/2-intact (BRCA1/2 wild-type non-BRCA1-methylated) OC. Studies used different methods to define BRCA1 methylation. Where BRCA1 methylation was determined using methylation-specific polymerase chain reaction and gel electrophoresis (n = 834), it was associated with improved survival (PFS: HR = 0.80, 95% CI = 0.66 to 0.97; P = .02; OS: HR = 0.80, 95% CI = 0.63 to 1.00; P = .05) on mixed-effects modeling. CONCLUSION BRCA1-methylated OC displays similar clinicopathological features to BRCA1-mutated OC but is not associated with survival. Heterogeneity within BRCA1 methylation assays influences associations. Refining these assays may better identify cases with silenced BRCA1 function and improved patient outcomes.
Collapse
Affiliation(s)
- Roshni D Kalachand
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Britta Stordal
- Department of Natural Sciences, Middlesex University, Hendon, London NW4 4BT, UK
| | - Stephen Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Beaux Lane House, Dublin, Ireland
| | - Benjamin Chandler
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Julie Cunningham
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ellen L Goode
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Ilary Ruscito
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.,Cell Therapy Unit and Laboratory of Tumor Immunology, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Elena I Braicu
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, European Competence Center for Ovarian Cancer, Campus Virchow Klinikum, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Atanas Ignatov
- Department of Gynecology and Obstetrics, University Medical Center, Regensburg, Germany
| | - Herbert Yu
- University of Hawaii Cancer Centre, Honolulu, HI, USA
| | - Dionyssios Katsaros
- AOU Citta della Salute and Department of Surgical Sciences, Gynecologic Oncology, University of Torino, Italy
| | - Gordon B Mills
- Department of Cell, Development and Cancer Biology Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Karen H Lu
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark S Carey
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Jolanta Kupryjanczyk
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Iwona K Rzepecka
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Agnieszka Podgorska
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Jessica N McAlpine
- Division of Gynecologic Oncology, Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - Ciaran O'Riain
- Department of Histopathology, Trinity College Dublin, Central Pathology Laboratory, St. James's Hospital, Dublin, Ireland
| | - Sharon O'Toole
- Department of Obstetrics and Gynaecology/Histopathology, Trinity College Dublin, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland.,Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Central Pathology Laboratory, St. James's Hospital, Dublin, Ireland.,Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin, Ireland
| | | | - David M Thomas
- Genomic Cancer Medicine, Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, Australia
| | - Katharina Prieske
- Department of Gynecology and Gynecologic Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Joosse
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linn Woelber
- Department of Gynecology and Gynecologic Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Parvesh Chaudhry
- Department of Radiotherapy, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Norman Häfner
- Department for Gynaecology and Reproductive Medicine, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Ingo B Runnebaum
- Department for Gynaecology and Reproductive Medicine, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Bryan T Hennessy
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.,Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland.,Our Lady of Lourdes Hospital, Drogheda, Ireland
| |
Collapse
|
19
|
Sharma A, Liu H, Herwig-Carl MC, Chand Dakal T, Schmidt-Wolf IGH. Epigenetic Regulatory Enzymes: mutation Prevalence and Coexistence in Cancers. Cancer Invest 2021; 39:257-273. [PMID: 33411587 DOI: 10.1080/07357907.2021.1872593] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic regulation is an important layer of transcriptional control with the particularity to affect the broad spectrum of genome. Over the years, largely due to the substantial number of recurrent mutations, there have been hundreds of novel driver genes characterized in various cancers. Additionally, the relative contribution of two dysregulated epigenomic entities (DNA methylation and histone modifications) that gradually drive the cancer phenotype remains in the research focus. However, a complex scenario arises when the disease phenotype does not harbor any relevant mutation or an abnormal transcription level. Although the cancer landscape involves the contribution of multiple genetic and non-genetic factors, herein, we discuss specifically the mutation spectrum of epigenetically-related enzymes in cancer. In addition, we address the coexistence of these two epigenetic entities in malignant human diseases, especially cancer. We suggest that the study of epigenetically-related somatic mutations in the early cellular differentiation stage of embryonic development might help to understand their later-staged footprints in the cancer genome. Furthermore, understanding the co-occurrence and/or inverse association of different disease types and redefining the general definition of "healthy" controls could provide insights into the genome reorganization.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Integrated Oncology, CIO Bonn, University Hospital Bonn, Bonn, Germany.,Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Hongde Liu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | | | - Tikam Chand Dakal
- Department of Biotechnology, Mohanlal Sukhadia University, Rajasthan, India
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, CIO Bonn, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
20
|
Gİrgİn B, KaradaĞ-Alpaslan M, KocabaŞ F. Oncogenic and tumor suppressor function of MEIS and associated factors. ACTA ACUST UNITED AC 2021; 44:328-355. [PMID: 33402862 PMCID: PMC7759197 DOI: 10.3906/biy-2006-25] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022]
Abstract
MEIS proteins are historically associated with tumorigenesis, metastasis, and invasion in cancer. MEIS and associated PBX-HOX proteins may act as tumor suppressors or oncogenes in different cellular settings. Their expressions tend to be misregulated in various cancers. Bioinformatic analyses have suggested their upregulation in leukemia/lymphoma, thymoma, pancreas, glioma, and glioblastoma, and downregulation in cervical, uterine, rectum, and colon cancers. However, every cancer type includes, at least, a subtype with high MEIS expression. In addition, studies have highlighted that MEIS proteins and associated factors may function as diagnostic or therapeutic biomarkers for various diseases. Herein, MEIS proteins and associated factors in tumorigenesis are discussed with recent discoveries in addition to how they could be modulated by noncoding RNAs or newly developed small-molecule MEIS inhibitors.
Collapse
Affiliation(s)
- Birkan Gİrgİn
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul Turkey.,Graduate School of Natural and Applied Sciences, Yeditepe University, İstanbul Turkey.,Meinox Pharma Technologies, İstanbul Turkey
| | - Medine KaradaĞ-Alpaslan
- Department of Medical Genetics, Faculty of Medicine, Ondokuz Mayıs University, Samsun Turkey
| | - Fatih KocabaŞ
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, İstanbul Turkey.,Graduate School of Natural and Applied Sciences, Yeditepe University, İstanbul Turkey.,Meinox Pharma Technologies, İstanbul Turkey
| |
Collapse
|
21
|
Jaworski JJ, Morgan RD, Sivakumar S. Circulating Cell-Free Tumour DNA for Early Detection of Pancreatic Cancer. Cancers (Basel) 2020; 12:E3704. [PMID: 33317202 PMCID: PMC7763954 DOI: 10.3390/cancers12123704] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/04/2020] [Indexed: 01/11/2023] Open
Abstract
Pancreatic cancer is a lethal disease, with mortality rates negatively associated with the stage at which the disease is detected. Early detection is therefore critical to improving survival outcomes. A recent focus of research for early detection is the use of circulating cell-free tumour DNA (ctDNA). The detection of ctDNA offers potential as a relatively non-invasive method of diagnosing pancreatic cancer by using genetic sequencing technology to detect tumour-specific mutational signatures in blood samples before symptoms manifest. These technologies are limited by a number of factors that lower sensitivity and specificity, including low levels of detectable ctDNA in early stage disease and contamination with non-cancer circulating cell-free DNA. However, genetic and epigenetic analysis of ctDNA in combination with other standard diagnostic tests may improve early detection rates. In this review, we evaluate the genetic and epigenetic methods under investigation in diagnosing pancreatic cancer and provide a perspective for future developments.
Collapse
Affiliation(s)
- Jedrzej J. Jaworski
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK;
| | - Robert D. Morgan
- Department of Medical Oncology, Christie NHS Foundation Trust, Manchester M20 4BX, UK;
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Shivan Sivakumar
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
- Department of Medical Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LE, UK
| |
Collapse
|
22
|
Chou YH, Tantoh DM, Wu MC, Tyan YS, Chen PH, Nfor ON, Hsu SY, Shen CY, Huang CN, Liaw YP. PM 2.5 exposure and DLEC1 promoter methylation in Taiwan Biobank participants. Environ Health Prev Med 2020; 25:68. [PMID: 33153431 PMCID: PMC7646067 DOI: 10.1186/s12199-020-00909-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Particulate matter (PM) < 2.5 μm (PM2.5) or fine PM is a serious public health concern. It affects DNA methylation and heightens carcinogenesis. Deleted in lung and esophageal cancer 1 (DLEC1) is a tumor suppressor gene. However, aberrant methylation of the gene is associated with several cancers. We evaluated the association between PM2.5 and DLEC1 promoter methylation in Taiwanese adults based on regular outdoor exercise. METHODS We obtained DNA methylation and exercise data of 496 participants (aged between 30 and 70 years) from the Taiwan Biobank (TWB) database. We also extracted PM2.5 data from the Air Quality Monitoring Database (AQMD) and estimated participants' exposure using residential addresses. RESULTS DLEC1 methylation and PM2.5 were positively associated: beta coefficient (β) = 0.114 × 10-3; p value = 0.046. The test for interaction between exercise and PM2.5 on DLEC1 methylation was significant (p value = 0.036). After stratification by exercise habits, PM2.5 and DLEC1 methylation remained significantly associated only among those who exercised regularly (β = 0.237 × 10-3; p value = 0.007). PM2.5 quartile-stratified analyses revealed an inverse association between regular exercise and DLEC1 methylation at PM2.5 < 27.37 μg/m3 (β = - 5.280 × 10-3; p value = 0.009). After combining exercise habits and PM2.5 quartiles, one stratum (i.e., regular exercise and PM2.5 < 27.37 μg/m3) was inversely associated with DLEC1 methylation (β = -5.160 × 10-3, p value = 0.007). CONCLUSIONS We found significant positive associations between PM2.5 and DLEC1 promoter methylation. Regular exercise at PM2.5 < 27.37 μg/m3 seemingly regulated DLEC1 promoter methylation.
