Identification of genes predicting chemoresistance and short survival in ovarian cancer
Original Article

Identification of genes predicting chemoresistance and short survival in ovarian cancer

Cong Wang1#, Cuilan Chen1#, Xiaoying Chen1, Jie Luo1, Yuting Su1, Xia Liu2, Fuqiang Yin1,3

1Life Sciences Institute, Guangxi Medical University, Nanning, China; 2Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Centre for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China; 3Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, China

Contributions: (I) Conception and design: X Liu, F Yin; (II) Administrative support: F Yin; (III) Provision of study materials or patients: F Yin; (IV) Collection and assembly of data: X Liu, C Wang, C Chen; (V) Data analysis and interpretation: X Liu, C Wang, C Chen, Y Su; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Fuqiang Yin, PhD. Professor, Life Sciences Institute, Guangxi Medical University, 22 Shuangyong Rd., Nanning 530021, China; Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning 530021, China. Email: yinfq@mail2.sysu.edu.cn; Xia Liu, PhD. Professor, Key Laboratory of Longevity and Ageing-Related Disease of Chinese Ministry of Education, Centre for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, 22 Shuangyong Rd., Nanning 530021, China. Email: realliuxia@sina.com.

Background: Ovarian cancer (OC) is a kind of lethiferous cancer in gynecology, and the development of chemoresistance is the brief reason for treatment failure. The genes which contribute to chemoresistance are often leading to short survival. Thus, this study aims to identify predictive markers for chemoresistance and survival from chemoresistant-related genes.

Methods: Coremine was used to retrieve of genes linked to OC chemoresistance. The relationship of genes with patient survival was analyzed in 489 OC patients of The Cancer Genome Atlas (TCGA) cohort, which the subgroup of 90 resistant and 197 sensitive samples was used to determine gene expression. Kaplan-Meier (KM) plotter of 1,816 OC patients with survival data was retrieved for survival analysis. Survival analysis was carried out by the R survival package in R (version 3.3.1). KM and receiver operating characteristic (ROC) curve were respectively used to access the ability of a gene to predict survival and chemoresistance.

Results: In this study, a group of genes potentially linked to OC chemoresistance was identified, which dysregulated in 90 chemoresistant tissues compared with 197 sensitive tissues. Of them, thirteen genes could predict chemoresistance in 1,347 patients, especially SOS1, MSH6, STAT5A were excellent for predicting chemoresistance to any drugs, platin and taxane, CASP2 and PARD6B for any drugs and platin, and HSP90AA1 and HSP90B1 for taxane. Meanwhile, 44 genes linked to OC chemoresistance could predict short overall survival (OS) and/or disease-free survival (DFS) in 489 OC patients, and 10 of them could predict short OS in large cohort of up to 1,657 patients. Finally, it is noteworthy that CASP2 was down-regulated in 90 chemoresistant samples, and low expression of the gene predicted chemoresistance in 1,347 patients, short OS and DFS in 489 patients, and short OS and progression-free survival (PFS) in 1,657 patients.

Conclusions: The identified genes specifically the CASP2 might be potentially used as predictive marker, prognostic marker and therapeutic target in management of OC.

Keywords: Ovarian cancer (OC); chemoresistance; predictive markers


Submitted Nov 22, 2023. Accepted for publication Jun 21, 2024. Published online Aug 06, 2024.

doi: 10.21037/tcr-23-2157


Highlight box

Key findings

• Our study found the CASP2 might be potentially used as predictive marker, prognostic marker and therapeutic target in management of ovarian cancer (OC).

What is known and what is new?

• The development of chemoresistance is the main reason for the treatment failure and low survival rate of advanced OC patients.

• The study employed large sample analyses to identify genes predicting chemoresistance and short survival from chemoresistant-related genes in OC. And we found CASP2 might be used as biomarker and therapeutic target in OC management.

What is the implication, and what should change now?

• The CASP2 might be used as biomarker and therapeutic target in OC management. Further research is required to elucidate its functions in OC.


Introduction

Ovarian cancer (OC) is a lethal gynecological malignancy, with about 19,710 new cases and 13,270 deaths every year in United States (1). The standard first-line treatment of OC is surgery followed with platinum and taxane centered chemotherapy (2). Although these therapies can achieve complete remission at the initial stage, most of patients are likely to suffer tumor recurrence predominantly owning to the emergence of chemoresistance, which finally leads to poor prognosis (3). Thus, the factors contribute to the chemoresistance are often the reason for short survival of the OC patients.

Lots of factors are participated in the modulation of chemoresistance and thus many kinds of molecules can be the potential biomarkers for chemoresistance and survival, such as microRNAs, cell cycle and mitosis-molecules, cancer stem cell related molecules, the immune response related molecules and other cancer-associated molecules (4). However, poor sensitivity and lack of specificity are the limitation for majority of biomarkers that have been studied (2). Therefore, there is an ongoing need to identify factors that affect chemoresistance and survival in OC.

Open data and bioinformatics can boost advancements in basic science (5), and reuse of open data is very powerful (6). For example, based on 2,579 tumors from The Cancer Genome Atlas (TCGA) of four gynecological types plus breast, five molecular subtypes have been developed to assess the survival status of patients (7). On the basis of big data of TCGA and Gene Expression Omnibus (GEO), we previously discovered a group of genes relevant to chemoresistance and outcome in OC (8), and developed the multi-gene prognostic signatures in liver cancer (9).

In the present study, based on big data mining and large sample analysis, we identified hundreds of chemoresistant-related genes in OC. The role of those genes in prediction of chemoresistance and short survival was evaluated. We present this article in accordance with the REMARK reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2157/rc).


