Skip to main content
Erschienen in: Journal of Ovarian Research 1/2016

Open Access 01.12.2016 | Research

A network-pathway based module identification for predicting the prognosis of ovarian cancer patients

verfasst von: Xin Wang, Shan-shan Wang, Lin Zhou, Li Yu, Lan-mei Zhang

Erschienen in: Journal of Ovarian Research | Ausgabe 1/2016

Abstract

Background

This study aimed to screen multiple genes biomarkers based on gene expression data for predicting the survival of ovarian cancer patients.

Methods

Two microarray data of ovarian cancer samples were collected from The Cancer Genome Atlas (TCGA) database. The data in the training set were used to construct Reactome functional interactions network, which then underwent Markov clustering, supervised principal components, Cox proportional hazard model to screen significantly prognosis related modules. The distinguishing ability of each module for survival was further evaluated by the testing set. Gene Ontology (GO) functional and pathway annotations were performed to identify the roles of genes in each module for ovarian cancer.

Results

The network based approach identified two 7-gene functional interaction modules (31: DCLRE1A, EXO1, KIAA0101, KIN, PCNA, POLD3, POLD2; 35: DKK3, FABP3, IRF1, AIM2, GBP1, GBP2, IRF2) that are associated with prognosis of ovarian cancer patients. These network modules are related to DNA repair, replication, immune and cytokine mediated signaling pathways.

Conclusions

The two 7-gene expression signatures may be accurate predictors of clinical outcome in patients with ovarian cancer and has the potential to develop new therapeutic strategies for ovarian cancer patients.

Background

Ovarian cancer is the most common lethal gynecologic malignancy in women worldwide, with an estimated 22,280 newly diagnosed cases and approximately 14,240 deaths in 2016 in the United States [1]. Due to the lack of specific symptoms and effective screening tests, approximately 70 % of ovarian cancer patients have been in advanced-stage (stage III or IV) when they are firstly diagnosed, leading to the 5-year survival rate of less than 30 % [2]. By contrast, patients who are diagnosed with early-stage (stage I or II) have a 5-year survival rate of up to 70–90 % [2]. These data indicate the importance to identify the sensitive biomarkers to early distinguish the patients with different prognosis, aiming to determine optimal treatment strategies.
In the past years, remarkable achievements have been obtained in the investigation of prognostic markers for ovarian cancer. For instance, a 10-gene signature (AEBP1, COL11A1, COL5A1, COL6A2, LOX, POSTN, SNAI2, THBS2, TIMP3, and VCAN) has been validated to be associated with poor overall survival in patients with high-grade serous ovarian cancer [3]. The presence of a BRCA1 or BRCA2 mutation is associated with a better prognosis in patients with invasive ovarian cancer [4]. A recent study has found that suppression of ABHD2 in OVCA420 cells increased phosphorylated p38 and ERK, platinum resistance, and side population cells, promoting a malignant phenotype and poor prognosis in serous ovarian cancer [5]. Furthermore, CD73 enhances ovarian tumor cell growth and expression of antiapoptotic BCL-2 family members, indicating a role of CD73 as a prognostic marker of patient survival in high-grade serous ovarian cancer [6]. Although the aforementioned genes have been shown to be correlated with the prognosis in ovarian cancer, their prognostic accuracy may be limited because the development of disease usually involves several genes and the interaction between them to form a complex pathway. Therefore, it is necessary to identify gene networks and pathways including multiple genes and their interactions, which can be achieved by Reactome functional interaction (FI) network construction as described previously [7, 8].
In the present study, we aimed to construct the Reactome FIs network to analyze the gene signatures that was significantly related to ovarian cancer patient survival based on gene expression profiling data extracted from The Cancer Genome Atlas (TCGA) database.

Methods

As the paper did not involve any human or animal, the ethical approval was not required.

Gene expression data

Two gene expression datasets with their corresponding clinical data (including survival status and time) for ovarian cancer samples were downloaded from TCGA database (https://​tcga-data.​nci.​nih.​gov/​tcga). Data of one gene expression dataset were produced from the BI-HT-HG-U133A platform, in which 536 samples were included and 12042 genes were expressed in each sample (defined as BI). The other gene expression profiling from 559 ovarian cancer patients was produced from the UNC-AgilentG4502A-07-3 microarray platform, in which 17814 genes were included (defined as U3). These two datasets were randomly divided into training (BI) or testing sets (U3).

Construction of Reactome FI network

The annotated FIs were extracted from five pathway databases, including Reactome [9], kyoto encyclopedia of genes and genomes (KEGG) [10], protein annotation through evolutionary relationship (Panther) [11], The Cancer Cell Map (http://​cancer.​cellmap.​org/​), and NCI Pathway Interaction Database (NCI-PID) [12]. The protein FIs were predicted by physical protein-protein interactions (PPIs) in human organisms (catalogued in the Biological General Repository for Interaction Datasets (BioGrid) [13], the Human Protein Reference Database (HPRD) [14] and IntACT [15]), model organisms (from IntAct [15] based on Ensembl Compara [16]), and protein domain–domain interactions (from PFam [17]). The naive Bayes classifier, a simple machine learning method [18], was used to score the probability that a protein pair-wise relationship reflects a functional pathway event, during which the annotated FIs were selected as positive training sets, whereas the predicted FIs were defined as negative training sets. Subsequently, the gene expression data of BI from the TCGA were mapped into the constructed Reactome FIs via co-expression relationships (calculated by Pearson correlation) to distribute the weight of each edge.

