Open Access

Dual‑regulated oncolytic adenovirus carrying ERCC1‑siRNA gene possesses potent antitumor effect on ovarian cancer cells

  • Authors:
    • Ting Zhao
    • Wei Ye
    • Rui Zhang
    • Xiaoyan Zhu
    • Qin Shi
    • Xiaofeng Xu
    • Weifeng Chen
    • Ling Xu
    • Yaping Meng
  • View Affiliations

  • Published online on: May 15, 2024     https://doi.org/10.3892/mmr.2024.13245
  • Article Number: 120
  • Copyright: © Zhao 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

Ovarian cancer is a multifactorial and deadly disease. Despite significant advancements in ovarian cancer therapy, its incidence is on the rise and the molecular mechanisms underlying ovarian cancer invasiveness, metastasis and drug resistance remain largely elusive, resulting in poor prognosis. Oncolytic viruses armed with therapeutic transgenes of interest offer an attractive alternative to chemical drugs, which often face innate and acquired drug resistance. The present study constructed a novel oncolytic adenovirus carrying ERCC1 short interfering (si)RNA, regulated by hTERT and HIF promoters, termed Ad‑siERCC1. The findings demonstrated that this oncolytic adenovirus effectively inhibits the proliferation, migration and invasion of ovarian cancer cells. Furthermore, the downregulation of ERCC1 expression by siRNA ameliorates drug resistance to cisplatin (DDP) chemotherapy. It was found that Ad‑siERCC1 blocks the cell cycle in the G1 phase and enhances apoptosis through the PI3K/AKT‑caspase‑3 signaling pathways in SKOV3 cells. The results of the present study highlighted the critical effect of oncolytic virus Ad‑siERCC1 in inhibiting the survival of ovarian cancer cells and increasing chemotherapy sensitivity to DDP. These findings underscore the potent antitumor effect of Ad‑siERCC1 on ovarian cancers in vivo.

Introduction

Ovarian cancer is one of the most common malignant tumors affecting female reproductive organs worldwide and it has the highest mortality rate among gynecological tumors (1,2). Due to the lack of early diagnostic methods and inconspicuous clinical symptoms, most women are diagnosed at an advanced stage (corresponding to stages III and IV), resulting in 5-year survival rates ranging from 26–42% (3,4). Despite significant advancements in medical science and technology, the combination of surgery and chemotherapy has improved the therapeutic outcomes for ovarian cancer in recent years, but unfortunately, there has been no significant improvement in the 5-year survival rate (5). Therefore, it is imperative to explore new strategies to enhance the treatment of ovarian cancer.

The current front-line standard of care for ovarian cancer involves surgery followed by platinum-taxane maintenance chemotherapy (6,7). However, chemotherapy resistance remains a significant factor contributing to the high mortality rate in ovarian cancer (5). Various factors, such as decreased drug accumulation, elevated glutathione levels, increased metallothionein and enhanced DNA repair ability, have been implicated in the development of tumor cell resistance to paclitaxel or platinum. Among these factors, the most crucial one is the enhanced tolerance and repair of DNA damage through the nucleotide-excision-repair (NER) pathway (8,9). The NER pathway is highly conserved and represents one of the major DNA repair mechanisms in mammalian cells that counteract the formation of genetic damage (8). Understanding the involvement of the NER pathway in chemotherapy resistance could provide valuable insights for developing novel therapeutic strategies to combat ovarian cancer.

Nucleotide excision repair cross-complementary gene 1 (ERCC1) is a gene responsible for recognizing DNA damage and cleaving the DNA chain (7). This gene plays a crucial role in repairing the damaged platinum and DNA compounds of tumor cells, directly affecting the ability of tumor tissues to restore replication and proliferation (10). A number of studies have indicated that ERCC1 serves as a marker for ovarian cancer resistance to platinum drugs (1113). Research findings have suggested that individuals with low ERCC1 expression tend to have an improved response to cisplatin chemotherapy compared with those with high expression levels (8,14,15). Moreover, using RNA interference (RNAi) to downregulate ERCC1 mRNA expression in gastric cancer cells resistant to cisplatin (DDP) has been shown to restore susceptibility to treatment (16). Given these findings, inhibiting ERCC1 expression emerges as a promising therapeutic strategy for improving the treatment of ovarian cancer. By targeting ERCC1, it may be possible to enhance the efficacy of platinum-based chemotherapy and overcome drug resistance in ovarian cancer patients.

Adenovirus vectors have emerged as one of the most widely studied and applied vectors for cancer gene therapy. They are designed to replicate preferentially in cancer cells and induce their destruction through the natural process of lytic virus replication (17). This approach allows for targeted delivery of the virus to tumor cells with specific genetic alterations or gene expression profiles, making them applicable only to tumors with the desired characteristics (18). However, despite the significant increase in the number of studies on oncolytic adenoviruses in recent years, research on ovarian cancer remains relatively limited. For example, according to a 2023 review by Lundstrom (19), only five out of ~150 studies involved ovarian cancer. This suggests that more exploration and research are needed in the field of ovarian cancer using oncolytic adenovirus therapy. In addition, the research mechanism of using oncolytic adenovirus vectors to carry therapeutic transgenes exhibits diversity. However, there has been no research report on exploring the use of oncolytic adenovirus vectors to target ERCC1 gene. Given the important role of ERCC1 gene in the resistance of ovarian cancer, exploring the use of oncolytic adenovirus vectors to carry interference RNA targeting ERCC1 gene is a promising direction.

Furthermore, it is highly desirable to develop a repertoire of vectors that can exploit microenvironmental constraints specific to tumor growth, in addition to cell intrinsic gene expression profiles or genetic alterations. Hypoxia, characterized by a reduction in O2 partial pressure, is a key element of the tumor microenvironment, commonly found in most solid tumors irrespective of their origin, location, or genetic makeup (20,21). Hypoxia plays a pivotal role in conferring resistance of cancer cells to radiotherapy and chemotherapy, promoting the selection of more aggressive tumor clones and facilitating metastasis predisposition (22,23). Thus, there is a critical need to devise innovative therapeutic strategies that can specifically target hypoxic regions within tumors. Hypoxia-inducible factor (HIF) is a heterodimeric transcription factor that governs cellular responses to hypoxia by binding to a hypoxia-response element (HRE) present within target genes (20,21). The HIF/HRE regulatory system becomes active under hypoxia or in response to genetic alterations during cell transformation. Therefore, harnessing the HIF/HRE system holds significant promise for selectively targeting therapeutic gene expression to tumor tissues.