Methods

Text mining

Coremine (http://www.coremine.com/medical/) was used for retrieval of genes potentially linked to chemoresistance in OC, by combination of “ovarian cancer” [‘Ovarian Neoplasms (alias Ovarian Cancer)’ (mesh); ‘Ovarian Neoplasms (alias Ovarian Cancer)’ (mesh); ‘Malignant neoplasm of ovary (alias Ovarian Cancer)’ (disease)] and “drug resistance” [‘drug resistance’ (mesh); ‘Drug Resistance, Neoplasm’ (mesh)] (P<0.05).

Data acquisition and large samples

TCGA ovarian cohort (10) of 489 OC patients with clinical data and gene expression was retrieved from cBioPortal (http://www.cbioportal.org/) (11,12), including “staging”, “grading”, “overall survival” and “primary treatment outcome”, in which a subgroup of 90 platinum resistant samples and 197 sensitive samples was included. Kaplan-Meier (KM) plotter (13) of 1,816 OC patients with survival data was retrieved for survival analysis, containing a subgroup of 1,656 patients of which the overall survival (OS) data were accessible, and 1,435 patients of whom the progression-free survival (PFS) data were available. The survival data of 1,816 OC patients with mRNA expressions were integrated from GEO (https://www.ncbi.nlm.nih.gov/geoprofiles/) (14,15) (GSE51373, GSE9891, GSE63885, GSE15622, GSE30161, GSE14764, GSE65986, GSE18520, GSE27651, GSE26712, GSE19829, GSE26193, GSE23554 and GSE3149) and the ovarian cohort of TCGA (10). The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013).

Statistical analysis

Statistical analysis was performed with SPSS (v 20.0). Student’s t-test was used to determine the significant variance between the two sets of data. Survival analyses were carried out by the R survival package in R (version 3.3.1). The survival curves were evaluated by the KM method, which gene expression was divided into low and high expression according to the median value, in accordance with a previous study (16). The univariate survival analysis was applied to calculate the hazard ratio (HR) and 95% confidence intervals. The biological processes annotation and pathway enrichment were subsequently performed. Gene expression in prediction of chemoresistance was analyzed by receiver operating characteristic (ROC) plotter (http://www.rocplot.org/) (17), in which the transcriptomic data of 1,816 OC patients were included. The relapse status at six months was used as a cutoff for determination of patient’s response to therapy. And patients relapsed within six months were considered as non-responders to drugs.


Results

Retrieval of genes that potentially affect OC chemoresistance

A total of 1,319 genes which potentially affect OC chemoresistance were retrieved from Coremine database by use of keywords “ovarian cancer” and “drug resistance” (P<0.05). A total of 1,298 genes potentially linked to chemoresistance in OC were obtained from a previous study (18). The above two groups of genes were combined and a total of 2,431 genes were obtained after deletion of duplicates. The transcript expression data of the 2,431 genes with clinical factors in 489 OC samples (with a subgroup of 90 chemoresistant samples and 197 sensitive samples) were retrieved to analyze the relationships of these genes with chemoresistance and prognosis, and 2,218 genes of which the data were available were used for subsequent analyses.

Identification of chemoresistant-related genes that predict chemoresistance

There were 301 genes differentially expressed in chemoresistant samples interacting with each other

Function annotation and enrichment were performed. There were 353 biological processes significantly annotated from the 301 genes and proteins [false discovery rate (FDR) <0.01]. And among the top 17 processes (FDR <1.0e−06), at least 185 genes (61.5%) were responded to stimulus, 166 (55.2%) were involved in regulation of biological processes, 118 (39.2%) were development related, and 115 (38.2%) were responded to chemical processes (Figure 1A). These results at least partially provide the links between those biological processes with chemoresistance in OC, specifically for the genes in response to stimulus.

Figure 1 The 301 genes differentially expressed in 90 chemoresistant samples in contrast to 197 sensitive samples according to TCGA ovarian cancer cohort. (A) Function annotation and enrichment analyses revealed the top 17 biological processes (FDR <1×10−6) that were differentially expressed, bubbles in the same cluster are represented by the same color; (B) enriched top 10 typical pathways that correlated with ovarian cancer chemoresistance. The X-axis represents the number of genes. TCGA, The Cancer Genome Atlas; FDR, false discovery rate.

Pathway-based identification of 26 novel and key genes which contributed to chemoresistance

Fifty-three pathways were significantly enriched from the 301 genes (FDR <0.01), and among the top 16 pathways (FDR <0.001), 10 of them were typical pathways playing critical roles in OC resistance, which including PI3K-Akt signaling, apoptosis, platinum chemoresistance signaling, Ras signaling, mammalian target of rapamycin (mTOR) signaling, ErbB signaling, etc. (Figure 1B).

Gene distribution on the above 10 pathways related to chemoresistance in OC was comprehensively analyzed. Combination of all the genes in the 10 pathways indicated that 51 of the 301 genes were distributed in the 10 pathways. Further analysis based on previous studies was performed to reveal the associations of the 51 genes with cancer development, and the results indicated that the roles of 26 genes in OC chemoresistance were less reported. The transcriptional levels of the 26 genes in 90 chemoresistant OC samples and 197 sensitive samples are shown in Figure 2. Among these, 14 genes including MSH6, CASP2, EIF2AK3, SOS1, EIF2AK2, FLT1, FZD5, HSP90AA1, HSP90B1, KDR, PAX6, PCK1, SDC1 and WNT7A were significantly down-regulated in 90 resistant samples, and 12 genes including PARD6B, AKT1S1, CALML3, CSF3, GNG7, IHH, NRG1, PIK3CD, RIN1, RPS6KA1, STAT5A and TBP were significantly up-regulated.

