Open Access

Glucose restriction combined with chemotherapy decreases telomere length and cancer antigen‑125 secretion in ovarian carcinoma

  • Authors:
    • Stephanie Antoun
    • David Atallah
    • Roula Tahtouh
    • Mona Diab Assaf
    • Malak Moubarak
    • Eliane Nasser Ayoub
    • Georges Chahine
    • George Hilal
  • View Affiliations

  • Published online on: December 20, 2019     https://doi.org/10.3892/ol.2019.11233
  • Pages: 1338-1350
  • Copyright: © Antoun 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

Although chemotherapy is the standard treatment for ovarian cancer (OC), recent studies have focused on its coupling with hypoglycemic drugs to decrease glucose availability. Similarly to cancer antigen 125 (Ca‑125), telomerase, the key protein for telomere lengthening, is overexpressed in 90% of OC cases. The aim of the present study was to investigate the effect of the combination of glucose restriction and chemotherapy on telomere length and Ca‑125 secretion in OC cells. SKOV‑3, OVCAR‑3 and Igrov‑1 cells were treated with 20 µM cisplatin and 100 nM paclitaxel for 48 h in three different glucose concentrations: i) 4.5 g/l, ii) 1 g/l and iii) 0.5 g/l. The same treatment was repeated once per week for 6 consecutive weeks. The surviving cells were considered platinum‑taxane escape (PTES) cells. The expression levels of telomerase and Ca‑125 in treated and PTES cells were quantified by qPCR, and Ca‑125 secretion by ELISA. Telomere length was evaluated by qPCR according to the Cawthon method. The modulation of Ca‑125 by telomerase was assessed using inhibitors, small interfering RNA and transfection with human telomerase reverse transcriptase (hTERT) vectors. The implication of phosphatidylinositol‑4,5‑bisphosphate 3‑kinase/protein kinase B/mechanistic target of rapamycin (PI3K/Akt/mTOR) in Ca‑125 modulation was investigated using specific inhibitors. An increase in hTERT and Ca‑125 expression levels (range, 1.5‑3 fold) was observed in short‑term treated cells. However, an opposite effect was detected in PTES cells, where the rate of decrease in the expression levels of hTERT and Ca‑125 reached 60% after treatment in 0.5 g/l glucose. Moreover, telomere length was decreased by 30% in cells treated with 0.5 g/l glucose. Inhibition of hTERT expression significantly decreased Ca‑125 secretion, suggesting a potential modulation of Ca‑125 by hTERT. The inhibition of the PI3K/Akt/mTOR pathway also decreased Ca‑125 secretion; however, the effect of this treatment was not enhanced when coupled with telomerase inhibitors. In conclusion, the combination of chemotherapy and glucose restriction was observed to decrease Ca‑125 secretion and telomerase expression leading to shortening in telomere length. Thus, decreasing glucose availability for OC cells during treatment may lead to a better clinical outcome and potentially improve the prognosis of patients with OC.

Introduction

Ovarian cancer (OC) is one of the most commonly diagnosed types of gynecological cancer worldwide and had the highest mortality rates among all types of cancer of the female reproductive system in 2017 (1). Despite the poorly understood etiology of this cancer type, certain risk factors have been established, which include genetic predisposition and age. By contrast, contraceptive administration and increased parity have been found to be major protective factors (2). The most common subtypes of OC include serous, mucinous, endometrioid and clear cell carcinoma (3). Since OC is asymptomatic during its early stages, it is most commonly diagnosed at late stages, leading to a poor prognosis and short overall survival time (4). In total, >80% of OC cases exhibit a positive response to standard chemotherapy along with platinum-based and taxane molecules (5); nevertheless, chemoresistance remains a major limitation in cancer treatment (6).

Although cancer antigen 125 (Ca-125) is the major biomarker used for diagnosis and disease monitoring in OC, this glycoprotein is also highly expressed in other types of malignancies, including breast cancer, mesothelioma, non-Hodgkin's lymphoma and gastric cancer (711), and is less expressed in some benign cases, including pregnancy and ovulatory cycles (12,13). Despite the functions of Ca-125 being poorly understood, certain studies have suggested the implication of this glycoprotein in the cell-mediated immune response. Ca-125 binds to natural killer (NK) cells via galectin-1 and other antibodies, leading to a reduction in CD16 expression and suppression of the NK cell cytotoxic responses (14,15). Moreover, Ca-125 is able to bind to mesothelin, a 40-kDa protein expressed by mesothelial cells and certain types of cancer cells, including epithelial OC cells. Taking into account that peritoneal cells express the membrane-bound form of mesothelin, this interaction between both proteins may increase the risk of metastasis of Ca-125-expressing tumor cells into the peritoneal cavity (16,17). Furthermore, overexpression of the mucin 16 gene coding for Ca-125 has been observed to increase the risk of resistance to certain chemotherapeutic agents, including cisplatin (18). Ca-125, which is modulated by various factors, regulates the activity of mechanistic target of rapamycin (mTOR) kinase, which in turn controls the expression of its downstream target c-MYC. In addition, knockdown of the MUC16 gene in pancreatic cancer cells has been shown to lead to a decrease in glucose uptake and lactate secretion, and to an alteration of cellular proliferation and metabolism (19).

In >80% of different types of cancer case, telomerase is expressed by cancer cells to ensure their immortalization (20). This enzyme promotes telomere length stabilization and elongation through de novo synthesis of repeats lost after DNA replication or oxidative stress (21). Recently, inhibitors of telomerase activity, including BIBR1532, have been investigated as potential adjuvants to platinum-based chemotherapy in OC cells in vitro (22). The catalytic subunit of telomerase, human telomerase reverse transcriptase (hTERT), regulates telomerase activity through the variation of its expression (23). In fact, hTERT mRNA expression is regulated by a number of proteins; however, the transcription factor c-MYC serves an essential role in the activation of this expression by forming a complex with the MYC-associated factor X protein and binding to the E boxes of the promoter region (24). The expression of c-MYC is linked to various signaling pathways, including the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (Akt)/mTOR, Wnt/β catenin and mitogen-activated protein kinase signaling pathways (25,26).

Increased glucose metabolism is a distinct characteristic of highly proliferative cells, including cancer cells, stem cells and immune cells (27). Even in the presence of oxygen, cancer cells tend to metabolize glucose into lactate instead of undergoing oxidative phosphorylation. This process is termed the Warburg effect (28). Since one of the eight hallmark characteristics of cancer cells is their ability to reprogram glucose metabolism (29), the effect of glucose restriction on cancer cell proliferation, apoptosis and response to treatment has been recently studied. In fact, glucose levels in cancer patients may be an important prognostic indicator. In OC, increased expression of glucose transporter 1 (GLUT1), a transmembrane protein responsible for glucose uptake, is related to shorter survival time in patients with OC (30). In addition, decreasing glucose availability for colon cancer cells has been reported to contribute to an increase in cell death (31). Moreover, the combination of glucose restriction and autophagy inhibition has been shown to result in decreased tumor growth and cancer cell proliferation (32). Furthermore, a decrease in telomerase activity and a higher response rate to the telomerase inhibitor BIBR-1532 was observed in breast cancer cells cultured in medium with low glucose concentration (33).