The present study genetically engineered an oncolytic adenovirus armed with ERCC1 short interfering (si)RNA, specifically regulated by hypoxia/HIF-dependent mechanisms. The experiments demonstrated that the recombinant adenovirus effectively induced sustained silencing of ERCC1 and downregulation of its oncogenic signaling in in vitro models of ovarian cancer. Moreover, the adenovirus exhibits targeted killing of cancer cells resistant to DDP. These findings present a promising avenue for developing novel adjuvant chemotherapy approaches for ovarian cancer.

Materials and methods

Cell lines and cell culture

The human ovarian cancer cell line SKOV3 was procured from the American Type Culture Collection. The 293 cells were acquired from Canada Microbix Biosystems Inc. All cell lines were cultured following the instructions provided by the suppliers in high-glucose Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% (v/v) fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.). The cultures were maintained at 37°C in a humidified atmosphere containing 5% CO2.

Recombinant oncolytic adenoviruses construction

The dual-regulated adenovirus Ad-ERCC1-siRNA-hTERT-E1A-HRE-E1B (Ad-ERCC1) and its control adenovirus Ad-negative control (NC)-siRNA-hTERT-E1A-HRE-E1B (Ad-NC) were constructed by Shanghai GeneChem Co., Ltd. Briefly, fragments of the specific siRNA of ERCC1 (ERCC1-siRNA) gene were obtained which were designed based on Genbank's ERCC1 gene sequence by gene synthesis conducted by Shanghai GeneChem Co., Ltd. The sequence of ERCC1-siRNA was 5′-ccAAGCCCTTATTCCGATCTA-3′ and the negative control sequence was 5′-TTCTCCGAACGTGTCACGT-3′. An overexpression vector was first constructed using PXC1 plasmid [PXC1 (hTERTp-E1A + HREp-E1B)], in which the human telomerase reverse transcriptase promoter (hTERT) was used to regulate the adenovirus ELA gene; the hypoxia regulatory element sequence (HRE) regulates the E1B gene. (Fig. 1A and B). ERCC1-siRNA and PXC1 were digested by BamHI and KpnI and then the products were combined by T4 DNA ligase to generate the PXC1-ERCC1-siRNA plasmid. The connected plasmids were transformed into competent cells, identified by PCR and sequenced and then extracted with a TIANprep Mini Plasmid Kit (Tiangen Biotech Co., Ltd.). The purified PXC1-ERCC1-siRNA plasmid was transfected into 293 cells with adenovirus packing plasmid (PBHGE3) using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) as follows: 5 µg of plasmid was added gently to the DMEM, the total volume adjusted to 50 µl and incubate at room temperature for 5 min. Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.; 10 µl) was mixed with 50 µl of DMEM and incubated at room temperature for 5 min. the two solutions were gently mixed without shaking and incubated at room temperature for 20 min to form a DNA/Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) transfection complex. The transfection complex was slowly added to the 293 cell mixing well and incubate at 37°C and in 5% CO2. At 6 h after incubation, the medium containing the transfection mixture was discarded and the cells rinsed with sterile PBS and gently shaken to wash away the residual transfection mixture. Then 5 ml of DMEM containing 10% FBS was slowly added and incubation continued at 37°C and in 5% CO2. At 10–15 days after transfection, single virus plaques appeared in 293 cells, which were collected and confirmed by PCR analysis using the forward and reverse primers. The confirmed recombinant adenovirus was designated Ad-ERCC1. Ad-ERCC1 was amplified in 293 cells and purified by ultracentrifugation on cesium chloride (CsCl) gradients (40,000 × g; 2 h; 4°C). Other viruses used were treated with the same method. EGFP gene was cloned into the viruses to determine the infectivity. Viral titer confirmation was performed by infecting 293 T cells using an End-point dilution method and was calculated by Spearman-Karber Method as follows: Viral titer=10 (x+0. 8) plaque-forming units (PFU)/ml) (x=the sum of positive rates of cytopathic effect in sequential dilution from 10−1 to 10−13).

Reverse transcription-quantitative (RT-q) PCR for mRNA analysis

For quantitative RT-PCR, total RNA was extracted from cells of different groups (6-well plate with 80% cell density) using SuperfecTRI, Total RNA Isolation Reagent (cat. no. 3101-400; Shanghai Pufei Biotechnology Co., Ltd.). cDNA was synthesized from 400 ng of RNA with the Promega M-MLV kit according to the manufacturer's protocol (cat. no. M1705; Promega Corporation). RT-qPCR) were performed in triplicate according to the kit protocol of Bulge-Loop miRNA qRT-PCR Starter Kit (cat. no. C10211-1; Guangzhou RiboBio Co., Ltd.) using a LightCycler 480 II (Roche Diagnostics) with SYBR green PCR reagents. Experiments were replicated four times and the data are shown as fold changes. The primer sequences were: ERCC1: 5′-CTACGCCGAATATGCCATCTC-3′, 3′-GTACGGGATTGCCCCTCTG-5′; GAPDH: 5′-TGACTTCAACAGCGACACCCA-3′, 3′-CACCCTGTTGCTGTAGCCAAA-5′. The PCR program included 95°C for 30 s, 40 cycles of 95°C for 5 sec and 60°C for 30 sec. The relative amount of mRNA for target gene was determined by the 2−ΔΔCq method (24) and presented as changes in fold in the target gene expression normalized to two endogenous reference genes (GAPDH).

