Lentivirus-mediated shRNA interference targeting vascular endothelial growth factor inhibits angiogenesis and progression of human pancreatic carcinoma

  • Authors:
    • Xin Zhao
    • Dong-Ming Zhu
    • Wen-Juan Gan
    • Zhi Li
    • Jiang-Lei Zhang
    • Hua Zhao
    • Jin Zhou
    • De-Chun Li
  • View Affiliations

  • Published online on: December 19, 2012     https://doi.org/10.3892/or.2012.2203
  • Pages: 1019-1026
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Angiogenesis is known to be essential to the survival, growth, invasion and metastasis of cancer cells. Vascular endothelial growth factor (VEGF) is an important factor regulating tumor angiogenesis. In the present study, we analyzed the effect of lentivirus-mediated shRNA interference targeting vascular endothelial growth factor (VEGF) on angiogenesis and progression in the pancreatic cancer cell line Patu8988 in vitro and in vivo. The study aimed to construct a recombinant lentivirus carrying targeted VEGF shRNA (LV-RNAi) to be used to transfect Patu8988 cells, and we investigated its anti-angiogenic and growth inhibitory effects on pancreatic cancer. VEGF expression was measured by RQ-PCR, western blotting and enzyme-linked immunosorbent assay (ELISA). In subcutaneous transplantation models, tumor volumes were determined, and the expression levels of VEGF and CD34 were assessed by immunohistochemistry. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) was used to determine apoptosis. In the orthotopic transplantation models, tumor volume and liver metastasis were determined. We successfully constructed LV-RNAi and confirmed that it knocked down the VEGF gene at the mRNA and protein levels in Patu8988 cells. In the subcutaneous transplantation models, tumors with low levels of VEGF expression exhibited reduced pancreatic carcinoma angiogenesis and growth, and the apoptotic index was significantly higher. In the orthotopic transplantation models, tumors with low levels of VEGF expression exhibited significantly reduced pancreatic carcinoma growth, but no significant difference was observed between the three mouse groups, LV-RNAi, LV-NC and the control, in regards to liver metastasis. In summary, lentivirus-mediated RNAi silencing of VEGF inhibited tumor angiogenesis and growth, and increased apoptosis of the pancreatic cancer cell line Patu8988. VEGF targeted gene silencing approach has the potential to serve as a novel treatment for pancreatic cancer.

Introduction

Pancreatic cancer is a highly lethal disease that is usually diagnosed at an advanced stage for which there is little or no effective therapy. It remains the fourth most common cause of cancer-related death in the Western world (1). Due to the aggressive natural history of this disease, most patients with pancreatic cancer present with local invasion or distant metastasis at the time of diagnosis, and less than 20% of patients are candidates for surgery with curative intent (2). The overall 5-year survival rates are reported to be below 5% (3). No adjuvant treatments have shown efficacy in improving survival to date. Thus, new approaches including gene therapy are definitely required to improve treatment results (4,5).

Angiogenesis is necessary for successful tumor growth (6,7), and inhibition of VEGF represents the most validated anti-angiogenic approach described thus far (8,9). VEGF is a key stimulating factor for angiogenesis of cancer, and it contributes to the malignant development and metastasis of tumors through many processes. In addition, VEGF is highly expressed in most human tumors (1014).

RNA interference (RNAi) has emerged as a powerful tool to induce lose-of-function phenotypes by post-transcriptional silencing of gene expression (15,16). Lentiviral vectors have provided a huge advance in technology and offer the means to achieve significant levels of gene transfer in vitro and in vivo(17,18).

In this study, we used the lentiviral vector mediating RNAi to deliver a specially designed small hairpin RNA for the human VEGF gene (LV-RNAi) into pancreatic carcinoma cell line Patu8988 to observe the gene therapeutic effects on angiogenesis and progression.

Materials and methods

Animals and cell lines

BALB/c nude mice were obtained from the Shanghai Experimental Animal Center (Shanghai, China) and maintained according to guidelines of the Animal Research Committee of Soochow University (Suzhou, China).

Human pancreatic cancer cell line Patu8988 was provided by Professor Chang-Geng Ruan, Jiangsu Institute of Hematology, and maintained in RPMI (Roswell Park Memorial Institute)-1640 medium (Gibco, USA) supplemented with 10% fetal bovine serum (FBS) at 37°C in a humidified atmosphere containing 5% CO2. The cultures were passaged 2 or 3 times weekly to maintain log-phase growth.

