A panel of 7 prognosis-related long non-coding RNAs to improve platinum-based chemoresistance prediction in ovarian cancer

  • Authors:
    • Jing Song
    • Wanfeng Zhang
    • Sen Wang
    • Kun Liu
    • Fangzhou Song
    • Longke Ran
  • View Affiliations

  • Published online on: May 11, 2018     https://doi.org/10.3892/ijo.2018.4403
  • Pages: 866-876
  • Copyright: © Song et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In order to study the role of long non-coding RNAs (lncRNAs) in predicting platinum-based chemoresistance in patients with high-grade serous ovarian carcinoma (HGS-OvCa), a=7-lncRNA signature was developed by analyzing 561 microarrays and 136 specimens from RNA-sequencing (RNA-seq) obtained from online databases [odds ratio (OR), 2.859; P<0.0001]. The upregulated lncRNAs (RP11-126K1.6, ZBED3-AS1, RP11-439E19.10 and RP11‑348N5.7) and downregulated lncRNAs [RNF144A-AS1, growth arrest specific 5 (GAS5) and F11-AS1] exhibited high sensitivity and specificity in predicting chemoresistance in the Gene Expression Omnibus and the Cancer Genome Atlas (area under curve >0.8). The lncRNA signature was independent of clinical characteristics and 4 HGS-OvCa molecular subtypes. This signature was negatively associated with disease-free survival (n=47; log-rank, P<0.01). Furthermore, the expression of the 7 lncRNAs was consistent with microarray (GSE63885, GSE51373, GSE15372 and GSE9891) and RNA-seq data. In in vitro experiments, ZBED3-AS1, F11-AS1 and GAS5 were differentially expressed in cell lines that are known to be resistant and non-resistant to platinum-based drugs, which was consistent with the results in the present study. This lncRNA signature may be used as a prognostic marker for predicting resistance to platinum-based chemotherapeutics in HGS-OvCa. These findings may contribute to individualized therapies in patients with HGS-OvCa in the future.

Introduction

In the past 30 years, ovarian cancer has exhibited the highest mortality rate of all types of lethal gynecological cancer worldwide, with a 5-year survival rate of <30% (1,2). The characteristics of ovarian carcinoma encompass the development of chemoresistance (3), recurrence (4) and poor prognosis (5), which pose huge challenges for drug development and therapeutics (6). Once chemoresistance occurs, patients may not only experience adverse effects, but they may also eventually relapse (7). Although ~75% of patients initially respond to the platinum-based chemotherapy, the majority relapse with resistance, which leads to treatment failure and causes >90% of mortalities (8). To date, patients with ovarian cancer have been defined as treatment-resistant if the duration of disease-free survival (DFS) or the platinum-free interval was <6 months, while patients with longer durations have been defined as treatment-sensitive (3,9). However, this approach cannot provide guidance for individualized treatment of patients with drug resistance.

Long non-coding (lnc)RNA is a type of RNA that has no protein coding potential (10). However, lncRNAs have been reported to be critical in multiple stages of tumorigenesis and progression, including apoptosis, DNA damage response and metastasis (11,12).

lncRNAs may also be potential markers for the prediction of platinum-based chemoresistance (11,12). LncRNA ABHD11 antisense RNA 1 (tail to tail) was demonstrated to promote the proliferation, invasion and migration of ovarian cancer cells (13). LncRNA nuclear enriched abundant transcript 1 was reported to interact with BAI1-associated protein 1, which may contribute to the sensitivity against gemcitabine in cholangiocarcinoma (14). The role of lncRNAs in drug resistance of high-grade serous ovarian cancer (HGS-OvCa) remains to be fully investigated. Therefore, examining the possibilities of individualized treatment in patients with chemoresistance to platinum-based therapeutics and ovarian cancer from the perspective of lncRNAs may be useful to improve our understanding of drug resistance (15,16). The present study investigated the role of lncRNAs in platinum-based chemo-resistance and survival in patients with HGS-OvCa in order to contribute to current research on individualized therapies.

Materials and methods

Training and validation datasets

Raw human microarray expression profiles were retrieved and downloaded from Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih. gov/geo/). First of all, the key words 'high-grade serous ovarian cancer' and 'chemotherapeutic resistance' were used to search for samples. Secondly, as more lncRNAs could be detected using the Affymetrix Human Genome U133 Plus2.0 micro-array (Affynetrix; Thermo Fisher Scientific, Inc., Waltham, MA, USA), the samples not from that platform were filtered. Finally, the samples with data associated with chemotherapeutic response were selected. In total, 328 microarray profiles with corresponding clinical information [i.e. age, tumor grade, International Federation of Gynecology and Obstetrics (FIGO) stage (17), residual tumor size and platinum sensitivity] were downloaded. The accession numbers of GEO for the datasets were as follows: GSE63885 (18) (n=70); GSE51373 (19) (n=28) and GSE15372 (20) (n=10). Another dataset GSE9891 (21) (n=220) was used as a GEO validation dataset. The expression levels were normalized using the robust multi-array average (RMA) algorithm (22). Batch effects were removed using the Combat algorithm (23).

The expression profiles and clinical information of 233 patients with HGS-OvCa were downloaded from The Atlas of Noncoding RNAs in Cancer (24) and cBioportal (25) databases, respectively, as The Cancer Genome Atlas (TCGA) validation dataset. The expression levels were processed using the same methods as aforementioned. All datasets were analyzed to determine the differences in expression (Table I).

Table I

Patient characteristics in GEO and TCGA datasets.

