Combined RNAi targeting human Stat3 and ADAM9 as gene therapy for non‑small cell lung cancer

Retraction in: /10.3892/ol.2022.13595

  • Authors:
    • Liang Chang
    • Fangchao Gong
    • Hongfei Cai
    • Zhihong Li
    • Youbin Cui
  • View Affiliations

  • Published online on: December 9, 2015     https://doi.org/10.3892/ol.2015.4018
  • Pages: 1242-1250
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Previous studies have demonstrated that human signal transducer and activator of transcription 3 (Stat3) and disintegrin and metalloproteinase 9 (ADAM9) are promising targets for RNA interference (RNAi)‑based gene therapy for human non‑small cell lung cancer (NSCLC). Thus, in the present study, the recombinant lentiviral (Lv) small hairpin (sh)RNA expression plasmids Lv/sh‑Stat3 and Lv/sh‑ADAM9, which targeted Stat3 and ADAM9, respectively, were constructed and subsequently infected into the A549 human NSCLC cell line. Cell proliferation, migration, invasion and apoptosis were determined in vitro in A549 cells following treatment with Lv/sh‑Stat3 or Lv/sh‑ADAM9 alone or in combination. In addition, the combined effect of Lv/sh‑Stat3 and Lv/sh‑ADAM9 gene therapy was evaluated in vivo using A549 xenograft models in nude mice. The in vitro experiments demonstrated that A549 cells treated with a combination of Lv/sh‑Stat3 and Lv/sh‑ADAM9 exhibited a significant additive effect in their cell proliferation, migration, invasion and apoptosis abilities, compared with A549 cells treated with Lv/sh‑Stat3 or Lv/sh‑ADAM9 alone. The in vivo experiments conducted in A549 xenograft tumor mouse models revealed that the combined treatment with Lv/sh‑Stat3 and Lv/sh‑ADAM9 exerted an additive effect on tumor growth inhibition, compared with the treatment with Lv/sh‑Stat3 or Lv/sh‑ADAM9 alone. These results suggested that combined RNAi gene therapy targeting human Stat3 and ADAM9 may be a novel and promising strategy for the treatment of NSCLC.

Introduction

Lung cancer is the most common cause of cancer-associated mortality globally, and non-small cell lung cancer (NSCLC) is responsible for ~85% of cases of lung cancer (1,2). Despite the wide utilization of chemotherapy and radiotherapy for the treatment of advanced NSCLC, patient outcomes remain poor, with only <15% of patients surviving >5 years following diagnosis (3). Metastasis is a major cause of morbidity and mortality in patients with NSCLC (4). Chemotherapy is the primary treatment for metastatic NSCLC (5). However, commonly used cytotoxic chemotherapeutic agents frequently display narrow therapeutic indices due to nonspecific cytotoxicity, undesirable side effects and intrinsic or acquired chemoresistance, which results in numerous cases of NSCLC being regarded as incurable (2). Therefore, the development of novel and effective therapeutic strategies for the treatment of NSCLC is required.

Signal transducer and activator of transcription 3 (Stat3), a member of the STAT family, is capable of regulating the expression of target genes implicated in cell cycle progression, apoptosis, promotion of cellular transformation and aberrant cell proliferation (6). Therefore, Stat3 represents an attractive target for therapeutic intervention. Previous studies using RNA interference (RNAi), dominant negative Stat3 and small molecule inhibitors have demonstrated that the inhibition of Stat3 signaling suppresses tumor cell proliferation and invasion, induces apoptosis in vitro and delays tumor growth in animal models of breast, myeloma, prostate, head and neck, liver, pancreatic and lung cancer (615). A recent study demonstrated that small interfering (si)RNA-mediated downregulation of Stat3 markedly inhibited NSCLC tumor growth and increased the sensitivity of tumor cells to certain drugs (16), which suggests that Stat3 may be a potential target for the treatment of NSCLC.

A number of studies have identified disintegrin and metalloproteinase 9 (ADAM9) as a potential target for anticancer therapy (17,18). A previous study on lung cancer demonstrated that the overexpression of ADAM9 was able to enhance the adhesion and invasion abilities of NSCLC cells by modulating certain adhesion molecules and altering the sensitivity of NSCLC cells to growth factors, thereby promoting their metastatic capacity to the brain (19). It has been previously demonstrated that ADAM9 RNAi-based gene therapy is capable of inhibiting adenoid cystic carcinoma cell growth and metastasis in vitro and in vivo (20). In addition, a previous study by Chang et al (21) revealed that downregulating the expression of ADAM9 in A549 tumor cells via an RNA silencing approach significantly inhibited cell proliferation, migration and invasion, and induced cell apoptosis in vitro, in addition to suppressing tumor growth in vivo in an experimental mouse model.