Lentiviral vectors for VEGF shRNA

Small hairpin RNA (shRNA) targeting human VEGF (GenBank, NM 001025366) was designed as follows. The underlined part in the sense strand is the target sequence of the VEGF gene which is 19-bp long, the italicized characters in the sense strand is the loop sequence of the hairpin. According to the sense strand, the antisense was also synthetized: sense, 5′-GATCCC(G)CCAT GAACTTTCTGCTGTCTTGATATCCGGACAGCAGAA AGTTCATGGTTTTTTCCAAC-3′; antisense, 3′-GG(C)GGT ACTTGAAAGACGACAGAACTATAGGCCTGTCGTCTT TCAAGTACCAAAAAAGGTTGAGCT-5′.

The recombinant lentivirus gene transfer vector targeting VEGF pGCSIL-GFP-VEGF (LV-RNAi) encoding the green fluorescent protein (GFP) sequence was constructed and gifted by Dr L. Li followed by chemically synthesized shRNAs and lentiviral vector constructed as previously described (19,20). The targeting sequence of the shRNA was confirmed by sequencing. The lentiviral vector pGCSIL-GFP-Negative (LV-NC) containing an invalid RNAi sequence (GeneChem, Shanghai) was used to monitor non-specific responses caused by heterologous siRNA. The LV-RNAi and the LV-NC were prepared to 5×109 Tu/ml (transfection units/ml).

Lentiviral vector transfection

Cells were subcultured at 5×104 cells/well into 6-well tissue culture plates overnight. The viral supernatant was then added into cells at a multiplicity of infection (MOI) of 10 with ENi.S and 5 μg/ml Polybrene. The infected cells were considered to be the LV-RNAi and the LV-NC group, respectively, and the Patu8988 cells without infection were considered as the control group. Flow cytometry was used to detect the transfection efficiency, and fluorescence microscopy was used to observe the cells which released fluorescence. The three groups mentioned above were used in the experiments below.

Real-time quantitative RT-PCR

Total RNA was collected using TRIzol reagent following the manufacturer’s instructions. The concentration and purity of the total RNA were detected with an ultraviolet spectrophotometer and then reversely transcribed into cDNA with MMLV. Total RNA (2 μg) was converted to cDNA in 40 μl and stored at −20°C until use. The transcriptional level of VEGF was analyzed using the MJ Research DNA Engine Opticon 2 System with SYBR-Green fluorochrome. The GAPDH gene was used as an internal control. PCR was carried out with cDNA derived from 50 ng of RNA, 1 unit Taq polymerase and reaction kits in a final volume of 25 μl. Each cycle of PCR included 15 sec of denaturation at 95°C, 20 sec of primer annealing at 58°C and 20 sec of extension/synthesis at 72°C. The primer sequences were as follows: 5′-GCTTTACTGCTGTACCTCCAC-3′ (sense) 5′-TCCAGGGCTTCATCGTTA-3′ (antisense) for VEGF (239 bp); 5′-GCAAGTTCAACGGCACAG-3′ (sense) 5′-GCCAGTAGACTCCACGACAT-3′ (antisense) for GAPDH (140 bp).

Western blotting

Cells were washed twice and lysed on ice. After centrifugation, the supernatants were collected. Protein concentrations were determined using the Bio-Rad DC Protein Assay system. The β-actin gene was used as an internal control. The following steps were as previously described (20).

Detecting VEGF levels in culture supernatants by ELISA

Cells were seeded in new cell culture bottles, and after a 72-h culture supernatants were collected and cell counting was performed. The expression of VEGF in the collected supernatants was tested by a human VEGF ELISA kit (R&D Co.) according to the handbook. VEGF concentration/cell count was considered as the VEGF expression level. VEGF expression level in samples of the control group was considered as 1, and the expression of VEGF in the other groups was calculated by comparison to the control. Duplicate wells were set, and the entire experiment was repeated twice.