Collapse
Affiliation(s)
- Ying-Hsiang Chou
- Institute of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
- School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan
| | - Disline Manli Tantoh
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Ming-Chi Wu
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
- School of Medical Informatics, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Yeu-Sheng Tyan
- School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
| | - Pei-Hsin Chen
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Oswald Ndi Nfor
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Shu-Yi Hsu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan
| | - Chao-Yu Shen
- School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung City, 40201, Taiwan.
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan.
| | - Chien-Ning Huang
- Institute of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan.
- Department of Internal Medicine, Chung-Shan Medical University Hospital, Taichung City, 40201, Taiwan.
| | - Yung-Po Liaw
- Department of Medical Imaging, Chung Shan Medical University Hospital, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan.
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, No. 110, Sec. 1 Jianguo N. Rd, Taichung City, 40201, Taiwan.
- Medical Imaging and Big Data Center, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan.
| |
Collapse
|
23
|
Lam D, Clark S, Stirzaker C, Pidsley R. Advances in Prognostic Methylation Biomarkers for Prostate Cancer. Cancers (Basel) 2020; 12:E2993. [PMID: 33076494 PMCID: PMC7602626 DOI: 10.3390/cancers12102993] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/24/2022] Open
Abstract
There is a major clinical need for accurate biomarkers for prostate cancer prognosis, to better inform treatment strategies and disease monitoring. Current clinically recognised prognostic factors, including prostate-specific antigen (PSA) levels, lack sensitivity and specificity in distinguishing aggressive from indolent disease, particularly in patients with localised intermediate grade prostate cancer. There has therefore been a major focus on identifying molecular biomarkers that can add prognostic value to existing markers, including investigation of DNA methylation, which has a known role in tumorigenesis. In this review, we will provide a comprehensive overview of the current state of DNA methylation biomarker studies in prostate cancer prognosis, and highlight the advances that have been made in this field. We cover the numerous studies into well-established candidate genes, and explore the technological transition that has enabled hypothesis-free genome-wide studies and the subsequent discovery of novel prognostic genes.
Collapse
Affiliation(s)
- Dilys Lam
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
| | - Susan Clark
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Clare Stirzaker
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| | - Ruth Pidsley
- Epigenetics Research Laboratory, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia; (D.L.); (S.C.); (C.S.)
- St. Vincent’s Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
24
|
Rose M, Burgess JT, O’Byrne K, Richard DJ, Bolderson E. PARP Inhibitors: Clinical Relevance, Mechanisms of Action and Tumor Resistance. Front Cell Dev Biol 2020; 8:564601. [PMID: 33015058 PMCID: PMC7509090 DOI: 10.3389/fcell.2020.564601] [Citation(s) in RCA: 412] [Impact Index Per Article: 82.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022] Open
Abstract
The Poly (ADP-ribose) polymerase (PARP) family has many essential functions in cellular processes, including the regulation of transcription, apoptosis and the DNA damage response. PARP1 possesses Poly (ADP-ribose) activity and when activated by DNA damage, adds branched PAR chains to facilitate the recruitment of other repair proteins to promote the repair of DNA single-strand breaks. PARP inhibitors (PARPi) were the first approved cancer drugs that specifically targeted the DNA damage response in BRCA1/2 mutated breast and ovarian cancers. Since then, there has been significant advances in our understanding of the mechanisms behind sensitization of tumors to PARP inhibitors and expansion of the use of PARPi to treat several other cancer types. Here, we review the recent advances in the proposed mechanisms of action of PARPi, biomarkers of the tumor response to PARPi, clinical advances in PARPi therapy, including the potential of combination therapies and mechanisms of tumor resistance.
Collapse
Affiliation(s)
- Maddison Rose
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Joshua T. Burgess
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kenneth O’Byrne
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
- Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Derek J. Richard
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Emma Bolderson
- Cancer & Ageing Research Program, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
25
|
Xin J, Wu Y, Wang X, Li S, Chu H, Wang M, Du M, Zhang Z. A transcriptomic study for identifying cardia- and non-cardia-specific gastric cancer prognostic factors using genetic algorithm-based methods. J Cell Mol Med 2020; 24:9457-9465. [PMID: 32649057 PMCID: PMC7417703 DOI: 10.1111/jcmm.15618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/31/2022] Open
Abstract
Gastric cancer (GC) is a heterogeneous tumour with numerous differences of epidemiologic and clinicopathologic features between cardia cancer and non-cardia cancer. However, few studies were performed to construct site-specific GC prognostic models. In this study, we identified site-specific GC transcriptomic prognostic biomarkers using genetic algorithm (GA)-based support vector machine (GA-SVM) and GA-based Cox regression method (GA-Cox) in the Cancer Genome Atlas (TCGA) database. The area under time-dependent receive operating characteristic (ROC) curve (AUC) regarding 5-year survival and concordance index (C-index) was used to evaluate the predictive ability of Cox regression models. Finally, we identified 10 and 13 prognostic biomarkers for cardia cancer and non-cardia cancer, respectively. Compared to traditional models, the addition of these site-specific biomarkers could notably improve the model preference (cardia: AUCtraditional vs AUCcombined = 0.720 vs 0.899, P = 8.75E-08; non-cardia: AUCtraditional vs AUCcombined = 0.798 vs 0.994, P = 7.11E-16). The combined nomograms exhibited superior performance in cardia and non-cardia GC survival prediction (C-indexcardia = 0.816; C-indexnoncardia = 0.812). We also constructed a user-friendly GC site-specific molecular system (GC-SMS, https://njmu-zhanglab.shinyapps.io/gc_sms/), which is freely available for users. In conclusion, we developed site-specific GC prognostic models for predicting cardia cancer and non-cardia cancer survival, providing more support for the individualized therapy of GC patients.
Collapse
Affiliation(s)
- Junyi Xin
- Department of Environmental GenomicsJiangsu Key Laboratory of Cancer Biomarkers, Prevention and TreatmentCollaborative Innovation Center for Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
- Department of Genetic ToxicologyThe Key Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Yanling Wu
- Department of Environmental GenomicsJiangsu Key Laboratory of Cancer Biomarkers, Prevention and TreatmentCollaborative Innovation Center for Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
- Department of Genetic ToxicologyThe Key Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Xiaowei Wang
- Department of Environmental GenomicsJiangsu Key Laboratory of Cancer Biomarkers, Prevention and TreatmentCollaborative Innovation Center for Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
- Department of Genetic ToxicologyThe Key Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Shuwei Li
- Department of Environmental GenomicsJiangsu Key Laboratory of Cancer Biomarkers, Prevention and TreatmentCollaborative Innovation Center for Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
- Department of Genetic ToxicologyThe Key Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Haiyan Chu
- Department of Environmental GenomicsJiangsu Key Laboratory of Cancer Biomarkers, Prevention and TreatmentCollaborative Innovation Center for Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
- Department of Genetic ToxicologyThe Key Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Meilin Wang
- Department of Environmental GenomicsJiangsu Key Laboratory of Cancer Biomarkers, Prevention and TreatmentCollaborative Innovation Center for Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
- Department of Genetic ToxicologyThe Key Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Mulong Du
- Department of BiostatisticsCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Zhengdong Zhang
- Department of Environmental GenomicsJiangsu Key Laboratory of Cancer Biomarkers, Prevention and TreatmentCollaborative Innovation Center for Cancer Personalized MedicineSchool of Public HealthNanjing Medical UniversityNanjingChina
- Department of Genetic ToxicologyThe Key Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| |
Collapse
|
26
|
ARMCX Family Gene Expression Analysis and Potential Prognostic Biomarkers for Prediction of Clinical Outcome in Patients with Gastric Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3575038. [PMID: 32685472 PMCID: PMC7345962 DOI: 10.1155/2020/3575038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/20/2020] [Indexed: 01/04/2023]
Abstract
Armadillo gene subfamily members (ARMCX1-6) are well-known to regulate protein-protein interaction involved in nuclear transport, cellular connection, and transcription activation. Moreover, ARMCX signals on cell pathways also implicated in carcinogenesis and tumor progression. However, little is known about the associations of the ARMCX subfamily members with gastric carcinoma. This study investigated the prognostic value of ARMCX subfamily mRNA expression levels with the prognosis of gastric carcinoma (GC). We retrieved the data of a total of 351 GC patients from TCGA database. Survival and gene set enrichment analyses were employed to explore the predictive value and underlying mechanism of ARMCX genes in GC. The multivariate survival analysis revealed that individually low expressions of ARMCX1 (adjusted P = 0.006, HR = 0.620, CI = 0.440 - 0.874) and ARMCX2 (adjusted P = 0.005, HR = 0.610, 95%CI = 0.432-0.861) were related to preferable overall survival (OS). The joint-effects analysis shown that combinations of low level expression of ARMCX1 and ARMCX2 were correlated with favorable OS (adjusted P = 0.003, HR = 0.563, 95%CI = 0.384-0.825). ARMCX1 and ARMCX2 were implicated in WNT and NF-kappaB pathways, and biological processes including cell cycle, apoptosis, RNA modification, DNA replication, and damage response. Our results suggest that mRNA expression levels of ARMCX subfamily are potential prognostic markers of GC.