The onset and progression of tumors is a complex multistep process (22). Therefore, it is difficult to treat a tumor using a single therapeutic gene (21,23). Stat3 and ADAM9 are promising targets for cancer gene therapy. However, to the best of our knowledge, the simultaneous targeting of these two genes as a therapeutic strategy for the treatment of lung cancer has not been reported thus far. Therefore, the aim of the present study was to evaluate the therapeutic potential of combined RNAi gene therapy targeting Stat3 and ADAM9 for the treatment of NSCLC in vitro and in vivo.

Materials and methods

Cell culture

The human NSCLC A549 and human embryonic kidney (HEK) 293T cell lines were obtained from the Cell Bank of Type Culture Collection of the Chinese Academy of Sciences of the Shanghai Institute of Cell Biology (Shanghai, China), and were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific Inc., Waltham, MA, USA) supplemented with heat-inactivated 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific Inc.) at 37°C in a humidified atmosphere containing 5% CO2.

Construction of Stat3 and ADAM9 small hairpin (sh)RNA lentiviral (Lv) vectors and cell infection

siRNA target design tools from Ambion® (Thermo Fisher Scientific, Inc.) were utilized to design Stat3-, ADAM9- and negative control Scramble-shRNA sequences. The synthesized oligonucleotides (Takara Bio, Dalian, China), which contained a specific target sequence, a loop, the reverse complementary sequence of the target sequence, a stop codon for the U6 promoter and two sticky ends, were annealed and ligated into the NheI/PacI-linearized pFH-L vector (Shanghai Hollybio, Shanghai, China). The target sequences in the oligonucleotides for suppressing the expression of Stat3 and ADAM9 were 5′-TTCAGACCCGTCAACAAA-3′ and 5′-GGCGGGATTAATGTGTTT-3′, respectively, while the sequence of the scramble shRNA was 5′-AATTCTCCGAACGTGTCACGT-3′. The sequences of the recombinant lentivirus-based shRNA-expressing vectors were confirmed by DNA sequencing (Takara Bio). These vectors were subsequently transfected alongside packaging vectors into HEK293T cells, using Lipofectamine™ 2000 (Invitrogen, Thermo Fisher Scientific, Inc.), in order to generate lentiviruses. At 72 h post-transfection, the lentiviruses were collected and purified using ultracentrifugation (Sigma 3–18K centrifuge; Sigma-Aldrich, St. Louis, MO, USA) at 17,000 × g for 30 min, and their titer was determined using a GenesysTM15 ultraviolet spectrophotometer (Thermo Fisher Scientific, Inc.).

For lentivirus infection, 3×105 A549 cells were cultured in 6-well plates and incubated for 24 h prior to be infected. Next, Stat3 shRNA lentivirus (Lv/sh-Stat3), ADAM9 shRNA lentivirus (Lv/sh-ADAM9) or scramble shRNA lentivirus (Lv/sh-Scramble) at a multiplicity of infection (MOI) of 100, or a combination of Lv/sh-Stat3 and Lv/sh-ADAM9 at an MOI of 100 (Lv/sh-Stat3 MOI, 50 and Lv/sh-ADAM9 MOI, 50), was added to the cells. Mock-infected A549 cells treated with PBS served as negative control. Following incubation for 3 days, the infected cells were observed under a fluorescence microscope (BX51; Olympus Corporation, Tokyo, Japan). The subsequent experiments were performed using viruses at the aforementioned MOIs, unless indicated otherwise.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