Subcutaneous transplantation model

A total of 18 BALB/c-nu mice, 5-weeks old and 20–24 g in weight, were bred in a specific pathogen-free (SPF) condition and maintained at a constant humidity and temperature (25–28°C). All mice underwent subcutaneous injection of a 200-μl cell suspension of Patu8988 cells (1.0×107) in the infra-axillary region, respectively. Two weeks later, the animals were randomly divided into three groups and intratumorally injected only once with 400 μl normal saline, 400 μl LV-NC or 400 μl LV-RNAi, respectively. The size of the tumors was measured in a blinded manner once a week with calipers, and the volume was determined using the simplified formula of a rotational ellipsoid (L × W2 × 0.5).

Immunohistochemical staining

Tumors were harvested from mice 5 weeks after treatment, and VEGF expression and microvessel density (MVD) of the tumor specimens were determined by immunohistochemistry. The tissue specimens fixed with formalin solution were embedded in paraffin wax, serially sectioned at 4 μm and immunohistochemically stained using the SP method according to the manufacturer’s instructions for the SP kit. The primary antibodies were diluted to 1:50 for VEGF (as mentioned above) and 1:100 for CD34 (Santa Cruz Biotechnology, Santa Cruz, CA, USA). Rectal cancer slides served as the positive control, and PBS was used to replace the primary antibodies to serve as the negative control. The expression of VEGF protein was scored semi-quantitatively. Sections were then evaluated for the presence of brown diaminobenzidine precipitates indicative of positive reactivity by microscopy. Ten visual fields (magnification, ×200) were counted for each section. The brown staining in or around the nucleus was considered as positive reactivity for VEGF. CD34 is used as a biomarker in endothelial cells for the identification of new blood microvessels. One lumen of blood vessels was assessed as one new blood capillary. The MVD value was determined based on Weidner’s method (21).

Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay

Apoptotic tumor cells were detected with the TUNEL method, using an in situ cell death detection kit (Roche Diagnostics, Mannheim, Germany). The assay was performed according to the manufacturer’s instructions. Briefly, after routine deparaffinization and treatment with H2O2 (3%), sections were digested with proteinase K (20 μg/ml, pH 7.4, 12 min) at 25°C and incubated with the reaction mixture (1:40, 60 min) at 37°C. Incorporated fluorescein was detected with horseradish peroxidase after a 30-min incubation at 37°C and subsequently dyed with DAB. Cell with brown-colored nuclei were assessed as positive apoptotic cells, and the number of apoptotic cells counted for 1,000 tumor cells in one section for at least 5 high power fields, was scored as the apoptotic index (AI).

Orthotopic transplantation pancreatic cancer model

The establishment of the infra-axillary subcutaneous transplantation tumor model was performed as described in the ‘Subcutaneous transplantation model’ section. After reaching a specific volume, the tumors were resected under aseptic environment and washed twice in antibiotic-containing RPMI-1640 to prevent possible infection. Necrotic tissues were removed, and the remaining viable tumor tissues were cut into small pieces of 1 mm3. Five-week-old BALB/c-nu mice, weighing 20–24 g, were anesthetized with urethane (4 ml/kg) by intramuscular injection. After the abdominal skin was sterilized, an incision was made in the upper left abdomen and the pancreas was exposed. Tumor pieces were attached to the pancreas using absorbable sutures. The pancreas was then returned to the peritoneum, and the abdominal wall and the skin were closed with silk sutures, respectively. The animals were allowed to recover for 24 h. Eighteen surviving mice were randomly divided into three groups (n=6) and intraperitoneally injected only once with 400 μl normal saline, 400 μl LV-NC or 400 μl LV-RNAi, respectively. All of the mice were sacrificed 6 weeks after treatment. Tumors were harvested from mice, and the volume was determined as previously described. Liver metastasis was also observed.

Statistical analysis

Statistical analysis was carried out using SAS 9.0 statistical software. Data are presented as the means ± standard deviation (SD). The Student’s t-test or ANOVA was used to compare the means of different groups. Chi-square test was used to compare categorical variables and clinical pathological correlation. The relationships among VEGF and MVD were investigated by Spearman-rank correlation. A P<0.05 was considered to indicate a statistically significant difference.

Results

Sequencing result and transfection efficiency of the lentiviral vector

The result of sequencing for the recombinant vector confirmed that the target sequences were constructed to the lentivirus system pGCSIL-GFP successively (Fig. 1). We used a lentiviral vector system to express shRNAs directed against VEGF. In addition, GFP was as used as a reporter gene. After a single exposure of Patu8988 cells to the encoding lentivirus at MOI of 10 for 120 h, the cells were examined by fluorescence microscopy (Fig. 2) and flow cytometry (Fig. 3). A high percentage (>90%) of transfectants expressed GFP, indicating high and stable transfection of the lentiviral vector system.