Table I

Patient characteristics in GEO and TCGA datasets.

CharacteristicsGEO training datasetGEO validation datasetTCGA validation datasetP-value
Sample size, n108328233
Mean age (SD), years59.3 (8.3)60.4 (9.8)59.8 (11.4)0.714
Tumor grade, n (%)<0.001
 G29 (12.9)105 (32.0)32 (13.7)
 G346 (65.7)170 (51.8)197 (84.5)
 G415 (21.4)15 (4.6)0 (0)
 Unknown38 (35.2)38 (11.6)4 (1.7)
FIGO stage, n (%)0.073
 II8 (7.4)34 (10.4)11(4.7)
 III78 (72.2)255 (77.7)190 (81.5)
 IV12 (11.1)29 (8.8)32 (13.7)
 Unknown10 (9.3)10 (3.0)0 (0)
Residual tumor size, n (%)<0.001
 R0 (1-10 mm)14 (13.0)14 (4.3)116 (49.8)
 R1 (10-20 mm)35 (32.4)35 (10.7)15 (6.4)
 R2 (>20 mm)21 (19.4)21 (6.4)41 (17.6)
 Unknown38 (35.2)258 (78.7)61 (26.2)
Platinum sensitivity, n (%)0.021
 Sensitive59 (54.6)217 (66.2)163 (70.0)
 Resistance49 (45.4)111 (33.8)70 (30.0)
 Molecular subtypes, n (%)0.114
Differentiated28 (25.9)77 (23.5)66 (28.3)
 Immunoreactive26 (24.1)94 (28.6)44 (18.9)
 Mesenchymal18 (16.7)59 (18.0)56 (24.0)
 Proliferative36 (33.3)98 (29.9)67 (28.8)

[i] P-values for the difference among the GEO and TCGA cohorts were calculated using analysis of variance (for age) and the χ2 test (for tumor grade, FIGO stage, residual tumor size, platinum sensitivity, and molecular subtypes). GEO, Gene Expression Omnibus; TCGA, The Cancer Genome Atlas; SD, standard deviation; FIGO, International Federation of Gynecology and Obstetrics.

Integration and probe annotation of lncRNA datasets

LNCipeida (26) is a publicly available database for annotated human lncRNA transcript sequences and structures. The public repository produced by Du et al (27) provides a resource of clinically relevant lncRNAs for the development of lncRNA biomarkers and the identification of lncRNA therapeutic targets. The present study integrated the records of lncRNAs from GENCODE (28), LNCipedia, NONCODE (29), TCGA and the public repository by Du et al (27). In total, 24,372 lncRNAs were included in the present study. To avoid inaccurate annotations, the probes were cross-referenced by gene name and Ensembl transcript ID (release 89). For multiple probes of a single lncRNA, the median expression of the unique transcript ID was preserved. Finally, the expression profiles of 6,955 lncRNAs were obtained.

Clustering and differential expression analysis

The platinum-based sensitivity of patients with HGS-OvCa varies greatly, and the DFS ranged from 180 days to several thousand days. In this context, unsupervised hierarchical clustering based on Pearson distance for platinum-sensitive samples were performed using the ConcensusClusterPlus package in R (30).

The differential expression analysis was performed using R/Limma package (31). The common differentially expressed lncRNAs between each sensitive subgroup and resistant group were screened [differential expressed (DE) set A]. The differentially expressed lncRNAs between the complete sensitive group and the resistant group were obtained (DE set B); DE sets A and B were then merged.

Univariate and multivariate logistic regression analysis and survival analysis

To investigate the platinum-based chemoresistance-associated lncRNAs and the clinical characteristics of patients with HGS-OvCa, an independent univariate logistic regression was performed on 6,955 lncRNAs and clinical characteristics. The lncRNAs and clinical characteristics with P<0.05 were considered statistically significant and associated with chemoresistance. The survival package in R (32) was used to investigate DFS-associated lncRNAs. The Kaplan-Meier survival curves of highly expressed and weakly expressed lncRNAs were constructed. The median expression level was used as a cut-off for determining high and low expression.

The lncRNAs and clinical characteristics that were statistically significant in the univariate logistic regression were used as candidate parameters for constructing the multivariate logistic regression model. A best multivariate logistic regression model was generated after fitting the model thousands of times. The expression levels of each lncRNA and its coefficient in the best multivariate logistic model were used to calculate the platinum-based chemoresistance risk score of patients with HGSC-OvCa. The risk score was computed by the following formula:

where n is the number of prognostic lncRNAs in the model, 'expri' is the expression level of lncRNAi and coefi is the estimated regression coefficient of lncRNAi in the multivariate logistic regression model. Patients who have higher risk scores are expected to have higher probability of platinum-based chemoresistance. The median risk score was used as a cut-off to divide the patients in the GEO validation dataset into low-risk and high-risk groups. Furthermore, the multivariate logistic regression analysis was performed to test whether the risk score was independent of clinical covariates.
Subtype classification of patients with HGS-OvCa

The 100-gene set containing four gene signatures corresponding to four HGS-OvCa subtypes (differentiated, immunoreactive, proliferative and mesenchymal) (33) were used to train the support vector machine for subtype classification. The univariate and multivariate logistic regression were performed to investigate whether the HGS-OvCa subtypes were associated with platinum-based chemoresistance, and whether these subtypes associated with our signature, respectively.