Figure 2 Twenty-six genes significantly and differentially expressed in chemoresistant samples in contrast to the sensitive samples, in accordance with TCGA ovarian cancer cohort. Sensitive: 197 platinum sensitive ovarian cancer samples; resistant: 90 resistant samples. *, P<0.05; **, P<0.01. TCGA, The Cancer Genome Atlas.

Large sample-based identification of 13 genes that predicted chemoresistance

The roles of the above 26 genes in prediction of chemoresistance in OC were evaluated in a large sample of 1,347 OC patients. As shown in Figure 3, 13 of the 26 genes were identified to be potential predictive biomarkers of chemoresistance in OC. Consistent with their expressions in chemoresistant samples (Figure 2), low expression of SOS1, CASP2, MSH6, HSP90AA1, HSP90B1 and FLT1, and high expression of PARD6B, STAT5A, RPS6KA1, RIN1, PIK3CD, CALML3 and NRG1, could predict the emergence of chemoresistance (Figure 3A). In particular, on one hand, low expression of SOS1, CASP2 and MSH6, and high expression of PARD6B and STAT5A could be more excellent for predicting drug resistance to any drugs and platin [P<0.01, area under the curve (AUC) >0.6] (Figure 3B); while low expression of SOS1, MSH6, HSP90AA1 and HSP90B1, and high expression of STAT5A could be more excellent for prediction of taxane resistance (P<0.01, AUC >0.6) (Figure 3C). On the other hand, three genes including SOS1, MSH6 and STAT5A were excellent for predicting chemoresistance to any drugs, platin and taxane (P<0.01, AUC >0.6).

Figure 3 Role of genes in prediction of chemoresistance in ovarian cancer. Transcriptome-level data of 1,347 ovarian cancer patients were included according to ROC plotter. The relapse status at 6 months was used as a cut off for definition of patient’s response to therapy, and those relapsed within the 6 months were considered as non-responders. (A) Abnormal expression of genes predicts chemoresistance to any drugs (includes platin, taxane, docetaxel, paclitaxel, gemcitabine, topotecan and avastin), in which 130 non-responders and 1,217 responders were included. (B) Abnormal expression of genes predicts chemoresistance to platin, in which 114 non-responders and 1,095 responders were included. (C) Abnormal expression of genes predicts chemoresistance to taxane, in which 81 non-responders and 807 responders were included. AUC, area under the curve; FPR, false positive rate; TPR, true positive rate; ROC, receiver operating characteristic.

Identification of chemoresistant-related genes for predicting short survival

Forty-four genes which dysregulated in chemoresistant samples could predict short OS and disease-free survival (DFS)

The roles of the 2,218 genes in prediction of DFS and OS were determined in 489 OC samples of TCGA cohort. Of which, 207 genes were related to DFS (P<0.05), 249 genes were related to OS (P<0.05), and collectively a total of 380 genes were related to DFS and/or OS. Then, an intersection of these 380 genes with the 301 genes dysregulated in 90 chemoresistant samples were performed, and total of 84 common genes were identified.

Further analyses on these 84 genes were performed. On one hand, the expression of the gene in chemoresistant samples should match the poor status of prognosis. For example, a gene would be retained if it was highly expressed in 90 chemoresistant samples in contrast to 197 sensitive samples, and the high expression predicted a short survival. On the other hand, a gene would be retained if its association with prognosis in OC was poorly studied. After the selection, total of 44 genes which significantly dysregulated in chemoresistant samples and predicted short survival were identified (Table 1).

Table 1

Forty-four genes which dysregulated in chemoresistant samples were relevant to DFS and OS in ovarian cancer

Gene Up/down-regulated in drug resistant samples* (P<0.05) High/low expression predicts poor survival# (P<0.05)
Up Down High Low DFS OS
CASP2
CHIT1
STAT1
MSH6
AADAC
CAPN13
GCH1
OR6F1
PHGDH
RNF148
SLAMF7
WDR45B
LAYN
PARD6B
GDF6
LIPC
TENM3
ALDH5A1
TREML2
MRS2
TRIM27
CXCR4
KCNE3
SPOCK2
RPL23
TCF15
ACSS3
NCOA1
KCNJ16
C16ORF89
HIPK1
LARP4
LGR5
PSMD1
VTCN1
EIF2AK3
SOS1
CRYAB
GAP43
ICAM5
CATSPERD
PSG1
RSL24D1
SNHG29

The expression of the gene in chemoresistant samples and their relationships with survival in ovarian cancer was determined based on the TCGA ovarian cohort: (*) 90 chemoresistant samples and 197 sensitive samples were used for determine gene expression, and (#) 489 samples were used for prognosis analysis. Kaplan-Meier method was used for survival analysis; gene expression was divided into low and high by the median value. DFS, disease-free survival; OS, overall survival; TCGA, The Cancer Genome Atlas.

Among the 44 genes, 17 of them were prominently associated with both OS and DFS in 489 OC patients. Among them, low expression of 12 genes (CASP2, CHIT1, STAT1, MSH6, AADAC, CAPN13, GCH1, OR6F1, PHGDH, RNF148, SLAMF7, and WDR45B) which were down-regulated in resistant samples were correlated with poor prognosis, and high expression of five genes (LAYN, PARD6B, GDF6, LIPC and TENM3) which were up-regulated in resistant samples predicted poor prognosis (Tables 1,2, Figure 4). Ten genes were only significantly associated with OS, among those, low expression of six genes (ALDH5A1, TREML2, MRS2, TRIM27, CXCR4 and KCNE3) which were down-regulated in resistant samples predicted short OS, and high expression of four genes (SPOCK2, RPL23, TCF15 and ACSS3) which were up-regulated in resistant samples associated with short OS (Tables 1,3). Seventeen genes were only significantly relevant to DFS, among those, low expression of 10 genes (NCOA1, KCNJ16, C16ORF89, HIPK1, LARP4, LGR5, PSMD1, VTCN1, EIF2AK3 and SOS1) which were down-regulated in resistant samples predicted short DFS and high expression of seven genes (GRYAB, GAP43, ICAM5, CATSPERD, PSG1, RSL24D1 and SNHG29) which were up-regulated in chemoresistant samples predicted short DFS (Tables 1,4).