Collapse
|
27
|
Baker LA, Holliday H, Roden D, Krisp C, Wu SZ, Junankar S, Serandour AA, Mohammed H, Nair R, Sankaranarayanan G, Law AMK, McFarland A, Simpson PT, Lakhani S, Dodson E, Selinger C, Anderson L, Samimi G, Hacker NF, Lim E, Ormandy CJ, Naylor MJ, Simpson K, Nikolic I, O'Toole S, Kaplan W, Cowley MJ, Carroll JS, Molloy M, Swarbrick A. Proteogenomic analysis of Inhibitor of Differentiation 4 (ID4) in basal-like breast cancer. Breast Cancer Res 2020; 22:63. [PMID: 32527287 PMCID: PMC7291584 DOI: 10.1186/s13058-020-01306-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Basal-like breast cancer (BLBC) is a poorly characterised, heterogeneous disease. Patients are diagnosed with aggressive, high-grade tumours and often relapse with chemotherapy resistance. Detailed understanding of the molecular underpinnings of this disease is essential to the development of personalised therapeutic strategies. Inhibitor of differentiation 4 (ID4) is a helix-loop-helix transcriptional regulator required for mammary gland development. ID4 is overexpressed in a subset of BLBC patients, associating with a stem-like poor prognosis phenotype, and is necessary for the growth of cell line models of BLBC through unknown mechanisms. METHODS Here, we have defined unique molecular insights into the function of ID4 in BLBC and the related disease high-grade serous ovarian cancer (HGSOC), by combining RIME proteomic analysis, ChIP-seq mapping of genomic binding sites and RNA-seq. RESULTS These studies reveal novel interactions with DNA damage response proteins, in particular, mediator of DNA damage checkpoint protein 1 (MDC1). Through MDC1, ID4 interacts with other DNA repair proteins (γH2AX and BRCA1) at fragile chromatin sites. ID4 does not affect transcription at these sites, instead binding to chromatin following DNA damage. Analysis of clinical samples demonstrates that ID4 is amplified and overexpressed at a higher frequency in BRCA1-mutant BLBC compared with sporadic BLBC, providing genetic evidence for an interaction between ID4 and DNA damage repair deficiency. CONCLUSIONS These data link the interactions of ID4 with MDC1 to DNA damage repair in the aetiology of BLBC and HGSOC.
Collapse
Affiliation(s)
- Laura A Baker
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Holly Holliday
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Daniel Roden
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Christoph Krisp
- Australian Proteome Analysis Facility (APAF), Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, Australia
- Mass Spectrometric Proteome Analysis, Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Sunny Z Wu
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Simon Junankar
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Aurelien A Serandour
- Cancer Research UK, The University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Hisham Mohammed
- Cancer Research UK, The University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Radhika Nair
- Rajiv Gandhi Centre for Biotechnology, Thycaud Post, Poojappura, Thiruvananthapuram, Kerala, 695014, India
| | - Geetha Sankaranarayanan
- Cancer Research UK, The University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Andrew M K Law
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Andrea McFarland
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - Peter T Simpson
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Sunil Lakhani
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Pathology Queensland, The Royal Brisbane and Women's Hospital, Herston, , Brisbane, QLD, Australia
| | - Eoin Dodson
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Christina Selinger
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Lyndal Anderson
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Goli Samimi
- National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Neville F Hacker
- School of Women's and Children's Health, University of New South Wales, and Gynaecological Cancer Centre, Royal Hospital for Women, Sydney, NSW, Australia
| | - Elgene Lim
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Christopher J Ormandy
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Matthew J Naylor
- School of Medical Sciences and Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kaylene Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Iva Nikolic
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Sandra O'Toole
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Warren Kaplan
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - Mark J Cowley
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Jason S Carroll
- Cancer Research UK, The University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Mark Molloy
- Australian Proteome Analysis Facility (APAF), Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, Australia
| | - Alexander Swarbrick
- The Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2052, Australia.
| |
Collapse
|
28
|
Singh A, Gupta S, Badarukhiya JA, Sachan M. Detection of aberrant methylation of HOXA9 and HIC1 through multiplex MethyLight assay in serum DNA for the early detection of epithelial ovarian cancer. Int J Cancer 2020; 147:1740-1752. [PMID: 32191343 DOI: 10.1002/ijc.32984] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/22/2020] [Accepted: 03/05/2020] [Indexed: 02/06/2023]
Abstract
Accumulated evidence revealed that aberrant CpG island hypermethylation plays an important role in carcinogenesis which can serve as a promising target for molecular detection in body fluids. Despite a myriad of attempts to diagnose ovarian cancer (OC) at an early stage, this clinical aim remains a major challenge. To date, no single biomarker is able to accurately detect early OC in either tissue or body fluid. Aberrant DNA methylation patterns in circulating DNA provide highly specific cancer signals. In our study, we establish a novel panel of methylation-specific genes for the development of a TaqMan based qPCR assay to quantify methylation levels. We analyzed promoter methylation of homeobox A9 (HOXA9) and hypermethylated in cancer 1 (HIC1) quantitatively in 120 tissue samples and in 70 matched serum cell-free DNA (CFDNA) of cancerous and noncancerous samples by MethyLight assay. HOXA9 and HIC1 methylation occurred in 82.3 and 80.0% of OC tissue samples in singleplex assay, thereby confirming that methylation was highly cancer-specific. When either or both gene promoter showed methylation, the sensitivity was 88.2% with a specificity of 88.6% in tissue samples. The combined sensitivity for this novel marker panel in serum CFDNA was 88.9% (area under the curve [AUC] = 0.95). In contrast, no hypermethylation was observed in serum from matched cancer-free control women. Our results confirm the elevated performance of novel epigenetic marker panel (HOXA9 and HIC1) when analyzed in tissue and matched serum samples. Our findings reveal the potential of this biomarker panel as a suitable diagnostic serum biomarker for early screening of OC.
Collapse
Affiliation(s)
- Alka Singh
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, India
| | - Sameer Gupta
- Department of Surgical Oncology, King George Medical University, Lucknow, India
| | | | - Manisha Sachan
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, India
| |
Collapse
|
29
|
Makabe T, Arai E, Hirano T, Ito N, Fukamachi Y, Takahashi Y, Hirasawa A, Yamagami W, Susumu N, Aoki D, Kanai Y. Genome-wide DNA methylation profile of early-onset endometrial cancer: its correlation with genetic aberrations and comparison with late-onset endometrial cancer. Carcinogenesis 2020; 40:611-623. [PMID: 30850842 PMCID: PMC6610171 DOI: 10.1093/carcin/bgz046] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 01/28/2019] [Accepted: 03/03/2019] [Indexed: 12/20/2022] Open
Abstract
The present study was performed to clarify the significance of DNA methylation alterations during endometrial carcinogenesis. Genome-wide DNA methylation analysis and targeted sequencing of tumor-related genes were performed using the Infinium MethylationEPIC BeadChip and the Ion AmpliSeq Cancer Hotspot Panel v2, respectively, for 31 samples of normal control endometrial tissue from patients without endometrial cancer and 81 samples of endometrial cancer tissue. Principal component analysis revealed that tumor samples had a DNA methylation profile distinct from that of control samples. Gene Ontology enrichment analysis revealed significant differences of DNA methylation at 1034 CpG sites between early-onset endometrioid endometrial cancer (EE) tissue (patients aged ≤40 years) and late-onset endometrioid endometrial cancer (LE) tissue, which were accumulated among 'transcriptional factors'. Mutations of the CTNNB1 gene or DNA methylation alterations of genes participating in Wnt signaling were frequent in EEs, whereas genetic and epigenetic alterations of fibroblast growth factor signaling genes were observed in LEs. Unsupervised hierarchical clustering grouped EE samples in Cluster EA (n = 22) and samples in Cluster EB (n = 12). Clinicopathologically less aggressive tumors tended to be accumulated in Cluster EB, and DNA methylation levels of 18 genes including HOXA9, HOXD10 and SOX11 were associated with differences in such aggressiveness between the two clusters. We identified 11 marker CpG sites that discriminated EB samples from EA samples with 100% sensitivity and specificity. These data indicate that genetically and epigenetically different pathways may participate in the development of EEs and LEs, and that DNA methylation profiling may help predict tumors that are less aggressive and amenable to fertility preservation treatment.