Markov clustering (MCL)

The gene/protein correlations in the Reactome FI network were input into the Reactome FI Cytoscape plugin (MCL) [7] to generate a sub-network for a list of selected network modules based on module size (≥7) and average correlation (Pearson correlation coefficient ≥0.25). To control the size of network modules generated from the MCL clustering, the inflation coefficient was set as 5.0.
The prognosis-related modules were further predicted based on the supervised principal components (superpc) [19] using the Superpc V1.05 software package under the programming environment R (http://​statweb.​stanford.​edu/​~tibs/​superpc/​). A module-based gene expression matrix was generated by using mean expression level of genes in each module across 536 ovarian cancer samples, and then underwent the superpc analysis. A 10-fold cross-validation curve was performed for estimating the best threshold. In addition, Cox proportional hazard (PH) model was also performed to correlate each module with survival data (p < 0.05), followed by Kaplan-Meier analysis to demonstrate the distinguishing ability of each module for survival.

Gene Ontology (GO) functional and pathway annotations

The genes in prognosis-related modules were subjected to the GO and pathway enrichment analyses to identify their roles in ovarian cancer. GO and pathway functional annotations were conducted for the survival-associated genes using the Reactome FI plug-in of Cytoscape [20]. False discovery rate (FDR) < 0.05 was used for a threshold to assess the statistical significance.

Results

Data information

Two datasets [BI-HT-HG-U133A (BI), and UNC-AgilentG4502A-07-3(U2)] were obtained from TCGA. The BI dataset contained 536 samples, and expression data of 12042 genes were included in each sample. The U2 dataset contained 559 samples, and expression data of 17814 genes were included in each sample. In this study, BI was used as the training dataset, and U2 was used as the test dataset (Fig. 1).

Analysis of the FI network and modules

Based on the BI dataset, a weighted FI network including 710 proteins and 9516 interactions were constructed. Subsequently, using MCL network clustering, a total of 41 modules were obtained, and the number of genes in each module ranged from 7 to 118. Furthermore, using the Superpc package with a threshold value of 0.73, 14 prognosis-related modules were identified from the 41 modules (Table 1). Afterwards, 6 significant modules (modules 6, 8, 20, 26, 31 and 35) with the p-value < 0.05 were identified from the 14 modules based on the Cox PH analysis (Table 2). These 6 modules were validated by the U2 dataset, and two modules (modules 31 and 35) were also significant in the U2 dataset. Thus, modules 31 and 35 were further analyzed.
Table 1
Superpc analysis for prognosis related modules according to 10-fold cross-validation method
Modules
Threshold
Cross-validation scores
1
0.050
9.667
6
0.119
9.096
8
0.187
7.953
12
0.255
7.146
14
0.324
8.551
19
0.392
7.371
20
0.460
6.903
25
0.529
8.649
26
0.597
8.848
27
0.665
9.558
28
0.734
10.394
31
0.802
10.177
35
0.870
7.547
36
0.939
7.414
Table 2
Cox proportional hazard analysis for prognosis related modules using the training (BI) and test datasets (U3)
Module
Size
BI
U3
Likelihood ratio
p-value
Likelihood ratio
p-value
26
9
9.41
0.002
0.32
0.574
20
10
6.91
0.009
3.58
0.058
31
7
7.43
0.006
4.45
0.035
35
7
6.19
0.013
6.25
0.012
6
22
5.96
0.015
2.57
0.109
8
21
6.22
0.013
0.67
0.414

Analysis of modules 31 and 35

A set of 7 genes (DCLRE1A, EXO1, KIAA0101, KIN, PCNA, POLD3, POLD2) were included in the module 31 (Fig. 2a), and 7 genes (DKK3, FABP3, IRF1, AIM2, GBP1, GBP2, IRF2) were included in the module 35 (Fig. 2b). Kaplan-Meier plot demonstrated that the gene expression in these two modules can significantly distinguish the patients with longer and shorter survivals (Fig. 3).
To further investigate the biological functions of the genes in modules 31 and 35, GO and pathway annotations were performed. The genes in module 31 were mainly related to the functions of DNA repair, DNA replication and cell cycle (Fig. 4). The genes in module 35 were significantly associated with functions about immune and cytokine or interferon mediated signaling pathways (Fig. 5).