Assessment of the VEGF silencing effect by RQ-PCR and western blotting

To detect the effect of VEGF silencing, real-time RQ-PCR and western blot analysis were performed to determine the mRNA and protein levels of VEGF following transfection. As shown in Fig. 4A, the relative VEGF mRNA expression (0.29±0.04, P<0.05) was significantly decreased in the LV-RNAi group compared with the LV-NC (0.93±0.07) and the control group (1.00±0.06), while no significant differences were noted between the LV-NC and control group (P>0.05). As shown in Fig. 4B, a 46-kDa protein band, VEGF protein, was detected in the control and the LV-NC group, but was weakly expressed in the LV-RNAi group.

VEGF expression levels in vitro

To detect the downregulation effect on VEGF expression, ELISA assay was performed. VEGF concentration/cell counting was considered as the VEGF expression level (Fig. 5). The VEGF expression level in the culture supernatants of the LV-RNAi group was 15.7±3.06 pg/ml/105cells, and in comparison to the control group (32.16±3.90 pg/ml/105cells), it was obviously inhibited (P<0.05), with a high inhibition efficiency (51.18%). There were no significant differences between the LV-NC and the control group.

Tumor growth in the subcutaneous transplantation model

All of the 18 mice developed detectable tumors at the beginning of this experiment. Inhibition of growth was observed more significantly in mice after treatment with LV-RNAi for 5 weeks, when compared to the LV-NC (3,000±430 mm3) or control group (3,100±480 mm3). The average tumor volume (1,630±250 mm3) in the LV-RNAi group was significantly lower than the other two groups (P<0.05) (Fig. 6). No significant differences were noted between the LV-NC and the control group.

Evaluation of expression of VEGF and CD34 by immunohistochemistry

In order to demonstrate the mechanism of the anti-angiogenic effect of LV-RNAi, the expression of VEGF and CD34 was assessed by immunohistochemistry in the nude mouse transplanted tumors after treatments. The LV-RNAi group exhibited downregulation of VEGF expression and a decrease in the MVD when compared to the LV-NC and control groups (P<0.05) (Figs. 7 and 8). There were no significant differences between the LV-NC and control group (P>0.05).

Apoptosis assay by TUNEL staining

The number of positive apoptotic tumor cells exhibiting brown nuclei in the TUNEL assay was determined (Fig. 9). Based on the TUNEL assay, we found that increased numbers of apoptotic pancreatic carcinoma cells were present in the subcutaneously transplanted tumors treated with LV-RNAi. When compared to the LV-NC (0.047±0.020) or control group (0.044±0.014), the AI (0.254±0.029) in the LV-RNAi group was significantly higher than that in the former two groups (P<0.05) (Fig. 10). These results indicate that inhibition of VEGF gene expression caused apoptotic cell death in pancreatic carcinoma cells in vivo.

Tumor growth and liver metastasis in the orthotopic transplantation pancreatic cancer model

All of the 18 mice developed orthotopic transplantation pancreatic carcinoma tumors in this experiment (Fig. 11A). Liver metastasis was detected (Fig. 11B) in the orthotopic transplantation pancreatic cancer model. Inhibition of growth was significantly observed in mice following treatment with LV-RNAi for 5 weeks, when compared to the mice treated with LV-NC (197±49 mm3) or normal saline (213±52 mm3). The average tumor volume (47±22 mm3, P<0.05) in the former group was significantly lower than that in the latter two groups (Fig. 11C). The number of cases of liver metastasis in the LV-RNAi group (1/6, 16.67%) was less than that in the LV-NC (3/6, 50.00%) or the control group (4/6, 66.67%), while there was no significant differences between the LV-NC and the control group (P=0.58).