Collapse
Affiliation(s)
- Takeshi Makabe
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan.,Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Eri Arai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Takuro Hirano
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan.,Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Nanako Ito
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | | | - Yoriko Takahashi
- Bioscience Department, Mitsui Knowledge Industry Co, Ltd, Tokyo, Japan
| | - Akira Hirasawa
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Nobuyuki Susumu
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan.,Department of Obstetrics and Gynecology, International University of Health and Welfare School of Medicine, Chiba, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Rusan M, Andersen RF, Jakobsen A, Steffensen KD. Circulating HOXA9-methylated tumour DNA: A novel biomarker of response to poly (ADP-ribose) polymerase inhibition in BRCA-mutated epithelial ovarian cancer. Eur J Cancer 2019; 125:121-129. [PMID: 31865042 DOI: 10.1016/j.ejca.2019.11.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 10/16/2019] [Accepted: 11/16/2019] [Indexed: 12/11/2022]
Abstract
AIM Poly (ADP-ribose) polymerase (PARP) inhibitors have emerged as a novel treatment option in BRCA-mutated ovarian cancer (OC); however, responses are variable and there is a lack of prognostic and predictive biomarkers. We therefore investigated whether homeobox A9 (HOXA9) promoter methylation in circulating tumour DNA (meth-ctDNA) can serve as a biomarker in patients with platinum-resistant BRCA-mutated OC, undergoing treatment with a PARP inhibitor. METHODS Patients (n = 32) were enrolled as part of a phase II trial testing veliparib in platinum-resistant BRCA-mutated OC. HOXA9 meth-ctDNA was determined at baseline and just before each treatment cycle using digital droplet polymerase chain reaction. Methylation status and change in methylation compared with baseline were correlated with overall survival (OS) and progression-free survival (PFS). RESULTS Detection of HOXA9 meth-ctDNA during treatment with a PARP inhibitor was associated with worse clinical outcomes. This association was apparent after the first cycle of treatment and maintained throughout treatment. After three treatment cycles, patients with detectable HOXA9 meth-ctDNA had a median PFS of 5.1 months compared with 8.3 months for patients without, and a median OS of 9.5 months compared with 19.4 months (p < 0.0001 and p = 0.002, respectively). Patients with detectable HOXA9 meth-ctDNA at baseline, but subsequent undetectable levels, had the most favourable clinical outcome, followed by patients with undetectable levels throughout. These associations were maintained in multivariate analysis. CONCLUSIONS Longitudinal monitoring of HOXA9 meth-ctDNA is clinically feasible and is strongly correlated to clinical outcomes (PFS, OS), suggesting that it may serve as a valuable predictive biomarker to inform clinical decision-making in the setting of platinum-resistant BRCA-mutated OC treated with a PARP inhibitor.
Collapse
Affiliation(s)
- Maria Rusan
- Department of Clinical Oncology, Vejle University Hospital, Vejle, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark.
| | - Rikke F Andersen
- Department of Biochemistry, Vejle University Hospital, Vejle, Denmark
| | - Anders Jakobsen
- Department of Clinical Oncology, Vejle University Hospital, Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Karina D Steffensen
- Department of Clinical Oncology, Vejle University Hospital, Vejle, Denmark; Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
31
|
Ciechomska M, Roszkowski L, Maslinski W. DNA Methylation as a Future Therapeutic and Diagnostic Target in Rheumatoid Arthritis. Cells 2019; 8:E953. [PMID: 31443448 PMCID: PMC6770174 DOI: 10.3390/cells8090953] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/28/2022] Open
Abstract
Rheumatoid arthritis (RA) is a long-term autoimmune disease of unknown etiology that leads to progressive joint destruction and ultimately to disability. RA affects as much as 1% of the population worldwide. To date, RA is not a curable disease, and the mechanisms responsible for RA development have not yet been well understood. The development of more effective treatments and improvements in the early diagnosis of RA is direly needed to increase patients' functional capacity and their quality of life. As opposed to genetic mutation, epigenetic changes, such as DNA methylation, are reversible, making them good therapeutic candidates, modulating the immune response or aggressive synovial fibroblasts (FLS-fibroblast-like synoviocytes) activity when it is necessary. It has been suggested that DNA methylation might contribute to RA development, however, with insufficient and conflicting results. Besides, recent studies have shown that circulating cell-free methylated DNA (ccfDNA) in blood offers a very convenient, non-invasive, and repeatable "liquid biopsy", thus providing a reliable template for assessing molecular markers of various diseases, including RA. Thus, epigenetic therapies controlling autoimmunity and systemic inflammation may find wider implications for the diagnosis and management of RA. In this review, we highlight current challenges associated with the treatment of RA and other autoimmune diseases and discuss how targeting DNA methylation may improve diagnostic, prognostic, and therapeutic approaches.
Collapse
Affiliation(s)
- Marzena Ciechomska
- Department of Pathophysiology and Immunology, National Institute of Geriatrics Rheumatology and Rehabilitation, 02-635 Warsaw, Poland.
| | - Leszek Roszkowski
- Department of Rheumatology, National Institute of Geriatrics Rheumatology and Rehabilitation, 02-635 Warsaw, Poland
| | - Wlodzimierz Maslinski
- Department of Pathophysiology and Immunology, National Institute of Geriatrics Rheumatology and Rehabilitation, 02-635 Warsaw, Poland
| |
Collapse
|
32
|
Delta tocotrienol in recurrent ovarian cancer. A phase II trial. Pharmacol Res 2019; 141:392-396. [DOI: 10.1016/j.phrs.2019.01.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 12/27/2022]
|
33
|
Feng LY, Chen CX, Li L. Hypermethylation of tumor suppressor genes is a risk factor for poor prognosis in ovarian cancer: A meta-analysis. Medicine (Baltimore) 2019; 98:e14588. [PMID: 30813180 PMCID: PMC6408028 DOI: 10.1097/md.0000000000014588] [Citation(s) in RCA: 6] [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] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE DNA methylation is the earliest and most studied epigenetic modification in cancer. The literature reported that the abnormal methylation level of multiple genes was associated with poor prognosis in ovarian cancer. However, due to a small sample size, the results reported in the literature vary widely. In this study, the correlation between aberrant methylation level of genes and poor prognosis of ovarian cancer was reviewed in order to clarify the role of DNA methylation in the prognosis of ovarian cancer. METHODS A systematic research of PubMed, EMbase, Cochrane Library, China Biology Medicine disc (CBMdisc), China National Knowledge Infrastructure (CNKI), Wanfang databases, and EMBASE was performed, and calculated the hazard ratio (HR) of overall survival (OS) and progression-free survival (PFS) and its 95% confidence interval. RESULTS HR of the OS obtained of target genes was 2.32 (95% CI: 1.54-3.48, P = .000); HR of the PFS obtained of target genes was 1.318 (95% CI: 0.848-2.050, P = .220). HR of OS achieved by tumor suppressor genes was 3.09 (95% CI 1.80 - 5.30, P = .000). CONCLUSION Hypermethylation of tumor suppressor genes indicate poor prognosis of ovarian cancer.