Considering the fact that most relapse cases in patients with OC occur due to chemoresistance, mechanisms aiding in reversing resistance or preventing its occurrence should be investigated. Several studies have proven the involvement of hTERT in cancer cell immortalization. Moreover, Ca-125 affects the response of cancer cells to chemotherapy and glucose restriction decreases cancer cell proliferation and viability (34). Based on these facts, the effect of chemotherapy combined with glucose restriction on the expression and activity of Ca-125 and telomerase was assessed in the present study. Additionally, the modulation of Ca-125 expression by hTERT and the possible involvement of the PI3K/Akt/mTOR signaling pathway were investigated.

Materials and methods

Cell culture and drugs

The present study was performed on 3 OC cell lines, namely the Igrov-1 (Institut Gustave Roussy), SKOV-3 and Ovcar-3 (both American Type Culture Collection) cell lines. SKOV-3 and Igrov-1 cells were cultured in 4.5 g/l DMEM supplemented with 10% FBS and 1% penicillin-streptomycin (PS; Sigma-Aldrich; Merck KGaA) according to the manufacturer's protocol, whereas Ovcar-3 cells was cultured in F12 medium supplemented with 20% FBS and 1% PS (Sigma-Aldrich; Merck KGaA). All cells were incubated in a humidified incubator at 37°C with 5% CO2. CDDP (Sigma-Aldrich; Merck KGaA) was freshly dissolved in 0.9% NaCl solution, whereas PTX (Sigma-Aldrich; Merck KGaA) was prepared in DMSO and stored at −20°C.

Short-term combinatorial treatment

The three OC cell types were seeded in 6-well plates (2×105 cells/well) and treated with 20 µM cisplatin (CDDP) and 100 nM paclitaxel (PTX) for 48 h at 37°C at 80% confluence. This treatment was performed in three different cell culture media: i) DMEM with 4.5 g/l glucose, ii) DMEM with 1 g/l glucose and iii) DMEM with 0.5 g/l glucose. The negative control corresponded to non-treated cells. The supernatant was subsequently collected and the treated cells were subjected to RNA extraction using Nucleospin RNA Extraction kit (Macherey-Nagel, GmbH).

Generation of platinum-taxane escape (PTES) cells (35)

To generate PTES cells, 1×105 SKOV-3, Ovcar-3 and Igrov-1 cells were seeded in 6-well plates and treated at 37°C with 20 µM CDDP for 1 h, followed by 100 nM PTX for 3 h once per week for 6 weeks. Prior to and during the treatment, the cells were exposed to glucose restriction for 48 h by administering three different glucose concentrations (4.5, 1 and 0.5 g/l glucose) in the culture media. After 6 weeks, the live cells were considered as PTES cells.

Determination of telomere length

The PTES cells remained in culture for 1 month after the treatment, and were subsequently harvested and subjected to DNA extraction using the Nucleospin DNA Extraction kit (Macherey-Nagel, GmbH) following the manufacturer's protocol. Telomere length was assessed according to the protocol proposed by R. Cawthon (36) Briefly, quantitative PCR (qPCR) was performed using the following primer pairs: Telo forward, 5′-CGGTTTGTTTGGGTTTGGGTTTGGGTTTGGGTTTGGGTT-3′ and reverse, 5′-GGCTTGCCTTACCCTTACCCTTACCCTTACCCTTACCCT-3′); and 36B4 (reference gene), forward, 5′-CAGCAAGTGGGAAGGTGTAATCC-3′ and reverse, 5′-CCCATTCTATCAACGGGTACAA-3′. The following thermocycling conditions were applied: For telomeres, 5 min at 95°C, followed by 18 cycles of 95°C for 15 sec, 54°C for 2 min and 72°C for 10 sec, and then 40°C for 30 sec; and for 36b4, 10 min at 95°C, followed by 30 cycles of 95°C for 15 sec, 58°C for 60 sec and 72°C for 10 sec, and then 40°C for 30 sec (37).

Treatment with hTERT and PI3K/Akt/mTOR inhibitors

A total of 1×105 SKOV-3, Ovcar-3 and Igrov-1 cells were seeded in 6-well plates. Once they reached 80% confluence, these cells were treated at 37°C for 48 h with the following inhibitors: Telomerase inhibitors BIBR-1532 (5 and 10 µM), costunolide (5 and 10 µM) and MST-312 (1 and 2 µM); PI3K inhibitors PI 828, wortmanin and GSK (10 µM); AKT inhibitor GSK 690693 (100 nM); and mTOR inhibitor rapamycin (200 nM) (all Tocris Bioscience). The negative control corresponded to non-treated cells maintained in the same conditions as treated cells. In order to indicate the concentrations that should be used for each inhibitor, a toxicity test was performed, and the concentrations were chosen according to the highest concentration that has no toxic effect, therefore no effect on cell viability.

hTERT silencing

Small interfering RNA (siRNA, 5 nmol) specific for hTERT (sense, 5′-GGAGCAAGUUGCAAAGCAUTT-3′ and antisense, 5′-AUGCUUUGCAACUUGCUCCAG-3′) with non-silencing (negative control) and cell death (positive control) siRNAs were used for hTERT silencing (all Qiagen Inc.). The SKOV-3, Ovcar-3 and Igrov-1 cells were seeded in 6-well plates at a density of 2×105 cells/well and transfected for 72 h with the siRNAs at a concentration of 20 µM using the HighPerfect transfection reagent (Qiagen, Inc.), according to the manufacturer's protocols. Knockdown efficacy of hTERT was confirmed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR).

Transfection with hTERT-wild type construct

The three cell lines were transfected with two types of vectors, pBabe-neo-hTERT (cat. no. 1774) and pBabe-neo (control; cat. no. 1767), gifted from Addgene, Inc. After plasmid purification from transformed bacteria using a GenElute HP Plasmid Maxiprep kit (Sigma-Aldrich; Merck KGaA), the transfection was performed using Attractene Transfection Reagent (Qiagen Inc.), according to the manufacturer's protocols. Briefly, the cells were seeded in 6-well plates at a density of 0.5×106 cells/well. Prior to seeding, 1.2 µg of plasmid, attractene and serum-free DMEM (4.5 g/l glucose; for SKOV-3 and Igrov-1), or F-12 for Ovcar-3, were mixed in the wells and incubated for 15 min at room temperature. Subsequently, cells were seeded in 6-well plates for 48 h at 37°C prior to subsequent experiments.