Following infection with the lentivirus constructs, total RNA was extracted from A549 cells 5 days later using TRIzol® (Invitrogen, Thermo Fisher Scientific, Inc.). Complementary DNA was synthesized from total RNA using M-MLV Reverse Transcriptase (Promega Corporation, Madison, WI, USA) with random primers (Takara Bio), according to the manufacturer's protocol. RT-qPCR analysis was performed using SYBR® Green PCR Master Mix Kit (Applied Biosystems, Thermo Fisher Scientific, Inc.) on CFX Connect™ Real-Time PCR Detection System (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The primer sequences utilized were as follows: Stat3 sense, 5′-ACCTGCAGCAATACCATTGAC-3′ and antisense, 5′-AAGGTGAGGGACTCAACTGC-3′; ADAM9 sense, 5′-TGTGGGAACAGTGTGTTCAAGGA-3′ and antisense, 5′-CCAATTCATGAGCAACAATGGAAG-3′; and β-actin sense, 5′-GTGGACATCCGCAAAGAC-3′ and antisense, 5′-AAAGGGTGTAACGCAACTA-3′. Amplification of Stat3, ADAM9 and β-actin was performed with 1 cycle at 95°C for 5 min, followed by 40 cycles of 95°C for 15 sec and 58°C for 60 sec. For relative quantification, 2−ΔΔCq was calculated and used as an indication of the relative expression levels of Stat3 and ADAM9, which were calculated by subtracting the Cq values of the control gene (β-actin) from the Cq values of the Stat3 and ADAM9 genes.

Western blotting

A549 cells were collected 5 days following infection with the lentivirus constructs, and were subsequently lysed in radioimmunoprecipitation assay lysis buffer (Sigma-Aldrich). Protein concentrations were determined using Protein Assay Dye Reagent Concentrate (Bio-Rad Laboratories, Inc.). An identical quantity of protein (20 µg/lane) was next loaded into each lane and separated by 10–15% sodium dodecyl sulfate-polyacrylamide gel (Bio-Rad Laboratories, Inc.) electrophoresis, prior to being transferred to polyvinylidene difluoride membranes (Bio-Rad Laboratories, Inc.). Blocking was performed using 5% skimmed milk for 2 h at room temperature. The membranes were subsequently incubated overnight at 4°C with the following antibodies: Monoclonal mouse anti-human Stat3 (cat. no. sc-8019; 1:2,000; Santa Cruz Biotechnology, Inc., Dallas, TX, USA), monoclonal mouse anti-human ADAM9 (cat. no. sc-23290; 1:2,000; Santa Cruz Biotechnology, Inc.), monoclonal mouse anti-human matrix metalloproteinase (MMP)-2 (cat. no. 13132; 1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA) and monoclonal mouse anti-human MMP-9 (cat. no. sc-21773; 1:2,000; Santa Cruz Biotechnology, Inc.). Monoclonal mouse anti-human β-actin (cat. no. sc-47778; 1:10,000; Santa Cruz Biotechnology, Inc.) served as a loading control. Following washing with PBS with Tween 20 (Sigma-Aldrich) twice and incubation with goat anti-mouse horseradish peroxidase-conjugated IgG secondary antibody (cat. no. sc-2005; 1:5,000; Santa Cruz Biotechnology, Inc.) for 2 h at room temperature, the blotted proteins were detected using an enhanced chemiluminescence system (EMD Millipore, Billerica, MA, USA).

Cell proliferation assay

In order to measure the effect of the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 on cell proliferation in NSCLC cells, a cell proliferation assay was performed with Cell Counting Kit-8 (CCK-8) (Dojindo Molecular Technologies, Inc., Kumamoto, Japan). In brief, 1×105 A549 cells in serum-free DMEM were infected with Lv/sh-Stat3 or Lv/sh-ADAM9 at an MOI of 100, or underwent combined infection with Lv/sh-Stat3 and Lv/sh-ADAM9 at an MOI of 100 (Lv/sh-Stat3 MOI, 50 and Lv/sh-ADAM9 MOI, 50). The virus-containing medium was removed 8 h later and replaced with fresh DMEM medium containing 10% FBS. At 48 h post-infection, cell proliferation was measured by CCK-8 assay, according to the manufacturer's protocol. The experiment was performed ≥3 times, and similar results were achieved with each replicate.

Apoptosis analysis

In order to measure the effect of the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 on the apoptosis of A549 cells, a terminal deoxynucleotidyl transferase 2′-deoxyuridine, 5′-triphosphate nick end labeling (TUNEL) assay was performed. In brief, 1×104 A549 cells in serum-free DMEM were infected with Lv/sh-Stat3 or Lv/sh-ADAM9 at an MOI of 100, or underwent combined infection with Lv/sh-Stat3 and Lv/sh-ADAM9 at an MOI of 100 (Lv/sh-Stat3 MOI, 50 and Lv/sh-ADAM9 MOI, 50) for 48 h. Cellular DNA fragmentation was measured with ApopTag® Red In Situ Apoptosis Detection Kit (EMD Millipore), according to the manufacturer's protocol. In order to quantify the number of apoptotic cells, TUNEL+ cells were counted using a confocal microscope (FV100; Olympus Corporation).