Discussion

In this study, a total of 41 modules were obtained from the FI network based on the expression data in the BI dataset. Using MCL network clustering, superpc modeling and Cox PH analysis, two modules, modules 31 and 35, were identified to be significantly associated with prognosis of ovarian cancer patients. Seven genes were included in the two modules (31: DCLRE1A, EXO1, KIAA0101, KIN, PCNA, POLD3, POLD2; 35: DKK3, FABP3, IRF1, AIM2, GBP1, GBP2, IRF2). Furthermore, the genes in module 31 were related to DNA repair or replication, whereas the genes in module 35 were associated with immune and cytokine interferon mediated signaling pathways.
DCLRE1, also known as SNM1A, belongs to a member of a small gene family that is characterized by a metallo-β-lactamase fold and an appended β-CASP domain that together are proposed to function as a DNA endonuclease to participate in DNA inter-strand cross-link repair [21]. DNA cross-link repair is beneficial to maintain genomic stability and enables cells to survive DNA damage, contributing to less risk of tumorigenesis [22]. However, recent studies indicate that the high efficiency of DNA cross-link repair may also promote the excessive proliferation of cells, driving tumor initiation and progression [2325]. Thus, down-regulation of DNA repair genes may be a promising target for anticancer therapy [26], which has been demonstrated by the study of Wu et al. [27]. Wu et al. have found that DCLRE1A is significantly decreased by bufalin, which promotes lung cancer apoptosis [27]. In addition, inhibition of DNA cross-link repair was also proved to reverse treatment resistance and improve the therapeutic efficacy [28].
EXO1 encodes exonuclease and plays important roles in mismatch repair by resecting the damaged strand. Similar to DCLRE1A, Exo1 is also shown to be higher expressed in tumor tissues than that in the normal tissues [29, 30]. A previous study has demonstrated that FOXM1 facilitates DNA repair through regulating direct transcriptional target EXO1 to protect ovarian cancer cells from cisplatin-mediated apoptosis, and attenuating EXO1 expression by small interfering RNA augments the cisplatin sensitivity of ovarian cancer cells [31]. POLD2 or POLD3 are both the subunits of DNA polymerase delta that possesses both polymerase and 3′ to 5′ exonuclease activity and plays a critical role in DNA replication and repair [32]. POLD2 was found to be increased in average 2.5- to almost 20-fold in moderately and poorly differentiated serous carcinomas of epithelial ovarian cancer, eventually leading to poor prognosis [33].
Furthermore, proliferating cell nuclear antigen (PCNA) is a ring-shaped homo-triomeric protein that functions as a necessary clamping platform to recruit numerous enzymes involved in DNA replication and repair, such as DNA polymerases, endonuclease, and DNA ligase, ultimately responsible for cell proliferation [34]. Therefore, PCNA is widely considered as a biomarker for cancer progression and prognosis. A recent study has found that PCNA was expressed in 52.2 % of gastric cancer patients, and positive expression of PCNA was significantly associated with poor 3-year disease-free survival (p = 0.035) [35]. KIAA0101 is a 15-kDa protein that has a conserved motif to bind to PCNA via a yeast two-hybrid system and thus involved in the regulation of DNA repair and cell proliferation [36]. Similar to PCNA, overexpression of KIAA0101 can promote growth and invasion of cancer cells [37] and predict poor prognosis in cancer patients [38, 39]. Collectively, these genes in the module 31 may play critical roles in the prognosis of ovarian cancer via regulation of DNA repair and cell proliferation.
In the module 35, 7 genes were included. Interferon regulatory factor 1 (IRF1) is a member of the interferon regulatory transcription factor (IRF) family, which can cause the inhibition of cell proliferation and stimulation of apoptosis [40]. IRF2 is a functional antagonist of IRF1 and may act as an oncogene, promoting the formation and progression of cancer [41]. A previous study has demonstrated that increased level of IRF1 is associated with both increased progression-free and overall survival of patients with ovarian carcinoma, and IRF1 is an independent predictor of platinum resistance and survival in high-grade serous ovarian carcinoma [42]. Furthermore, IRF1 directly mediates the interferon-γ (IFN-γ)-induced apoptosis via the activation of caspase-1 gene expression in IFN-γ-sensitive ovarian cancer cells [43]. However, in a recent study of ovarian cancer, IRF-1 was identified to be up-regulated in ovarian cancer samples compared with healthy ovarian tissue although strong expression of IRF-1 predicted improved disease-free survival and overall survival [44]. This finding may be attributed to a compensation or adaptation mechanism. Further study indicated the IRF1 seemed to play a key role in the transcriptional activation of interferon-inducible guanylate binding proteins (GBP1 and GBP2) [45], which subsequently induces T-lymphocyte immune response against the cancer cell spreading and proliferation [46]. Therefore, GBP1 and GBP2 may be also tumor suppressor genes and associated with better prognosis [47].
AIM2 is another human IFN-inducible protein, which forms the AIM2 inflammasome with an adaptor protein ASC upon sensing foreign cytoplasmic double-stranded DNA [48]. The activated AIM2 inflammasome in macrophages promotes the proteolytic cleavage and secretion of pro-inflammatory cytokines (IL-1β and IL-18) through the activation of caspase-1, leading to cell senescence, apoptosis and preventing cancer progression [49]. Thereby, AMI2 may be also correlated with excellent prognosis [50, 51].