RT-qPCR

To assess the effect of CDDP + PTX treatment on the mRNA expression of hTERT, Ca-125 and certain molecules [interleukin (IL)-6, IL-8, bone morphogenetic protein 2 (BMP2) and excision repair cross-complementation group 1 (ERCC1)] involved in chemoresistance, total RNA was extracted from the PTES and 48-h treated cells using Nucleospin RNA extraction kit. In addition, cells treated with hTERT and PI3K/Akt/mTOR inhibitors underwent RNA extraction using Nucleospin RNA extraction kit. Total RNA (40 ng) was reverse transcribed into cDNA using the iScript cDNA synthesis kit (Bio-Rad Laboratories, Inc.) according to the manufacturer's instruction. Subsequently, the mRNA expression levels of hTERT, Ca-125, IL-6, IL-8, BMP2 and ERCC1, glucose transporters 1 and 3 (GLUT-1 and GLUT-3) and hypoxia inducible subunit α (HIF1α), and the internal reference gene GAPDH were quantified by qPCR using the QuantiFast SYBR Green PCR kit (Qiagen, Inc.) using specific primers for each gene (Table I). cDNA amplification was performed following a PCR program of 40 cycles, with denaturation at 95°C for 10 sec and annealing at 58°C (IL-6, IL-8, BMP-2 and Ca-125) or 60°C for 30 sec, followed by elongation at 72°C for 10 sec (hTERT, ERCC1 and GAPDH) using a Rotor-Gene qPCR cycler (Qiagen GmbH). The mRNA levels were quantified using the 2−ΔΔCq method, where the control was normalized to 1 and the treated samples were compared with their control (38).

Table I.

List of primer sequences.

Table I.

List of primer sequences.

GenePrimer sequence (5′-3′)
hTERTF: CGGAAGAGTGTCTGGAGCAA
R: CTCCCACGACGTAGTCCATG
Ca-125F: CTGCATGTACTCCCATCTCTTCAA
GAGAGAGATGGGAGTAGATGCAG
R: CTGCATCTACTCCCATCTCTCTC
TTGAAGAGATGGGAGTAGATGCAG
IL-6F: TCAATATTAGAGTCTCAACCCCCA
R: TTCTCTTTCGTTCCCGGTGG
IL-8F: CCACCGGAGCACTCCATAAG
R: GATGGTTCCTTCCGGTGGTT
BMP2F: TTTCAATGGACGTGTCCCCG
R: AGCAGCAACGCTAGAAGAA
ERCC1F: AGGCACAAGTAACAGGCTCAC
R: AAGGTCGTAATTCCTTTGCAC
GAPDHF: TGAGCCAGATAGGCTGGAA
R: TAACGCAGGCGATGTTGTC
GLUT-3F: CCCAGATCTTTGGTCTGGAA
R: AAGGGCTGCACTTTGTAGGA
GLUT-1F: GATGATGCGGGAGAAGAAGG
R: AAGACAGCGTTGATGCCAGAC
HIF1αF: TATGAGCCAGAAGAACTTTTAGGC
R: CACCTCTTTTGGCAAGCATCCTG

[i] hTERT, human telomerase reverse transcriptase; Ca-125, cancer antigen 125; IL-6, interleukin-6; IL-8, interleukin-8; BMP2, bone morphogenetic protein 2; ERCC1, excision repair cross-complementation group 1; GLUT-3, glucose transporter 3; GLUT-1, glucose transporter 1; HIF1α, hypoxia inducible factor subunit α; F, forward; R, reverse.

Measurement of Ca-125 in cell supernatant

Ca-125 levels secreted into the supernatant by treated and control SKOV-3, Ovcar-3 and Igrov-1 cells (5×105 cells) were quantified using the DuoSet Human Ca-125/MUC16 ELISA Kit (R&D Systems, Inc; cat. no. DY990) according to the manufacturer's protocols. The supernatant was collected after all treatments were performed and then assayed. The optical density, which is proportional to Ca-125 concentration, was measured using an ELISA reader (Thermo Fisher Scientific, Inc.) at 450 nm and the results were normalized according to the number of cells.

Statistical analysis

All data are presented as the mean ± SD and each experiment was repeated at least three times. All data were assessed with SPSS software using one-way ANOVA followed by the LSD post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Short-term combination of chemotherapy and glucose restriction

To study the effect of glucose restriction on the efficacy of chemotherapy, a 48-h short-term treatment was performed on the SKOV-3, Ovcar-3 and Igrov-1 cell lines using three different glucose concentrations. The comparison between the control and treated cells in the same glucose conditions revealed an increase in the mRNA expression of hTERT in the treated cells (range, 1.5–2.5 fold; Fig. 1A). Moreover, this short-term treatment also lead to an increase in Ca-125 mRNA levels (range, 1.5–3 fold; Fig. 1B). As presented in Fig. 1C, an approximate 2-fold increase was observed in the secretion of Ca-125 in the Ovcar-3 cells; however, Ca-125 secretion by the SKOV-3 and Igrov-1 cells was undetectable by ELISA. Nevertheless, no significant difference was observed among the cells treated in different glucose conditions, indicating that the expression of both proteins was not affected by the decrease in glucose levels during the treatment.

Figure 1.

Effect of 48-h combinatory treatment of CDDP, PTX and glucose restriction on hTERT and Ca-125 expression and secretion. SKOV-3, Ovcar-3 and Igrov-1 cells were seeded in 6 well-plates. These cells were treated with three different glucose concentrations (4.5, 1 and 0.5 g/l) with 20 µM CDDP and 100 nM PTX for 48 h. Supernatant was collected following the treatment in order to quantify secreted Ca-125 using ELISA. The cells were then harvested, and RNA was isolated for hTERT and Ca-125 mRNA quantification by reverse transcription-quantitative polymerase chain reaction. (A and B) hTERT and Ca-125 mRNA expression levels were found to be increased in all treated cells; these results did not appear to be significantly affected by the different glucose levels. (C) A similar effect was observed with regard to Ca-125 secretion in the treated Ovcar-3 cells. The values displayed in this figure refer to values adjusted to 10,000 cells, since cell number usually affects the levels of secreted proteins. Cell count was performed following cell harvesting and values were normalized to 10,000 cells. Each value represents the mean from three different experiments. The control group was set to 1, and the remaining values were compared with the control. The results are presented as the mean ± SD. *P<0.05 vs. control group, and **P<0.01 vs. control group. CTL, control; CDDP, cisplatin; PTX, paclitaxel; hTERT, human telomerase reverse transcriptase; Ca-125, cancer antigen 125; NT, non-treated cells.