Classifier performance evaluation

To assess the sensitivity and specificity of the risk scores in prediction of chemoresistance in patients with HGS-OvCa, ROC (34) and ROCR package (35) in R software were used to construct the receiver operating characteristic curve and perform the area under curve (AUC) analysis, respectively. Furthermore, the predictive performance of the risk score and three signatures previously developed by Zhou et al (36), Liu et al (37) and TCGA mRNA signatures (38) was compared.

Verification of the expression level of 7 lncRNAs

Microarrays and RNA-seq data were used to validate the expression of lncRNAs. The RNA-seq data of 136 patients with HGS-OvCa and platinum-based chemosensitivity was derived from the Gaucher Disease Advisory Committee (39). The reads per kilobase per million mapped reads were obtained by chromosome location (GRCH37, hg19) of lncRNAs and normalized using Z-scores.

First, the gene expression in the Cancer Cell Line Encyclopedia (CCLE) (40) database had been pre-processed using the RMA algorithm. The chemotherapeutic status of 52 ovarian cancer cell lines was investigated. The data regarding the cell lines of clear cell, granulosa cell, undifferentiated cell, mixed cell, endometrioid cell, Brenner cell, mucinous cell and cross-contaminated cell were filtered. The cell lines that were treated with cisplatin or carboplatin under-went >3 cycles of treatment. The cell lines exhibited resistance to platinum and the degree of resistance varied. Secondly, gene expression levels in 46 ovarian cancer cell lines with or without cisplatin treatment were obtained from GSE47856 (41). To investigate the changes in the expression of lncRNAs that was induced by platinum, cell lines from the CCLE were analyzed using the Integrative Genomics Viewer (IGV) software (version 2.3) (42). In addition, cell lines from GSE47856 with ≥3 replicates were analyzed. For multi-probe lncRNA, the median expression was calculated.

Functional prediction analysis

To investigate the biological processes and cellular components implicated in the drug resistance mechanisms, the enriched Gene Ontology (GO) (43) terms of 7 lncRNAs were identified using the NONCODE data-base and were visualized using Cytoscape (version 3.2.1) (44). The potential functions of the lncRNAs were further analyzed using non-coding RNA in the Drug Resistance (ncDR) data-base (45); P<0.05 was used to identify resistant compounds of lncRNAs in cancer.

Statistical analysis

The empirical Bayes method and a two-tailed Student's t-test were used in differential expression analysis. The log-rank test was used for survival analysis, with Kaplan-Meier curves. Clinical characteristics and molecular subtypes of three datasets were tested using the χ2 tests or two-way analysis of variance, where appropriate. In multivariate logistic regression, the χ2 test was used to add or filter features and avoid overdispersion. The features were also filtered using the methods of Watt et al (46). P<0.05 was considered to indicate a statistically significant difference. All statistical analyses were conducted using the R software (version 3.2.1).

Results

Cluster analysis of chemosensitive patients reveals three subgroups

The results of unsupervised hierarchical clustering were obtained when clustering center k was set from 2 to 5 (Fig. 1). The specimens were perfectly divided into two clusters (Fig. 1A); a third cluster was observed when k=3 (Fig. 1B). However, fourth stable clusters were not found when the number of cluster centers k continued to increase (Fig. 1C and D). These results indicate that there may be three stable subgroups with different expression patterns in chemosensitive patients.

Identification and assessment of chemoresistance-associated lncRNA signature

In the present study, 386 differential expressed lncRNAs were obtained subsequent to merging 262 lncRNAs in DE set A and 365 lncRNAs in DE set B (Fig. 2A). In univariate regression, 337 lncRNAs (288 plus 43 plus 7 minus 1) were associated with platinum-based chemoresistance after merging the lncRNA transcripts corresponding to the same lncRNA gene, 50 lncRNAs (43 plus 7) (Fig. 2B) were associated with DFS (log-rank P<0.05). Next, a 7-lncRNA signature was identified (Table II and Fig. 2C), which was associated with platinum-based chemoresistance in the univariate and multivariate logistic regression model. The risk scores based on the 7-lncRNA signature for the prediction of platinum-based chemoresistance in patients with HGS-OvCa were also calculated. According to the risk scores, patients in the GEO training dataset were divided into low-score and high-score groups using the median risk score as a cut-off in the multivariate logistic regression model (Fig. 2D). The high-score group exhibited a higher probability of drug resistance [univariate model: Odds ratio (OR), 2.675; 95% confidence interval (CI), 1.841-3.897; P=6.36×10−7; multivariate model: OR, 2.859; 95% CI, 1.801-5.257; P=9.36×10−5). The results of Kaplan-Meier curves also indicated that the high risk score was significantly associated with the poor survival of patients with HGS-OvCa and drug resistance (n=47; log-rank, P<0.05; Fig. 2E).

Table II

Univarate and multivariate logistic regression model of 7 long non-coding RNAs in the Gene Expression Omnibus training dataset.

Table II

Univarate and multivariate logistic regression model of 7 long non-coding RNAs in the Gene Expression Omnibus training dataset.