Discussion

Pancreatic adenocarcinoma is one of the deadliest human malignancies, accounting for more than 20% of gastrointestinal cancer-related deaths (22,23). At the time of diagnosis, the disease has often progressed to an advanced stage at which surgical resection is often not a viable option and at which time tumors are highly resistant to conventional chemotherapy and radiation treatments (24,25). The resistance of pancreatic adenocarcinoma to conventional treatment strategies has led to a search for novel targeted therapies that may be useful in eradicating this disease. Gene therapy has been recently emphasized for its contribution to a more favorable patient prognosis (26). In this study, we chose a lentiviral vector since it displays high efficiency in gene delivery and is expressed long-term. In addition, the vector can integrate genes into non-dividing cells with little immunologic reaction (17,18). Our results showed that a high gene transduction efficiency (>90%) was achieved at day 5 after exposure to lentiviral vectors, suggesting gene integration.

Small hairpin RNA (shRNA) mimic natural RNAi in ways that synthetic siRNA oligonucleotides do not (27). shRNA expression vector systems have been established to induce RNA interference (RNAi) in mammalian cells (28). Although these vectors provide certain advantages over chemically synthesized siRNAs, some disadvantages remain, including transient shRNA expression and low transfection efficiency, especially in non-dividing primary cells. To overcome these limitations, shRNA delivery systems using retroviral vectors (29), adenoviral vectors (30) and, more recently, lentiviral vectors (31) have been reported and proven safe for humans. Lentiviral vectors encoding antisense targeting sequences have been used in clinical trials with no obvious side effects (32,33). Lentivirus-delivered shRNAs are capable of specific, highly stable and functional silencing of gene expression in a variety of human cells including primary non-dividing cells and also in transgenic mice (34,35).

Tumor growth relies on angiogenesis, the formation of new blood vessels, to receive an adequate supply of oxygen and nutrients (6,7). In the absence of a blood vascular network, tumors are restrained in size due to limits in the diffusion of oxygen. Therefore, angiogenesis is an important process for growth, progression and metastasis of solid tumors (36). VEGF plays a central role in tumor angiogenesis; it is expressed in most tumors, often at substantially increased levels (37), which makes it a critical target for cancer therapy (3842). Experiments with neutralizing antibodies and other inhibitors have demonstrated that blockade of the VEGF pathway is sufficient to significantly suppress angiogenesis associated with solid tumor growth in many models. Subcutaneous and orthotopic models have been used to test the effects of inhibitors of the VEGF/VEGFR pathway on the growth of a variety of tumor cell lines (43). Angiogenesis in pancreatic carcinoma is based on the same fundamental principles of activation, proliferation and migration of endothelial cells. The expression of VEGF and CD34 in pancreatic carcinomas was demonstrated to be significantly higher than that in normal pancreatic tissue samples, respectively (44,45).

In the present study, we used lentivirus-mediated shRNA expression system targeting the VEGF gene to downregulate gene expression. After successful lentivirus-mediated VEGF RNA interference, the mRNA and protein expression of the VEGF gene was virtually knocked down in vitro as detected by real-time RT-PCR, western blot analysis and ELISA, respectively. Decreased expression of VEGF in the human pancreatic carcinoma cell line and in the xenografted tumors contributed to decreased angiogenesis, growth and metastasis. CD34 is a cell surface marker of progenitor cells and is frequently used as a new vessel marker and an indicator of microvessel density in tissues (46,47). Immunostaining assays revealed VEGF and CD34 in tumors were significantly decreased after LV-RNAi transfection. As shown in the subcutaneous and orthotopic xenografted pancreatic cancer in mice, downregulation of VEGF was found to lead to the suppression of cancer growth, resulting in reduced tumor size.

Additionally, overexpression of VEGF has been shown to be associated with enhanced tumorigenicity and tumor metastatic potential (8,48). Evidence now indicates that the direct receptor-mediated effects of VEGF on tumors such as VEGF induction of phosphatidylinositol-3′-kinase (PI3K) and Akt and enhancement of cell survival (49,50), affect clinical outcomes more decisively than do changes in blood flow and/or oxygenation (51). As is now recognized for co-expression of the kinase-impaired HER3 with HER2, (52) co-expression of kinase-defective VEGFR-1 with active VEGFR-2 increases Akt signaling (53) thus enhancing cell survival and tumor invasion (48). The pro-apoptotic effect of VEGF signaling inhibition is also apparent in vitro(54), confirming that the pro-apoptotic action is at least in part independent of the blood supply. In the TUNEL assay of the subcutaneous xenografted tumors, we found that decreased VEGF expression increased cell apoptosis of pancreatic carcinoma cells. Moreover, VEGF-dependent signaling cascades were found to increase cell motility via Src or Fak inducible tyrosine phosphorylation of adhesion substrates (55) and such motility is directly inducible in vitro by VEGF (56). In the orthotopic transplantation pancreatic cancer model, we found that decreased VEGF expression reduced the liver metastasis rate, although there was no significant difference detected by statistics which may have been due to the small sample size. Thus, more animal experiments nust be performed to further confirm the inhibitory effects of metastasis mediated by lentivirus-mediated shRNA interference targeting VEGF in pancreatic cancer in vivo. Therefore, the anticancer effects induced by lentivirus-mediated shRNA interference targeting VEGF require further investigation, and this will be the focus of our interest in future research.