Detection of the inhibition rate in ovarian cancer cells by MTT assay

The cancer cell inhibition rate was evaluated by a standard MTT assay. Briefly, SKOV3 cells were plated in 96-well plates at a density of 2×104 cells/well. After incubation for 24 h, the cells were infected with of different multiplicity of infection (MOI)s of Ad-siERCC1 (0, 0.001, 0.01, 0.1, 1, 5, 10, 50, 100 and 500 for 48 or 72 h) or different concentrations of DDP (0, 2, 4, 6, 8, 10, 12, 14, 16 and 18 µmol for 24 h) or both (10 MOI Ad-siERCC1 first for 24 h, 10 µmol DDP for anther 24 h). After treatment, 20 µl (5 mg/ml) MTT solution (cat. no. JT343; Genview Corp.) was added to each well. After 4 h incubation at 37°C, the supernatant was discarded and 100 µl DMSO was loaded in every well for 2–5 min. Blank control wells were also set. The optical density (OD) of each well was measured with a Microplate Reader (cat. no. M2009PR; Tecan infinite; Tecan Group, Ltd.) at 490 nm. The cell growth inhibitory rate (I%) was calculated according to the following equation: inhibitory rate%=(ODcontrol-ODsample)/(ODcontrol-ODblank) × 100%, where ODcontrol is the absorbance of the untreated cells, ODsample is the absorbance of the cells exposed to Ad-siERCC1, DDP or both and Ablank is the absorbance of the media. A total of three reduplicate wells were measured at each group and every experiment was performed at least 3 times.

Wound healing migration assay

Cells were counted in a Neubauer chamber slide using the trypan blue exclusion method. Viable cells were plated at 3×105 cells/well in 6-well culture plates using growth media containing 10% FBS for 24 h. The cells were washed with DPBS and pretreated with either DDP or recombinant adenovirus in serum-free media. To control wells, only serum-free media was added. The cells were scratched perpendicular to the horizontal line with a 200 µl sterile pipette tip to create a cell-free wound area. After washing away suspended cells, fresh serum-free media was added and images were captured immediately (time 0 h) using an Olympus CX41 inverted microscope (Olympus Corporation). The cells were cultured with 37°C and 5% CO2 and images were captured again after 24 h at the same position. The measurement of cell scratch was done by ImageJ software (version 1.47; National Institutes of Health). Within each assay the experiments were performed in triplicates. Data shown are representative of minimum three independent experiments.

Matrigel invasion assay

A total of 3×105 cells in 100 µl serum-free medium were seeded in a Transwell chamber (MilliporeSigma) with a coating of Matrigel (MilliporeSigma). The Matrigel was placed on ice and thawed overnight at 4°C in the freezer. It was applied diluted with DMEM at 1:5 to the insert cell growth surface at 150–200 µl/cm2 at 37°C for 30 min. Briefly, SKOV3 cells were seeded into the upper chamber in a serum-free medium and treated with either DDP or recombinant adenovirus. A volume of 0.5 ml of medium containing 10% FBS was added to the lower chamber. After 24 h of incubation, the upper surface of the membrane was wiped off with a cotton swab. Cells that invaded the lower surface of the porous membrane were fixed with 4% formaldehyde in PBS containing 4% sucrose for 10 min at room temperature and stained with 0.1% crystal violet for 10 min at room temperature. Stained cells were counted in 20 random fields per filter, in a total of three filters (n=3). Invasion was presented as percentage of invasion=(number treated cells/number of control cells) ×100.

Cell cycle analysis

SKOV3 cells were transfected with Ad at an MOI of 10 for 6 h. Following DDP treatments for 24 h, cells were detached with trypsinization, washed with Dulbecco's phosphate buffered saline (DPBS) and the cells collected. For assessment of DNA contents, cells were stained with propidium iodide (PI; 50 µg/ml; cat. no. P4170; MilliporeSigma) with 0.5 mg/ml RNase (cat. no. EN0531; Thermo Fisher Scientific, Inc.) in DPBS + 0.1% Tween (pH 7.4) in the dark for 30 min and then monitored by a fluorescence-activated cell sorter (FACS) using a BD C6 PLUS (BD Biosciences). The FACS data were analyzed using NovoExpress (ACEA Biosciences, Inc.) to calculate the fraction of cells in G1, S and G2 phases.

Cell apoptosis assay

Annexin V-APC staining and flow cytometry analysis were used to detect cell apoptosis. Annexin V Apoptosis Detection Kit APC (cat. no. 88-8007; eBioscience; Thermo Fisher Scientific, Inc.) was used to measure cell apoptosis according to the manufacturer's instructions. Briefly, after washing with PBS, cells were resuspended in Annexin-V binding buffer, stained with the Annexin V-APC for 15 min at room temperature in the dark and analyzed using FACSCalibur (C6 PLUS; BD Biosciences) and the apoptotic rate was calculated as the numbers of early + late apoptotic cells/total numbers of cell.

Protein extraction and immunoblotting

Cells were harvested and re-suspended in a RIPA buffer (Beyotime Institute of Biotechnology) containing protease inhibitor cocktail (cat. no. GK10014; GlpBio). After determining the protein concentration using an Enhanced BCA Protein Assay kit (Beyotime Institute of Biotechnology), the obtained lysates (20 µg/well) were subjected to 4–10% SDS-PAGE to separate the proteins, which were subsequently transferred to PVDF membranes (cat. no. A29562259; Cytiva). The membranes were incubated with the following primary antibodies: anti-PI3K (1:1,000; cat. no. 4257; Cell Signaling Technology, Inc.), phosphorylated (p)-Akt (Ser473; 1:1,000; cat. no. 4060; Cell Signaling Technology, Inc.), anti-AKT (1:1,000; cat. no. ab179463; Abcam), anti-caspase-3 (1:1,000; cat. no. sc-7272; Santa Cruz Biotechnology, Inc.), cleaved caspase-3 (1:1,000; cat. no. 9661S; Cell Signaling Technology, Inc.), GAPDH (1:2,000; cat. no. sc-32233; Santa Cruz Biotechnology), β-Actin (1:2,000; cat. no. sc-8432; Santa Cruz Biotechnology). After blocking for 1 h at room temperature in non-fat milk in TBST (Tris-buffered saline with 0.1% Tween-20), membranes were incubated overnight at 4°C with primary antibodies in blocking buffer. After rinses with TBST, membranes were incubated with horseradish peroxidase-conjugated secondary antibodies (goat anti-rabbit, cat. no. 7074; goat anti-mouse, cat. no. 7076; Cell Signaling Technology, Inc.) for 1.5 h at room temperature and detected using 20X LumiGLO reagent and 20X peroxide (cat. no. 7003; Cell Signaling Technology, Inc.) and film exposure. Optical densities of the bands from the original image were measured with NIH ImageJ software (version 1.47; National Institutes of Health).