Transcript IDGene nameChromosome in GRCh38 (strand)Univariate regression
Multivariate regression
OR (95% CI)P-valueOR (95% CI)P-value
ENST00000426122RNF144A-AS1 chr2:6913627-6918667 (−)0.26 (0.080-0.797)0.0220.088 (0.017- 0.388)0.026
ENST00000434796GAS5 chr1:173863980-173864901 (−)0.476 (0.221-0.914)0.0410.1898 (0.062- 0.521)0.021
ENST00000455503RP11-126K1.6 chr1:151346967-151348027 (+)2.297 (1.177-4.740)0.0183.1549 (1.224- 9.360)0.025
ENST00000508110F11-AS1 chr4:186423728-186500997 (−)0.039 (0.003-0.472)0.0140.009101 (0.0003- 0.208)0.005
ENST00000515419ZBED3-AS1 chr5:77086798-77147745 (+)3.842 (1.173-14.310)0.0345.624 (1.097- 30.824)0.003
ENST00000567832RP11-439E19.10 chr1:246772301-246775772 (+)43.995 (5.072-49.635)0.00136.666 (2.372- 826.504)0.014
ENST00000603915RP11-348N5.7 chr10:110910596-110912244 (+)4.658 (1.068-22.672)0.0411.194 (1.470-107.695)0.002

[i] OR, odds ratio; 95% CI, 95% confidence interval of OR.

Risk score can be used as an independent predictor for drug resistance

In the multivariate logistic regression model of the training datasets, platinum-based chemosensitivity was a dependent variable. Platinum-based chemosensitivity has multiple covariates, including tumor stage, grade, tumor size, molecular subtypes and risk score. However, the results indi-cated that the risk score was an independent variable when adjusting using the covariates as aforementioned (OR, 2.859; 95% CI, 1.801-5.257; P=9.36×10−5; Table III).

Table III

Univariate and multivariate logistic regression models in GEO and TCGA datasets.

Table III

Univariate and multivariate logistic regression models in GEO and TCGA datasets.

VariableTraining set (GSE63885, GSE15372, GSE51373; n=108)
GEO validation set (GSE9891; n=220)
TCGA validation set (n=23)
Univariate regression
Multivariable regression
Univariate regression
Multivariable regression
Univariate regression
Multivariable regression
P-valueOR (95% CI)P-valueOR (95% CI)P-valueOR (95% CI)P-valueOR (95% CI)P-valueOR (95% CI)P-valueOR (95% CI)
IncRNA signature (high/low) 6.36×10−7a2.675
(1.841-3.897)
9.36×10−5a2.859
(1.801-5.257)
4.68×10−14a4.190
(2.974-4.685)
1.15×10−13a4.21
(2.967-6.366)
2.33×10−5a2.315
(1.852-2.956)
2.65×10−5a2.514
(1.817-3.354)
Age (mean, 62 years)0.3590.978
(0.930-1.026)
0.991.001
(0.929-1.081)
0.9620.999
(0.964-1.035)
0.9610.999
(0.947-1.053)
0.281.014
(0.989-1.039)
0.3211.014
(0.986-1.045)
FI GO stage (ref=11)
 III0.9927.572
(3.361-21.245)
0.9933.438
(1.625-5.218)
0.9671.032
(0.245-5.164)
0.6951.481
(0.219-1.204)
4.95×10−3a7.077
(1.964-33.252)
0.196.254
(1.443-34.206)
 IV0.9925.111
(1.252-20.579)
0.9931.615
(0.354-3.548)
0.7941.250
(0.239-7.412)
0.2623.869
(0.381-4.602)
0.023a5.867
(1.381-31.421)
0.266.38
(1.213-41.476)
Tumor grade (ref=G2)
 G30.2070.341
(0.047-1.589)
0.4860.429
(0.313-4.219)
0.4892.192
(0.311-4.656)
0.22811.131
(0.469-65.332)
0.2640.259
(0.012-2.274)
0.3670.357
(0.013-3.258)
 G40.0840.192
(0.023-1.111)
0.1140.105
(0.048-1.509)
0.1476.011
(0.677-9.967)
0.05660.499
(1.707-73.757)
0.9680.954
(0.047-7.642)
0.7261.555
(0.068-14.942)
Residual tumor size (ref=R0, 1-10 mm)
R1 (11-20 mm)0.2070.424
(0.101-1.533)
0.1550.225
(0.035-1.543)
0.2012.361
(0.652-9.946)
0.1014.071
(0.819-25.049)
0.9821.016
(0.249-4.152)
0.5581.598
(0.331-7.899)
 R2 (>20 mm)0.1690.364
(0.079-1.472)
0.2420.282
(0.029-2.182)
0.1922.751
(0.679-12.741)
0.0854.932
(0.869-35.164)
0.4451.136
(0.359-3.559)
0.9421.049
(0.282-3.835)
Histology subtypes (ref, differentiated)
 Immunoreactive0.2210.407
(0.089-1.642)
0.4042.81
(0.249-3.531)
0.4331.389
(0.614-3.201)
0.8370.885
(0.274-2.860)
0.4091.469
(0.601-3.791)
0.3771.549
(0.597-4.221)
 Proliferative0.04a0.221
(0.047-0.889)
0.8551.26
(0.106-1.644)
0.02a2.428
(1.134-5.385)
0.3881.651
(0.533-5.269)
0.7920.905
(0.428-1.904)
0.7740.885
(0.382-2.041)
 Mesenchymal0.1620.333
(0.066-1.505)
0.631.793
(0.171-2.157)
0.2411.681
(0.707-4.058)
0.5851.397
(0.422-4.725)
0.5840.802
(0.362-1.781)
0.9640.98
(0.414-2.336)

a P<0.05. OR, odds ratio; 95 CI%, 95% confidence interval; GEO, Gene Expression Omnibus; TCGA, The Cancer Genome Atlas; lncRNA, long non-coding RNA.