In conclusion, our findings indicate that lentivirus-mediated shRNA interference targeting VEGF potently suppressed angiogenesis, growth and increased cell apoptosis in the Patu8988 pancreatic cancer cell line in vivo. Our findings support the theory that lentivirus-mediated shRNA interference targeting VEGF may be a promising mothod for the treatment of pancreatic carcinoma.

Acknowledgements

This study was supported by a grant from the Post-graduate Scientific Research Innovation Project of the Education Department of Jiangsu Province (no. CXZZ11_0125), China, and the Science and Technology Research Project of the Science and Technology Bureau of Suzhou City (no. SYS201120), China.

References

1 

Jemal A, Siegel R, Ward E, et al: Cancer statistics, 2007. CA Cancer J Clin. 57:43–66. 2007. View Article : Google Scholar

2 

Neoptolemos JP, Cunningham D, Friess H, et al: Adjuvant therapy in pancreatic cancer: historical and current perspectives. Ann Oncol. 14:675–692. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Hidalgo M: Pancreatic cancer. N Engl J Med. 362:1605–1617. 2010. View Article : Google Scholar

4 

Ghaneh P, Costello E and Neoptolemos JP: Biology and management of pancreatic cancer. Postgrad Med J. 84:478–497. 2008. View Article : Google Scholar

5 

Pan X, Sheng W, Zhu Q, et al: Inhibition of pancreatic carcinoma growth by adenovirus-mediated human interleukin-24 expression in animal model. Cancer Biother Radiopharm. 23:425–434. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Folkman J: Tumor angiogenesis: a possible control point in tumor growth. Ann Intern Med. 82:96–100. 1975. View Article : Google Scholar : PubMed/NCBI

7 

Kerbel RS: Tumor angiogenesis. New Engl J Med. 358:2039–2049. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Dai J and Rabie A: VEGF: an essential mediator of both angiogenesis and endochondral ossification. J Dent Res. 86:937–950. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Midgley R and Kerr D: Bevacizumab: current status and future directions. Ann Oncol. 16:999–1004. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Weidner N, Folkman J, Pozza F, et al: Tumor angiogenesis: a new significant and independent prognostic indicator in early-stage breast carcinoma. J Natl Cancer Inst. 84:1875–1887. 1992. View Article : Google Scholar : PubMed/NCBI

11 

Olson TA, Mohanraj D, Carson LF, et al: Vascular permeability factor gene expression in normal and neoplastic human ovaries. Cancer Res. 54:276–280. 1994.PubMed/NCBI

12 

Seo Y, Baba H, Fukuda T, et al: High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer. 88:2239–2245. 2000. View Article : Google Scholar : PubMed/NCBI

13 

El-Houseini ME, Abdel-Azim SA, El-Desouky GI, et al: Clinical significance of vascular endothelial growth factor (VEGF) in sera of patients with pediatric malignancies. J Egypt Natl Canc Inst. 16:57–61. 2004.PubMed/NCBI

14 

Rutkowski P, Kamińska J, Kowalska M, et al: Cytokine and cytokine receptor serum levels in adult bone sarcoma patients: correlations with local tumor extent and prognosis. J Surg Onco1. 84:151–159. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Merritt WM, BarEli M and Sood AK: The dicey role of dicer: implications for RNAi therapy. Cancer Res. 70:2571–2574. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Agrawal N, Dasaradhi PV, Mohmmed A, et al: RNA interference: biology, mechanism, and applications. Microbiol Mol Biol Rev. 67:657–685. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Buchschacher GL and Wong-Staal F: Development of lentiviral vectors for gene therapy for human diseases. Blood. 95:2499–2504. 2000.PubMed/NCBI

18 

Naldini L: Medicine. A comeback for gene therapy. Science. 326:805–806. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Li L, Zhang R, Cen JN, et al: Construction and identification of lentiviral vector-mediated RNA interference of VEGF gene. Soochow Univ J Med Sci. 28:20–22. 2008.(abstract in English).