Generation of PTES cells

For the development of OC cell lines with double resistance to cisplatin and paclitaxel, the SKOV-3, Ovcar-3 and Igrov-1 cell lines were treated with CDDP and PTX once per week for 6 consecutive weeks. The doses of the chemotherapeutic agents (20 µM CDDP and 100 nM PTX) were chosen according to the peak plasma levels reached when 100 mg/m2 of CDDP and 175 mg/m2 of PTX were administered intravenously in patients with ovarian cancer (35). The cells were treated with CDDP for 1 h and PTX for 3 h. The treatment durations correspond to the half-life of these chemotherapeutic agents in the human body (35). The treatment was performed in culture media of the three cell lines with the following glucose concentrations, which correspond to the physiological alterations observed in blood glucose levels: i) 4.5 g/l glucose, high; ii) 1 g/l glucose, low; and (iii) 0.5 g/l glucose, fasting.

After 6 weeks of treatment, the remaining living cells were considered as PTES cells. Six PTES cell types were generated: PTES SKOV-3 cultured in three different glucose concentrations and PTES Igrov-1 cultured in these same conditions. Despite their resistance to clinically relevant concentrations of cisplatin, PTES Ovcar-3 cells could not be generated. In order to confirm the decrease in the sensitivity of PTES cells to additional chemotherapeutic challenges, these cells were subjected to a single exposure of higher doses of CDDP, PTX and a combination of both treatments in the same glucose conditions as the initial treatment. According to the obtained results, SKOV-3 and Igrov-1 PTES cells required higher concentrations of the chemotherapeutic treatment to reach the same levels of proliferation observed in the parental cells treated with lower doses (Fig. 2). The chemoresistance profile of the PTES cells was evaluated by quantifying the mRNA expression levels of the IL-6, IL-8 and BMP2 proteins, implicated in environment-mediated drug resistance, and the ERCC1 protein, which is directly involved in DNA repair (39). The obtained results revealed a significant increase in the expression of these mRNAs in all PTES cells, indicating that these cells may have developed resistance to the chemotherapeutic treatment (Fig. 3). Additional investigations concerning the effect of the combination of chemotherapy with induced hypo-, normo- and hyperglycemia on the expression of GLUT1, GLUT3 and HIF1α were performed by measuring the mRNA expression levels of these genes in control and PTES cells. The results revealed a significant increase in the expression of both glucose transporters in PTES cells treated with the three glucose concentrations, with no significant difference between them (Fig. 4A and B). However, HIF1α expression level was significantly increased only in SKOV-3 4.5 PTES cells (Fig. 4C).

Decrease of telomere length and hTERT and Ca-125 expression in PTES cells

Next, the effect of this combined long-term (6 weeks) treatment on the expression of hTERT, the catalytic subunit of telomerase, was assessed in all PTES cell lines. The results demonstrated a 20% decrease in the hTERT mRNA expression levels of PTES SKOV-3 cells treated with 4.5 g/l of glucose; however, this decrease reached 40% with 0.5 g/l of glucose. A similar effect was observed in the PTES Igrov-1 cells, where the reduction ranged from 15% with 4.5 g/l glucose to 60% with fasting glucose concentrations (Fig. 5A). Telomere length was subsequently evaluated to assess the effect of this reduction on telomerase expression. A non-significant decrease was observed in PTES SKOV-3 and Igrov-1 cultured with 4.5 and 1 g/l glucose. However, the decrease in telomere length was greater in the both PTES cell lines treated with fasting glucose concentrations (Fig. 5B). Additionally, the Ca-125 mRNA expression levels were quantified in chemosensitive and chemoresistant cells. It was observed that Ca-125 expression decreased in a similar manner as the hTERT expression; however, the reduction ranged between 60 and 80% in the PTES SKOV-3 cells and between 65 and 85% in the PTES Igrov-1 cells (Fig. 5C).

Effect of hTERT inhibition on Ca-125 expression and secretion

Based on the aforementioned results, a parallel change in the expression of hTERT and Ca-125 was observed after both short-term and long-term treatments. Taking into consideration that telomerase regulates the expression of various genes, the role of hTERT in the modulation of Ca-125 expression and secretion was investigated. Thus, chemosensitive cells from the three cell lines were treated with three telomerase inhibitors: BIBR1532 at 5 and 10 µM, costunolide at 5 and 10 µM and MST-312 at 1 and 2 µM. The concentrations of these inhibitors were selected according to a dose-response curve, which demonstrated that they did not exert any cytotoxic effects. The results indicated a significant decrease in the mRNA and protein expression of Ca-125 in the three cell lines after treatment with all three inhibitors compared with the control. However, the highest effect was observed with BIBR1532 at 5 µM for SKOV-3 and Ovcar-3 and at 10 µM for Igrov-1 (Fig. 6A and B). Transfection of all three cell lines with hTERT siRNA was performed to confirm the direct involvement of hTERT in Ca-125 modulation and to exclude the possibility of extratelomeric action of the hTERT inhibitors. In order to make sure that the transfection was successful, hTERT mRNA expression was assessed in control and transfected cells. The results (Fig. 7A) demonstrated a significant decrease (~90%) in hTERT expression level in transfected cells compared with control cells. As expected, the mRNA expression pattern obtained by gene silencing was similar to that obtained after treatment with the inhibitors (Fig. 6C and D). A significant decrease was observed in the mRNA expression of Ca-125 in the three cell lines, and in the protein expression of Ca-125 in Ovcar-3 cell line following hTERT silencing.

Figure 6.

Effect of hTERT inhibition and overexpression on Ca-125 mRNA and protein expression. Cells from the three cell lines were cultured in 6-well plates. At 80% confluence, these cells were treated with telomerase inhibitors BIBR1532 (5 and 10 µM), costunolide (5 and 10 µM) and MST-312 (1 and 2 µM). Next, the supernatant was collected, and the cells were harvested for RNA extraction. Ca-125 mRNA expression was assayed by reverse transcription-quantitative polymerase chain reaction and protein secretion levels by ELISA. (A and B) A significant decrease was observed after treatment with all inhibitors compared with the control; however, the greatest effect was caused by BIBR1532. In addition, the cells were transfected with siRNA for hTERT and pBabe-neo-hTERT to evaluate the effect of both gene silencing and gene overexpression on Ca-125. (C and D) The effect of sihTERT was similar to the effect of hTERT inhibitors. (E and F) However, transfection with pBabe-neo-hTERT caused a significant increase in both mRNA and protein levels of Ca-125 compared with the control. The values displayed in this figure refer to values adjusted to 10,000 cells, since cell number usually affects the levels of secreted proteins. Cell count was performed following cell harvesting and values were normalized to 10,000 cells. Each experiment was performed at least three times, and the values included in these graphs represent the mean from these experiments. The results are presented as the mean ± SD. *P<0.05 vs. control group, and **P<0.01 vs. control group. CTL, control; hTERT, human telomerase reverse transcriptase; sihTERT, siRNA for hTERT; siRNA, small interfering RNA; Ca-125, cancer antigen 125.