Furthermore, the activity of caspase-3, −8 and −9 was determined as an additional indicator of apoptosis, using the corresponding Caspase Colorimetric Protease Assay Kit (EMD Millipore), as previously described (24). The relative caspase activity of the control group was normalized to 100.

Wound-healing assay

A wound-healing assay was performed to assess the effect of the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 on cell migration. Briefly, A549 cells infected with Lv/sh-Stat3 or Lv/sh-ADAM9 alone at an MOI of 100, A549 cells that had undergone combined infection with Lv/sh-Stat3 and Lv/sh-ADAM9 at an MOI of 100 (Lv/sh-Stat3 MOI, 50 and Lv/sh-ADAM9 MOI, 50) and untreated cells were incubated in 6-cm dishes, at a density of 1.5×106 cells/dish, and cultured for 24 h. A linear wound was then created by scratching the monolayer of confluent cells with a 100-µl pipette tip. The monolayer of scratched cells was next washed with phosphate-buffered saline (PBS), and 24 h later, the area of migration was evaluated under a light microscope (X51; Olympus Corporation). All experiments were performed in triplicate.

Transwell migration assay

Transwell migration chambers (8-µm pore filter; BD Biosciences, Franklin Lakes, NJ, USA) were coated with Matrigel™ Basement Membrane Matrix (BD Biosciences) and incubated at 37°C for 4 h to allow solidification. Following 24-h incubation with Lv/sh-Stat3 and Lv/sh-ADAM9 alone or in combination, 2×105 A549 cells suspended in serum-free DMEM were added to the upper chamber, and DMEM containing 10% FBS was added to the lower chamber as a chemoattractant. Non-invading cells were gently removed 48 h later, using cotton swabs (Sigma-Aldrich), and invasive cells located on the lower surface of the chamber were then stained with 0.1% crystal violet (Sigma-Aldrich) dissolved in 20% methanol (Sigma-Aldrich). Cell invasiveness was determined by counting the number of penetrating cells in ten random high-power fields under a phase-contrast microscope (A19.2703; Nikon Corporation, Tokyo, Japan) at ×200 magnification.

Tumor xenograft assay

A total of 50 male BALB/c nude mice (~6-week-old) were purchased from the Animal Center of Norman Bethune College of Medicine of Jilin University (Changchun, China). All animals were maintained in pathogen-free conditions at room temperature with free access to food and water and exposed to a 12 h light/dark cycle, in accordance with the protocols approved by the ethics committee of the Disease Model Research Center of Jilin University.

In vitro cultured A549 cells were harvested, and a tumorigenic dose of 2×106 cells was injected intraperitoneally into BALB/c mice. Tumor volume was calculated using the following formula: Volume = length × width2/2. When tumors reached an average volume of 110 mm3, the mice were randomly divided into control (untreated group), Lv/sh-Scramble, Lv/sh-Stat3, Lv/sh-ADAM9 and combined Lv/sh-Stat3 plus Lv/sh-ADAM9 groups (n = 10 mice/group). In the treated groups, the animals were injected once a week for 3 weeks with 5×108 plaque-forming units of Lv/sh-Scramble, Lv/sh-Stat3, Lv/sh-ADAM9 or combined Lv/sh-Stat3 and Lv/sh-ADAM9, diluted in 20 µl PBS. The control group was administered 20 µl PBS once a week for 3 weeks. Tumor volume was measured prior to injection, and at 7, 14 and 21 days post-injection. On day 21 of the treatment, the animals were sacrificed, and the tumors were resected and weighed. A sample of tumor tissue from each group was immediately fixed for TUNEL analysis, according to the manufacturer's protocol.

Statistical analysis

Data are expressed as the mean ± standard deviation. Statistical differences between groups were evaluated using analysis of variance, followed by Tukey's post hoc test. GraphPad Prism version 5.0 (GraphPad Software, Inc., La Jolla, CA, USA) and SPSS® version 19.0 (IBM SPSS, Armonk, NY, USA) for Windows® 7.0 (Microsoft Corporation, Redmond, WA, USA) were used for statistical analyses. P<0.05 was considered to indicate a statistically significant difference.