Collapse
|
34
|
Asphaug L, Melberg HO. The Cost-Effectiveness of Multigene Panel Testing for Hereditary Breast and Ovarian Cancer in Norway. MDM Policy Pract 2019; 4:2381468318821103. [PMID: 30746499 PMCID: PMC6360477 DOI: 10.1177/2381468318821103] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 11/21/2018] [Indexed: 12/20/2022] Open
Abstract
Background. Expansion of routine genetic testing for hereditary breast and ovarian cancer from conventional BRCA testing to a multigene test could improve diagnostic yield and increase the opportunity for cancer prevention in both identified carriers and their relatives. We use an economic decision model to assess whether the current knowledge on non-BRCA mutation prevalence, cancer risk, and patient preferences justifies switching to a multigene panel for testing of early-onset breast cancer patients. Methods. We evaluated routine testing by BRCA testing, a 7-gene panel, and a 14-gene panel using individual-level simulations of annual health state transitions over a lifetime perspective. Breast and ovarian cancer incidence is reduced and posttreatment survival is improved when high-risk mutations are detected and risk-reducing treatment offered. Most model inputs were synthesized from published literature. Intermediate health outcomes included breast and ovarian cancer incidence rates, along with organ-specific cancer mortality. Cost-effectiveness outcomes were health sector costs and quality-adjusted life years. Results. Intermediate health outcomes improved by testing with multigene panels. At a cost-effectiveness threshold of $77,000, a 7-gene panel test with five non-BRCA genes was the optimal strategy with an incremental cost-effectiveness ratio of $53,310 per quality-adjusted life year compared to BRCA-only testing. Limitations. Unable to stratify carriers to specific mutations within genes, we can only make predictions on the gene level, with combined risk estimates for known variants. As mutation prevalence is the absolute upper bound of returns to more expansive testing, the rarity of modelled mutations makes analysis outcomes sensitive to model implementation. Conclusions. A 7-gene panel to diagnose hereditary breast and ovarian cancer in early-onset breast cancer patients can be a cost-effective alternative to current BRCA-only testing in Norway.
Collapse
Affiliation(s)
- Lars Asphaug
- Department of Health Management and Health Economics, University of Oslo, Oslo, Norway
| | - Hans Olav Melberg
- Department of Health Management and Health Economics, University of Oslo, Oslo, Norway
| |
Collapse
|
35
|
Hentze JL, Høgdall CK, Høgdall EV. Methylation and ovarian cancer: Can DNA methylation be of diagnostic use? Mol Clin Oncol 2019; 10:323-330. [PMID: 30847169 DOI: 10.3892/mco.2019.1800] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 12/04/2018] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer is a silent killer and, due to late diagnosis and frequent chemo resistance in patients, the primary cause of fatality amongst the various types of gynecological cancer. The discovery of a specific and sensitive biomarker for ovarian cancer could improve early diagnosis, thereby saving lives. Biomarkers could also improve treatment, by predicting which patients will benefit from specific treatment strategies. DNA methylation is an epigenetic mechanism, and 'methylation imbalance' is characteristic of cancer. Previous research suggests that changes in DNA methylation can be used diagnostically, and that they may predict resistance to treatment. This paper gives an up-to-date overview of research investigating the potential of DNA methylation-based markers for diagnostics, prognostics, screening and prediction of drug resistance for ovarian cancer patients. DNA methylation cancer-biomarkers may be useful for cancer treatment, particularly since they are chemically stable and since cancer-associated changes in methylation typically precedes tumor growth. DNA methylation markers could improve diagnosis and treatment and might even be used for screening in the future. Furthermore, DNA methylation biomarkers could facilitate the development of precision medicine. However, at this point no biomarkers for ovarian cancer have a sufficient combination of sensitivity and specificity in a clinical setting. A reason for this is that most studies have focused on a single or a few methylation sites. More large screenings and genome-wide studies must be performed to increase the chance of identifying a DNA methylation marker which can identify ovarian cancer.
Collapse
Affiliation(s)
- Julie L Hentze
- Department of Pathology, Herlev Hospital, University of Copenhagen, 2730 Herlev, Denmark
| | - Claus K Høgdall
- Department of Gynecology, The Juliane Marie Centre, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Estrid V Høgdall
- Department of Pathology, Herlev Hospital, University of Copenhagen, 2730 Herlev, Denmark
| |
Collapse
|
36
|
Lu JJ, Yuan Z. Application of DNA methylation in early diagnosis and treatment of pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2019; 27:13-19. [DOI: 10.11569/wcjd.v27.i1.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most malignant gastrointestinal tumors, characterized by a poor prognosis. Most of the patients have an advanced disease at the time of diagnosis and lose the opportunity of radical surgery, resulting in a 5-year survival rate of less than 5%. Circulating tumor DNA, whose concentration in plasma of patients with pancreatic adenocarcinoma is higher than that in health controls, carries specific gene mutation and aberrant DNA methylation. Epigenetic change is one of the important characteristics of cell carcinogenesis. DNA methylation is an early event in tumorigenesis, which is more helpful for early diagnosis than gene mutation and can be observed in each stage of PC. Therefore, the detection of aberrant DNA methylation in the promoter region in patients with PC may be a non-invasive method for early cancer detection, predicting prognosis, and monitoring recurrence. In the present review, we discuss the recent advances in the study of DNA methylation in the early diagnosis of PC, and the potential application value in the treatment of PC.
Collapse
Affiliation(s)
- Jia-Jun Lu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zhou Yuan
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
37
|
|
38
|
Guo W, Zhu L, Yu M, Zhu R, Chen Q, Wang Q. A five-DNA methylation signature act as a novel prognostic biomarker in patients with ovarian serous cystadenocarcinoma. Clin Epigenetics 2018; 10:142. [PMID: 30446011 PMCID: PMC6240326 DOI: 10.1186/s13148-018-0574-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/28/2018] [Indexed: 12/25/2022] Open
Abstract
Background Ovarian cancer is the most fatal tumor of the female reproductive system and the fifth leading cause of cancer death among women in the USA. The prognosis is poor due to the lack of biomarkers for treatment options. Results The methylation array data of 551 patients with ovarian serous cystadenocarcinoma (OSC) in The Cancer Genome Atlas (TCGA) database were assessed in this study to explore the methylation biomarkers associated with prognosis and improve the prognosis of patients. These patients were divided into training (first two thirds) and validation datasets (remaining one third). A five-DNA methylation signature was found to be significantly associated with the overall survival of patients with OSC using the Cox regression analysis in the training dataset. The Kaplan–Meier analysis showed that the five-DNA methylation signature could significantly distinguish the high- and low-risk patients in both training and validation sets. The receiver operating characteristic (ROC) analysis further confirmed that the five-DNA methylation signature exhibited high sensitivity and specificity to predict the prognostic survival of patients. Also, the five-DNA methylation signature was not only applicable in patients of different ages, stages, histologic grade, and size of residual tumor after surgery but also more accurate in predicting OSC prognosis compared with known biomarkers. Conclusions This five-DNA methylation signature demonstrated the potential of being a novel independent prognostic indicator and served as an important tool for guiding the clinical treatment of OSC to improve outcome prediction and management for patients. Hence, the findings of this study might have potential clinical significance. Electronic supplementary material The online version of this article (10.1186/s13148-018-0574-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenna Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Liucun Zhu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Minghao Yu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Rui Zhu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qihan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Qiang Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
39
|
Lin HW, Fu CF, Chang MC, Lu TP, Lin HP, Chiang YC, Chen CA, Cheng WF. CDH1, DLEC1 and SFRP5 methylation panel as a prognostic marker for advanced epithelial ovarian cancer. Epigenomics 2018; 10:1397-1413. [PMID: 30324802 DOI: 10.2217/epi-2018-0035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM To investigate the CDH1, DLEC1 and SFRP5 gene methylation panel for advanced epithelial ovarian carcinoma (EOC). MATERIALS & METHODS One hundred and seventy-seven advanced EOC specimens were evaluated by methylation-specific PCR. We also used The Cancer Genome Atlas dataset to evaluate the panel. RESULTS The presence of two or more methylated genes was significant in recurrence (hazard ratio [HR]: 1.91 [1.33-2.76]; p = 0.002) and death (HR: 1.96 [1.26-3.06]; p = 0.006) in our cohort. In The Cancer Genome Atlas dataset, the presence of two or three methylated genes was significant in death (HR: 1.59 [1.15-2.18]; p = 0.0047) and close to the significance level in recurrence (HR: 1.37 [0.99-1.88]; p = 0.058). CONCLUSION The CDH1, DLEC1 and SFRP5 methylation panel is a potential prognostic biomarker for advanced EOC.