Statistical analysis

All data are expressed as mean ± SEM. The statistical comparisons for two groups were made with unpaired Independent-Samples t test. Multiple comparisons were made with one-way ANOVA followed by between-group comparisons using the Bonferroni comparisons or Dunnett's T3 comparisons post hoc tests, according to whether the data were normally distributed or not, using SPSS 13.0 software (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Recombinant adenovirus can effectively inhibit the proliferation of ovarian cancer cells

In the present study, the recombinant adenovirus effectively infected and killed tumor cells (Fig. 1C). Subsequently, the transfection efficiency of the recombinant adenovirus on ovarian cancer cells was evaluated. Cultured SKOV3 cells were infected with the recombinant adenovirus expressing green fluorescent protein (GFP) with ERCC1-siRNA (Ad-ERCC1-siRNA, Ad-siERCC1) or the empty vector (Ad-NC-siRNA, Ad-NC) at varying MOI levels ranging from 1–10,000. It was determined that the most efficient MOI value was 10 (Fig. 2A). At MOI=10, the mRNA and protein levels of ERCC1 were validated using RT-qPCR and immunoblotting techniques, revealing a decrease in ERCC1 mRNA (~60% less than control; Independent-Samples t test; t(6)=36.842; P<0.001; Fig. 2B) and protein (>3-fold less than control) (Independent-Samples t test; t(6)=24.036; P<0.001; Fig. 2C) upon Ad-siERCC1 treatment.

Oncolytic viruses specifically target and replicate within tumor cells, ultimately lysing them and releasing viral progeny that can propagate among tumors, eventually leading to the destruction of all tumors and thereby inhibiting tumor cell proliferation and survival (16). Considering this, it was further assessed using MTT assay whether the recombinant adenovirus would induce inhibition of cancer cell survival following transduction. As demonstrated in Fig. 2D, the recombinant adenovirus caused a significant increase in cell mortality from MOI 5 (~47.87% for 48 h and 62.49% for 72 h) to MOI 500 [~94.03% for 48 h and 95.43% for 72 h; one-way ANOVA; 48 h: F (9,20)=362.834; P<0.001; vs. the control group, LOG (5): P<0.001; LOG (10): P<0.001; LOG (50): P<0.001; LOG (100): P<0.001; LOG (500): P<0.001; 72 h: F (9,20)=313.047; P<0.001; vs. the control group, LOG (5): P<0.001; LOG (10): P<0.001; LOG (50): P<0.001; LOG (100): P<0.001; LOG (500): P<0.001].

ERCC1 silencing can significantly enhance chemosensitivity to DDP of ovarian cancer cells

DDP is a widely used anticancer drug in combination regimens and it exerts its activity by inducing the formation of various types of DNA adducts, leading to the inhibition of DNA synthesis, function and transcription (25). According to our previous study (26), a systematic inhibition of SKOV3 cell survival was observed with increasing concentrations of DDP from 0–18 µM after 24 h of treatment, as determined by MTT assay (one-way ANOVA; F (9,30)=258.978; P<0.001; Fig. 3A). Markedly, the inhibitory effect had a significantly higher expression of 18.05, 24.98, 29.18, 51.73 and 59.75% for 10,12,14,16,18 µM DDP (vs. the 0 µM DDP group, 10 µM DDP: P=0.001; 12 µM DDP: P=0.001; 14 µM DDP: P=0.002; 16 µM DDP: P=0.001; 18 µM DDP: P=0.001). Therefore, this concentration was chosen for subsequent experiments. Subsequently, the inhibitory rates were compared among DDP, Ad-NC and Ad-siERCC1. The results demonstrated that the recombinant adenovirus [both Ad-NC (38.63%) and Ad-siERCC1 (40%)] exhibited greater effectiveness in inducing cell mortality vs. DDP alone (10.7%; one-way ANOVA; F (2,9)=258.978, P<0.001; vs. DDP group, Ad-NC: P<0.001; Ad-siERCC1: P<0.001, Fig. 3B).

Resistance, however, has limited the efficacy of these drugs in most ovarian cancer patients. Among the various mechanisms contributing to cisplatin (DDP) resistance, enhanced tolerance and repair of DNA damage through the NER pathway have been recognized as the most crucial resistance mechanism to platinum drugs (7,8). To further investigate whether ERCC1 silencing could enhance the cancer cell-killing effect following chemotherapy, MTT assays were performed on SKOV3 cells treated with recombinant adenovirus and DDP. As depicted in Fig. 3C, in the presence of DDP, cancer cells transduced with Ad-siERCC1 (74.19%) exhibited significantly lower cell viability compared with those transduced with Ad-NC (56.85%) six days post-transduction (Independent-Samples t test; t(6)=−7.420; P<0.001). Additionally, cell cycle analysis revealed that SKOV3 cells in all groups were arrested in the G1 phase when compared with the control group (control group: 46.60%; Ad-NC: 63.79%; Ad-siERCC1: 66.05%; DDP: 68.42%; DDP + Ad-NC: 70.56%; DDP + Ad-siERCC1: 75.12%; one-way ANOVA, G1: F (5,12)=148.758, P<0.001; vs. the control group, Ad-NC: P<0.001; Ad-siERCC1: P<0.001; DDP: P<0.001; DDP + Ad-NC: P<0.001; DDP + Ad-siERCC1: P<0.001). However, the G1 phase cell block was more pronounced in the Ad-siERCC1 combined with cisplatin group (vs. DDP group, DDP + Ad-siERCC1: P=0.001; vs. DDP + Ad-NC group, DDP + Ad-siERCC1: P=0.027) and the proportion of cells in the G2/M phase was the lowest (Fig. 3D and E; one-way ANOVA, S: F (5,12)=16.442, P<0.001; vs. the control group, Ad-NC: P=0.036; Ad-siERCC1: P=0.001; DDP: P<0.001; DDP + Ad-NC: P<0.001; DDP + Ad-siERCC1: P<0.001; G2/M: F (5,12)=3.697, P=0.029; vs. the control group, DDP + Ad-siERCC1: P<0.036).