Results

Combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 inhibits Stat3 and ADAM9 expression in A549 cells

Lv/sh-Stat3 and Lv/sh-ADAM9, alone or in combination, were infected into A549 cells. At 5-days post-infection, the mRNA and protein expression levels of Stat3 and ADAM9 were determined using RT-qPCR and western blotting, respectively. The mRNA and protein expression levels of Stat3 were observed to be reduced in the Lv/sh-Stat3 and combined treatment groups, while the mRNA and protein expression levels of ADAM9 were reduced in the Lv/sh-ADAM9 and combined treatment groups, compared with the control and Lv/sh-Scramble groups (P<0.05; Fig. 1). These results suggested that the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 was able to specifically and significantly inhibit the expression of Stat3 and ADAM9 in A549 cells.

Combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 inhibits cell proliferation in A549 cells

In order to assess the effect of the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 on cell proliferation, a CCK-8 assay was performed on A549 cells at 48 h post-infection. Fig. 2 reveals that the treatment with Lv/sh-Scramble did not alter cell proliferation (P>0.05), while the treatment with Lv/sh-Stat3 or Lv/sh-ADAM9 alone significantly inhibited cell proliferation, compared with the untreated and Lv/sh-Scramble-treated groups. Combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 demonstrated an additive effect on the inhibition of cell proliferation, compared with treatment with Lv/sh-Stat3 or Lv/sh-ADAM9 alone (P<0.05; Fig. 2).

Combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 induces cell apoptosis in A549 cells

In order to investigate the effect of the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 on the apoptosis of A549 cells, a TUNEL assay was performed. As demonstrated in Fig. 3A, treatment with Lv/sh-Stat3 or Lv/sh-ADAM9 alone significantly increased cell apoptosis, compared with the control and Lv/sh-Scramble groups. Furthermore, a significant enhancement of apoptosis was observed in A549 cells treated with a combination of Lv/sh-Stat3 and Lv/sh-ADAM9.

The effects of the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 on the activity of caspase-3, −8 and −9 were analyzed in A549 cells. As revealed in Fig. 3B–D, the activity of caspase-3, −8 and −9 was significantly increased in the groups treated with Lv/sh-Stat3 or Lv/sh-ADAM9 alone, compared with that of the control and Lv/sh-Scramble groups (P<0.05). The combined treatment group exhibited the most significant increase in activity, compared with the Lv/sh-Stat3 or Lv/sh-ADAM9 alone treatment groups.

Combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 inhibits cell migration and invasion in A549 cells

A wound-healing assay was performed to investigate the effects of the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 on the migration ability of A549 cells. As indicated in Fig. 4A, A549 cells infected with Lv/sh-Stat3 or Lv/sh-ADAM9 alone migrated significantly less than A549 cells infected with Lv/sh-Scramble (P<0.05). The most remarkable reduction in migration was observed in A549 cells infected with a combination of Lv/sh-Stat3 and Lv/sh-ADAM9, compared with A549 cells treated with Lv/sh-Stat3 or Lv/sh-ADAM9 alone (P<0.05; Fig. 4A).

Subsequently, the ability of the Lv/sh-Stat3 plus Lv/sh-ADAM9 combined treatment to reduce the invasiveness of A549 cells was investigated by transwell assay. The invasion ability of A549 cells was significantly reduced in the Lv/sh-Stat3 and Lv/sh-ADAM9 alone treatment groups, compared with the control and Lv/sh-Scramble groups (P<0.05; Fig. 4B). A synergistic inhibition of invasion was observed in A549 cells subjected to combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 (Fig. 4B).

MMP-2 and MMP-9 participate in cell migration and invasion

The protein expression levels of MMP-2 and MMP-9 in A549 cells infected with Lv/sh-Stat3 and Lv/sh-ADAM9, alone or in combination, were investigated by western blotting. The results revealed a marked reduction in the protein levels of MMP-2 and MMP-9 in A549 cells treated with Lv/sh-Stat3 or Lv/sh-ADAM9 alone, compared with the control and Lv/sh-Scramble groups (P<0.05; Fig. 4C and D). The group subjected to combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 displayed the most significant reduction in the expression levels of MMP-2 and MMP-9, compared with the groups subjected to treatment with Lv/sh-Stat3 or Lv/sh-ADAM9 alone (P<0.05; Fig. 4C and D).

Combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 suppresses tumor growth in vivo

In order to test the potential utility of combined Lv/sh-Stat3 and Lv/sh-ADAM9 gene therapy for the treatment of NSCLS, pre-established A549 xenograft tumors were treated for 21 days with Lv/sh-Stat3 and Lv/sh-ADAM9, alone or in combination. On day 7 following the termination of the treatment, the animals were sacrificed, and the volume and weight of the tumors were determined. The tumor volume in the Lv/sh-Stat3 and Lv/sh-ADAM9 groups was significantly reduced at various time points, compared with the Lv/sh-Scramble and PBS control groups (P<0.05; Fig. 5A and B). The combined treatment group displayed a significant reduction in tumor volume, compared with the Lv/sh-Stat3 alone and the Lv/sh-ADAM9 alone groups (P<0.05; Fig. 5B). In addition, the tumor weight was reduced following treatment with Lv/sh-Stat3 or Lv/sh-ADAM9 alone (P<0.05), compared with tumors treated with Lv/sh-Scramble (Fig. 5A and C). Notably, the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 was more effective than the treatment with Lv/sh-Stat3 or Lv/sh-ADAM9 alone at an identical total dose of virus (P<0.05; Fig. 5A and C). Furthermore, the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 exerted an effect on tumor tissue cell apoptosis in vivo, as determined by TUNEL assay. As revealed in Fig. 5D, low levels of apoptosis were detected in the control and Lv/sh-Scramble groups, and a small number of apoptotic cells was observed in the Lv/sh-Stat3 alone and Lv/sh-ADAM9 alone groups, while significantly increased apoptosis was noticed in A549 cells following combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 (P<0.05; Fig. 5D). These results indicate that the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 was able to suppress tumor growth of NSCLC in mouse models.

Discussion

It has been previously proposed that gene therapy targeting human Stat3 or ADAM9 alone may inhibit tumor growth (21,23). However, to the best of our knowledge, the present study is the first report demonstrating that combined RNAi gene therapy targeting simultaneously human Stat3 and ADAM9 in NSCLC cells may lead to synergistic effects on cell proliferation and apoptosis in vitro. The most notable finding of the present study is that the majority of mice injected with NSCLC cells that received combined RNAi gene therapy targeting human Stat3 and ADAM9 experienced tumor growth inhibition. Therefore, this approach represents a novel strategy for the treatment of NSCLC, which, if adopted in clinics, may improve the therapeutic outcome of patients with NSCLC.

The development and progression of cancer involves multiple genes and factors (25). Therefore, targeting a number of genes simultaneously via combined treatment may be more effective for inhibiting cancer growth than targeting individual genes (24,2629). Hu et al (30) reported a considerable additive effect in tumor growth inhibition of hepatocellular carcinoma following combined RNAi gene therapy targeting human telomerase reverse transcriptase and epidermal growth factor receptor with pegylated immuno-lipopolyplexes as a gene carrier. Chen et al (31) identified that combined molecular-targeted repression of the cyclin D1 and B cell lymphoma (Bcl)-xL genes demonstrated increased effectiveness as a therapeutic strategy for the treatment of NSCLC in terms of promoting cell apoptosis and reducing cell proliferation, compared with the downregulation of cyclin D1 or Bcl-xL alone. Lai et al (32) identified that combined treatment with aptamer nucleic acid (aptNCL)-SLUGsiRNA and aptNCL-neuropilin 1siRNA was able to synergistically suppress lung cancer cell invasion, tumor growth and angiogenesis, via cancer-specific targeting combined with gene-specific silencing. Similarly, the results of the present study revealed that combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 was able to significantly inhibit the growth of NSCLC tumors in vitro and in vivo.

Metastasis is a major cause of morbidity and mortality in cancer (33). The process of tumor metastasis is complex and involves a series of events, including epithelial-mesenchymal transition, cancer cell migration, invasion, intravasation into the systemic circulation and subsequent adhesion to endothelial cells, followed by extravasation, colonization of distant organs and induction of angiogenesis (34). These consecutive events result in alterations in a large number of genes and signaling pathways (35). ADAM9, a member of the ADAM family, appears to be involved in lung cancer progression and metastasis (19,20). A previous study demonstrated that the downregulation of endogenous ADAM9 by RNAi was capable of inhibiting adenoid cystic carcinoma cell growth and metastasis in vitro and in vivo (20). In addition, previous studies have confirmed that Stat3 is involved in cancer migration and invasion, and blocking the activity of Stat3 has been observed to inhibit cancer cell migration and invasion (616). The results of the aforementioned studies suggest that gene therapy targeting human Stat3 or ADAM9 alone may be able to inhibit cancer cell migration and invasion. However, to the best of our knowledge, the present study constitutes the first report to demonstrate that the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 exerted an additive effect on the inhibition of NSCLC cell migration and invasion.