Collapse
Affiliation(s)
- Han-Wei Lin
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 10055, Taiwan
| | - Chi-Feng Fu
- Department of Obstetrics & Gynecology, E-da Cancer Hospital, Kaohsiung 82445, Taiwan
| | - Ming-Cheng Chang
- Department of Obstetrics & Gynecology, College of Medicine, National Taiwan University, Taipei 10041, Taiwan.,Institute of Nuclear Energy Research, Atomic Energy Council, Executive Yuan, Taoyuan 32546, Taiwan
| | - Tzu-Pin Lu
- Institute of Epidemiology & Preventive Medicine, College of Public Health, National Taiwan University, Taipei 10055, Taiwan
| | - Hsiu-Ping Lin
- Department of Obstetrics & Gynecology, College of Medicine, National Taiwan University, Taipei 10041, Taiwan
| | - Ying-Cheng Chiang
- Department of Obstetrics & Gynecology, College of Medicine, National Taiwan University, Taipei 10041, Taiwan
| | - Chi-An Chen
- Department of Obstetrics & Gynecology, College of Medicine, National Taiwan University, Taipei 10041, Taiwan
| | - Wen-Fang Cheng
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 10055, Taiwan.,Department of Obstetrics & Gynecology, College of Medicine, National Taiwan University, Taipei 10041, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| |
Collapse
|
40
|
Klymenko Y, Nephew KP. Epigenetic Crosstalk between the Tumor Microenvironment and Ovarian Cancer Cells: A Therapeutic Road Less Traveled. Cancers (Basel) 2018; 10:E295. [PMID: 30200265 PMCID: PMC6162502 DOI: 10.3390/cancers10090295] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022] Open
Abstract
Metastatic dissemination of epithelial ovarian cancer (EOC) predominantly occurs through direct cell shedding from the primary tumor into the intra-abdominal cavity that is filled with malignant ascitic effusions. Facilitated by the fluid flow, cells distribute throughout the cavity, broadly seed and invade through peritoneal lining, and resume secondary tumor growth in abdominal and pelvic organs. At all steps of this unique metastatic process, cancer cells exist within a multidimensional tumor microenvironment consisting of intraperitoneally residing cancer-reprogramed fibroblasts, adipose, immune, mesenchymal stem, mesothelial, and vascular cells that exert miscellaneous bioactive molecules into malignant ascites and contribute to EOC progression and metastasis via distinct molecular mechanisms and epigenetic dysregulation. This review outlines basic epigenetic mechanisms, including DNA methylation, histone modifications, chromatin remodeling, and non-coding RNA regulators, and summarizes current knowledge on reciprocal interactions between each participant of the EOC cellular milieu and tumor cells in the context of aberrant epigenetic crosstalk. Promising research directions and potential therapeutic strategies that may encompass epigenetic tailoring as a component of complex EOC treatment are discussed.
Collapse
Affiliation(s)
- Yuliya Klymenko
- Cell, Molecular and Cancer Biology Program, Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA.
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN 46617, USA.
| | - Kenneth P Nephew
- Cell, Molecular and Cancer Biology Program, Medical Sciences, Indiana University School of Medicine, Bloomington, IN 47405, USA.
- Department of Cellular and Integrative Physiology and Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
41
|
Losi L, Fonda S, Saponaro S, Chelbi ST, Lancellotti C, Gozzi G, Alberti L, Fabbiani L, Botticelli L, Benhattar J. Distinct DNA Methylation Profiles in Ovarian Tumors: Opportunities for Novel Biomarkers. Int J Mol Sci 2018; 19:ijms19061559. [PMID: 29882921 PMCID: PMC6032431 DOI: 10.3390/ijms19061559] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 01/16/2023] Open
Abstract
Aberrant methylation of multiple promoter CpG islands could be related to the biology of ovarian tumors and its determination could help to improve treatment strategies. DNA methylation profiling was performed using the Methylation Ligation-dependent Macroarray (MLM), an array-based analysis. Promoter regions of 41 genes were analyzed in 102 ovarian tumors and 17 normal ovarian samples. An average of 29% of hypermethylated promoter genes was observed in normal ovarian tissues. This percentage increased slightly in serous, endometrioid, and mucinous carcinomas (32%, 34%, and 45%, respectively), but decreased in germ cell tumors (20%). Ovarian tumors had methylation profiles that were more heterogeneous than other epithelial cancers. Unsupervised hierarchical clustering identified four groups that are very close to the histological subtypes of ovarian tumors. Aberrant methylation of three genes (BRCA1, MGMT, and MLH1), playing important roles in the different DNA repair mechanisms, were dependent on the tumor subtype and represent powerful biomarkers for precision therapy. Furthermore, a promising relationship between hypermethylation of MGMT, OSMR, ESR1, and FOXL2 and overall survival was observed. Our study of DNA methylation profiling indicates that the different histotypes of ovarian cancer should be treated as separate diseases both clinically and in research for the development of targeted therapies.
Collapse
Affiliation(s)
- Lorena Losi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
- Unit of Pathology, Azienda Ospedaliero-Universitaria Policlinico, 41124 Modena, Italy.
| | - Sergio Fonda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Sara Saponaro
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
- Institute of Pathology, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| | - Sonia T Chelbi
- Institute of Pathology, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| | - Cesare Lancellotti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Gaia Gozzi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Loredana Alberti
- Institute of Pathology, Lausanne University Hospital, 1011 Lausanne, Switzerland.
| | - Luca Fabbiani
- Unit of Pathology, Azienda Ospedaliero-Universitaria Policlinico, 41124 Modena, Italy.
| | - Laura Botticelli
- Unit of Pathology, Azienda Ospedaliero-Universitaria Policlinico, 41124 Modena, Italy.
| | - Jean Benhattar
- Institute of Pathology, Lausanne University Hospital, 1011 Lausanne, Switzerland.
- Aurigen, Centre de Génétique et Pathologie, 1004 Lausanne, Switzerland.
| |
Collapse
|
42
|
Wang H, Cui M, Zhang S, He J, Song L, Chen Y. Relationship between RAS Association Domain Family Protein 1A Promoter Methylation and the Clinicopathological Characteristics in Patients with Ovarian Cancer: A Systematic Meta-Analysis. Gynecol Obstet Invest 2017; 83:349-357. [PMID: 29130987 DOI: 10.1159/000484245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/13/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND To investigate the relationship between RAS association domain family protein 1A (RASSF1A) promoter methylation and the clinical features, and the survival of ovarian cancer patients. METHODS A comprehensive literature search was conducted in the PubMed, Embase, EBSCO, and Cochrane Library databases. The overall ORs with their 95% CIs were calculated in this meta-analysis. RESULTS Finally 17 relevant publications with 1,108 ovarian cancer samples were available for the current meta-analysis. RASSF1A promoter methylation had a significantly higher level in ovarian cancer than in low malignant potential (LMP) tumors. No significant relationship was observed between RASSF1A promoter methylation and the clinicopathological characteristics in ovarian cancer. Two studies reported that RASSF1A promoter methylation was not correlated with the survival of patients with ovarian cancer. CONCLUSIONS Our findings suggest that the use of RASSF1A promoter methylation could distinguish ovarian cancer and LMP tumors. -RASSF1A promoter methylation may not be correlated with the clinical features and the survival of ovarian cancer patients. More studies with large sample sizes are essential in the future.
Collapse
Affiliation(s)
- Hong Wang
- Department of Obstetrics and Gynaecology, Affiliated Hospital of Beihua University, Jilin, China
| | - Manhua Cui
- Department of Obstetrics and Gynaecology, The Second Hospital of Jilin University, Changchun, China
| | - Shuangli Zhang
- Department of Obstetrics and Gynaecology, 307 Hospital of the people's Liberation Army, Beijing, China
| | - Jie He
- Department of Obstetrics and Gynaecology, Affiliated Hospital of Beihua University, Jilin, China
| | - Li Song
- Department of Obstetrics and Gynaecology, Affiliated Hospital of Beihua University, Jilin, China
| | - Ying Chen
- Department of Obstetrics and Gynaecology, Affiliated Hospital of Beihua University, Jilin, China
| |
Collapse
|
43
|
Zhao H, Xu H, Xue L. Regulatory network involving miRNAs and genes in serous ovarian carcinoma. Oncol Lett 2017; 14:6259-6268. [PMID: 29113276 DOI: 10.3892/ol.2017.6927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/23/2017] [Indexed: 12/19/2022] Open
Abstract
Serous ovarian carcinoma (SOC) is one of the most life-threatening types of gynecological malignancy, but the pathogenesis of SOC remains unknown. Previous studies have indicated that differentially expressed genes and microRNAs (miRNAs) serve important functions in SOC. However, genes and miRNAs are identified in a disperse form, and limited information is known about the regulatory association between miRNAs and genes in SOC. In the present study, three regulatory networks were hierarchically constructed, including a differentially-expressed network, a related network and a global network to reveal associations between each factor. In each network, there were three types of factors, which were genes, miRNAs and transcription factors that interact with each other. Focus was placed on the differentially-expressed network, in which all genes and miRNAs were differentially expressed and therefore may have affected the development of SOC. Following the comparison and analysis between the three networks, a number of signaling pathways which demonstrated differentially expressed elements were highlighted. Subsequently, the upstream and downstream elements of differentially expressed miRNAs and genes were listed, and a number of key elements (differentially expressed miRNAs, genes and TFs predicted using the P-match method) were analyzed. The differentially expressed network partially illuminated the pathogenesis of SOC. It was hypothesized that if there was no differential expression of miRNAs and genes, SOC may be prevented and treatment may be identified. The present study provided a theoretical foundation for gene therapy for SOC.