To assess the effect of the recombinant adenoviruses on SKOV3 cell migration, a scratch wound-healing migration assay was performed. The change in scratch width reflected cell mobility. Ad-siERCC1 combined with DDP resulted in the most potent inhibition of cell migration to 54.69%. (one-way ANOVA, F (3,16)=24.970, P<0.001; vs. the control group, DDP: P=0.038; DDP + Ad-NC: P=0.024; DDP + Ad-siERCC1: P=0.001; vs. DDP group, DDP + Ad-siERCC1: P=0.034; Vs. DDP + Ad-NC group, DDP + Ad-siERCC1: P=0.035; Fig. 4A and B). In the Transwell invasion assay, it was observed that the recombinant adenovirus and DDP inhibited the migration ability of SKOV3 cells (one-way ANOVA, F (3,16)=69.035, P<0.001; vs. the control group, DDP: P=0.001; DDP + Ad-NC: P=0.001; DDP + Ad-siERCC1: P=0.001; Fig. 4C and D). Moreover, in the presence of DDP, the effect of Ad-siERCC1 was significantly superior to the DDP group (control group: 100.00%; DDP: 53.62%; DDP + Ad-NC: 56.18%; DDP + Ad-siERCC1: 29.93%; vs. DDP group, DDP + Ad-siERCC1: P=0.034; vs. DDP + Ad-NC group, DDP + Ad-siERCC1: P=0.021). These findings indicated that the recombinant adenovirus significantly enhanced the inhibitory effect on migration and invasion of ovarian cancer cells and Ad-siERCC1 can improve the chemotherapy sensitivity of DDP.

Recombinant adenovirus improves drug resistance to DDP by enhancing apoptosis through the PI3K/AKT-caspase-3 signaling pathways in ovarian cancer cells. Flow cytometry with Annexin V-APC single-dye method was used to detect cells arrested in early apoptosis. As shown in Fig. 5A and B (one-way ANOVA, F (5,18)=892.196, P<0.001), recombinant adenovirus and/or DDP induced apoptosis in SKOV3 cells. The mean fluorescence intensity was 0.61± 0.07 on control cells, 0.65±0.03 on Ad-NC cells, 1.11±0.11 on Ad-siERCC1 cells, 2.05±0.11 on DDP cells, 1.96±0.14 on DDP + Ad-NC cells and 5.68±0.21 on DDP + Ad-siERCC1 cells. (Vs. the control group, Ad-NC: P<0.001; Ad-siERCC1: P<0.001; DDP: P<0.001; DDP + Ad-NC: P<0.001; DDP + Ad-siERCC1: P<0.001) and Ad-siERCC1 significantly enhanced the apoptotic effect of DDP (>2.7-fold more than control; vs. DDP group, DDP + Ad-siERCC1: P<0.001; vs. DDP + Ad-NC group, DDP + Ad-siERCC1: P<0.001). To gain further insights into this phenomenon, immunoblotting was performed to examine the proteins involved in apoptosis. As depicted in Fig. 5C (one-way ANOVA, F (5,18)=14.040, P<0.001; vs. the control group, Ad-NC: P=0.019; Ad-siERCC1: P=0.006; DDP: P=0.002; DDP + Ad-NC: P=0.002; DDP + Ad-siERCC1: P<0.001; vs. DDP group, P=0.034; vs. DDP + Ad-NC group, P=0.037). PI3K were significantly suppressed by recombinant adenovirus and/or DDP and Ad-siERCC1 greatly enhanced the effect of DDP (DDP + Ad-siERCC1: 0.55±0.10 vs. DDP: 0.76±0.05). Recombinant adenovirus and/or DDP reduced the phosphorylation level of p-AKT. Ad-siERCC1 enhanced the effect of DDP on p-AKT [DDP + Ad-siERCC1: 0.56±0.11 vs. DDP: 0.77±0.05; one-way ANOVA, p-AKT: F (5,18)=16.062, P<0.001; vs. the control group, Ad-siERCC1: P=0.003; DDP: P=0.003; DDP + Ad-NC: P=0.001; DDP + Ad-siERCC1: P<0.001; vs. DDP group, P=0.012; vs. DDP + Ad-NC group, DDP + Ad-siERCC1: P=0.035. AKT: F (5,18)=0.107, DDP + Ad-siERCC1: P=0.989]. Concurrently, procaspase-3 levels were notably reduced after incubation with recombinant adenovirus and/or DDP (control: 1.03±0.11 vs. DDP: 0.77±0.05 vs. DDP + Ad-NC: 0.71±0.04 vs. DDP + Ad-siERCC1: 0.56±0.08), whereas the cleaved caspase-3, the activated form of procaspase-3, exhibited a substantial increase (control: 0.21±0.04 VS DDP: 0.44±0.08 vs. DDP + Ad-NC: 0.49±0.04 vs. DDP + Ad-siERCC1: 0.52±0.08; one-way ANOVA, procaspase-3: F (5,18)=36.689, P<0.001; vs. the control group, DDP: P<0.001; DDP + Ad-NC: P<0.001; DDP + Ad-siERCC1: P<0.001; vs. DDP group, P=0.003. cleaved caspase-3: F (5,18)=22.502, P<0.001; vs. the control group, DDP: P=0.001; DDP + Ad-NC: P<0.001; DDP + Ad-siERCC1: P<0.001, Fig. 5E). These results indicated that recombinant adenovirus ameliorates drug resistance to DDP by promoting apoptosis through the PI3K/AKT-caspase-3 signaling pathways in ovarian cancer cells.

Discussion

The present study developed a recombinant oncolytic adenovirus regulated by the hTERT/HIF dual system, which effectively inhibited the proliferation of ovarian cancer cells. Furthermore, using siRNA technology to downregulate ERCC1 gene expression improved drug resistance to DDP by enhancing apoptosis through the PI3K/AKT-caspase-3 signaling pathways in ovarian cancer cells.