The results of the univariate and multivariate logistic regression analysis for the GEO validation set (n=220) and the TCGA validation set (n=233) confirmed that the risk score of the training set was associated with platinum-based chemore-sistance of patients with HGS-OvCa (univariate model of the GEO dataset: OR, 4.190; 95% CI, 2.974-4.685; P<4.68×10−14; multivariate model of the GEO dataset: OR, 4.210; 95% CI, 2.967-6.366; P<1.15×10−13; univariate model of TCGA dataset: OR, 2.315; 95% CI, 1.852-2.956; P=2.33×10−5; and multivariate model of TCGA dataset: OR, 2.514; 95% CI, 1.817-3.354; P=2.65×10−5; Table III). Furthermore, clinical characteristics and HGS-OvCa subtypes (differentiated, immunoreactive, proliferative and mesenchymal) were not associated with platinum-based resistance in two validation datasets. Collectively, these results suggest that risk score has the potential to be an independent predictor for chemoresistance of HGS-OvCa cancer.

Predictive lncRNA signature performance evaluation

The risk score of the 7-lncRNA signature indicated a high sensitivity and specificity in predicting platinum-based chemoresistance in the GEO training (AUC=0.848), GEO validation (AUC=0.901) and TCGA validation (AUC=0.818) datasets (Fig. 3A). Compared with the lncRNA signature of the study by Liu et al (37), the lncRNA signature developed in the study by Zhou et al (36) and the TCGA mRNA signature, the 7-lncRNA signature exhibited an improved predictive performance (Fig. 3B–D).

Verification of the expression level of 7 lncRNAs in microarray and RNA-seq data

A total of 7 lncRNAs were observed in the GEO validation dataset, while F11-AS1 and RP11-348N5.7 were not observed in the TCGA validation dataset and RNA-seq dataset, respectively. However, all of the 7 lncRNAs were covered by validation datasets. Notably, the expression levels of 7 lncRNAs were concordant in at least two validation datasets (P<0.05; Fig. 3E).

Predicted function of the 7 lncRNAs may be associated with drug resistance mechanisms

With several cycles of cisplatin or carboplatin treatment, ZBED1-AS1 was upregulated, while F11-AS1 was downregulated by platinum in OV56 and OVKATE cell lines (P<0.05; Fig. 3F). Growth arrest specific 5 (GAS5) was downregulated by cisplatin in A2008, DOV13B and HeyA8 cell lines (P<0.05; Fig. 3G). This indicates that these lncRNAs may be involved in drug resistance mechanisms. Therefore, the functional prediction analysis of 7 lncRNAs based on NONCODE database was further performed. The results showed that a total of 42 GO terms were involved in the biological functions of 7 lncRNAs, which were distributed in 'biological processes' (91.3%) and 'cellular components' (8.7%). Furthermore, 16 of the 42 GO terms was associated with >1 lncRNA (Fig. 4). Based on ncDR, the aberrant regulation of lncRNAs was predicted to be resistant to dozens of compounds across various cancer types. For example, the upregulation of ZBED3-AS1 is associated with resistance to cisplatin, dasatinib and bicalutamide in low-grade glioma. The downregulation of RNF144A-AS1 is associated with resistance to dacinostat in sarcoma, and the downregulation of GAS5 is associated with resistance to cisplatin and cytarabine in pancreatic adenocarcinoma.

Discussion

The compilation of multiple datasets offers a unique opportunity to discover transcriptional variation in HGS-OvCa. This type of combinatorial computational analysis must take the context of numerous confounding factors, including distinct platforms, different protocols of sample collection and processing, and different normalization algorithms of expression level, which may not associate with tumor biology or consider cancer behavior (45). Therefore, in the present study, the independent analysis of multiple datasets was performed, and batch effect was removed. Furthermore, the verification of the expression levels of the identified lncRNAs was conducted using multiple datasets and cell lines. The results from multiple datasets were concordant and verified that the 4 upregulated lncRNAs (RP11-126K1.6, ZBED3-AS1, RP11-439E19.10 and RP11-348N5.7) and 3 downregulated lncRNAs (RNF144A-AS1, GAS5 and F11-AS1) were differentially expressed in response to platinum. In addition, not only the 7-lncRNA signature, but also the individual lncRNAs themselves were associated with the prognosis of patients with HGS-OvCa.

The molecular mechanisms of drug resistance in ovarian cancer are mainly involved in apoptosis evasion, escape from immune surveillance and natural killer cell-mediated oncolysis, promotion of DNA repair, and sustained angiogenesis, proliferation, invasion and metastasis (6,47,48). To date, the majority of the lncRNAs reported have not been functionally characterized in drug-resistant ovarian cancer.

GAS5 has been reported as a biomarker of drug resistance in non-small cell lung cancer (49) and breast cancer (50). The downregulation of GAS5 promotes the proliferation, migration and invasion of ovarian cancer cells, and inhibits apoptosis, which leads to a poor prognosis (51,52). From the results of functional prediction in the present study, the downregulation of RNF144A-AS1 and GAS5, and the upregulation of RP11-126K1.6, RP11-439E19.10 and RP11-348N5.7 in patients with ovarian cancer may contribute to platinum resistance by affecting the regulation of tumor growth. RNF144A-AS1, F11-AS1, ZBED3-AS1, RP11-439E19.10 and RP11-348N5.7 were predicted to participate in the acute-phase response. The acute-phase response serves as a core of the innate immune response, which can be triggered by inflammation (53). It has been confirmed that inflammation and immunogenic-tumor microenvironment interactions will increase the risk of ovarian tumor initiation and progression by overexpression of several proinflammatory cytokines (54,55). For example, the complex formed by the binding of the proinflammatory cytokine inter-leukin-6 (IL-6) to its receptor IL-6R activates Janus kinase and various downstream effectors, including signal transducer and activator of transcription 3 and mitogen-activated protein kinase, which are critical for cell proliferation, apoptosis evasion, survival and drug resistance in ovarian cancer (6,56,57). Taken together, the present results indicated that the 5 lncRNAs (RNF144A-AS1, F11-AS1, ZBED3-AS1, RP11-439E19.10 and RP11-348N5.7) may interact with proinflammatory cytokines to promote ovarian tumor initiation and progression. Additionally, the ncDR database also indicated that the identified lncRNAs were involved in resistance to platinum-based chemotherapeu-tics across various cancer types.

