Tumor suppressor p53 is a multifunctional transcription factor that regulates diverse cell fates, including apoptosis and autophagy in tumor biology. p53 overexpression enhances the antitumor activity of oncolytic adenoviruses; however, the molecular mechanism of this occurrence remains unclear. We previously developed a tumor-specific replication-competent oncolytic adenovirus, OBP-301, that kills human osteosarcoma cells, but some human osteosarcoma cells were OBP-301–resistant. In this study, we investigated the antitumor activity of a p53-expressing oncolytic adenovirus, OBP-702, and the molecular mechanism of the p53-mediated cell death pathway in OBP-301–resistant human osteosarcoma cells. The cytopathic activity of OBP-702 was examined in OBP-301–sensitive (U2OS and HOS) and OBP-301–resistant (SaOS-2 and MNNG/HOS) human osteosarcoma cells. The molecular mechanism in the OBP-702–mediated induction of two cell death pathways, apoptosis and autophagy, was investigated in OBP-301–resistant osteosarcoma cells. The antitumor effect of OBP-702 was further assessed using an orthotopic OBP-301–resistant MNNG/HOS osteosarcoma xenograft tumor model. OBP-702 suppressed the viability of OBP-301–sensitive and -resistant osteosarcoma cells more efficiently than OBP-301 or a replication-deficient p53-expressing adenovirus (Ad-p53). OBP-702 induced more profound apoptosis and autophagy when compared with OBP-301 or Ad-p53. E1A-mediated miR-93/106b upregulation induced p21 suppression, leading to p53-mediated apoptosis and autophagy in OBP-702–infected cells. p53 overexpression enhanced adenovirus-mediated autophagy through activation of damage-regulated autophagy modulator (DRAM). Moreover, OBP-702 suppressed tumor growth in an orthotopic OBP-301–resistant MNNG/HOS xenograft tumor model. These results suggest that OBP-702–mediated p53 transactivation is a promising antitumor strategy to induce dual apoptotic and autophagic cell death pathways via regulation of miRNA and DRAM in human osteosarcoma cells. Mol Cancer Ther; 12(3); 314–25. ©2012 AACR.

Osteosarcoma is one of the most common malignant tumors in young children (1, 2). Current treatment strategies, which consist of multi-agent chemotherapy and aggressive surgery, have significantly improved the cure rate and prognosis of patients with osteosarcoma. In fact, over the past 30 years, the 5-year survival rate has increased from 10% to 70% (3–5). Even in patients with osteosarcoma with metastases at diagnosis, the 5-year survival rate has reached 20% to 30% in response to chemotherapy and surgical removal of primary and metastatic tumors (6). However, treatment outcomes for patients with osteosarcomas have further improved over the last few years. Therefore, the development of novel therapeutic strategies is required to improve the clinical outcomes in patients with osteosarcomas.

Tumor-specific replication-competent oncolytic viruses are being developed as novel anticancer therapy, in which the promoters of cancer-related genes are used to regulate virus replication in a tumor-dependent manner. More than 85% of all human cancers express high telomerase activity to maintain the length of the telomeres during cell division, whereas normal somatic cells seldom show this enhanced telomerase activity (7, 8). Telomerase activity has also been detected in 44% to 81% of bone and soft-tissue sarcomas (9, 10). Telomerase activation is closely correlated with the expression of the human telomerase reverse transcriptase (hTERT) gene (11). On the basis of these data, we previously developed a telomerase-specific replication-competent oncolytic adenovirus OBP-301 (Telomelysin) in which the hTERT gene promoter drives the expression of the E1A and E1B genes (12). A phase I clinical trial of OBP-301, which was conducted in the United States on patients with advanced solid tumors, indicated that OBP-301 was well tolerated by patients (13). Recently, we reported that OBP-301 efficiently killed human bone and soft-tissue sarcoma cells (14, 15). However, some osteosarcoma cell lines were not sensitive to the antitumor effect of OBP-301. Therefore, to efficiently eliminate tumor cells with OBP-301, its antitumor effects need to be enhanced.