Conclusion

Based on gene expression profiling data, two 7-gene functional interaction modules were identified to be likely associated with prognosis of ovarian cancer patients. These network modules were related to DNA repair, replication, immune and cytokine mediated signaling pathways. However, further experimental studies are required to confirm these genes in the modules.

Acknowledgements

None.

Funding

None.

Availability of data and material

The raw data were collected and analyzed by the Authors, and are not ready to share their data because the data have not been published.

Authors’ contributions

XW,SSW and LZ carried out all the operations,designed and applied the technique. And they had also critically reviewed the final draft of the manuscript. LY and LMZ collected the data,wrote the manuscript,conceived of the study,participated in its design and coordination and found and organized the literature. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):10–29.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):10–29.CrossRef
2.
Zurück zum Zitat Baldwin LA, Huang B, Miller RW, Tucker T, Goodrich ST, Podzielinski I, et al. Ten-year relative survival for epithelial ovarian cancer. Obstet Gynecol. 2012;120(3):612–8.CrossRefPubMed Baldwin LA, Huang B, Miller RW, Tucker T, Goodrich ST, Podzielinski I, et al. Ten-year relative survival for epithelial ovarian cancer. Obstet Gynecol. 2012;120(3):612–8.CrossRefPubMed
3.
Zurück zum Zitat Cheon DJ, Tong Y, Sim MS, Dering J, Berel D, Cui X, et al. A collagen-remodeling gene signature regulated by TGFβ signaling is associated with metastasis and poor survival in serous ovarian cancer. Clin Cancer Res. 2014;20(3):711–23.CrossRefPubMed Cheon DJ, Tong Y, Sim MS, Dering J, Berel D, Cui X, et al. A collagen-remodeling gene signature regulated by TGFβ signaling is associated with metastasis and poor survival in serous ovarian cancer. Clin Cancer Res. 2014;20(3):711–23.CrossRefPubMed
4.
Zurück zum Zitat Mclaughlin JR, Rosen B, Moody J, Pal T, Fan I, Shaw PA, et al. Long-term ovarian cancer survival associated with mutation in BRCA1 or BRCA2. J Natl Cancer Inst. 2013;105(2):141–8.CrossRefPubMedPubMedCentral Mclaughlin JR, Rosen B, Moody J, Pal T, Fan I, Shaw PA, et al. Long-term ovarian cancer survival associated with mutation in BRCA1 or BRCA2. J Natl Cancer Inst. 2013;105(2):141–8.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Yamanoi K, Matsumura N, Murphy SK, Baba T, Abiko K, Hamanishi J, et al. Suppression of ABHD2, identified through a functional genomics screen, causes anoikis resistance, chemoresistance and poor prognosis in ovarian cancer. Oncotarget. 2016 Yamanoi K, Matsumura N, Murphy SK, Baba T, Abiko K, Hamanishi J, et al. Suppression of ABHD2, identified through a functional genomics screen, causes anoikis resistance, chemoresistance and poor prognosis in ovarian cancer. Oncotarget. 2016
6.
Zurück zum Zitat Turcotte M, Spring K, Pommey S, Chouinard G, Cousineau I, George J, et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 2015;75(21):4494–503.CrossRefPubMed Turcotte M, Spring K, Pommey S, Chouinard G, Cousineau I, George J, et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 2015;75(21):4494–503.CrossRefPubMed
8.
9.
Zurück zum Zitat Vastrik I, D'Eustachio P, Schmidt E, Joshi-Tope G, Gopinath G, Croft D, et al. Reactome: a knowledge base of biologic pathways and processes. Genome Biol. 2007;8(3):R39.CrossRefPubMedPubMedCentral Vastrik I, D'Eustachio P, Schmidt E, Joshi-Tope G, Gopinath G, Croft D, et al. Reactome: a knowledge base of biologic pathways and processes. Genome Biol. 2007;8(3):R39.CrossRefPubMedPubMedCentral
10.
11.
Zurück zum Zitat Mi H, Dong Q, Muruganujan A, Gaudet P, Lewis S, Thomas PD. PANTHER version 7: improved phylogenetic trees, orthologs and collaboration with the Gene Ontology Consortium. Nucleic Acids Res. 2010;38 suppl 1:D204–10.CrossRefPubMed Mi H, Dong Q, Muruganujan A, Gaudet P, Lewis S, Thomas PD. PANTHER version 7: improved phylogenetic trees, orthologs and collaboration with the Gene Ontology Consortium. Nucleic Acids Res. 2010;38 suppl 1:D204–10.CrossRefPubMed