In conclusion, the present study provides evidence that the combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 in A549 cells is able to inhibit cell proliferation, migration and invasion, and induce cell apoptosis in vitro. In addition, the present study demonstrated that combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 was able to synergistically suppress the growth of tumors in mouse models. Therefore, the results of the present study suggest that combined treatment with Lv/sh-Stat3 and Lv/sh-ADAM9 may be a novel and effective therapeutic strategy for the treatment of human lung cancer.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Reungwetwattana T, Weroha SJ and Molina JR: Oncogenic pathways, molecularly targeted therapies, and highlighted clinical trials in non-small-cell lung cancer (NSCLC). Clin Lung Cancer. 13:252–266. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Temel JS, Greer JA, Muzikansky A, et al: Early palliative care for patients with metaSTATic non-small-cell lung cancer. N Engl J Med. 363:733–742. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Yano T, Okamoto T, Fukuyama S and Maehara Y: Therapeutic strategy for postoperative recurrence in patients with non-small cell lung cancer. World J Clin Oncol. 5:1048–1054. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Hsieh FC, Cheng G and Lin J: Evaluation of potential Stat3-regulated genes in human breast cancer. Biochem Biophys Res Commun. 335:292–299. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Catlett-Falcone R, Landowski TH, Oshiro MM, et al: Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 10:105–115. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Grandis JR, Drenning SD, Zeng Q, et al: Constitutive activation of Stat3 signaling abrogates apoptosis in squamous cell carcinogenesis in vivo. Proc Natl Acad Sci USA. 97:4227–4232. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Yan X, Fraser M, Qiu Q and Tsang BK: Over-expression of PTEN sensitizes human ovarian cancer cells to cisplatin-induced apoptosis in a p53-dependent manner. Gynecol Oncol. 102:348–355. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Epling-Burnette PK, Liu JH, Catlett-Falcone R, et al: Inhibition of Stat3 signaling leads to apoptosis of leukemic large granular lymphocytes and decreased Mcl-1 expression. J Clin Invest. 107:351–362. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Mora LB, Buettner R, Seigne J, et al: Constitutive activation of Stat3 in human prostate tumors and cell lines: Direct inhibition of Stat3 signaling induces apoptosis of prostate cancer cells. Cancer Res. 62:6659–6666. 2002.PubMed/NCBI

12 

Scholz A, Heinze S, Detjen KM, et al: Activated signal transducer and activator of transcription 3 (Stat3) supports the malignant phenotype of human pancreatic cancer. Gastroenterology. 125:891–905. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Kanda N, Seno H, Konda Y, et al: Stat3 is constitutively activated and supports cell survival in association with survivin expression in gastric cancer cells. Oncogene. 23:4921–4929. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Li L and Shaw PE: Autocrine-mediated activation of Stat3 correlates with cell proliferation in breast carcinoma lines. J Biol Chem. 277:17397–17405. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Song L, Turkson J, Karras JG, Jove R and Haura EB: Activation of Stat3 by receptor tyrosine kinases and cytokines regulates survival in human non-small cell carcinoma cells. Oncogene. 22:4150–4165. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Kulesza DW, Carré T, Chouaib S and Kaminska B: Silencing of the transcription factor Stat3 sensitizes lung cancer cells to DNA damaging drugs, but not to TNFα- and NK cytotoxicity. Exp Cell Res. 319:506–516. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Peduto L: ADAM9 as a potential target molecule in cancer. Curr Pharm Des. 15:2282–2287. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Duffy MJ, McKiernan E, O'Donovan N and McGowan PM: Role of ADAMs in cancer formation and progression. Clin Cancer Res. 15:1140–1144. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Shintani Y, Higashiyama S, Ohta M, et al: Overexpression of ADAM9 in non-small cell lung cancer correlates with brain metastasis. Cancer Res. 64:4190–4196. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Xu Q, Liu X, Cai Y, Yu Y and Chen W: RNAi-mediated ADAM9 gene silencing inhibits metastasis of adenoid cystic carcinoma cells. Tumour Biol. 31:217–224. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Chang L, Gong F and Cui Y: RNAi-mediated A disintegrin and metalloproteinase 9 gene silencing inhibits the tumor growth of non-small lung cancer in vitro and in vivo. Mol Med Rep. 12:1197–1204. 2015.PubMed/NCBI