Effect of hTERT overexpression on Ca-125 expression and secretion

After investigating the effect of hTERT inhibition, the SKOV-3, Ovcar-3 and Igrov-1 cells were transfected with pBabe-neo-hTERT to evaluate the effect of telomerase overexpression on Ca-125 expression and secretion. In order to make sure that the transfection was successful, hTERT mRNA expression was assessed in control and transfected cells. The results (Fig. 7B) demonstrated a significant 25–30 fold increase in hTERT expression level in transfected cells compared with control cells. The results indicated a significant increase affecting both the mRNA and protein levels of Ca-125 after 48 h of transfection. Notably, an increase of ~30% in the SKOV-3 and Ovcar-3 cells, and an increase of 50% in Igrov-1 cells was observed with regards to Ca-125 mRNA expression, compared with the control (Fig. 6E and F).

Effect of PI3K/Akt/mTOR signaling pathway inhibition on Ca-125 expression and secretion

As aforementioned, previous studies have investigated the potential link between Ca-125 and the protein mTOR. Since this protein is implicated in the PI3K/Akt/mTOR signaling pathway and a mutual modulation links hTERT to this pathway (40), the regulation of Ca-125 expression and secretion by PI3K/Akt/mTOR was examined. The SKOV-3, Ovcar-3 and Igrov-1 cells were treated with various inhibitors specific to proteins in this pathway for 48 h. The decrease in Ca-125 mRNA expression reached 60% after treatment with PI3K and Akt inhibitors (PI828, GSK 2126458 and Wortmanin for PI3K, and GSK 69029 for Akt) in the Igrov-1 cells and 40% with the mTOR inhibitor rapamycin (Fig. 8A). mRNA expression and protein secretion in the treated Ovcar-3 cells exhibited similar results (Fig. 8A and B, respectively). Thus, the PI3K/Akt/mTOR signaling pathway was indicated as a potential regulator of Ca-125 in OC cells.

Effect of the combination of telomerase and PI3K/Akt/mTOR inhibitors on Ca-125

hTERT and the PI3K/Akt/mTOR pathway are both regulators of Ca-125 in OC cells. Taking into account that telomerase is an activator of this signaling pathway, the possible regulation of Ca-125 by hTERT via PI3K/Akt/mTOR was investigated. The PTES cells were treated with telomerase inhibitors (BIBR-1532, Costunolide and MST-312) combined with PI3K, Akt and mTOR inhibitors for 48 h. However, this combination did not potentiate the effects observed on Ca-125 mRNA and protein levels in all three cell lines (Fig. 9).

Discussion

Resistance to chemotherapy is a major limitation in the treatment of various types of cancer, including OC. Several mechanisms are responsible for the onset and development of chemoresistance, including the activation of DNA repair mechanisms (41), the development of sequential genetic alterations inducing transient environment-mediated drug resistance (42) and the presence of cancer-initiating cells embedded within the tumor (43). To overcome chemoresistance, a previous study focused on the development of new strategies and treatment combinations, including the establishment of second-line chemotherapy or coupling standard treatment regimens with immunotherapy (44). Another recently explored strategy is based on the targeting of cancer cell metabolism to improve its response to therapeutics. A previous study demonstrated the effect of targeting certain glycolytic enzymes to overcome resistance to trastuzumab in breast cancer cells (45). Furthermore, a previous study on breast cancer highlighted the potential use of lactate dehydrogenase A as a target to reverse resistance to PTX (46). Thus, glucose availability and metabolism in cancer serve important roles in the sensitivity of cells to chemotherapeutic drugs. Based on these facts, the effect of chemotherapy combined with glucose restriction on the development of chemoresistance was evaluated in the present study. Moreover, the outcome of this combinatorial treatment on the immortality of cancer cells was investigated by studying telomerase expression and activity, and the ability of these cells to secrete the serum marker Ca-125. The PTES cells were initially treated with the chemotherapeutic agents CDDP and PTX for 48 h in 3 different glucose concentrations. The three concentrations were chosen so as to mimic the physiology of a patient following high, low and fasting glucose diets. Following this short-term treatment, expression of both hTERT and Ca-125 increased significantly. A similar effect on hTERT expression was observed in a previous study after treatment of hepatocellular carcinoma cells with low doses of cisplatin for 24 h. It was reported that increased expression of the transcription factor c-MYC resulted in hTERT upregulation (47). In addition, higher Ca-125 levels after short-term treatment may be explained by the implication of this protein in the modulation of the response of OC cells to genotoxic drugs, including cisplatin (48). However, no significant difference was detected among the cells treated with various glucose concentrations. This may be explained by the short period of exposure to glucose restriction and cell adaptation that requires additional time for metabolic flux changes.

After the long-term treatment of cells with specific concentrations of CDDP and PTX, the expression levels of IL-6, IL-8, BMP2 and ERCC1, which are implicated in the initiation and development of resistance to chemotherapy, were evaluated. The increased expression of all these proteins observed in the PTES cells compared with the control explains the ability of these cells to survive 6 weeks of treatment. These proteins are associated with various mechanisms implicated in chemoresistance. For instance, ERCC1 is involved in the nucleotide excision repair mechanism. Interleukin expression corresponds to the environment-mediated drug resistance developed by the PTES cells (49). However, no significant difference was observed in the expression of these markers among the PTES cells treated with various glucose concentrations. Thus, decreasing glucose availability during treatment may not directly affect the ability of cancer cells to develop chemoresistance.

Following several cellular divisions, the telomeric ends of the chromosomes reach a critical length, leading to the activation of apoptosis. However, telomerase serves a key role in the elongation of these telomeres in cancer cells. To assess the effect of the combinatorial treatment on the immortalization of the PTES cells, the mRNA expression of the catalytic subunit hTERT was examined in the present study. Previous studies revealed that telomerase expression was increased in osteosarcoma cisplatin-resistant cells, thus leading to the suppression of cisplatin-induced apoptosis (50). However, hTERT expression was significantly decreased in the PTES cells in the present study. This decrease was greater in the PTES cells treated with fasting glucose concentrations. To further investigate the effect of this reduction, the length of the telomeres in control and resistant cells was assessed. A decrease in telomere length was observed; however, the shortening of telomeres was statistically significant only in the PTES cells treated with fasting glucose concentrations. The non-significant results for 4.5 and 1 g/l glucose concentrations may be explained by the fact that slight decreases affecting telomerase expression and availability do not necessarily lead to a shortening in telomeres, since telomerase has the ability to elongate the chromosomal telomeric ends, even if present in low quantities (51). Based on these findings, it may be suggested that the combination of chemotherapy and glucose restriction could lead to a decrease in both hTERT expression and telomere length in PTES cells, which in turn may result in a decrease in cancer cell immortalization.