Cancer gene therapy is defined as the treatment of malignant tumors via the introduction of a therapeutic tumor suppressor gene or the abrogation of an oncogene. The tumor suppressor p53 gene has an attractive tumor suppressor profile as a potent therapeutic transgene for induction of cell-cycle arrest, senescence, apoptosis, and autophagy (16). Dual cell death pathways, such as apoptosis and autophagy, induced by p53 transactivation are mainly involved in the suppression of tumor initiation and progression. However, among the p53 downstream target genes, p21, which is most rapidly and strongly induced during the DNA damage response, mainly induces cell-cycle arrest through suppression of apoptotic and autophagic cell death pathways (17, 18). Thus, p21 suppression may be a more effective strategy for the induction of apoptotic and autophagic cell death pathways in tumor cells, particularly when the tumor suppressor p53 gene is overexpressed in tumor cells in response to cancer gene therapy.

A p53-expressing replication-deficient adenovirus (Ad-p53, Advexin) has previously been reported to induce an antitumor effect in the in vitro and in vivo settings (19, 20) as well as in some clinical studies (21–24). We recently reported that combination therapy with OBP-301 and Ad-p53 resulted in a more profound antitumor effect than monotherapy with either OBP-301 or Ad-p53 (25). Moreover, we generated armed OBP-301 expressing the wild-type p53 tumor suppressor gene (OBP-702) and showed that OBP-702 suppressed the viability of various types of epithelial malignant cells more efficiently than did OBP-301 (26). OBP-702 induced a more profound apoptotic cell death effect than Ad-p53, likely via adenoviral E1A-mediated suppression of anti-apoptotic p21 in human epithelial malignant cells. However, it remained unclear whether OBP-702 efficiently induces an antitumor effect in human nonepithelial malignant cells, including osteosarcomas.

In the present study, we investigated the in vitro cytopathic efficacy of the p53-expressing telomerase-specific replication-competent oncolytic adenovirus, OBP-702, in human osteosarcoma cells, and we compared the induction level of apoptotic and autophagic cell deaths in OBP-301–resistant human osteosarcoma cells infected with OBP-301, OBP-702, and Ad-p53. The molecular mechanism by which OBP-702 mediates induction of apoptosis and autophagy was also investigated. Finally, the in vivo antitumor effect of OBP-702 was evaluated using an orthotopic OBP-301–resistant human osteosarcoma xenograft tumor model.

Cell lines

The human osteosarcoma cell lines, HOS and SaOS-2, were kindly provided by Dr. Satoru Kyo (Kanazawa University, Ishikawa, Japan). These cells were propagated as monolayer cultures in Dulbecco's Modified Eagle's Medium. The human osteosarcoma cell line, U2OS, was obtained from the American Type Culture Collection and was grown in McCoy's 5a Medium. The human osteosarcoma cell line, MNNG/HOS, was purchased from DS Pharma Biomedical and was maintained in Eagle's Minimum Essential Medium containing 1% nonessential amino acids. All media were supplemented with 10% FBS, 100 U/mL penicillin, and 100 mg/mL streptomycin. The normal human lung fibroblast (NHLF) cell line, NHLF, was obtained from TaKaRa Biomedicals. NHLF cells were propagated as monolayer culture in the medium recommended by the manufacturer. Although cell lines were not authenticated by the authors, cells were immediately expanded after receipt and stored in liquid N2. Cells were not cultured for more than 5 months following resuscitation. The cells were maintained at 37°C in a humidified atmosphere with 5% CO2.

Recombinant adenoviruses

The recombinant telomerase-specific replication-competent adenovirus OBP-301 (Telomelysin), in which the promoter element of the hTERT gene drives the expression of E1A and E1B genes, was previously constructed and characterized (12, 27). For OBP-301–mediated induction of exogenous p53 gene expression, we recently generated OBP-702, in which a human wild-type p53 gene expression cassette was inserted into the E3 region (Supplementary Fig. S1; ref. 26). Ad-p53 is a replication-defective adenovirus serotype 5 vector with a p53 gene expression cassette at the E1 region (19, 20). Recombinant viruses were purified by ultracentrifugation using cesium chloride step gradients, their titers were determined by a plaque-forming assay using 293 cells and they were stored at −80°C.