12.
Zurück zum Zitat Schaefer CF, Anthony K, Krupa S, Buchoff J, Day M, Hannay T, et al. PID: the pathway interaction database. Nucleic Acids Res. 2009;37 suppl 1:D674–9.CrossRefPubMed Schaefer CF, Anthony K, Krupa S, Buchoff J, Day M, Hannay T, et al. PID: the pathway interaction database. Nucleic Acids Res. 2009;37 suppl 1:D674–9.CrossRefPubMed
13.
Zurück zum Zitat Chatr-Aryamontri A, Breitkreutz B-J, Heinicke S, Boucher L, Winter A, Stark C, et al. The BioGRID interaction database: 2013 update. Nucleic Acids Res. 2013;41(D1):D816–23.CrossRefPubMed Chatr-Aryamontri A, Breitkreutz B-J, Heinicke S, Boucher L, Winter A, Stark C, et al. The BioGRID interaction database: 2013 update. Nucleic Acids Res. 2013;41(D1):D816–23.CrossRefPubMed
14.
Zurück zum Zitat Prasad TK, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, et al. Human protein reference database—2009 update. Nucleic Acids Res. 2009;37 suppl 1:D767–72.CrossRef Prasad TK, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, et al. Human protein reference database—2009 update. Nucleic Acids Res. 2009;37 suppl 1:D767–72.CrossRef
15.
Zurück zum Zitat Kerrien S, Alam-Faruque Y, Aranda B, Bancarz I, Bridge A, Derow C, et al. IntAct—open source resource for molecular interaction data. Nucleic Acids Res. 2007;35 suppl 1:D561–5.CrossRefPubMed Kerrien S, Alam-Faruque Y, Aranda B, Bancarz I, Bridge A, Derow C, et al. IntAct—open source resource for molecular interaction data. Nucleic Acids Res. 2007;35 suppl 1:D561–5.CrossRefPubMed
16.
Zurück zum Zitat Flicek P, Aken BL, Ballester B, Beal K, Bragin E, Brent S, et al. Ensembl’s 10th year. Nucleic Acids Res. 2010;38 suppl 1:D557–62.CrossRefPubMed Flicek P, Aken BL, Ballester B, Beal K, Bragin E, Brent S, et al. Ensembl’s 10th year. Nucleic Acids Res. 2010;38 suppl 1:D557–62.CrossRefPubMed
17.
Zurück zum Zitat Finn RD, Mistry J, Schuster-Böckler B, Griffiths-Jones S, Hollich V, Lassmann T, et al. Pfam: clans, web tools and services. Nucleic Acids Res. 2006;34 suppl 1:D247–51.CrossRefPubMed Finn RD, Mistry J, Schuster-Böckler B, Griffiths-Jones S, Hollich V, Lassmann T, et al. Pfam: clans, web tools and services. Nucleic Acids Res. 2006;34 suppl 1:D247–51.CrossRefPubMed
18.
Zurück zum Zitat Murakami Y, Mizuguchi K. Applying the Naïve Bayes classifier with kernel density estimation to the prediction of protein–protein interaction sites. Bioinformatics. 2010;26(15):1841–8.CrossRefPubMed Murakami Y, Mizuguchi K. Applying the Naïve Bayes classifier with kernel density estimation to the prediction of protein–protein interaction sites. Bioinformatics. 2010;26(15):1841–8.CrossRefPubMed
20.
Zurück zum Zitat Smoot ME, Ono K, Ruscheinski J, Wang P-L, Ideker T. Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics. 2011;27(3):431–2.CrossRefPubMed Smoot ME, Ono K, Ruscheinski J, Wang P-L, Ideker T. Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics. 2011;27(3):431–2.CrossRefPubMed
22.
Zurück zum Zitat Ahkter S, Richie CT, Zhang N, Behringer RR, Zhu C, Legerski RJ. Snm1-deficient mice exhibit accelerated tumorigenesis and susceptibility to infection. Mol Cell Biol. 2005;25(22):10071–8.CrossRefPubMedPubMedCentral Ahkter S, Richie CT, Zhang N, Behringer RR, Zhu C, Legerski RJ. Snm1-deficient mice exhibit accelerated tumorigenesis and susceptibility to infection. Mol Cell Biol. 2005;25(22):10071–8.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Herrera M, Dominguez G, Garcia JM, Peña C, Jimenez C, Silva J, et al. Differences in repair of DNA cross-links between lymphocytes and epithelial tumor cells from colon cancer patients measured in vitro with the comet assay. Clin Cancer Res. 2009;15(17):5466–72.CrossRefPubMed Herrera M, Dominguez G, Garcia JM, Peña C, Jimenez C, Silva J, et al. Differences in repair of DNA cross-links between lymphocytes and epithelial tumor cells from colon cancer patients measured in vitro with the comet assay. Clin Cancer Res. 2009;15(17):5466–72.CrossRefPubMed
24.