Ca-125 is a key regulator of the metastasis of OC cells into the peritoneal cavity due to its strong affinity to mesothelin. Moreover, the sensitivity of OC cells to cisplatin has been narrowly linked to the expression of the surface marker Ca-125. Based on these facts, the changes in the expression of Ca-125 were investigated in both PTES and control groups. A decrease was observed in Ca-125 expression, which was lowest after treatment with fasting glucose concentrations. These findings are contradictory to those of previous studies stating that increased expression of the Ca-125 gene leads to a decrease in the sensitivity of cells to genotoxic drugs, including cisplatin. Other studies have demonstrated that increased levels of Ca-125 at the end of the chemotherapeutic treatment are considered as a marker of poor prognosis and are associated with shorter progression-free and overall survival rates (52). The results of the present study revealed that the combination of chemotherapy with fasting glucose levels during the treatment resulted in the lowest Ca-125 expression in all three types of PTES cells. Thus, this combination may contribute to improved survival rates in patients with OC, coupled with decreased levels of cancer cell immortalization due to the shortening of telomeres.

Based on the aforementioned results, telomerase and Ca-125 expression varied similarly after both short- and long-term treatments. To explain the low levels of Ca-125 observed after 6 weeks of chemotherapy, the possible regulation of Ca-125 gene expression by hTERT was investigated. In addition to its canonical function, one of the non-canonical functions of hTERT is its ability to regulate the expression of several genes in cancer (53). The decrease observed after the inhibition of hTERT by inhibitors (BIBR-1532, Costunolide and MST-312) and the silencing of the gene by hTERT siRNA demonstrated that hTERT regulates Ca-125 expression in OC. Moreover, the overexpression of Ca-125 after transfection of the PTES cells with pBabe-neo-hTERT verified the association between both proteins. A previous study reported that Ca-125 serum levels were higher in patients with telomerase-positive OC compared with patients with telomerase-negative OC (54). Moreover, a clinical study linking hTERT expression in ovarian biopsies and serum Ca-125 to the grade and stage of OC revealed a linear relationship between these two biomarkers (55). Therefore, it may be postulated that the decrease in hTERT expression in PTES cells may account for the decline in Ca-125 expression and secretion.

Telomerase regulates the expression of several genes either directly or via its ability to control various signaling pathways, including the PI3K/Akt/mTOR pathway. Previous studies have highlighted the modulation of Ca-125 expression by mTOR via the transcription factor c-MYC; therefore, the possible implication of the PI3K/Akt/mTOR pathway in the modulation of Ca-125 by hTERT was investigated. The results observed after the treatment with inhibitors of the pathway alone indicate a possible implication of this pathway in the regulation of Ca-125 expression. However, when combined with hTERT inhibitors, the treatment did not significantly potentiate the effect of the signaling pathway inhibitors when used as a mono-treatment. Thus, other signaling pathways may be involved in the regulation of Ca-125 by hTERT. A possible pathway may be the nuclear factor-κB pathway. In fact, mesothelin, a protein modulated by the activation of this pathway, interacts with Ca-125 to promote peritoneal metastasis (56).

To the best of our knowledge, the present study was the first to demonstrate the effect of chemotherapy combined with glucose restriction on the immortalization and metastasis of OC cells. When neoadjuvant or adjuvant chemotherapy was administered under fasting glucose conditions, telomerase expression was found to significantly decrease, thus contributing to the shortening of chromosomal telomeric ends, which in turn decreases the immortalization of cancer cells. Since telomerase serves an essential role in cisplatin resistance, this decrease may improve the response to chemotherapy. Moreover, this combined treatment was demonstrated to decrease Ca-125 expression and secretion, which could contribute to improved prognosis, since it is able to reduce the risk of peritoneal metastasis. Furthermore, since the combination of chemotherapy with hypoglycemic drugs is currently used in the treatment of various cancer types, including OC, the present study indicates a potential advantage in using this treatment regimen in both neoadjuvant and adjuvant forms.

Acknowledgements

Not applicable.

Funding

This study was funded by the Research Council of Saint Joseph University (grant no. FM 302).

Availability of data and materials

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

Authors' contributions

SA performed cell culture, cell treatments, RT-qPCR, ELISA and transfections, participated in the design of the study and wrote the manuscript. DA participated in the design of the study and manuscript. RT supervised and participated in the experimental work (cell culture and treatments), troubleshooting and data analysis. MDA made substantial contributions to analysis and interpretation of data. MM participated in the design of the manuscript. ENA and GH were responsible for the clinical data interpretation. GC made substantial contributions to interpretation of clinical data. GH designed the study, provided guidance and edited the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethical Committee of Saint Joseph University (Beirut, Lebanon).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

BMP2

bone morphogenetic protein 2

Ca-125

cancer antigen 125

CDDP

cisplatin

ERCC1

excision repair cross-complementation group 1

hTERT

human telomerase reverse transcriptase

IL-6

interleukin-6

IL-8

interleukin-8

MAPK

mitogen-activated protein kinase

mTOR

mechanistic target of rapamycin

NK

natural killer

PI3K

phosphatidylinositol-4,5-bisphosphate 3-kinase

PTES

platinum-taxane escape

PTX

paclitaxel

RT-qPCR

reverse transcription-quantitative polymerase chain reaction

siRNA

small interfering RNA

References

1 

National Cancer Institute: Cancer Statistics, . https://www.cancer.gov/about-cancer/understanding/statisticsMay 18–2017

2 

McLemore MR, Miaskowski C, Aouizerat BE, Chen LM and Dodd MJ: Epidemiologic and genetic factors associated with ovarian cancer. Cancer Nurs. 32:281–290. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Vajpeyi R: WHO Classification of Tumours: Pathology and Genetics of Tumours of the Breast and Female Genital Organs. J Clin Pathol. 58:671–672. 2005.PubMed/NCBI

4 

Jelovac D and Armstrong DK: Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J Clin. 61:183–203. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Kim A, Ueda Y, Naka T and Enomoto T: Therapeutic strategies in epithelial ovarian cancer. J Exp Clin Cancer Res. 31:142012. View Article : Google Scholar : PubMed/NCBI

6 

Argento M, Hoffman P and Gauchez AS: Ovarian cancer detection and treatment: Current situation and future prospects. Anticancer Res. 28:3135–3138. 2008.PubMed/NCBI