Cell viability assay

Cells were seeded on 96-well plates at a density of 1 × 103 cells/well 24 hours before viral infection. All cell lines were infected with OBP-702 at multiplicity of infections (MOI) of 0, 0.1, 1, 10, 50, or 100 plaque-forming units (PFU)/cell. Cell viability was determined on days 1, 2, 3, and 5 after virus infection using Cell Proliferation Kit II (Roche Molecular Biochemicals). The 50% inhibiting dose (ID50) value of OBP-702 for each cell line was calculated using cell viability data obtained on day 5 after virus infection.

Time-lapse confocal laser microscopy

GFP-expressing MNNG/HOS (MNNG/HOS-GFP) cells were established by stable transfection with GFP expression plasmid using Lipofectamine LTX (Invitrogen). MNNG/HOS-GFP and NHLF cells were seeded in 35-mm glass-based dishes at a density of 1 × 105 cells/dish 24 hours before infection and were infected with OBP-702 at an MOI of 10 PFU/cell for 72 hours. Phase-contrast and fluorescence time-lapse recordings were obtained to concomitantly analyze cell morphology and GFP expression using an inverted FV10i confocal laser scanning microscopy (OLYMPUS).

Western blot analysis

SaOS-2 and MNNG/HOS cells, seeded in a 100-mm dish at a density of 1 × 105 cells/dish, were infected with OBP-301, OBP-702, or Ad-p53 at the indicated MOIs. In contrast, SaOS-2 cells were transfected with 10 nmol/L miR-93 (Ambion), miR-106b (Ambion), or control miRNA (Ambion) 24 hours before Ad-p53 infection and infected with Ad-p53 at an MOI of 100 for 48 hours. Whole-cell lysates were prepared in a lysis buffer [50 mmol/L Tris-HCl (pH 7.4), 150 mmol/L NaCl, 1% Triton X-100] containing a protease inhibitor cocktail (Complete Mini; Roche) at the indicated time points. Proteins were electrophoresed on 6% to 15% SDS-PAGE and were transferred to polyvinylidene difluoride membranes (Hybond-P; GE Health Care). Blots were blocked with 5% non-fat dry milk in TBS-T (Tris-buffered saline and 0.1% Tween-20, pH 7.4). The primary antibodies used were: rabbit anti-PARP polyclonal antibody (pAb; Cell Signaling Technology), mouse anti-p53 monoclonal antibody (mAb; Calbiochem), mouse anti-p21WAF1 mAb (Calbiochem), rabbit anti-E2F1 pAb (Santa Cruz Biotechnology), mouse anti-Ad5 E1A mAb (BD PharMingen), rabbit anti-microtubule-associated protein 1 light chain 3 (LC3) pAb [Medical & Biological Laboratories (MBL)], mouse anti-p62 mAb (MBL), rabbit anti-damage-regulated autophagy modulator (DRAM) pAb (Abgent), and mouse anti-β-actin mAb (Sigma-Aldrich).

Flow cytometric analysis

To analyze the active caspase-3 expression, cells were incubated for 20 minutes on ice in Cytofix/Cytoperm solution (BD Biosciences), labeled with phycoerythrin (PE)-conjugated rabbit anti-active caspase-3 mAb (BD Biosciences) for 30 minutes, and then analyzed using FACS array (BD Biosciences).

To evaluate the sub-G1 population, which is a apoptosis indicator, in SaOS-2 cells after virus infection, SaOS-2 cells were seeded in a 100-mm dish at a density of 1 × 106 cells/dish 24 hours before viral infection and were infected with mock, OBP-301, Ad-p53, or OBP-702 at an MOI of 10 PFUs/cell for 48 hours. Cells were trypsinized and resuspended in original supernatant to ensure that both attached and nonattached cells were analyzed. Cells stained with propidium iodide were analyzed using FACS array (BD Biosciences).

Quantitative real-time reverse transcription PCR analysis

To evaluate the expressions of miR-93 and miR-106b in tumor cells after OBP-702 infection, SaOS-2 and MNNG/HOS cells were seeded on 6-well plates at a density of 2 × 105 cells/well 24 hours before viral infection and were infected with OBP-702 at MOIs of 0, 1, 5, 10, 50, or 100 PFU/cell. Three days after virus infection, total RNA was extracted from the cells using a miRNeasy Mini Kit (Qiagen). The concentration and quality of RNA were assessed using a Nanodrop spectrophotometer. cDNA was synthesized from 10 ng of total RNA using the TaqMan MicroRNA Reverse Transcription Kit (Applied Biosystems), and quantitative real-time reverse transcription (RT)-PCR was carried out using the Applied Biosystems StepOnePlus real-time PCR System. The expressions of miR-93 and miR-106b were defined from the threshold cycle (Ct), and relative expression levels were calculated using the |$2^{- \Delta \Delta C_{\rm t}}$| method after normalization with reference to the expression of U6 small nuclear RNA.