Table 2

Kaplan-Meier analyses revealed that seventeen genes were relevant to overall survival and disease-free survival in ovarian cancer, based on TCGA cohort of 489 patients

mRNA expression Expression level Disease-free survival Overall survival
Estimate Standard error 95% confidence interval P Estimate Standard error 95% confidence interval P
Lower Upper Lower Upper
CASP2 High 18.960 1.127 16.751 21.169 0.002 48.720 2.501 43.817 53.623 <0.001
Low 14.460 1.080 12.342 16.578 36.890 2.217 32.545 41.235
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
CHIT1 High 18.040 0.864 16.347 19.733 0.02 47.570 2.223 43.212 51.928 0.006
Low 15.280 1.036 13.250 17.310 40.380 2.656 35.174 45.586
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
STAT1 High 17.970 0.869 16.267 19.673 0.03 47.370 2.423 42.620 52.120 0.03
Low 14.720 1.396 11.984 17.456 41.000 2.997 35.126 46.874
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
MSH6 High 19.120 0.993 17.173 21.067 0.01 45.300 3.434 38.569 52.031 0.04
Low 15.150 0.931 13.326 16.974 40.970 2.933 35.220 46.720
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
AADAC High 17.840 1.158 15.571 20.109 0.02 48.290 3.223 41.973 54.607 0.001
Low 16.000 1.021 13.999 18.001 39.560 2.730 34.210 44.910
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
CAPN13 High 17.970 0.852 16.299 19.641 0.045 47.670 2.468 42.833 52.507 0.041
Low 15.380 1.044 13.333 17.427 37.910 2.935 32.157 43.663
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
GCH1 High 18.960 1.264 16.483 21.437 0.01 47.670 2.331 43.101 52.239 0.049
Low 15.080 1.063 12.996 17.164 40.970 2.778 35.525 46.415
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
OR6F1 High 17.970 1.274 15.474 20.466 0.007 45.110 3.046 39.139 51.081 0.03
Low 15.640 1.018 13.644 17.636 41.360 2.862 35.750 46.970
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
PHGDH High 19.120 1.009 17.143 21.097 0.003 44.880 1.928 41.100 48.660 0.02
Low 15.110 0.705 13.728 16.492 39.360 2.760 33.951 44.769
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
RNF148 High 19.150 1.271 16.658 21.642 0.02 48.290 2.795 42.812 53.768 <0.001
Low 15.110 1.128 12.900 17.320 36.240 2.347 31.639 40.841
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
SLAMF7 High 18.960 1.439 16.140 21.780 <0.001 47.370 2.753 41.974 52.766 0.007
Low 14.780 0.927 12.963 16.597 41.530 2.247 37.127 45.933
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
WDR45B High 18.170 1.669 14.899 21.441 0.02 48.750 3.009 42.852 54.648 0.005
Low 16.130 0.788 14.586 17.674 39.360 3.042 33.398 45.322
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
LAYN High 16.690 1.245 14.250 19.130 0.01 40.970 2.386 36.293 45.647 0.044
Low 17.510 1.042 15.468 19.552 47.370 2.732 42.016 52.724
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
PARD6B High 14.690 0.788 13.146 16.234 0.02 38.410 2.599 33.317 43.503 0.044
Low 18.660 0.853 16.987 20.333 47.670 2.345 43.075 52.265
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
GDF6 High 15.440 1.405 12.685 18.195 0.050 39.850 3.080 33.813 45.887 0.02
Low 17.640 0.968 15.743 19.537 47.510 2.155 43.286 51.734
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
LIPC High 15.280 0.630 14.044 16.516 0.005 39.360 2.581 34.301 44.419 0.004
Low 19.150 1.016 17.158 21.142 49.020 3.563 42.037 56.003
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940
TENM3 High 15.440 1.146 13.194 17.686 0.03 39.360 3.224 33.041 45.679 0.040
Low 17.970 1.543 14.945 20.995 44.880 2.200 40.568 49.192
All 16.850 0.743 15.394 18.306 43.790 2.117 39.640 47.940

Gene expression was divided into low (L) and high (H) by the median value. TCGA, The Cancer Genome Atlas.

Figure 4 Kaplan-Meier analyses determined that seventeen genes were relevant to overall survival and disease-free survival in ovarian cancer, based on TCGA cohort of 489 patients. Gene expression was divided into low (L) and high (H) by the median value. DFS, disease-specific survival; OS, overall survival; TCGA, The Cancer Genome Atlas.