7 

Morgado M, Sutton MN, Simmons M, Warren CR, Lu Z, Constantinou PE, Liu J, Francis LL, Conlan RS, Bast RC Jr and Carson DD: Tumor necrosis factor-α and interferon-γ stimulate MUC16 (CA125) expression in breast, endometrial and ovarian cancers through NFκB. Oncotarget. 7:14871–14884. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Norum LF, Erikstein B and Nustad K: Elevated CA125 in breast cancer--A sign of advanced disease. Tumour Biol. 22:223–228. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Cheng X, Gou HF, Liu JY, Luo DY and Qiu M: Clinical significance of serum CA125 in diffuse malignant mesothelioma. SpringerPlus. 5:3682016. View Article : Google Scholar : PubMed/NCBI

10 

Namikawa T, Kawanishi Y, Fujisawa K, Munekage E, Iwabu J, Munekage M, Maeda H, Kitagawa H, Kobayashi M and Hanazaki K: Serum carbohydrate antigen 125 is a signifcant prognostic marker in patients with unresectable advanced or recurrent gastric cancer. Surg Today. 48:388–394. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Memar B, Aledavood A, Shahidsales S, Ahadi M, Farzadnia M, Raziee H, Noori S, Tayebi-Meybodi N, Amouian S and Mohtashami S: The Prognostic Role of Tumor Marker CA-125 in B-Cell non-Hodgkin's Lymphoma. Iran J Cancer Prev. 8:42–46. 2015.PubMed/NCBI

12 

Baalbergen A, Janssen JW and van der Weiden RM: CA-125 levels are related to the likelihood of pregnancy after in vitro fertilization and embryo transfer. Am J Reprod Immunol. 43:21–24. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Bon GG, Kenemans P, Dekker JJ, Hompes PG, Verstraeten RA, van Kamp GJ and Schoemaker J: Fluctuations in CA 125 and CA 15-3 serum concentrations during spontaneous ovulatory cycles. Hum Reprod. 14:566–570. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Das S and Batra K: Understanding the unique attributes of MUC16 (CA125): Potential implications in targeted therapy. Cancer Res. 75:4669–4674. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Hoogstad-van Evert JS, Maas RJ, Van der Meer J, Cany J, Van der Steen S, Jansen JH, Miller JS, Bekkers R, Hobo W, Massuger L and Dolstra H: Peritoneal NK cells are responsive to IL-15 and percentages are correlated with outcome in advanced ovarian cancer patients. Oncotarget. 9:34810–34820. 2018.PubMed/NCBI

16 

Su Y, Tatzel K, Wang X, Belt B, Binder P, Kuroki L, Powell MA, Mutch DG, Hawkins WG and Spitzer D: Mesothelin's minimal MUC16 binding moiety converts TR3 into a potent cancer therapeutic via hierarchical binding events at the plasma membrane. Oncotarget. 7:31534–31549. 2016. View Article : Google Scholar : PubMed/NCBI

17 

He X, Wang L, Riedel H, Wang K, Yong Y, Dinu CZ and Rojanasakul Y: Mesothelin promotes epithelial-to-mesenchymal transition and tumorigenicity of human lung cancer and mesothelioma cells. Mol Cancer. 16:632017. View Article : Google Scholar : PubMed/NCBI

18 

Chanvorachote P, Luanpitpong S, Chunhacha P, Promden W and Sriuranpong V: Expression of CA125 and cisplatin susceptibility of pleural effusion-derived human lung cancer cells from a Thai patient. Oncol Lett. 4:252–256. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Shukla SK, Gunda V, Abrego J, Haridas D, Mishra A, Souchek J, Chaika NV, Yu F, Sasson AR, Lazenby AJ, et al: MUC16-mediated activation of mTOR and c-MYC reprograms pancreatic cancer metabolism. Oncotarget. 6:19118–19131. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Victorelli S and Passos J: Telomeres and cell senescence - size matters not. EBioMedicine. 21:14–20. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Shay JW and Wright WE: Senescence and immortalization: Role of telomeres and telomerase. Carcinogenesis. 26:867–874. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Meng E, Taylor B, Ray A, Shevde LA and Rocconi RP: Targeted inhibition of telomerase activity combined with chemotherapy demonstrates synergy in eliminating ovarian cancer spheroid-forming cells. Gynecol Oncol. 124:598–605. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Greider CW: Regulating telomere length from the inside out: The replication fork model. Genes Dev. 30:1483–1491. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Koh CM, Khattar E, Leow SC, Liu CY, Muller J, Ang WX, Li Y, Franzoso G, Li S, Guccione E and Tergaonkar V: Telomerase regulates MYC-driven oncogenesis independent of its reverse transcriptase activity. J Clin Invest. 125:2109–2122. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Zhu J, Blenis J and Yuan J: Activation of PI3K/Akt and MAPK pathways regulates Myc-mediated transcription by phosphorylating and promoting the degradation of Mad1. Proc Natl Acad Sci U S A. 105:6584–6589. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Zhang S, Li Y, Wu Y, Shi K, Bing L and Hao J: Wnt/β-catenin signaling pathway upregulates c-Myc expression to promote cell proliferation of p19 teratocarcinoma cells. Anat Rec (Hoboken). 295:2104–2113. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Aunoble B, Sanches R, Didier E and Bignon Y: Major oncogenes and tumor suppressor genes involved in epithelial ovarian cancer (Review). Int J Oncol. 16:567–576. 2000.PubMed/NCBI

28 

Vander Heiden MG, Cantley LC and Thompson CB: Understanding the warburg effect: The metabolic requirements of cell proliferation. Science. 324:1029–1033. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Ma Y, Wang W, Idowu MO, Oh U, Wang XY, Temkin SM and Fang X: Ovarian cancer relies on glucose transporter 1 to fuel glycolysis and growth: Anti-tumor activity of BAY-876. Cancers (Basel). 11:E332018. View Article : Google Scholar : PubMed/NCBI

31 

Fang S and Fang X: Advances in glucose metabolism research in colorectal cancer. Biomed Rep. 5:289–295. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Schroll MM, LaBonia GJ, Ludwig KR and Hummon AB: Glucose restriction combined with autophagy inhibition and chemotherapy in HCT 116 spheroids decreases cell clonogenicity and viability regulated by tumor suppressor genes. J Proteome Res. 16:3009–3018. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Wardi L, Alaaeddine N, Raad I, Sarkis R, Serhal R, Khalil C and Hilal G: Glucose restriction decreases telomerase activity and enhances its inhibitor response on breast cancer cells: Possible extra-telomerase role of BIBR 1532. Cancer Cell Int. 14:602014. View Article : Google Scholar : PubMed/NCBI

34 

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

35 

Gamarra-Luques CD, Hapon MB, Goyeneche AA and Telleria CM: Resistance to cisplatin and paclitaxel does not affect the sensitivity of human ovarian cancer cells to antiprogestin-induced cytotoxicity. J Ovarian Res. 7:452014. View Article : Google Scholar : PubMed/NCBI