Ovarian cancer predominantly implants and metastasizes in the pelvic cavity. However, effective drug concentration delivery intravenously to the pelvic cavity is challenging, resulting in poor therapeutic outcomes. Additionally, the resistance of tumor cells to platinum drugs hinders the anti-tumor effect, leading to a low 5-year postoperative survival rate (3,4). Thus, the development of a new treatment strategy that targets tumors effectively and reverses drug resistance is a crucial research objective in gynecologic oncology. Tumor-specific proliferative oncolytic adenovirus carriers have garnered attention due to their high tumor targeting and lethality. These carriers infect tumor cells, replicate, multiply, lyse and kill tumor cells, releasing more virus that can infect other tumor cells, resulting in a chain killing reaction. Replication and proliferation are limited to tumor cells, with minimal damage to normal host tissues (16). The present study constructed a recombinant oncolytic adenovirus regulated by the hTERT/HIF dual system, expressing GFP with ERCC1-siRNA (Ad-siERCC1). It was demonstrated that Ad-siERCC1 effectively inhibited the proliferation, migration, invasion and survival of ovarian cancer cells. Furthermore, compared with DDP alone, Ad-siERCC1 significantly increased the inhibitory effect on SKOV3 cells, possibly due to its amelioration of drug resistance to DDP.

Ovarian cancer is a heterogeneous disease categorized into several major morphological subtypes: Epithelial carcinoma, germ cell tumor, sex cord-stromal tumor and Krukenberg's tumor (2,25,27). The most common subtype is serous ovarian carcinoma (28). The present study focused on the inhibition ratio of the recombinant oncolytic adenovirus (Ad-siERCC1) on SKOV3 cells, derived from serous cystadenoma carcinoma. The results revealed that Ad-siERCC1 had a marked ability to kill tumor cells. However, the effect of Ad-siERCC1 on other subtypes of ovarian tumors was not assessed. Considering that the subtype of ovarian cancer varies with the age of onset and epidemiological differences exist between races and countries due to various factors, including genetics and economic factors (2,25,27), it is essential to investigate the effect of Ad-siERCC1 on different subtypes of ovarian cancer in future research.

Viral vectors in gene therapy in the treatment of ovarian cancer, according to a review by Lundstrom in 2023 (19); the five projects listed involve four vector systems, including p-dpp-VSVMP [a nanoparticle that carries the gene for the matrix protein of the vesicular stomatitis virus (VSVMP) and encapsulates paclitaxel] (29), VSVMP (the gene for VSVMP enclosed in a lipid complex) (30), SIN AR339 (an RNA virus belonging to the Togavirus genus) (31) and MV-CEA (a measles virus Edmonston vaccine strain expressing the CEA peptide) (32). The p-dpp-VSVMP was found to cause tumor regression in both subcutaneous and orthotopic ovarian cancer mouse models. When combined with the JAK1/2 inhibitor ruxolitinib, the treatment outcome was further improved. The VSVMP system demonstrated a high tumor suppression rate of 87–98% in mouse tumor xenografts models, leading to an increase in survival. These non-replicating vector particles induce apoptosis in cancer cells by disrupting cell cytoskeletal elements and suppressing host cell gene expression. Another study (31) found that intraperitoneal injection of the oncolytic adenovirus SIN AR339 led to tumor cell killing and regression in ovarian cancer. In clinical applications, the MV-CEA particle has been evaluated in a phase I clinical trial for recurrent ovarian cancer patients (32). This treatment showed no dose-limiting toxicities and all nine patients who received the treatment achieved disease stabilization. The median overall survival rate for these patients was 12.15 months, twice the expected survival time. However, this treatment is only suitable for patients with normal CEA levels. Compared to these studies, our research still has a long way to go, is focusing on exploring the application of ERCC1 gene in oncolytic adenovirus therapy. It is well known that ERCC1 is the most critical factor contributing to platinum resistance in ovarian cancer and is one of the most studied DNA repair genes (33). ERCC1, located on human chromosome 19, is involved in the cleavage and recognition of DNA chains damaged by platinum-based drugs. The level of ERCC1 mRNA reflects the ability of tumor tissue to repair DNA helix twist induced by platinum-based drugs and serves as an important indicator of tumor patients prognosis and the effectiveness of platinum-based chemotherapy (12). In the present study, interference with ERCC1 gene expression significantly improved the efficiency of DDP chemotherapy, consistent with numerous studies showing that higher ERCC1 expression in patients receiving platinum-based chemotherapy is associated with worse chemotherapy outcomes and patient prognosis (8,14,15). However, Bösmüller et al (34) reported that ERCC1 expression is not related to platinum chemosensitivity in ovarian cancer patients. The discrepancies could be attributed to limited small-scale and small-sample research on chemotherapy and ERCC1 gene or protein expression in ovarian cancer, lacking large-sample multicenter clinical studies.

Although oncolytic viruses are considered a promising new class of anticancer drugs, they present unique challenges. Oncolytic viruses are live viruses that proliferate upon administration, resulting in variable effective doses (35). Currently, limited data are available on correlating viral dose with in vivo replicative potential and therapeutic response. Further investigations relating viral replication to clinical response are crucial. Moreover, while a number of clinical trials have shown that replication-defective and replication-competent adenovirus vectors are safe and have therapeutic activity (3639), the replicative potential of oncolytic viruses and immunoreaction require further study. Additionally, the safety and immunological response to oncolytic viruses still need to be fully evaluated. The present study constructed a novel Dual-regulated oncolytic adenovirus carrying ERCC1-siRNA gene that can infect and replicate in ovarian cancer cells. This recombinant oncolytic adenoviruses effectively suppressed cancer cell proliferation, especially when combined with DDP. The findings of the present study suggested that the recombinant oncolytic adenovirus exerts its anti-cancer effect by manipulating the PI3K/Akt signaling pathway, a well-known survival and growth pathway in cancer cells (40,41). However, further investigation is required to confirm these results and to address any potential safety concerns or off-target effects. Overall, oncolytic viruses show great potential in the treatment of ovarian cancer, but more research is needed to optimize their use in clinical settings.

Acknowledgements

Not applicable.