Table 3

Kaplan-Meier analyses revealed that ten genes were correlated with overall survival in ovarian cancer, based on TCGA cohort of 489 patients

mRNA expression Expression level Overall survival
Estimate Standard error 95% confidence interval P
Lower Upper
ALDH5A1 High 48.290 4.063 40.326 56.254 0.002
Low 39.560 2.748 34.173 44.947
All 43.790 2.117 39.640 47.940
TREML2 High 49.580 2.877 43.942 55.218 <0.001
Low 36.340 2.287 31.857 40.823
All 43.790 2.117 39.640 47.940
MRS2 High 44.980 2.983 39.133 50.827 0.045
Low 41.000 2.290 36.513 45.487
All 43.790 2.117 39.640 47.940
TRIM27 High 49.810 4.298 41.387 58.233 0.004
Low 39.360 2.686 34.096 44.624
All 43.790 2.117 39.640 47.940
KCNE3 High 47.510 2.085 43.423 51.597 0.04
Low 39.000 2.227 34.635 43.365
All 43.790 2.117 39.640 47.940
CXCR4 High 48.030 3.115 41.925 54.135 0.04
Low 39.850 2.723 34.512 45.188
All 43.790 2.117 39.640 47.940
SPOCK2 High 38.410 2.886 32.754 44.066 0.01
Low 48.720 3.001 42.838 54.602
All 43.790 2.117 39.640 47.940
RPL23 High 41.000 2.268 36.556 45.444 0.02
Low 47.670 2.346 43.072 52.268
All 43.790 2.117 39.640 47.940
TCF15 High 39.850 2.037 35.858 43.842 0.02
Low 48.030 3.258 41.645 54.415
All 43.790 2.117 39.640 47.940
ACSS3 High 40.970 2.799 35.483 46.457 0.044
Low 45.300 2.743 39.923 50.677
All 43.790 2.117 39.640 47.940

Gene expression was divided into low (L) and high (H) by the median value. TCGA, The Cancer Genome Atlas.

Table 4

Kaplan-Meier analyses showed that seventeen genes were correlated to disease-free survival in ovarian cancer, based on TCGA cohort of 489 patients

mRNA expression Expression level Disease-free survival
Estimate Standard error 95% confidence interval
Lower Upper P
NCOA1 High 18.660 1.214 16.280 21.040 0.003
Low 15.280 1.180 12.967 17.593
All 16.850 0.743 15.394 18.306
KCNJ16 High 17.640 1.391 14.914 20.366 0.02
Low 16.300 1.371 13.613 18.987
All 16.850 0.743 15.394 18.306
C16ORF89 High 18.140 1.413 15.370 20.910 0.02
Low 15.410 0.746 13.948 16.872
All 16.850 0.743 15.394 18.306
HIPK1 High 18.100 1.126 15.894 20.306 <0.001
Low 14.720 0.968 12.823 16.617
All 16.850 0.743 15.394 18.306
LARP4 High 18.170 1.312 15.598 20.742 0.03
Low 15.380 1.238 12.953 17.807
All 16.850 0.743 15.394 18.306
LGR5 High 17.970 1.308 15.406 20.534 0.02
Low 15.540 1.245 13.099 17.981
All 16.850 0.743 15.394 18.306
PSMD1 High 18.860 0.871 17.154 20.566 0.003
Low 14.780 0.921 12.975 16.585
All 16.850 0.743 15.394 18.306
VTCN1 High 17.840 1.151 15.584 20.096 0.03
Low 15.640 1.432 12.833 18.447
All 16.850 0.743 15.394 18.306
EIF2AK3 High 18.170 1.296 15.630 20.710 0.047
Low 15.540 1.008 13.564 17.516
All 16.850 0.743 15.394 18.306
SOS1 High 18.170 1.000 16.209 20.131 0.001
Low 14.720 0.933 12.892 16.548
All 16.850 0.743 15.394 18.306
CRYAB High 15.640 0.953 13.773 17.507 0.046
Low 18.040 1.284 15.524 20.556
All 16.850 0.743 15.394 18.306
GAP43 High 14.460 1.248 12.013 16.907 0.02
Low 18.170 1.092 16.030 20.310
All 16.850 0.743 15.394 18.306
ICAM5 High 16.130 1.111 13.953 18.307 0.03
Low 17.840 0.986 15.908 19.772
All 16.850 0.743 15.394 18.306
CATSPERD High 15.150 1.177 12.842 17.458 0.01
Low 18.140 1.110 15.964 20.316
All 16.850 0.743 15.394 18.306
PSG1 High 14.880 1.100 12.724 17.036 0.02
Low 18.860 1.051 16.799 20.921
All 16.850 0.743 15.394 18.306
RSL24D1 High 15.380 1.284 12.864 17.896 0.04
Low 18.040 1.178 15.732 20.348
All 16.850 0.743 15.394 18.306
SNHG29 High 16.300 1.192 13.963 18.637 0.008
Low 18.040 1.153 15.779 20.301
All 16.850 0.743 15.394 18.306

Gene expression was divided into low (L) and high (H) by the median value. TCGA, The Cancer Genome Atlas.

Large sample-based identification of 10 genes which significantly predicted short OS

The roles of the above 44 genes in prediction of prognosis were further verified in large samples of 1,816 OC patients, which included a subgroup of 1,656 specimens with OS data, and 1,435 specimens with PFS data. Firstly, among the 17 genes which were relevant to OS in TCGA cohort of 489 patients (Figure 4, Table 3), 10 of them (CASP2, CH1T1, SPOCK2, TREML2, RPL23, TCF15, ALDH5A1, MRS2, TRIM27 and STAT1) were consistently related to OS in 1,656 OC samples (Figure 5A), of which, four genes including CASP2, CHIT1, SPOCK2 and TREML2 were also significantly associated with PFS (Figure 5A). The relationships of the 17 genes associated with DFS in the 489 patients (Table 4) were also submitted to the analysis, and five of them including CRYAB, GAP43, NCOA1, ICAM5 and KCNJ16 were consistently associated with prognosis in the 1,816 patients, and the former three were associated with both OS and PFS (Figure 5B).

Figure 5 Genes correlated with OS and PFS in 1,816 ovarian cancer patients based on the KM plotters collection. Gene expression was divided into low (L) and high (H) by the median value. (A) Genes related to OS in 489 patients of TCGA cohort consistently predicted OS and/or PFS in 1,816 patients of KM plotter collection. (B) Genes related to DFS in 489 patients was also relevant to OS and/or PFS in 1,816 patients. OS, overall survival; PFS, progression-free survival; KM, Kaplan-Meier; TCGA, The Cancer Genome Atlas.