Collapse
Affiliation(s)
- Haiyan Zhao
- College of Computer Science and Technology, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Hao Xu
- College of Computer Science and Technology, Jilin University, Changchun, Jilin 130012, P.R. China.,Zhuhai Laboratory of Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, Department of Computer Science and Technology, Zhuhai College of Jilin University, Zhuhai, Guangdong 519041, P.R. China
| | - Luchen Xue
- College of Software, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
44
|
Rezk NA, Mohamed RH, Alnemr AA, Harira M. Promoter Methylation of RASSF1A Gene in Egyptian Patients with Ovarian Cancer. Appl Biochem Biotechnol 2017; 185:153-162. [PMID: 29098560 DOI: 10.1007/s12010-017-2648-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 10/26/2017] [Indexed: 12/14/2022]
Abstract
Ovarian malignancy is diagnosed in nearly a fourth of a million women internationally every year. Methylation of RASSF1A tumor suppressor gene prompts its inactivation in diseases. In this study, the RASSF1A promoter methylation was detected by methylated-specific PCR and investigated serum RASSF1A protein level through enzyme-linked immunosorbant assay in 160 Egyptian patients with ovarian cancer and 160 healthy controls. The present work proved that there was a higher frequency of RASSF1A methylation and a decrease in its serum level in patients with ovarian cancer compared to controls as well as in the high-grade tumor patients compared to low grade ones and also in advanced ovarian tumor stage compared to early stages. Our study exhibited that RASSF1A promoter hypermethylation and its protein levels may be a reliable and sensitive tool for diagnosing and monitoring of ovarian malignancy patients.
Collapse
Affiliation(s)
- Noha A Rezk
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Rasha H Mohamed
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Amr AbdAlmohsen Alnemr
- Obstetrics and Gynecology Departments, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mervat Harira
- Obstetrics and Gynecology Departments, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
45
|
Giannopoulou L, Chebouti I, Pavlakis K, Kasimir-Bauer S, Lianidou ES. RASSF1A promoter methylation in high-grade serous ovarian cancer: A direct comparison study in primary tumors, adjacent morphologically tumor cell-free tissues and paired circulating tumor DNA. Oncotarget 2017; 8:21429-21443. [PMID: 28206954 PMCID: PMC5400595 DOI: 10.18632/oncotarget.15249] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 11/11/2016] [Indexed: 12/12/2022] Open
Abstract
The RASSF1A promoter is frequently methylated in high-grade serous ovarian cancer (HGSC). We examined RASSF1A promoter methylation in primary tumors, adjacent morphologically tumor cell-free tissues and corresponding circulating tumor DNA (ctDNA) samples of patients with HGSC, using a real-time methylation specific PCR (real-time MSP) and a methylation-sensitive high-resolution melting analysis (MS-HRMA) assay for the detection and semi-quantitative estimation of methylation, respectively. Two groups of primary HGSC tumor FFPE samples were recruited (Group A n=67 and Group B n=61), along with matched adjacent morphologically tumor cell-free tissues (n=58) and corresponding plasma samples (n=59) for group B. Using both assays, RASSF1A promoter was found highly methylated in primary tumors of both groups, and at lower percentages in the adjacent morphologically tumor cell-free tissues. Interestingly, RASSF1A promoter methylation was also observed in ctDNA by real-time MSP. Overall survival (OS) was significantly associated with RASSF1A promoter methylation in primary tumor samples using MS-HRMA (P=0.023). Our results clearly indicate that RASSF1A promoter is methylated in adjacent tissue surrounding the tumor in HGSC patients. We report for the first time that RASSF1A promoter methylation provides significant prognostic information in HGSC patients.
Collapse
Affiliation(s)
- Lydia Giannopoulou
- Analysis of Circulating Tumor Cells Laboratory, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, University Campus, Athens, 15771, Greece
| | - Issam Chebouti
- Department of Gynecology and Obstetrics, University Hospital of Essen, University of Duisburg-Essen, Essen, D-45122, Germany
| | - Kitty Pavlakis
- Pathology Department, IASO Women's Hospital, 15123, Marousi, Athens, Greece
| | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital of Essen, University of Duisburg-Essen, Essen, D-45122, Germany
| | - Evi S Lianidou
- Analysis of Circulating Tumor Cells Laboratory, Laboratory of Analytical Chemistry, Department of Chemistry, University of Athens, University Campus, Athens, 15771, Greece
| |
Collapse
|
46
|
El Bairi K, Kandhro AH, Gouri A, Mahfoud W, Louanjli N, Saadani B, Afqir S, Amrani M. Emerging diagnostic, prognostic and therapeutic biomarkers for ovarian cancer. Cell Oncol (Dordr) 2017; 40:105-118. [PMID: 27981507 DOI: 10.1007/s13402-016-0309-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In spite of various treatment options currently available, ovarian cancer (OC) still remains a leading cause of death in women world-wide. Diagnosis at an early stage is one of the most important factors that determines survival. Current clinical diagnostic tools have, however, a limited efficacy in early OC detection. Therefore, there is a critical need for new (early) diagnostic biomarkers and tools. Through advances in genomic, proteomic and metabolomic techniques, several novel molecular OC biomarkers have recently been identified. These biomarkers are currently subject to validation. In addition, integration of genomic, proteomic and metabolomic data, in conjunction with epidemiologic and clinical data, is considered essential for obtaining useful results. Interesting recent work has already shown that specific diagnostic biomarkers, such as BRCA mutations, may have profound therapeutic implications. Here, we review the current state of OC research through literature and database searches, with a focus on various recently identified biomarkers via different technologies for the (early) diagnosis, prognosis and treatment of OC. CONCLUSIONS Multi-biomarker panels accompanied by a meticulous determination of their sensitivity and specificity, as well their validation, using multivariate analyses will be critical for its clinical application, including early OC detection and tailor-made OC treatment.
Collapse
Affiliation(s)
- Khalid El Bairi
- Faculty of Medicine and Pharmacy, Oujda, Morocco.
- Independent Research Team in Cancer Biology and Bioactive Compounds, Mohammed 1st University, Oujda, Morocco.
| | - Abdul Hafeez Kandhro
- Department of Biochemistry, Healthcare Molecular and Diagnostic Laboratory, Hyderabad, Pakistan
| | - Adel Gouri
- Laboratory of Medical Biochemistry, Ibn Rochd University Hospital, Annaba, Algeria
| | - Wafaa Mahfoud
- Laboratory of Biology and Health, URAC-34, Faculty of Science Ben Msik, University Hassan II, Mohammedia, Casablanca, Morocco
| | | | - Brahim Saadani
- IVF center IRIFIV, Clinique des Iris, Casablanca, Morocco
| | - Said Afqir
- Department of Medical Oncology, Mohamed 1st University Hospital, Oujda, Morocco
| | - Mariam Amrani
- Equipe de Recherche ONCOGYMA, Faculty of Medicine, Pathology Department, National Institute of Oncology, Université Mohamed V, Rabat, Morocco
| |
Collapse
|
47
|
Next-Generation Sequencing Approach in Methylation Analysis of HNF1B and GATA4 Genes: Searching for Biomarkers in Ovarian Cancer. Int J Mol Sci 2017; 18:ijms18020474. [PMID: 28241454 PMCID: PMC5344006 DOI: 10.3390/ijms18020474] [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: 02/03/2017] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 12/31/2022] Open
Abstract
DNA methylation is well-known to be associated with ovarian cancer (OC) and has great potential to serve as a biomarker in monitoring response to therapy and for disease screening. The purpose of this study was to investigate methylation of HNF1B and GATA4 and correlate detected methylation with clinicopathological characteristic of OC patients. The study group consisted of 64 patients with OC and 35 control patients. To determine the most important sites of HNF1B and GATA4, we used next-generation sequencing. For further confirmation of detected methylation of selected regions, we used high-resolution melting analysis and methylation-specific real-time polymerase chain reaction (PCR). Selected regions of HNF1B and GATA4 were completely methylation free in all control samples, whereas methylation-positive pattern was observed in 32.8% (HNF1B) and 45.3% (GATA4) of OC samples. Evaluating both genes together, we were able to detect methylation in 65.6% of OC patients. We observed a statistically significant difference in HNF1B methylation between samples with different stages of OC. We also detected subtype specific methylation in GATA4 and a decrease of methylation in late stages of OC. The combination of unmethylated HNF1B and methylated GATA4 was associated with longer overall survival. In our study, we employed innovative approach of methylation analysis of HNF1B and GATA4 to search for possible epigenetic biomarkers. We confirmed the significance of the HNF1B and GATA4 hypermethylation with emphasis on the need of selecting the most relevant sites for analysis. We suggest selected CpGs to be further examined as a potential positive prognostic factor.