Funding

The present study was supported by Jiading District Health Commission of Shanghai Youth fund (grant no. 2018-QN-01).

Availability of data and materials

The data generated in the present study are included in the figures of this article.

Authors' contributions

WY, YM, LX, TZ initiated and directed the study; TZ, RZ, XZ, QS performed the research and wrote the first draft of the manuscript; TZ, XX, WC analyzed the data. WY and YM confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Yang L, Xie HJ, Li YY, Wang X, Liu XX and Mai J: Molecular mechanisms of platinum-based chemotherapy resistance in ovarian cancer (Review). Oncol Rep. 47:822022. View Article : Google Scholar : PubMed/NCBI

2 

Penny SM: Ovarian cancer: An overview. Radiol Technol. 91:561–575. 2020.PubMed/NCBI

3 

Wallis B, Bowman KR, Lu P and Lim CS: The challenges and prospects of p53-Based therapies in ovarian cancer. Biomolecules. 13:1592023. View Article : Google Scholar : PubMed/NCBI

4 

Terp SK, Stoico MP, Dybkær K and Pedersen IS: Early diagnosis of ovarian cancer based on methylation profiles in peripheral blood cell-free DNA: A systematic review. Clin Epigenetics. 15:242023. View Article : Google Scholar : PubMed/NCBI

5 

Chandrasekaran A and Elias KM: Synthetic lethality in ovarian cancer. Mol Cancer Ther. 20:2117–2128. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Zhang XW, Wu YS, Xu TM and Cui MH: CAR-T cells in the treatment of ovarian cancer: A promising cell therapy. Biomolecules. 13:4652023. View Article : Google Scholar : PubMed/NCBI

7 

Moufarrij S, Dandapani M, Arthofer E, Gomez S, Srivastava A, Lopez-Acevedo M, Villagra A and Chiappinelli KB: Epigenetic therapy for ovarian cancer: Promise and progress. Clin Epigenetics. 11:72019. View Article : Google Scholar : PubMed/NCBI

8 

Zhang C, Gao S and Hou J: ERCC1 expression and platinum chemosensitivity in patients with ovarian cancer: A meta-analysis. Int J Biol Markers. 35:12–19. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Jiang M, Jia K, Wang L, Li W, Chen B, Liu Y, Wang H, Zhao S, He Y and Zhou C: Alterations of DNA damage response pathway: Biomarker and therapeutic strategy for cancer immunotherapy. Acta Pharm Sin B. 11:2983–2994. 2021. View Article : Google Scholar : PubMed/NCBI

10 

Hamilton G and Rath B: Pharmacogenetics of platinum-based chemotherapy in non-small cell lung cancer: Predictive validity of polymorphisms of ERCC1. Expert Opin Drug Metab Toxicol. 14:17–24. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Zazuli Z, Vijverberg S, Slob E, Liu G, Carleton B, Veltman J, Baas P, Masereeuw R and Maitland-van der Zee AH: Genetic variations and cisplatin nephrotoxicity: A systematic review. Front Pharmacol. 9:11112018. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Tang N, Lyu D, Zhang Y and Liu H: Association between the ERCC1 polymorphism and platinum-based chemotherapy effectiveness in ovarian cancer: A meta-analysis. BMC Womens Health. 17:432017. View Article : Google Scholar : PubMed/NCBI

14 

Rosell R, Taron M, Camps C and López-Vivanco G: Influence of genetic markers on survival in non-small cell lung cancer. Drugs Today (Barc). 39:775–786. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Lenz HJ: Pharmacogenomics and colorectal cancer. Adv Exp Med Biol. 587:211–231. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Li W, Jie Z, Li Z, Liu YI, Gan Q, Mao Y and Wang X: ERCC1 siRNA ameliorates drug resistance to cisplatin in gastric carcinoma cell lines. Mol Med Rep. 9:2423–2428. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Wilson JM and Engelhardt J: Adenovirus vectors for gene therapy. Biotechnol Adv. 15:7691997. View Article : Google Scholar

18 

Berkey SE, Thorne SH and Bartlett DL: Oncolytic virotherapy and the tumor microenvironment. Adv Exp Med Biol. 1036:157–172. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Lundstrom K: Viral vectors in gene therapy: Where do we stand in 2023? Viruses. 15:6982023. View Article : Google Scholar : PubMed/NCBI

20 

Stella GM, Marchiò C, Bari E, Ferrarotti I, Bertuccio FR, Di Gennaro A, Abbott DM, Putignano P, Campo I, Torre ML and Corsico AG: The genes-stemness-secretome interplay in malignant pleural mesothelioma: Molecular dynamics and clinical hints. Int J Mol Sci. 24:34962023. View Article : Google Scholar : PubMed/NCBI

21 

Strapcova S, Takacova M, Csaderova L, Martinelli P, Lukacikova L, Gal V, Kopacek J and Svastova E: Clinical and pre-clinical evidence of carbonic anhydrase IX in pancreatic cancer and its high expression in pre-cancerous lesions. Cancers (Basel). 12:20052020. View Article : Google Scholar : PubMed/NCBI

22 

Bao MH and Wong CC: Hypoxia, metabolic reprogramming and drug resistance in liver cancer. Cells. 10:17152021. View Article : Google Scholar : PubMed/NCBI

23 

Kopecka J, Salaroglio IC, Perez-Ruiz E, Sarmento-Ribeiro AB, Saponara S, De Las Rivas J and Riganti C: Hypoxia as a driver of resistance to immunotherapy. Drug Resist Updat. 59:1007872021. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Loren P, Saavedra N, Saavedra K, De Godoy Torso N, Visacri MB, Moriel P and Salazar LA: Contribution of MicroRNAs in chemoresistance to cisplatin in the top five deadliest cancer: An updated review. Front Pharmacol. 13:8310992022. View Article : Google Scholar : PubMed/NCBI