Discussion

In this study, a total of 2,218 genes which potentially affected chemoresistance in OC were identified based on text mining, and 301 of them were significantly dysregulated in 90 chemoresistant OC samples in contrast to 197 sensitive samples. Analysis by biological processes annotation suggested those genes could potentially respond to stimulus (Figure 1A). It has been proved that response to stimulus is implicated in regulation of chemoresistance in cancers (19,20). Furthermore, pathway enrichment of the 301 genes was performed and 10 pathways such as PI3K-Akt signaling, apoptosis, and platinum chemoresistance signaling were significantly enriched (Figure 1B), which are typical pathways involved in modulation of chemoresistance in OC. The results strongly supported the relevance of those genes with chemoresistance. Intersection of all genes of the 10 pathways with the 301 genes identified 51 genes, which included some typical chemoresistant related genes in OC, such as AKT1, PIK3CA and MAPK1 (21). Among the 51 genes, the associations of 26 genes (Figure 2) with chemoresistance in OC have been rarely reported. The results above suggested that, probably via interactions with the typical 10 drug-resistant pathways in OC, the 26 genes might be new targets for management of OC, particularly in chemoresistant patients.

Because of the heterogeneity of OC, identifying predictive biomarkers are important for the selection of suitable treatments to improve patient survival (22). The use of gene expression signatures of key pathways that contribute to chemoresistance as predictive biomarkers is a reasonable approach (22). Several genes were previously identified to predict chemoresistance. For example, a low-RAS signature was shown to associate with sensitivity to the AKT inhibitor MK2206 (23), and BRCAness gene-expression in OC was associated with responsiveness to platinum-based chemotherapy (24,25). Despite of those findings, the biomarkers for prediction of chemoresistance are still less understood. In this study, the above identified 26 genes were distributed in 10 chemoresistant-related pathways, and based on the large sample analysis, 13 of them were potentially the predictive biomarkers for chemoresistance (Figure 3). Especially the low expression of SOS1, CASP2 and MSH6, and high expression of PARD6B and STAT5A were excellent for predicting chemoresistance to any drugs and platin; low expression of SOS1, MSH6, HSP90AA1 and HSP90B1, and high expression of STAT5A were excellent for predicting taxane resistance. The associations of SOS1, CASP2, PARD6B, STAT5A, HSP90AA1 and HSP90B1 with chemoresistance in OC were rarely known, although a research suggested that the silence of MSH6 in Saccharomyces cerevisiae could increase the strain resistance to cisplatin, doxorubicin, and carboplatin (26). This is consistent with our findings. However, the roles of those genes in prediction of chemoresistance in OC have not been reported so far.

Chemoresistance is a main factor that contributes to short survival in OC. Thus, further analyses of 2,218 genes in 489 OC patients were performed to identify the candidates that affected both chemoresistance and survival, and 44 genes were identified to be relevant to OS and/or DFS (Table 1), and their relationships with chemoresistance and survival were limited. The correlation of the above 44 genes with survival of OC patients was further verified in large cohort of up to 1,816 specimens, and 10 genes including CASP2, CH1T1, SPOCK2, TREML2, RPL23, TCF15, ALDH5A1, MRS2, TRIM27 and STAT1 were consistently related to OS (Figure 5). The results are consistent with the findings in previous studies. For example, low expression of CASP2 associated with poor OS in gastric carcinoma (27), and high expression of SPOCK2 predicted poor prognosis in OC (28).

Among all the genes identified in this study, CASP2 was the only one that the results were positive in all of the analyses. In the TCGA cohort of 489 OC patients, CASP2 was significantly down-regulated in 90 chemoresistant samples in contrast to 197 sensitive samples, and its low expression predicted short DFS and OS. In large cohort up to thousands of OC patients, CASP2 expression was consistently lower in chemoresistant samples and its low expression predicted chemoresistance to any drugs and platin, and short OS and PFS as well. CASP2 is one of CASPS, which often acts as intrinsic initiators of apoptosis (29,30). CASP2 plays important roles in apoptotic as well as nonapoptotic processes including apoptosis, cell cycle, autophagy, DNA repair, regulation of oxidant levels and lipid biosynthesis (31). The roles of CASP2 in cancer remain a matter of controversy, because the gene normally produces two mRNA splice variants CASP2L and CASP2S. CASP2L normally promotes apoptosis, while CASP2S normally inhibits apoptosis (32). In OC, CASP2 is the target of miR-383, which is overexpressed in OC cells and samples. High level of miR-383 and low expression of the CASP2 improve cell invasion, cell cycle progression and cell proliferation (33). These results are basically consistent with the findings in this study.

Taken together, 13 genes that affected chemoresistance and predicted chemoresistance in OC were identified, especially the six excellent candidate genes SOS1, CASP2, PARD6B, STAT5A, HSP90AA1 and HSP90B1. A total of 44 genes which potentially contributed to chemoresistance and related to prognosis were identified, especially 10 genes including CASP2, CH1T1, SPOCK2, TREML2, RPL23, TCF15, ALDH5A1, MRS2, TRIM27 and STAT1 were consistently related to overall survival in a group of 1,656 patients. Finally, it is noteworthy that CASP2 was the only gene that the results were positively and consistently in all analyses. The genes discovered in this study might be developed to be predictive markers, prognostic markers and therapeutic targets in the clinical management of OC. Studies have shown that, the caspase-2 regulatory mechanism can induce OC cell death (34), Han et al. found that casp-2S affects cellular apoptosis through its interaction with membrane-associated cytoskeletal Fodrin protein (35). Future work will investigate the function of CASP2.