20 

Li L, Zhang R, Cen JN, et al: Lentivirus-mediated RNA interference targeting vascular endothelial growth factor gene enhances the sensitivity of K562 cells to STI 571. Chin J Pathophysiol. 25:1122–1126. 2009.(abstract in English).

21 

Weidner N: Intratumor microvessel density as a prognostic factor in cancer. Am J Pathol. 147:9–19. 1995.PubMed/NCBI

22 

Korc M: Pathways for aberrant angiogenesis in pancreatic cancer. Mol Cancer. 2:2–8. 2003. View Article : Google Scholar

23 

Shi X, Friess H, Kleeff J, et al: Pancreatic cancer: factors regulating tumor development, maintenance and metastasis. Pancreatol. 1:517–524. 2001. View Article : Google Scholar : PubMed/NCBI

24 

Keleg S, Büchler P, Ludwig R, et al: Invasion and metastasis in pancreatic cancer. Mol Cancer. 2:142003. View Article : Google Scholar

25 

MacKenzie MJ: Molecular therapy in pancreatic adenocarcinoma. Lancet Oncol. 5:541–549. 2004. View Article : Google Scholar

26 

Jimeno A and Hidalgo M: Molecular biomarkers: their increasing role in the diagnosis, characterization, and therapy guidance in pancreatic cancer. Mol Cancer Ther. 5:787–796. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Snove O and Rossi JJ: Expressing short hairpin RNAs in vivo. Nat Methods. 3:689–695. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Brummelkamp TR, Bernards R and Agami R: A system for stable expression of short interfering RNAs in mammalian cells. Science. 296:550–553. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Brummelkamp TR, Bernards R and Agami R: Stable suppression of tumorigenicity by virus-mediated RNA interference. Cancer Cell. 2:243–247. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Xia H, Mao Q, Paulson HL, et al: siRNA-mediated gene silencing in vitro and in vivo. Nat Biotechnol. 20:1006–1010. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Qin XF, An DS, Chen IS, et al: Inhibiting HIV-1 infection in human T cells by lentiviral-mediated delivery of small interfering RNA against CCR5. Proc Natl Acad Sci USA. 100:183–188. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Manilla P, Rebello T, Afable C, et al: Regulatory considerations for novel gene therapy products: a review of the process leading to the first clinical lentiviral vector. Hum Gene Ther. 16:17–25. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Bank A, Dorazio R and Leboulch P: A phase I/II clinical trial of beta-globin gene therapy for beta-thalassemia. Ann NY Acad Sci. 1054:308–316. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Nishitsuji H, Ikeda T, Miyoshi H, et al: Expression of small hairpin RNA by lentivirus-based vector confers efficient and stable gene-suppression of HIV-1 on human cells including primary non-dividing cells. Microbes Infect. 6:76–85. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Rubinson DA, Dillon CP, Kwiatkowski AV, et al: A lentivirus-based system to functionally silence genes in primary mammalian cells, stem cells and transgenic mice by RNA interference. Nat Genet. 33:401–406. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Folkman J: Tumor angiogenesis: therapeutic implications. N Engl J Med. 285:1182–1186. 1971. View Article : Google Scholar : PubMed/NCBI

37 

Kim KJ, Li B, Winer J, et al: Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumor growth in vivo. Nature. 362:841–844. 1993. View Article : Google Scholar : PubMed/NCBI

38 

Sullivan LA and Brekken RA: The VEGF family in cancer and antibody-based strategies for their inhibition. MAbs. 2:165–175. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Dvorak HF: Vascular permeability factor/vascular endothelial growth factor: a critical cytokine in tumor angiogenesis and a potential target for diagnosis and therapy. J Clin Oncol. 20:4368–4380. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Roskoski R Jr: Vascular endothelial growth factor (VEGF) signaling in tumor progression. Critical Rev Oncol Hematol. 62:179–213. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Presta LG, Chen H, O’Connor SJ, et al: Humanization of an anti-VEGF monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res. 57:4593–4599. 1997.PubMed/NCBI