The present study has a number of limitations. Once multiple-transcript probes have been identified as candidate features of the multivariate logistic regression model, multi-collinearity could be a concern for the accuracy of predictors. Multicollinearity does not reduce the predictive reliability of the entire signature. In the present study, there was no collinearity between other lncRNAs and the 7 candidate lncRNAs. In addition, lncRNA records vary in well-known biological databases. Although in the present study the lncRNAs records from multiple databases were re-integrated for lncRNA identification, the omission of identification of lncRNAs may still occur.

In the present study, a 7-lncRNA signature for predicting platinum-based chemoresistance in patients with HGS-OvCa was developed. The signature is independent of clinical characteristics and molecular subtypes of HGS-OvCa, and can be used as a novel prognostic marker, with the potential to be a therapeutic molecular target in the future. Further clinical studies are required for validating the lncRNAs identified in the present study and for investigating the underlying mechanisms of drug resistance in HGS-OvCa.

Acknowledgments

Not applicable.

Funding

The present study was supported by a grant from the Chongqing Science and Technology Foundation of Yuzhong District (no. 20150107).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

JS designed the study, analyzed and interpreted the data, and was a major contributor in writing the manuscript. WZ made critical advices to the conception and design of the study. SW, KL and FS contributed to data collection. LR designed the study, supervised the project and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no conflict of interests.

References

1 

Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T and Thun MJ: Cancer statistics, 2008. CA Cancer J Clin. 58:71–96. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Bast RC Jr, Hennessy B and Mills GB: The biology of ovarian cancer: New opportunities for translation. Nat Rev Cancer. 9:415–428. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Alvero AB, Chen R, Fu HH, Montagna M, Schwartz PE, Rutherford T, Silasi DA, Steffensen KD, Waldstrom M, Visintin I, et al: Molecular phenotyping of human ovarian cancer stem cells unravels the mechanisms for repair and chemoresistance. Cell Cycle. 8:158–166. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Fotopoulou C, Richter R, Braicu IE, Schmidt SC, Neuhaus P, Lichtenegger W and Sehouli J: Clinical outcome of tertiary surgical cytoreduction in patients with recurrent epithelial ovarian cancer. Ann Surg Oncol. 18:49–57. 2011. View Article : Google Scholar

5 

Husseinzadeh N: Status of tumor markers in epithelial ovarian cancer has there been any progress? A review Gynecol Oncol. 120:152–157. 2011. View Article : Google Scholar

6 

Hiss D: Optimizing molecular-targeted therapies in ovarian cancer: The renewed surge of interest in ovarian cancer biomarkers and cell signaling pathways. J Oncol. 2012:7379812012. View Article : Google Scholar : PubMed/NCBI

7 

Kaye SB, Colombo N, Monk BJ, Tjulandin S, Kong B, Roy M, Chan S, Filipczyk-Cisarz E, Hagberg H, Vergote I, et al: Trabectedin plus pegylated liposomal doxorubicin in relapsed ovarian cancer delays third-line chemotherapy and prolongs the platinum-free interval. Ann Oncol. 22:49–58. 2011. View Article : Google Scholar

8 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, Yan PS, Huang TH and Nephew KP: Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 68:4311–4320. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Lipovich L, Johnson R and Lin CY: MacroRNA underdogs in a microRNA world: Evolutionary, regulatory, and biomedical significance of mammalian long non-protein-coding RNA. Biochim Biophys Acta. 1799:597–615. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Wang H, Huo X, Yang XR, He J, Cheng L, Wang N, Deng X, Jin H, Wang N, Wang C, et al: STAT3-mediated upregulation of lncRNA HOXD-AS1 as a ceRNA facilitates liver cancer metastasis by regulating SOX4. Mol Cancer. 16:1362017. View Article : Google Scholar : PubMed/NCBI

12 

Cui Z, Ren S, Lu J, Wang F, Xu W, Sun Y, Wei M, Chen J, Gao X, Xu C, et al: The prostate cancer-up-regulated long noncoding RNA PlncRNA-1 modulates apoptosis and proliferation through reciprocal regulation of androgen receptor. Urol Oncol. 31:1117–1123. 2013. View Article : Google Scholar

13 

Wu DD, Chen X, Sun KX, Wang LL, Chen S and Zhao Y: Role of the lncRNA ABHD11-AS1 in the tumorigenesis and progression of epithelial ovarian cancer through targeted regulation of RhoC. Mol Cancer. 16:1382017. View Article : Google Scholar :

14 

Parasramka M, Yan IK, Wang X, Nguyen P, Matsuda A, Maji S, Foye C, Asmann Y and Patel T: BAP1 dependent expression of long non-coding RNA NEAT-1 contributes to sensitivity to gemcitabine in cholangiocarcinoma. Mol Cancer. 16:222017. View Article : Google Scholar : PubMed/NCBI