OC cells interact with their surrounding microenvironment through a complex communication mechanism, impacting the tumor’s response to drugs. OC cells can change the composition of their surrounding microenvironment (immune cells, stromal cells and vascular endothelial cells, etc.) through various means, such as releasing cytokines, VEGF and other angiogenic factors (36), secreting exosomes (37), etc., thus forming a favorable environment that promotes tumor growth and invasion. The poorly metabolized tumor microenvironment affects clinical prognosis by forming a barrier to tumor-infiltrating immune cells (38).

The genes related to chemotherapy resistance in OC show some correlation with tumor immune infiltration, tumor mutation burden, and microsatellite instability. Some studies suggest that chemo-resistant cancer cells often have higher levels of tumor immune infiltration and tumor mutation burden (39-41). Additionally, microsatellite instability is also associated with chemotherapy resistance. An increase in mutation burden and microsatellite instability could potentially impact tumor growth, replication, and treatment response (42). Future work will explore the association between CASP2 and tumor immune invasion, tumor mutation burden, and microsatellite instability to provide reference for OC treatment strategies.


Conclusions

The identified genes specifically the CASP2 might be potentially used as predictive markers, prognostic markers and therapeutic targets in management of OC.


Acknowledgments

Funding: This work was supported by the National Natural Science Foundation of China (Nos. 81860458, 81903644, and 82260721), Guangxi Natural Science Foundation (Nos. 2024GXNSFDA010045, 2021GXNSFAA075002, and 2018GXNSFAA281227), Program Foundation of Key Laboratory of High-Incidence Tumor Prevention and Treatment, Ministry of Education (Nos. GKE-ZZ202148, and GKE-ZZ202017) and Advanced Innovation Teams and Xinghu Scholars Program of Guangxi Medical University, Innovation Project of Guangxi Graduate Education (No. YCSW2023237).


Footnote

Reporting Checklist: The authors have completed the REMARK reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2157/rc

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2157/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2157/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013).