42 

Ferrara N: Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev. 25:581–611. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Ferrara N: VEGF-A: a critical regulator of blood vessel growth. Eur Cytokine Netw. 20:158–163. 2009.PubMed/NCBI

44 

Itakura J, Ishiwata T, Shen B, et al: Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer. Int J Cancer. 85:27–34. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Sun HC, Qiu ZJ, Liu J, et al: Expression of hypoxia-inducible factor-1 alpha and associated proteins in pancreatic ductal adenocarcinoma and their impact on prognosis. Int J Oncol. 30:1359–1367. 2007.PubMed/NCBI

46 

Krause DS, Fackler MJ, Civin CI, et al: CD34: structure, biology, and clinical utility. Blood. 87:1–13. 1996.PubMed/NCBI

47 

Nielsen JS and McNagny KM: Novel functions of the CD34 family. J Cell Sci. 121:3683–3692. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Girling JE and Rogers PA: Regulation of endometrial vascular remodelling: role of the vascular endothelial growth factor family and the angiopoietin-TIE signalling system. Reproduction. 138:883–893. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Wu Y, Hooper AT, Zhong Z, et al: The vascular endothelial growth factor receptor (VEGFR-1) supports growth and survival of human breast carcinoma. Int J Cancer. 119:1519–1529. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Graells J, Vinyals A, Figueras A, et al: Overproduction of VEGF concomitantly expressed with its receptors promotes growth and survival of melanoma cells through MAPK and PI3K signaling. J Invest Dermatol. 123:1151–1161. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Mercurio AM, Lipscomb EA and Bachelder RE: Non-angiogenic functions of VEGF in breast cancer. J Mammary Gland Biol Neoplasia. 10:283–290. 2005. View Article : Google Scholar : PubMed/NCBI

52 

Sergina NV, Rausch M, Wang D, et al: Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature. 445:437–441. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Wu Y, Zhong Z, Huber J, et al: Anti-vascular endothelial growth factor receptor-1 antagonist antibody as a therapeutic agent for cancer. Clin Cancer Res. 12:6573–6584. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Krause S, Förster Y, Kraemer K, et al: Vascular endothelial growth factor antisense pretreatment of bladder cancer cells significantly enhances the cytotoxicity of mitomycin C, gemcitabine and cisplatin. J Urol. 174:328–331. 2005. View Article : Google Scholar

55 

Lesslie DP, Summy JM, Parikh NU, et al: Vascular endothelial growth factor receptor-1 mediates migration of human colorectal carcinoma cells by activation of Src family kinases. Br J Cancer. 94:1710–1717. 2006.PubMed/NCBI

56 

Wey JS, Fan F, Gray MJ, et al: Vascular endothelial growth factor receptor-1 promotes migration and invasion in pancreatic carcinoma cell lines. Cancer. 104:427–438. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 29 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao X, Zhu D, Gan W, Li Z, Zhang J, Zhao H, Zhou J and Li D: Lentivirus-mediated shRNA interference targeting vascular endothelial growth factor inhibits angiogenesis and progression of human pancreatic carcinoma. Oncol Rep 29: 1019-1026, 2013
APA
Zhao, X., Zhu, D., Gan, W., Li, Z., Zhang, J., Zhao, H. ... Li, D. (2013). Lentivirus-mediated shRNA interference targeting vascular endothelial growth factor inhibits angiogenesis and progression of human pancreatic carcinoma. Oncology Reports, 29, 1019-1026. https://doi.org/10.3892/or.2012.2203
MLA
Zhao, X., Zhu, D., Gan, W., Li, Z., Zhang, J., Zhao, H., Zhou, J., Li, D."Lentivirus-mediated shRNA interference targeting vascular endothelial growth factor inhibits angiogenesis and progression of human pancreatic carcinoma". Oncology Reports 29.3 (2013): 1019-1026.
Chicago
Zhao, X., Zhu, D., Gan, W., Li, Z., Zhang, J., Zhao, H., Zhou, J., Li, D."Lentivirus-mediated shRNA interference targeting vascular endothelial growth factor inhibits angiogenesis and progression of human pancreatic carcinoma". Oncology Reports 29, no. 3 (2013): 1019-1026. https://doi.org/10.3892/or.2012.2203