Zurück zum Zitat Kauffmann A, Rosselli F, Lazar V, Winnepenninckx V, Mansuet-Lupo A, Dessen P, et al. High expression of DNA repair pathways is associated with metastasis in melanoma patients. Oncogene. 2008;27(5):565–73.CrossRefPubMed Kauffmann A, Rosselli F, Lazar V, Winnepenninckx V, Mansuet-Lupo A, Dessen P, et al. High expression of DNA repair pathways is associated with metastasis in melanoma patients. Oncogene. 2008;27(5):565–73.CrossRefPubMed
26.
Zurück zum Zitat Abbotts R, Thompson N, Madhusudan S. DNA repair in cancer: emerging targets for personalized therapy. Cancer Manag Res. 2014;6:77.PubMedPubMedCentral Abbotts R, Thompson N, Madhusudan S. DNA repair in cancer: emerging targets for personalized therapy. Cancer Manag Res. 2014;6:77.PubMedPubMedCentral
27.
Zurück zum Zitat Wu S-H, Hsiao Y-T, Chen J-C, Lin J-H, Hsu S-C, Hsia T-C, et al. Bufalin alters gene expressions associated DNA damage, cell cycle, and apoptosis in human lung cancer NCI-H460 cells in vitro. Molecules. 2014;19(5):6047–57.CrossRefPubMed Wu S-H, Hsiao Y-T, Chen J-C, Lin J-H, Hsu S-C, Hsia T-C, et al. Bufalin alters gene expressions associated DNA damage, cell cycle, and apoptosis in human lung cancer NCI-H460 cells in vitro. Molecules. 2014;19(5):6047–57.CrossRefPubMed
28.
Zurück zum Zitat Ledermann JA, Gabra H, Jayson GC, Spanswick VJ, Rustin GJ, Jitlal M, et al. Inhibition of carboplatin-induced DNA interstrand cross-link repair by gemcitabine in patients receiving these drugs for platinum-resistant ovarian cancer. Clin Cancer Res. 2010;16(19):4899–905.CrossRefPubMedPubMedCentral Ledermann JA, Gabra H, Jayson GC, Spanswick VJ, Rustin GJ, Jitlal M, et al. Inhibition of carboplatin-induced DNA interstrand cross-link repair by gemcitabine in patients receiving these drugs for platinum-resistant ovarian cancer. Clin Cancer Res. 2010;16(19):4899–905.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Ioana M, Angelescu C, Burada F, Mixich F, Riza A, Dumitrescu T, et al. MMR gene expression pattern in sporadic colorectal cancer. J Gastrointestin Liver Dis. 2010;19(2):155–9.PubMed Ioana M, Angelescu C, Burada F, Mixich F, Riza A, Dumitrescu T, et al. MMR gene expression pattern in sporadic colorectal cancer. J Gastrointestin Liver Dis. 2010;19(2):155–9.PubMed
30.
Zurück zum Zitat Kretschmer C, Sterner-Kock A, Siedentopf F, Schoenegg W, Schlag PM, Kemmner W. Identification of early molecular markers for breast cancer. Mol Cancer. 2011;10(1):15.CrossRefPubMedPubMedCentral Kretschmer C, Sterner-Kock A, Siedentopf F, Schoenegg W, Schlag PM, Kemmner W. Identification of early molecular markers for breast cancer. Mol Cancer. 2011;10(1):15.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Zhou J, Wang Y, Yin X, He Y, Chen L, Wang W, et al. FOXM1 modulates cisplatin sensitivity by regulating EXO1 in ovarian cancer. PLoS One. 2014;9(5):e96989–9. Zhou J, Wang Y, Yin X, He Y, Chen L, Wang W, et al. FOXM1 modulates cisplatin sensitivity by regulating EXO1 in ovarian cancer. PLoS One. 2014;9(5):e96989–9.
32.
Zurück zum Zitat Tian X, Swenberg J, Nakamura J. POLD3 is required for DNA damage response to endogenous and exogenous DNA damage in human cells. Cancer Res. 2013;73(8 Supplement):1281–1. Tian X, Swenberg J, Nakamura J. POLD3 is required for DNA damage response to endogenous and exogenous DNA damage in human cells. Cancer Res. 2013;73(8 Supplement):1281–1.
33.
Zurück zum Zitat Elgaaen BV, Haug KBF, Wang J, Olstad OK, Fortunati D, Onsrud M, et al. POLD2 and KSP37 (FGFBP2) correlate strongly with histology, stage and outcome in ovarian carcinomas. 2010. Elgaaen BV, Haug KBF, Wang J, Olstad OK, Fortunati D, Onsrud M, et al. POLD2 and KSP37 (FGFBP2) correlate strongly with histology, stage and outcome in ovarian carcinomas. 2010.
34.
Zurück zum Zitat Essers J, Theil AF, Baldeyron C, van Cappellen WA, Houtsmuller AB, Kanaar R, et al. Nuclear dynamics of PCNA in DNA replication and repair. Mol Cell Biol. 2005;25(21):9350–9.CrossRefPubMedPubMedCentral Essers J, Theil AF, Baldeyron C, van Cappellen WA, Houtsmuller AB, Kanaar R, et al. Nuclear dynamics of PCNA in DNA replication and repair. Mol Cell Biol. 2005;25(21):9350–9.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Li N, Deng W, Ma J, Wei B, Guo K, Shen W, et al. Prognostic evaluation of Nanog, Oct4, Sox2, PCNA, Ki67 and E-cadherin expression in gastric cancer. Med Oncol. 2015;32(1):1–9.CrossRef Li N, Deng W, Ma J, Wei B, Guo K, Shen W, et al. Prognostic evaluation of Nanog, Oct4, Sox2, PCNA, Ki67 and E-cadherin expression in gastric cancer. Med Oncol. 2015;32(1):1–9.CrossRef