In vivo orthotopic MNNG/HOS xenograft tumor model

Animal experimental protocols were approved by the Ethics Review Committee for Animal Experimentation of Okayama University School of Medicine (Okayama, Japan). MNNG/HOS cells (5 × 106 cells per site) were inoculated into the tibias of female athymic nude mice aged 6 to 7 weeks (CLEA Japan). Palpable tumors developed within 21 days and were permitted to grow to approximately 5 to 6 mm in diameter. At that stage, a 50-μL volume of solution containing OBP-702, OBP-301, or Ad-p53 at a dose of 1 × 108 PFU or PBS was injected into the tumors for 3 cycles every 2 days. Tumor volume was monitored by computed tomographic (CT) imaging once a week after virus infection.

Three-dimensional computed tomography imaging

The tumor volume and formation of osteolytic lesions were evaluated using three-dimensional CT (3D-CT) imaging (ALOKA Latheta LCT-200; Hitachi Aloka Medical). The tumor volume was calculated by INTAGE Realia software (Cybernet Systems).

Histopathologic analysis

Tumors were fixed in 10% neutralized formalin and embedded in paraffin blocks. Sections were stained with hematoxylin/eosin and analyzed by light microscopy.

Statistical analysis

Data are expressed as means ± SD. Student t test was used to compare differences between groups. Statistical significance was defined as P < 0.05.

In vitro cytopathic efficacy of OBP-702 against human osteosarcoma cell lines

To evaluate the in vitro cytopathic activity of OBP-702, we used the 2 OBP-301–sensitive human osteosarcoma cells (HOS and U2OS) and the 2 OBP-301–resistant human osteosarcoma cells (SaOS-2 and MNNG/HOS) that were recently described (14). The cell viability of each cell was assessed over 5 days after infection using the XTT assay. OBP-702 infection suppressed the viability of OBP-301–sensitive and -resistant cells in dose- and time-dependent manners (Fig. 1A and B). When the ID50 values of OBP-702 in all 4 human osteosarcoma cells were compared with those of OBP-301 calculated in a previous report (14), all cell lines were more sensitive to OBP-702 than to OBP-301 (Table 1). The ID50 values of OBP-702 were also lower than those of Ad-p53 (Supplementary Fig. S2). However, OBP-702 did not exhibit any cytopathic effect in NHLF cells (Fig. 1B). When GFP-expressing MNNG/HOS-GFP cells were cocultured with human normal NHLF cells, OBP-702 infection showed a cytopathic effect (confirmed by observation of round-shaped morphologic changes) in MNNG/HOS-GFP cells but not in NHLF cells (Fig. 1C). These results indicate that OBP-702 was more cytopathic than OBP-301 for human osteosarcoma cells but was not cytopathic for normal human cells.

Figure 1.

In vitro cytopathic effect of OBP-702 in human osteosarcoma cell lines. A, OBP-301–sensitive (HOS and U2OS) and OBP-301–resistant (SaOS-2 and MNNG/HOS) human osteosarcoma cells were infected with OBP-702 at the indicated MOI, and cell viability was quantified over 5 days using the XTT assay. Cell viability was calculated relative to that of the mock-infected group on each day, which was set at 1.0. Cell viability data are expressed as mean values ± SD (n = 5). B, four human osteosarcoma cells and one normal fibroblast NHLF cell were seeded 24 hours before viral infection and were infected with OBP-702 at the indicated MOIs, and cell viability was examined on day 5 using the XTT assay. Cell viability was calculated relative to that of the mock-infected group, which was set at 1.0. Cell viability data are expressed as mean ± SD (n = 5). C, time lapse images of cytopathic effect of OBP-702 in coculture of GFP-expressing MNNG/HOS cells with human normal fibroblast NHLF cells. MNNG/HOS-GFP cells coincubated with NHLF cells were recorded for 72 hours after OBP-702 infection at an MOI of 10. Three images on the left are low magnification and one image on the right is high magnification of the area outlined by a white square. Left scale bars, 100 μm. Right scale bar, 10 μm.