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel RL, Miller KD, Wagle NS, et al. Cancer statistics, 2023. CA Cancer J Clin 2023;73:17-48. [Crossref] [PubMed]
  2. Chandra A, Pius C, Nabeel M, et al. Ovarian cancer: Current status and strategies for improving therapeutic outcomes. Cancer Med 2019;8:7018-31. [Crossref] [PubMed]
  3. Xiang Y, Chen YJ, Yan YB, et al. MiR-186 bidirectionally regulates cisplatin sensitivity of ovarian cancer cells via suppressing targets PIK3R3 and PTEN and upregulating APAF1 expression. J Cancer 2020;11:3446-53. [Crossref] [PubMed]
  4. Davidson B. Biomarkers of drug resistance in ovarian cancer - an update. Expert Rev Mol Diagn 2019;19:469-76. [Crossref] [PubMed]
  5. Mangul S, Martin LS, Langmead B, et al. How bioinformatics and open data can boost basic science in countries and universities with limited resources. Nat Biotechnol 2019;37:324-6. [Crossref] [PubMed]
  6. Rung J, Brazma A. Reuse of public genome-wide gene expression data. Nat Rev Genet 2013;14:89-99. [Crossref] [PubMed]
  7. Berger AC, Korkut A, Kanchi RS, et al. A Comprehensive Pan-Cancer Molecular Study of Gynecologic and Breast Cancers. Cancer Cell 2018;33:690-705.e9. [Crossref] [PubMed]
  8. Yin F, Yi S, Wei L, et al. Microarray-based identification of genes associated with prognosis and drug resistance in ovarian cancer. J Cell Biochem 2019;120:6057-70. [Crossref] [PubMed]
  9. Liu M, Liu X, Liu S, et al. Big Data-Based Identification of Multi-Gene Prognostic Signatures in Liver Cancer. Front Oncol 2020;10:847. [Crossref] [PubMed]
  10. Integrated genomic analyses of ovarian carcinoma. Nature 2011;474:609-15. [Crossref] [PubMed]
  11. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401-4. [Crossref] [PubMed]
  12. Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1. [Crossref] [PubMed]
  13. Gyorffy B, Lánczky A, Szállási Z. Implementing an online tool for genome-wide validation of survival-associated biomarkers in ovarian-cancer using microarray data from 1287 patients. Endocr Relat Cancer 2012;19:197-208. [Crossref] [PubMed]
  14. Barrett T, Wilhite SE, Ledoux P, et al. NCBI GEO: archive for functional genomics data sets--update. Nucleic Acids Res 2013;41:D991-5. [Crossref] [PubMed]
  15. Barrett T, Suzek TO, Troup DB, et al. NCBI GEO: mining millions of expression profiles--database and tools. Nucleic Acids Res 2005;33:D562-6. [Crossref] [PubMed]
  16. Henderson MJ, Haber M, Porro A, et al. ABCC multidrug transporters in childhood neuroblastoma: clinical and biological effects independent of cytotoxic drug efflux. J Natl Cancer Inst 2011;103:1236-51. [Crossref] [PubMed]
  17. Fekete JT, Ősz Á, Pete I, et al. Predictive biomarkers of platinum and taxane resistance using the transcriptomic data of 1816 ovarian cancer patients. Gynecol Oncol 2020;156:654-61. [Crossref] [PubMed]
  18. Lloyd KL, Cree IA, Savage RS. Prediction of resistance to chemotherapy in ovarian cancer: a systematic review. BMC Cancer 2015;15:117. [Crossref] [PubMed]
  19. Liu D, Wang L, Zhong R, et al. Parallel microfluidic networks for studying cellular response to chemical modulation. J Biotechnol 2007;131:286-92. [Crossref] [PubMed]
  20. Lahiani MH, Eassa S, Parnell C, et al. Carbon nanotubes as carriers of Panax ginseng metabolites and enhancers of ginsenosides Rb1 and Rg1 anti-cancer activity. Nanotechnology 2017;28:015101. [Crossref] [PubMed]
  21. Liu X, Gao Y, Lu Y, et al. Oncogenes associated with drug resistance in ovarian cancer. J Cancer Res Clin Oncol 2015;141:381-95. [Crossref] [PubMed]
  22. Cheaib B, Auguste A, Leary A. The PI3K/Akt/mTOR pathway in ovarian cancer: therapeutic opportunities and challenges. Chin J Cancer 2015;34:4-16. [Crossref] [PubMed]
  23. Loboda A, Nebozhyn M, Klinghoffer R, et al. A gene expression signature of RAS pathway dependence predicts response to PI3K and RAS pathway inhibitors and expands the population of RAS pathway activated tumors. BMC Med Genomics 2010;3:26. [Crossref] [PubMed]
  24. Konstantinopoulos PA, Spentzos D, Karlan BY, et al. Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer. J Clin Oncol 2010;28:3555-61. [Crossref] [PubMed]
  25. Weberpals JI, Clark-Knowles KV, Vanderhyden BC. Sporadic epithelial ovarian cancer: clinical relevance of BRCA1 inhibition in the DNA damage and repair pathway. J Clin Oncol 2008;26:3259-67. [Crossref] [PubMed]
  26. Durant ST, Morris MM, Illand M, et al. Dependence on RAD52 and RAD1 for anticancer drug resistance mediated by inactivation of mismatch repair genes. Curr Biol 1999;9:51-4. [Crossref] [PubMed]
  27. Wang Z, Ni F, Yu F, et al. Prognostic significance of mRNA expression of CASPs in gastric cancer. Oncol Lett 2019;18:4535-54. [Crossref] [PubMed]
  28. Lou W, Ding B, Zhong G, et al. Dysregulation of pseudogene/lncRNA-hsa-miR-363-3p-SPOCK2 pathway fuels stage progression of ovarian cancer. Aging (Albany NY) 2019;11:11416-39. [Crossref] [PubMed]
  29. Kim YR, Kim KM, Yoo NJ, et al. Mutational analysis of CASP1, 2, 3, 4, 5, 6, 7, 8, 9, 10, and 14 genes in gastrointestinal stromal tumors. Hum Pathol 2009;40:868-71. [Crossref] [PubMed]
  30. Kumar S. Mechanisms mediating caspase activation in cell death. Cell Death Differ 1999;6:1060-6. [Crossref] [PubMed]
  31. Tiwari M, Sharma LK, Vanegas D, et al. A nonapoptotic role for CASP2/caspase 2: modulation of autophagy. Autophagy 2014;10:1054-70. [Crossref] [PubMed]
  32. Solier S, Logette E, Desoche L, et al. Nonsense-mediated mRNA decay among human caspases: the caspase-2S putative protein is encoded by an extremely short-lived mRNA. Cell Death Differ 2005;12:687-9. [Crossref] [PubMed]
  33. Liu J, Dou Y, Sheng M. Inhibition of microRNA-383 has tumor suppressive effect in human epithelial ovarian cancer through the action on caspase-2 gene. Biomed Pharmacother 2016;83:1286-94. [Crossref] [PubMed]
  34. Yang CS, Matsuura K, Huang NJ, et al. Fatty acid synthase inhibition engages a novel caspase-2 regulatory mechanism to induce ovarian cancer cell death. Oncogene 2015;34:3264-72. [Crossref] [PubMed]
  35. Han C, Zhao R, Kroger J, et al. Caspase-2 short isoform interacts with membrane-associated cytoskeleton proteins to inhibit apoptosis. PLoS One 2013;8:e67033. [Crossref] [PubMed]
  36. Nunes SC. Tumor Microenvironment - Selective Pressures Boosting Cancer Progression. Adv Exp Med Biol 2020;1219:35-49. [Crossref] [PubMed]
  37. Nakamura K, Sawada K, Kinose Y, et al. Exosomes Promote Ovarian Cancer Cell Invasion through Transfer of CD44 to Peritoneal Mesothelial Cells. Mol Cancer Res 2017;15:78-92. [Crossref] [PubMed]
  38. Kao KC, Vilbois S, Tsai CH, et al. Metabolic communication in the tumour-immune microenvironment. Nat Cell Biol 2022;24:1574-83. [Crossref] [PubMed]
  39. Fan S, Gao X, Qin Q, et al. Association between tumor mutation burden and immune infiltration in ovarian cancer. Int Immunopharmacol 2020;89:107126. [Crossref] [PubMed]
  40. Gao C, Li H, Liu C, et al. Tumor Mutation Burden and Immune Invasion Characteristics in Triple Negative Breast Cancer: Genome High-Throughput Data Analysis. Front Immunol 2021;12:650491. [Crossref] [PubMed]
  41. Wang ZM, Xu QR, Kaul D, et al. Significance of tumor mutation burden and immune infiltration in thymic epithelial tumors. Thorac Cancer 2021;12:1995-2006. [Crossref] [PubMed]
  42. Chen J, Apizi A, Wang L, et al. TCGA database analysis of the tumor mutation burden and its clinical significance in colon cancer. J Gastrointest Oncol 2021;12:2244-59. [Crossref] [PubMed]
Cite this article as: Wang C, Chen C, Chen X, Luo J, Su Y, Liu X, Yin F. Identification of genes predicting chemoresistance and short survival in ovarian cancer. Transl Cancer Res 2024;13(8):4354-4371. doi: 10.21037/tcr-23-2157

Download Citation