36.
Zurück zum Zitat Yu P, Huang B, Shen M, Lau C, Chan E, Michel J, et al. p15^ P^ A^ F, a novel PCNA associated factor with increased expression in tumor tissues. Oncogene. 2001;20(4):484–9.CrossRefPubMed Yu P, Huang B, Shen M, Lau C, Chan E, Michel J, et al. p15^ P^ A^ F, a novel PCNA associated factor with increased expression in tumor tissues. Oncogene. 2001;20(4):484–9.CrossRefPubMed
37.
Zurück zum Zitat Jain M, Zhang L, Patterson EE, Kebebew E. KIAA0101 is overexpressed, and promotes growth and invasion in adrenal cancer. PLoS One. 2011;6(11):e26866.CrossRefPubMedPubMedCentral Jain M, Zhang L, Patterson EE, Kebebew E. KIAA0101 is overexpressed, and promotes growth and invasion in adrenal cancer. PLoS One. 2011;6(11):e26866.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Kato T, Daigo Y, Aragaki M, Ishikawa K, Sato M, Kaji M. Overexpression of KIAA0101 predicts poor prognosis in primary lung cancer patients. Lung Cancer. 2012;75(1):110–8.CrossRefPubMed Kato T, Daigo Y, Aragaki M, Ishikawa K, Sato M, Kaji M. Overexpression of KIAA0101 predicts poor prognosis in primary lung cancer patients. Lung Cancer. 2012;75(1):110–8.CrossRefPubMed
39.
Zurück zum Zitat Su X, Zhang T, Cheng P, Zhu Y, Li H, Li D, et al. KIAA0101 mRNA overexpression in peripheral blood mononuclear cells acts as predictive marker for hepatic cancer. Tumor Biol. 2014;35(3):2681–6.CrossRef Su X, Zhang T, Cheng P, Zhu Y, Li H, Li D, et al. KIAA0101 mRNA overexpression in peripheral blood mononuclear cells acts as predictive marker for hepatic cancer. Tumor Biol. 2014;35(3):2681–6.CrossRef
40.
Zurück zum Zitat Bouker KB, Skaar TC, Riggins RB, Harburger DS, Fernandez DR, Zwart A, et al. Interferon regulatory factor-1 (IRF-1) exhibits tumor suppressor activities in breast cancer associated with caspase activation and induction of apoptosis. Carcinogenesis. 2005;26(9):1527–35.CrossRefPubMed Bouker KB, Skaar TC, Riggins RB, Harburger DS, Fernandez DR, Zwart A, et al. Interferon regulatory factor-1 (IRF-1) exhibits tumor suppressor activities in breast cancer associated with caspase activation and induction of apoptosis. Carcinogenesis. 2005;26(9):1527–35.CrossRefPubMed
41.
Zurück zum Zitat Cui L, Deng Y, Rong Y, Lou W, Mao Z, Feng Y, et al. IRF-2 is over-expressed in pancreatic cancer and promotes the growth of pancreatic cancer cells. Tumor Biol. 2012;33(1):247–55.CrossRef Cui L, Deng Y, Rong Y, Lou W, Mao Z, Feng Y, et al. IRF-2 is over-expressed in pancreatic cancer and promotes the growth of pancreatic cancer cells. Tumor Biol. 2012;33(1):247–55.CrossRef
42.
Zurück zum Zitat Cohen S, Mosig R, Moshier E, Pereira E, Rahaman J, Prasad-Hayes M, et al. Interferon regulatory factor 1 is an independent predictor of platinum resistance and survival in high-grade serous ovarian carcinoma. Gynecol Oncol. 2014;134(3):591–8.CrossRefPubMed Cohen S, Mosig R, Moshier E, Pereira E, Rahaman J, Prasad-Hayes M, et al. Interferon regulatory factor 1 is an independent predictor of platinum resistance and survival in high-grade serous ovarian carcinoma. Gynecol Oncol. 2014;134(3):591–8.CrossRefPubMed
43.
Zurück zum Zitat Kim EJ, Lee JM, Namkoong SE, Um SJ, Park JS. Interferon regulatory factor-1 mediates interferon-γ-induced apoptosis in ovarian carcinoma cells. J Cell Biochem. 2002;85(2):369–80.CrossRefPubMed Kim EJ, Lee JM, Namkoong SE, Um SJ, Park JS. Interferon regulatory factor-1 mediates interferon-γ-induced apoptosis in ovarian carcinoma cells. J Cell Biochem. 2002;85(2):369–80.CrossRefPubMed
44.
Zurück zum Zitat Zeimet AG, Reimer D, Wolf D, Fiegl H, Concin N, Wiedemair A, et al. Intratumoral interferon regulatory factor (IRF)-1 but not IRF-2 is of relevance in predicting patient outcome in ovarian cancer. Int J Cancer. 2009;124(10):2353–60.CrossRefPubMed Zeimet AG, Reimer D, Wolf D, Fiegl H, Concin N, Wiedemair A, et al. Intratumoral interferon regulatory factor (IRF)-1 but not IRF-2 is of relevance in predicting patient outcome in ovarian cancer. Int J Cancer. 2009;124(10):2353–60.CrossRefPubMed