Collapse
|
48
|
Hossain T, Mahmudunnabi G, Masud MK, Islam MN, Ooi L, Konstantinov K, Hossain MSA, Martinac B, Alici G, Nguyen NT, Shiddiky MJA. Electrochemical biosensing strategies for DNA methylation analysis. Biosens Bioelectron 2017; 94:63-73. [PMID: 28259051 DOI: 10.1016/j.bios.2017.02.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 12/31/2022]
Abstract
DNA methylation is one of the key epigenetic modifications of DNA that results from the enzymatic addition of a methyl group at the fifth carbon of the cytosine base. It plays a crucial role in cellular development, genomic stability and gene expression. Aberrant DNA methylation is responsible for the pathogenesis of many diseases including cancers. Over the past several decades, many methodologies have been developed to detect DNA methylation. These methodologies range from classical molecular biology and optical approaches, such as bisulfite sequencing, microarrays, quantitative real-time PCR, colorimetry, Raman spectroscopy to the more recent electrochemical approaches. Among these, electrochemical approaches offer sensitive, simple, specific, rapid, and cost-effective analysis of DNA methylation. Additionally, electrochemical methods are highly amenable to miniaturization and possess the potential to be multiplexed. In recent years, several reviews have provided information on the detection strategies of DNA methylation. However, to date, there is no comprehensive evaluation of electrochemical DNA methylation detection strategies. Herein, we address the recent developments of electrochemical DNA methylation detection approaches. Furthermore, we highlight the major technical and biological challenges involved in these strategies and provide suggestions for the future direction of this important field.
Collapse
Affiliation(s)
- Tanvir Hossain
- Department of Biochemistry & Molecular Biology, Shahjalal University of Science & Technology, Sylhet 3114, Bangladesh
| | - Golam Mahmudunnabi
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science & Technology, Sylhet 3114, Bangladesh
| | - Mostafa Kamal Masud
- Department of Biochemistry & Molecular Biology, Shahjalal University of Science & Technology, Sylhet 3114, Bangladesh; Institute for Superconducting and Electronic Materials, Australian Institute for Innovative Materials (AIIM), University of Wollongong, Squires Way, Innovation Campus, North Wollongong, NSW 2519, Australia; Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia
| | - Md Nazmul Islam
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia; School of Natural Sciences, Griffith University (Nathan Campus), Nathan, QLD 4111, Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, School of Biological Sciences, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Konstantin Konstantinov
- Institute for Superconducting and Electronic Materials, Australian Institute for Innovative Materials (AIIM), University of Wollongong, Squires Way, Innovation Campus, North Wollongong, NSW 2519, Australia
| | - Md Shahriar Al Hossain
- Institute for Superconducting and Electronic Materials, Australian Institute for Innovative Materials (AIIM), University of Wollongong, Squires Way, Innovation Campus, North Wollongong, NSW 2519, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Gursel Alici
- ARC Centre of Excellence for Electromaterials Science, University of Wollongong, Northfields Avenue, Wollongong, NSW 2522, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia
| | - Muhammad J A Shiddiky
- Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia; School of Natural Sciences, Griffith University (Nathan Campus), Nathan, QLD 4111, Australia.
| |
Collapse
|
49
|
El Bairi K, Kandhro AH, Gouri A, Mahfoud W, Louanjli N, Saadani B, Afqir S, Amrani M. Emerging diagnostic, prognostic and therapeutic biomarkers for ovarian cancer. CELLULAR ONCOLOGY (DORDRECHT) 2016. [PMID: 27981507 DOI: 10.1007/s13402-016-0309-1] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND In spite of various treatment options currently available, ovarian cancer (OC) still remains a leading cause of death in women world-wide. Diagnosis at an early stage is one of the most important factors that determines survival. Current clinical diagnostic tools have, however, a limited efficacy in early OC detection. Therefore, there is a critical need for new (early) diagnostic biomarkers and tools. Through advances in genomic, proteomic and metabolomic techniques, several novel molecular OC biomarkers have recently been identified. These biomarkers are currently subject to validation. In addition, integration of genomic, proteomic and metabolomic data, in conjunction with epidemiologic and clinical data, is considered essential for obtaining useful results. Interesting recent work has already shown that specific diagnostic biomarkers, such as BRCA mutations, may have profound therapeutic implications. Here, we review the current state of OC research through literature and database searches, with a focus on various recently identified biomarkers via different technologies for the (early) diagnosis, prognosis and treatment of OC. CONCLUSIONS Multi-biomarker panels accompanied by a meticulous determination of their sensitivity and specificity, as well their validation, using multivariate analyses will be critical for its clinical application, including early OC detection and tailor-made OC treatment.
Collapse
Affiliation(s)
- Khalid El Bairi
- Faculty of Medicine and Pharmacy, Oujda, Morocco. .,Independent Research Team in Cancer Biology and Bioactive Compounds, Mohammed 1st University, Oujda, Morocco.
| | - Abdul Hafeez Kandhro
- Department of Biochemistry, Healthcare Molecular and Diagnostic Laboratory, Hyderabad, Pakistan
| | - Adel Gouri
- Laboratory of Medical Biochemistry, Ibn Rochd University Hospital, Annaba, Algeria
| | - Wafaa Mahfoud
- Laboratory of Biology and Health, URAC-34, Faculty of Science Ben Msik, University Hassan II, Mohammedia, Casablanca, Morocco
| | | | - Brahim Saadani
- IVF center IRIFIV, Clinique des Iris, Casablanca, Morocco
| | - Said Afqir
- Department of Medical Oncology, Mohamed 1st University Hospital, Oujda, Morocco
| | - Mariam Amrani
- Equipe de Recherche ONCOGYMA, Faculty of Medicine, Pathology Department, National Institute of Oncology, Université Mohamed V, Rabat, Morocco
| |
Collapse
|
50
|
El Bairi K, Kandhro AH, Gouri A, Mahfoud W, Louanjli N, Saadani B, Afqir S, Amrani M. Emerging diagnostic, prognostic and therapeutic biomarkers for ovarian cancer. CELLULAR ONCOLOGY (DORDRECHT) 2016. [PMID: 27981507 DOI: 10.1007/s13402-016-0309-1]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND In spite of various treatment options currently available, ovarian cancer (OC) still remains a leading cause of death in women world-wide. Diagnosis at an early stage is one of the most important factors that determines survival. Current clinical diagnostic tools have, however, a limited efficacy in early OC detection. Therefore, there is a critical need for new (early) diagnostic biomarkers and tools. Through advances in genomic, proteomic and metabolomic techniques, several novel molecular OC biomarkers have recently been identified. These biomarkers are currently subject to validation. In addition, integration of genomic, proteomic and metabolomic data, in conjunction with epidemiologic and clinical data, is considered essential for obtaining useful results. Interesting recent work has already shown that specific diagnostic biomarkers, such as BRCA mutations, may have profound therapeutic implications. Here, we review the current state of OC research through literature and database searches, with a focus on various recently identified biomarkers via different technologies for the (early) diagnosis, prognosis and treatment of OC. CONCLUSIONS Multi-biomarker panels accompanied by a meticulous determination of their sensitivity and specificity, as well their validation, using multivariate analyses will be critical for its clinical application, including early OC detection and tailor-made OC treatment.
Collapse
Affiliation(s)
- Khalid El Bairi
- Faculty of Medicine and Pharmacy, Oujda, Morocco. .,Independent Research Team in Cancer Biology and Bioactive Compounds, Mohammed 1st University, Oujda, Morocco.
| | - Abdul Hafeez Kandhro
- Department of Biochemistry, Healthcare Molecular and Diagnostic Laboratory, Hyderabad, Pakistan
| | - Adel Gouri
- Laboratory of Medical Biochemistry, Ibn Rochd University Hospital, Annaba, Algeria
| | - Wafaa Mahfoud
- Laboratory of Biology and Health, URAC-34, Faculty of Science Ben Msik, University Hassan II, Mohammedia, Casablanca, Morocco
| | | | - Brahim Saadani
- IVF center IRIFIV, Clinique des Iris, Casablanca, Morocco
| | - Said Afqir
- Department of Medical Oncology, Mohamed 1st University Hospital, Oujda, Morocco
| | - Mariam Amrani
- Equipe de Recherche ONCOGYMA, Faculty of Medicine, Pathology Department, National Institute of Oncology, Université Mohamed V, Rabat, Morocco
| |
Collapse
|