36 

Cawthon R: Telomere measurement by quantitative PCR. Nucleic Acids Res. 30:e472002. View Article : Google Scholar : PubMed/NCBI

37 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Rao X, Huang X, Zhou Z and Lin X: An improvement of the 2(-delta delta CT) method for quantitative real-time polymerase chain reaction data analysis. Biostat Bioinforma Biomath. 3:71–85. 2013.PubMed/NCBI

39 

Zhou M, Zhao Y, Ding Y, Liu H, Liu Z, Fodstad O, Riker AI, Kamarajugadda S, Lu J, Owen LB, et al: Warbug effect in chemosensitivity: Targeting lactate dehydrogenase-A re-sensitizes taxol-resistant cancer cells to taxol. Mol Cancer. 9:332010. View Article : Google Scholar : PubMed/NCBI

40 

Glasgow C, Pacheco-Rodriguez G, Steagall WK, Haughey ME, Julien-Williams PA, Stylianou MP, Gochuico BR and Moss J: CA-125 in disease progression and treatment of lymphangioleiomyomatosis. Chest. 153:339–348. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Liang C, Qin Y, Zhang B, Ji S, Shi S, Xu W, Liu J, Xiang J, Liang D, Hu Q, et al: Oncogenic KRAS targets MUC16/CA125 in pancreatic ductal adenocarcinoma. Mol Cancer Res. 15:201–212. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Muallem MZ, Braicu I, Nassir M, Richter R, Sehouli J and Arsenic R: ERCC1 expression as a predictor of resistance to platinum-based chemotherapy in primary ovarian cancer. Anticancer Res. 34:393–399. 2014.PubMed/NCBI

43 

Meads MB, Gatenby RA and Dalton WS: Environment-mediated drug resistance: A major contributor to minimal residual disease. Nat Rev Cancer. 9:665–674. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Bapat SA, Mali AM, Koppikar CB and Kurrey NK: Stem and progenitor-like cells contribute to the aggressive behavior of human epithelial ovarian cancer. Cancer Res. 65:3025–3059. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Kigawa J: New strategy for overcoming resistance to chemotherapy of ovarian cancer. Yonago Acta Med. 56:43–50. 2013.PubMed/NCBI

46 

Zhao Y, Liu H, Liu Z, Ding Y, LeDoux SP, Wilson GL, Voellmy R, Lin Y, Lin W, Nahta R, et al: Overcoming trastuzumab resistance in breast cancer by targeting dysregulated glucose metabolism. Cancer Res. 71:4585–4597. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Guo XL, Ma NN, Zhou FG, Zhang L, Bu XX, Sun K, Song JR, Li R, Zhang BH, Wu MC and Wei LX: Up-regulation of hTERT expression by low-dose cisplatin contributes to chemotherapy resistance in human hepatocellular cancer cells. Oncol Rep. 22:549–556. 2009.PubMed/NCBI

48 

Boivin M, Lane D, Piché A and Rancourt C: CA125 (MUC16) tumor antigen selectively modulates the sensitivity of ovarian cancer cells to genotoxic drug-induced apoptosis. Gynecol Oncol. 115:407–413. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Bonneau C, Rouzier R, Geyl C, Cortez A, Castela M, Lis R, Daraï E and Touboul C: Predictive markers of chemoresistance in advanced stages epithelial ovarian carcinoma. Gynecol Oncol. 136:112–120. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Zhang Z, Yu L, Dai G, Xia K, Liu G, Song Q, Tao C, Gao T and Guo W: Telomerase reverse transcriptase promotes chemoresistance by suppressing cisplatin-dependent apoptosis in osteosarcoma cells. Sci Rep. 7:70702017. View Article : Google Scholar : PubMed/NCBI

51 

Sotillo-Piñeiro E, Sierrasesúmaga L and Patiño-García A: Telomerase activity and telomere length in primary and metastatic tumors from pediatric bone cancer patients. Pediatr Res. 55:231–235. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Tian C, Markman M, Zaino R, Ozols RF, McGuire WP, Muggia FM, Rose PG, Spriggs D and Armstrong DK: CA-125 change following chemotherapy in prediction of treatment outcome among advanced mucinous and clear cell epithelial ovarian cancers: A gynecologic oncology group study. Cancer. 115:1395–1403. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Li Y and Tergaonkar V: Noncanonical functions of telomerase: Implications in telomerase-targeted cancer therapies. Cancer Res. 74:1639–1644. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Sapi E, Okpokwasili NI and Rutherford T: Detection of telomerase-positive circulating epithelial cells in ovarian cancer patients. Cancer Detect Prev. 26:158–167. 2002. View Article : Google Scholar : PubMed/NCBI

55 

Maraei AA, Hatta AZ, Shiran MS and Tan GC: Human telomerase reverse transcriptase expression in ovarian tumors. Indian J Pathol Microbiol. 55:187–191. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Bharadwaj U, Marin-Muller C, Li M, Chen C and Yao Q: Mesothelin confers pancreatic cancer cell resistance to TNF-α-induced apoptosis through Akt/PI3K/NF-κB activation and IL-6/Mcl-1 overexpression. Mol Cancer. 10:1062011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 19 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Antoun S, Atallah D, Tahtouh R, Assaf MD, Moubarak M, Ayoub EN, Chahine G and Hilal G: Glucose restriction combined with chemotherapy decreases telomere length and cancer antigen‑125 secretion in ovarian carcinoma. Oncol Lett 19: 1338-1350, 2020
APA
Antoun, S., Atallah, D., Tahtouh, R., Assaf, M.D., Moubarak, M., Ayoub, E.N. ... Hilal, G. (2020). Glucose restriction combined with chemotherapy decreases telomere length and cancer antigen‑125 secretion in ovarian carcinoma. Oncology Letters, 19, 1338-1350. https://doi.org/10.3892/ol.2019.11233
MLA
Antoun, S., Atallah, D., Tahtouh, R., Assaf, M. D., Moubarak, M., Ayoub, E. N., Chahine, G., Hilal, G."Glucose restriction combined with chemotherapy decreases telomere length and cancer antigen‑125 secretion in ovarian carcinoma". Oncology Letters 19.2 (2020): 1338-1350.
Chicago
Antoun, S., Atallah, D., Tahtouh, R., Assaf, M. D., Moubarak, M., Ayoub, E. N., Chahine, G., Hilal, G."Glucose restriction combined with chemotherapy decreases telomere length and cancer antigen‑125 secretion in ovarian carcinoma". Oncology Letters 19, no. 2 (2020): 1338-1350. https://doi.org/10.3892/ol.2019.11233