22 

Wei EK, Wolin KY and Colditz GA: Time course of risk factors in cancer etiology and progression. J Clin Oncol. 28:4052–4057. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Chiu HC, Chou DL, Huang CT, et al: Suppression of Stat3 activity sensitizes gefitinib-resistant non small cell lung cancer cells. Biochem Pharmacol. 81:1263–1270. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Liu S, Zhang W, Liu K, Wang Y, Ji B and Liu Y: Synergistic effects of co-expression plasmid-based ADAM10-specific siRNA and GRIM-19 on hepatocellular carcinoma in vitro and in vivo. Oncol Rep. 32:2501–2510. 2014.PubMed/NCBI

25 

Roberti A: LaS ala D and Cinti C: Multiple genetic and epigenetic interacting mechanisms contribute to clonally selection of drug-resistant tumors: Current views and new therapeutic prospective. J Cell Physiol. 207:571–581. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Wang GM, Ren ZX, Wang PS, et al: Plasmid-based Stat3-specific siRNA and GRIM-19 inhibit the growth of thyroid cancer cells in vitro and in vivo. Oncol Rep. 32:573–580. 2014.PubMed/NCBI

27 

Zhang L, Gao L, Li Y, et al: Effects of plasmid-based Stat3-specific short hairpin RNA and GRIM-19 on PC-3M tumor cell growth. Clin Cancer Res. 14:559–568. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Li X, Li Y, Hu J, Wang B, Zhao L, Ji K, Guo B, Yin D, Du Y, Kopecko DJ, et al: Plasmid-based E6-specific siRNA and co-expression of wild-type p53 suppresses the growth of cervical cancer in vitro and in vivo. Cancer Lett. 335:242–250. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Li L, Yu C, Ren J, et al: Synergistic effects of eukaryotic coexpression plasmid carrying LKB1 and FUS1 genes on lung cancer in vitro and in vivo. J Cancer Res Clin Oncol. 140:895–907. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Hu Y, Shen Y, Ji B, Wang L, Zhang Z and Zhang Y: Combinational RNAi gene therapy of hepatocellular carcinoma by targeting human EGFR and TERT. Eur J Pharm Sci. 42:387–391. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Chen Y, Cao Y, Yang D, et al: Increase of the therapeutic effect on non-small-cell lung cancer cells with combination treatment of shRNA against Cyclin D1 and Bcl-xL in vitro. Exp Ther Med. 3:255–260. 2012.PubMed/NCBI

32 

Lai WY, Wang WY, Chang YC, et al: Synergistic inhibition of lung cancer cell invasion, tumor growth and angiogenesis using aptamer-siRNA chimeras. Biomaterials. 35:2905–2914. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Hanibuchi M, Kim SJ, Fidler IJ and Nishioka Y: The molecular biology of lung cancer brain metastasis, An overview of current comprehensions and future perspectives. J Med Invest. 61:241–253. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Nguyen DX, Bos PD and Massagué J: Metastasis. From dissemination to organ-specific colonization. Nat Rev Cancer. 9:274–284. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Bacac M and Stamenkovic I: Metastatic cancer cell. Annu Rev Pathol. 3:221–247. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2016
Volume 11 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
Chang L, Gong F, Cai H, Li Z and Cui Y: Combined RNAi targeting human Stat3 and ADAM9 as gene therapy for non‑small cell lung cancer Retraction in /10.3892/ol.2022.13595. Oncol Lett 11: 1242-1250, 2016
APA
Chang, L., Gong, F., Cai, H., Li, Z., & Cui, Y. (2016). Combined RNAi targeting human Stat3 and ADAM9 as gene therapy for non‑small cell lung cancer Retraction in /10.3892/ol.2022.13595. Oncology Letters, 11, 1242-1250. https://doi.org/10.3892/ol.2015.4018
MLA
Chang, L., Gong, F., Cai, H., Li, Z., Cui, Y."Combined RNAi targeting human Stat3 and ADAM9 as gene therapy for non‑small cell lung cancer Retraction in /10.3892/ol.2022.13595". Oncology Letters 11.2 (2016): 1242-1250.
Chicago
Chang, L., Gong, F., Cai, H., Li, Z., Cui, Y."Combined RNAi targeting human Stat3 and ADAM9 as gene therapy for non‑small cell lung cancer Retraction in /10.3892/ol.2022.13595". Oncology Letters 11, no. 2 (2016): 1242-1250. https://doi.org/10.3892/ol.2015.4018