45.
Zurück zum Zitat Ramsauer K, Farlik M, Zupkovitz G, Seiser C, Kröger A, Hauser H, et al. Distinct modes of action applied by transcription factors STAT1 and IRF1 to initiate transcription of the IFN-γ-inducible gbp2 gene. Proc Natl Acad Sci. 2007;104(8):2849–54.CrossRefPubMedPubMedCentral Ramsauer K, Farlik M, Zupkovitz G, Seiser C, Kröger A, Hauser H, et al. Distinct modes of action applied by transcription factors STAT1 and IRF1 to initiate transcription of the IFN-γ-inducible gbp2 gene. Proc Natl Acad Sci. 2007;104(8):2849–54.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Godoy P, Cadenas C, Hellwig B, Marchan R, Stewart J, Reif R, et al. Interferon-inducible guanylate binding protein (GBP2) is associated with better prognosis in breast cancer and indicates an efficient T cell response. Breast Cancer. 2014;21(4):491–9.CrossRefPubMed Godoy P, Cadenas C, Hellwig B, Marchan R, Stewart J, Reif R, et al. Interferon-inducible guanylate binding protein (GBP2) is associated with better prognosis in breast cancer and indicates an efficient T cell response. Breast Cancer. 2014;21(4):491–9.CrossRefPubMed
47.
Zurück zum Zitat Britzen-Laurent N, Lipnik K, Ocker M, Naschberger E, Schellerer VS, Croner RS, et al. GBP-1 acts as a tumor suppressor in colorectal cancer cells. Carcinogenesis. 2013;34(1):153–62.CrossRefPubMed Britzen-Laurent N, Lipnik K, Ocker M, Naschberger E, Schellerer VS, Croner RS, et al. GBP-1 acts as a tumor suppressor in colorectal cancer cells. Carcinogenesis. 2013;34(1):153–62.CrossRefPubMed
48.
Zurück zum Zitat Fernandesalnemri T, Yu JW, Datta P, Wu J, Alnemri ES. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature. 2009;458(7237):509–13.CrossRef Fernandesalnemri T, Yu JW, Datta P, Wu J, Alnemri ES. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature. 2009;458(7237):509–13.CrossRef
49.
Zurück zum Zitat Ponomareva L, Liu H, Duan X, Dickerson E, Shen H, Panchanathan R, et al. AIM2, an IFN-inducible cytosolic DNA sensor, in the development of benign prostate hyperplasia and prostate cancer. Mol Cancer Res. 2013;11(10):1193–202.CrossRefPubMed Ponomareva L, Liu H, Duan X, Dickerson E, Shen H, Panchanathan R, et al. AIM2, an IFN-inducible cytosolic DNA sensor, in the development of benign prostate hyperplasia and prostate cancer. Mol Cancer Res. 2013;11(10):1193–202.CrossRefPubMed
50.
Zurück zum Zitat Dihlmann S, Tao S, Echterdiek F, Herpel E, Jansen L, Chang-Claude J, et al. Lack of Absent in Melanoma 2 (AIM2) expression in tumor cells is closely associated with poor survival in colorectal cancer patients. Int J Cancer. 2014;135(10):2387–96.CrossRefPubMed Dihlmann S, Tao S, Echterdiek F, Herpel E, Jansen L, Chang-Claude J, et al. Lack of Absent in Melanoma 2 (AIM2) expression in tumor cells is closely associated with poor survival in colorectal cancer patients. Int J Cancer. 2014;135(10):2387–96.CrossRefPubMed
51.
Zurück zum Zitat Liu R, Truax AD, Chen L, Hu P, Li Z, Chen J, et al. Expression profile of innate immune receptors, NLRs and AIM2, in human colorectal cancer: correlation with cancer stages and inflammasome components. Oncotarget. 2015;6(32):33456–69. Liu R, Truax AD, Chen L, Hu P, Li Z, Chen J, et al. Expression profile of innate immune receptors, NLRs and AIM2, in human colorectal cancer: correlation with cancer stages and inflammasome components. Oncotarget. 2015;6(32):33456–69.
Metadaten
Titel
A network-pathway based module identification for predicting the prognosis of ovarian cancer patients
verfasst von
Xin Wang
Shan-shan Wang
Lin Zhou
Li Yu
Lan-mei Zhang
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Journal of Ovarian Research / Ausgabe 1/2016
Elektronische ISSN: 1757-2215
DOI
https://doi.org/10.1186/s13048-016-0285-0

Weitere Artikel der Ausgabe 1/2016

Journal of Ovarian Research 1/2016 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.