15 

Yap TA, Carden CP and Kaye SB: Beyond chemotherapy: Targeted therapies in ovarian cancer. Nat Rev Cancer. 9:167–181. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Rowan K: Intraperitoneal therapy for ovarian cancer: Why has it not become standard? J Natl Cancer Inst. 101:775–777. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Benedet JL, Bender H, Jones H III, Ngan HY and Pecorelli S: FIGO staging classifications and clinical practice guidelines in the management of gynecologic cancers. Int J Gynaecol Obstet. 70:209–262. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Lisowska KM, Olbryt M, Dudaladava V, Pamuła-Piłat J, Kujawa K, Grzybowska E, Jarząb M, Student S, Rzepecka IK, Jarząb B, et al: Gene expression analysis in ovarian cancer - faults and hints from DNA microarray study. Front Oncol. 4:62014. View Article : Google Scholar : PubMed/NCBI

19 

Koti M, Gooding RJ, Nuin P, Haslehurst A, Crane C, Weberpals J, Childs T, Bryson P, Dharsee M, Evans K, et al: Identification of the IGF1/PI3K/NF κB/ERK gene signalling networks associated with chemotherapy resistance and treatment response in high-grade serous epithelial ovarian cancer. BMC Cancer. 13:5492013. View Article : Google Scholar

20 

Li M, Balch C, Montgomery JS, Jeong M, Chung JH, Yan P, Huang TH, Kim S and Nephew KP: Integrated analysis of DNA methylation and gene expression reveals specific signaling pathways associated with platinum resistance in ovarian cancer. BMC Med Genomics. 2:342009. View Article : Google Scholar : PubMed/NCBI

21 

Tothill RW, Tinker AV, George J, Brown R, Fox SB, Lade S, Johnson DS, Trivett MK, Etemadmoghadam D, Locandro B, et al: Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin Cancer Res. 14:5198–5208. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Sugio A, Yang B, Zhu T and White FF: Two type III effector genes of Xanthomonas oryzae pv. oryzae control the induction of the host genes OsTFIIAgamma1 and OsTFX1 during bacterial blight of rice. Proc Natl Acad Sci USA. 104:10720–10725. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Miller JA, Cai C, Langfelder P, Geschwind DH, Kurian SM, Salomon DR and Horvath S: Strategies for aggregating gene expression data: The collapseRows R function. BMC Bioinformatics. 12:3222011. View Article : Google Scholar : PubMed/NCBI

24 

Li J, Han L, Roebuck P, Diao L, Liu L, Yuan Y, Weinstein JN and Liang H: TANRIC: An interactive open platform to explore the function of lncRNAs in cancer. Cancer Res. 75:3728–3737. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, et al: Integrative analysis of complex cancer genomics and clinical profiles using the cBio-Portal. Sci Signal. 6:pl12013. View Article : Google Scholar

26 

Volders PJ, Helsens K, Wang X, Menten B, Martens L, Gevaert K, Vandesompele J and Mestdagh P: LNCipedia: A database for annotated human lncRNA transcript sequences and structures. Nucleic Acids Res. 41:D246–D251. 2013. View Article : Google Scholar :

27 

Du Z, Fei T, Verhaak RG, Su Z, Zhang Y, Brown M, Chen Y and Liu XS: Integrative genomic analyses reveal clinically relevant long noncoding RNAs in human cancer. Nat Struct Mol Biol. 20:908–913. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Harrow J, Frankish A, Gonzalez JM, Tapanari E, Diekhans M, Kokocinski F, Aken BL, Barrell D, Zadissa A, Searle S, et al: GENCODE: The reference human genome annotation for The ENCODE Project. Genome Res. 22:1760–1774. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Liu C, Bai B, Skogerbø G, Cai L, Deng W, Zhang Y, Bu D, Zhao Y and Chen R: NONCODE: An integrated knowledge database of non-coding RNAs. Nucleic Acids Res. 33:D112–D115. 2005. View Article : Google Scholar :

30 

Wilkerson MD and Hayes DN: ConsensusClusterPlus: A class discovery tool with confidence assessments and item tracking. Bioinformatics. 26:1572–1573. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 3:1–25. 2004. View Article : Google Scholar

32 

Hakulinen T and Abeywickrama KH: A computer program package for relative survival analysis. Comput Programs Biomed. 19:197–207. 1985. View Article : Google Scholar : PubMed/NCBI

33 

Verhaak RG, Tamayo P, Yang JY, Hubbard D, Zhang H, Creighton CJ, Fereday S, Lawrence M, Carter SL, Mermel CH, et al Cancer Genome Atlas Research Network: Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J Clin Invest. 123:517–525. 2013.

34 

Duda RO, Hart PE and Stork DG: Pattern Classification. China Machine Press; 2012

35 

Sing T, Beerenwinkel N and Lengauer T: Learning mixtures of localized rules by maximizing the area under the ROC curve. In: ROCAI-2004. Hernández-Orallo José: 1st International Workshop on ROC Analysis in Artificial Intelligence; Valencia. pp. 89–96. 2004

36 

Zhou M, Sun Y, Sun Y, Xu W, Zhang Z, Zhao H, Zhong Z and Sun J: Comprehensive analysis of lncRNA expression profiles reveals a novel lncRNA signature to discriminate nonequivalent outcomes in patients with ovarian cancer. Oncotarget. 7:32433–32448. 2016.PubMed/NCBI

37 

Liu R, Zeng Y, Zhou CF, Wang Y, Li X, Liu ZQ, Chen XP, Zhang W and Zhou HH: Long noncoding RNA expression signature to predict platinum-based chemotherapeutic sensitivity of ovarian cancer patients. Sci Rep. 7:182017. View Article : Google Scholar : PubMed/NCBI

38 

Network TCGA; Cancer Genome Atlas Research Network: Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Tomczak K, Czerwińska P and Wiznerowicz M: The Cancer Genome Atlas (TCGA): An immeasurable source of knowledge. Contemp Oncol (Pozn). 19:A68–A77. 2015.