26 

Zhao T, Bai J, Zou Q, Chen F and Xie Y: Insulin in combination with cisplatin induces the apoptosis of ovarian cancer cells via p53 and JNK activation. Mol Med Rep. 16:9095–9101. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Lheureux S, Gourley C, Vergote I and Oza AM: Epithelial ovarian cancer. Lancet. 393:1240–1253. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Lheureux S, Braunstein M and Oza AM: Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA Cancer J Clin. 69:280–304. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Long J, Yang Y, Kang T, Zhao W, Cheng H, Wu Y, Du T, Liu B, Li Y, Luo F and Gou M: Ovarian cancer therapy by VSVMP gene mediated by a paclitaxel-enhanced nanoparticle. ACS Appl Mater Interfaces. 9:39152–39164. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Zhong Q, Wen YJ, Yang HS, Luo H, Fu AF, Yang F, Chen LJ, Chen X, Qi XR, Lin HG, et al: Efficient inhibition of cisplatin-resistant human ovarian cancer growth and prolonged survival by gene transferred vesicular stomatitis virus matrix protein in nude mice. Ann Oncol. 19:1584–1591. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Unno Y, Shino Y, Kondo F, Igarashi N, Wang G, Shimura R, Yamaguchi T, Asano T, Saisho H, Sekiya S and Shirasawa H: Oncolytic viral therapy for cervical and ovarian cancer cells by Sindbis virus AR339 strain. Clin Cancer Res. 11:4553–4560. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Galanis E, Hartmann LC, Cliby WA, Long HJ, Peethambaram PP, Barrette BA, Kaur JS, Haluska PJ Jr, Aderca I, Zollman PJ, et al: Phase I trial of intraperitoneal administration of an oncolytic measles virus strain engineered to express carcinoembryonic antigen for recurrent ovarian cancer. Cancer Res. 70:875–882. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Chebouti I, Kuhlmann JD, Buderath P, Weber S, Wimberger P, Bokeloh Y, Hauch S, Kimmig R and Kasimir-Bauer S: ERCC1-expressing circulating tumor cells as a potential diagnostic tool for monitoring response to platinum-based chemotherapy and for predicting post-therapeutic outcome of ovarian cancer. Oncotarget. 8:24303–24313. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Bösmüller H, Haitchi-Petnehazy S, Webersinke G, Marschon R, Roithmeier F, Stummvoll W, Fehm T, Klier-Richter M, Bonzheim I, Staebler A and Fend F: Intratumoral lymphocyte density in serous ovarian carcinoma is superior to ERCC1 expression for predicting response to platinum-based therapy. Virchows Arch. 459:183–191. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Innao V, Rizzo V, Allegra AG, Musolino C and Allegra A: oncolytic viruses and hematological malignancies: A new class of immunotherapy drugs. Curr Oncol. 28:159–183. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Chintala NK, Choe JK, McGee E, Bellis R, Saini JK, Banerjee S, Moreira AL, Zauderer MG, Adusumilli PS and Rusch VW: Correlative analysis from a phase I clinical trial of intrapleural administration of oncolytic vaccinia virus (Olvi-vec) in patients with malignant pleural mesothelioma. Front Immunol. 14:11129602023. View Article : Google Scholar : PubMed/NCBI

37 

Vijver SV, Danklmaier S, Pipperger L, Gronauer R, Floriani G, Hackl H, Das K and Wollmann G: Prediction and validation of murine MHC class I epitopes of the recombinant virus VSV-GP. Front Immunol. 13:11007302023. View Article : Google Scholar : PubMed/NCBI

38 

Cohn DE, Sill MW, Walker JL, O'Malley D, Nagel CI, Rutledge TL, Bradley W, Richardson DL, Moxley KM and Aghajanian C: Randomized phase IIB evaluation of weekly paclitaxel versus weekly paclitaxel with oncolytic reovirus (Reolysin) in recurrent ovarian, tubal, or peritoneal cancer: An NRG Oncology/Gynecologic Oncology Group study. Gynecol Oncol. 146:477–483. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Moreno V, Barretina-Ginesta MP, García-Donas J, Jayson GC, Roxburgh P, Vázquez RM, Michael A, Antón-Torres A, Brown R, Krige D, et al: Safety and efficacy of the tumor-selective adenovirus enadenotucirev with or without paclitaxel in platinum-resistant ovarian cancer: A phase 1 clinical trial. J Immunother Cancer. 9:e0036452021. View Article : Google Scholar : PubMed/NCBI

40 

Liao H, Zhang L, Lu S, Li W and Dong W: KIFC3 promotes proliferation, migration, and invasion in colorectal cancer via PI3K/AKT/mTOR signaling pathway. Front Genet. 13:8489262022. View Article : Google Scholar : PubMed/NCBI

41 

Hinz N and Jücker M: Distinct functions of AKT isoforms in breast cancer: A comprehensive review. Cell Commun Signal. 17:1542019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2024
Volume 30 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao T, Ye W, Zhang R, Zhu X, Shi Q, Xu X, Chen W, Xu L and Meng Y: Dual‑regulated oncolytic adenovirus carrying <em>ERCC1</em>‑siRNA gene possesses potent antitumor effect on ovarian cancer cells. Mol Med Rep 30: 120, 2024
APA
Zhao, T., Ye, W., Zhang, R., Zhu, X., Shi, Q., Xu, X. ... Meng, Y. (2024). Dual‑regulated oncolytic adenovirus carrying <em>ERCC1</em>‑siRNA gene possesses potent antitumor effect on ovarian cancer cells. Molecular Medicine Reports, 30, 120. https://doi.org/10.3892/mmr.2024.13245
MLA
Zhao, T., Ye, W., Zhang, R., Zhu, X., Shi, Q., Xu, X., Chen, W., Xu, L., Meng, Y."Dual‑regulated oncolytic adenovirus carrying <em>ERCC1</em>‑siRNA gene possesses potent antitumor effect on ovarian cancer cells". Molecular Medicine Reports 30.1 (2024): 120.
Chicago
Zhao, T., Ye, W., Zhang, R., Zhu, X., Shi, Q., Xu, X., Chen, W., Xu, L., Meng, Y."Dual‑regulated oncolytic adenovirus carrying <em>ERCC1</em>‑siRNA gene possesses potent antitumor effect on ovarian cancer cells". Molecular Medicine Reports 30, no. 1 (2024): 120. https://doi.org/10.3892/mmr.2024.13245