40 

Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, Wilson CJ, Lehár J, Kryukov GV, Sonkin D, et al: The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 483:603–607. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Miow QH, Tan TZ, Ye J, Lau JA, Yokomizo T, Thiery JP and Mori S: Epithelial-mesenchymal status renders differential responses to cisplatin in ovarian cancer. Oncogene. 34:1899–1907. 2015. View Article : Google Scholar

42 

Thorvaldsdóttir H, Robinson JT and Mesirov JP: Integrative Genomics Viewer (IGV): High-performance genomics data visualization and exploration. Brief Bioinform. 14:178–192. 2013. View Article : Google Scholar :

43 

Consortium GO: The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res. 32:D258–D261. 2004. View Article : Google Scholar :

44 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Brannon AR, Haake SM, Hacker KE, Pruthi RS, Wallen EM, Nielsen ME and Rathmell WK: Meta-analysis of clear cell renal cell carcinoma gene expression defines a variant subgroup and identifies gender influences on tumor biology. Eur Urol. 61:258–268. 2012. View Article : Google Scholar

46 

Watt JP, O'Brien KL, Benin AL, McCoy SI, Donaldson CM, Reid R, Schuchat A, Zell ER, Hochman M, Santosham M, et al: Risk factors for invasive pneumococcal disease among Navajo adults. Am J Epidemiol. 166:1080–1087. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Dhillon KK, Swisher EM and Taniguchi T: Secondary mutations of BRCA1/2 and drug resistance. Cancer Sci. 102:663–669. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Sakai W, Swisher EM, Karlan BY, Agarwal MK, Higgins J, Friedman C, Villegas E, Jacquemont C, Farrugia DJ, Couch FJ, et al: Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature. 451:1116–1120. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Zhang N, Yang GQ, Shao XM and Wei L: GAS5 modulated autophagy is a mechanism modulating cisplatin sensitivity in NSCLC cells. Eur Rev Med Pharmacol Sci. 20:2271–2277. 2016.PubMed/NCBI

50 

Li J, Huang H, Li Y, Li L, Hou W and You Z: Decreased expression of long non-coding RNA GAS5 promotes cell proliferation, migration and invasion, and indicates a poor prognosis in ovarian cancer. Oncol Rep. 36:3241–3250. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Gao J, Liu M, Zou Y, Mao M, Shen T, Zhang C, Song S, Sun M, Zhang S, Wang B, et al: Long non-coding RNA growth arrest-specific transcript 5 is involved in ovarian cancer cell apoptosis through the mitochondria-mediated apoptosis pathway. Oncol Rep. 34:3212–3221. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Nagini S: Breast Cancer: Current molecular therapeutic targets and new players. Anticancer Agents Med Chem. 17:152–163. 2017. View Article : Google Scholar

53 

Cray C, Zaias J and Altman NH: Acute phase response in animals: A review. Comp Med. 59:517–526. 2009.PubMed/NCBI

54 

Clendenen TV, Lundin E, Zeleniuch-Jacquotte A, Koenig KL, Berrino F, Lukanova A, Lokshin AE, Idahl A, Ohlson N, Hallmans G, et al: Circulating inflammation markers and risk of epithelial ovarian cancer. Cancer Epidemiol Biomarkers Prev. 20:799–810. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Kulbe H, Thompson R, Wilson JL, Robinson S, Hagemann T, Fatah R, Gould D, Ayhan A and Balkwill F: The inflammatory cytokine tumor necrosis factor-alpha generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Res. 67:585–592. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Bollrath J and Greten FR: IKK/NF-kappaB and STAT3 pathways: Central signalling hubs in inflammation-mediated tumour promotion and metastasis. EMBO Rep. 10:1314–1319. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Duan Z, Foster R, Bell DA, Mahoney J, Wolak K, Vaidya A, Hampel C, Lee H and Seiden MV: Signal transducers and activators of transcription 3 pathway activation in drug-resistant ovarian cancer. Clin Cancer Res. 12:5055–5063. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 53 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song J, Zhang W, Wang S, Liu K, Song F and Ran L: A panel of 7 prognosis-related long non-coding RNAs to improve platinum-based chemoresistance prediction in ovarian cancer. Int J Oncol 53: 866-876, 2018
APA
Song, J., Zhang, W., Wang, S., Liu, K., Song, F., & Ran, L. (2018). A panel of 7 prognosis-related long non-coding RNAs to improve platinum-based chemoresistance prediction in ovarian cancer. International Journal of Oncology, 53, 866-876. https://doi.org/10.3892/ijo.2018.4403
MLA
Song, J., Zhang, W., Wang, S., Liu, K., Song, F., Ran, L."A panel of 7 prognosis-related long non-coding RNAs to improve platinum-based chemoresistance prediction in ovarian cancer". International Journal of Oncology 53.2 (2018): 866-876.
Chicago
Song, J., Zhang, W., Wang, S., Liu, K., Song, F., Ran, L."A panel of 7 prognosis-related long non-coding RNAs to improve platinum-based chemoresistance prediction in ovarian cancer". International Journal of Oncology 53, no. 2 (2018): 866-876. https://doi.org/10.3892/ijo.2018.4403