Figure 1.

In vitro cytopathic effect of OBP-702 in human osteosarcoma cell lines. A, OBP-301–sensitive (HOS and U2OS) and OBP-301–resistant (SaOS-2 and MNNG/HOS) human osteosarcoma cells were infected with OBP-702 at the indicated MOI, and cell viability was quantified over 5 days using the XTT assay. Cell viability was calculated relative to that of the mock-infected group on each day, which was set at 1.0. Cell viability data are expressed as mean values ± SD (n = 5). B, four human osteosarcoma cells and one normal fibroblast NHLF cell were seeded 24 hours before viral infection and were infected with OBP-702 at the indicated MOIs, and cell viability was examined on day 5 using the XTT assay. Cell viability was calculated relative to that of the mock-infected group, which was set at 1.0. Cell viability data are expressed as mean ± SD (n = 5). C, time lapse images of cytopathic effect of OBP-702 in coculture of GFP-expressing MNNG/HOS cells with human normal fibroblast NHLF cells. MNNG/HOS-GFP cells coincubated with NHLF cells were recorded for 72 hours after OBP-702 infection at an MOI of 10. Three images on the left are low magnification and one image on the right is high magnification of the area outlined by a white square. Left scale bars, 100 μm. Right scale bar, 10 μm.

Close modal
Table 1.

Comparison of ID50 values of OBP-301 and OBP-702 in various human osteosarcoma cell lines

ID50 valuea (MOI)
Cell linesSensitivity to OBP-301Cell typeRelative hTERT mRNA expressionOBP-301OBP-702Ratiob (OBP-702/OBP-301)
SaOS-2 Resistant ALT Negative 98.1 5.5 0.06 
MNNG/HOS Resistant Non-ALT 97.3 6.7 0.07 
U2OS Sensitive ALT 0.3 38.2 1.2 0.03 
HOS Sensitive Non-ALT 4.3 43.0 4.5 0.10 
ID50 valuea (MOI)
Cell linesSensitivity to OBP-301Cell typeRelative hTERT mRNA expressionOBP-301OBP-702Ratiob (OBP-702/OBP-301)
SaOS-2 Resistant ALT Negative 98.1 5.5 0.06 
MNNG/HOS Resistant Non-ALT 97.3 6.7 0.07 
U2OS Sensitive ALT 0.3 38.2 1.2 0.03 
HOS Sensitive Non-ALT 4.3 43.0 4.5 0.10 

aThe ID50 values of OBP-702 and OBP-301 were calculated from the data of XTT assay on day 5 after infection.

bThe ratio was calculated from the division of the ID50 value of OBP-702 by the ID50 value of OBP-301.

Increased induction of apoptosis by OBP-702 when compared with OBP-301 or Ad-p53

We next investigated whether OBP-702 induces more profound apoptosis when compared with OBP-301 or Ad-p53. OBP-301–resistant SaOS-2 and MNNG/HOS cells were infected with OBP-702, OBP-301, or Ad-p53, and apoptosis was assessed by Western blot and flow cytometric analyses. Western blot analysis showed that SaOS-2 cells exhibited the cleavage of PARP after infection with OBP-702 (>5 MOIs) or Ad-p53 (>50 MOIs), whereas MNNG/HOS cells had the cleavage of PARP after infection with OBP-702 (>5 MOIs) but not Ad-p53 (Fig. 2A). In contrast, OBP-301 did not induce apoptosis (data not shown). Furthermore, flow cytometric analysis showed that OBP-702 infection (10 MOIs) significantly increased the percentage of apoptotic cells with active caspase-3 when compared with Ad-p53 or OBP-301 at same doses in SaOS-2 and MNNG/HOS cells (Fig. 2B and C). Cell-cycle analysis also showed that OBP-702 (10 MOIs) induced the highest percentages of sub-G1 population in SaOS-2 cells when compared with Ad-p53 or OBP-301 at same doses (Fig. 2D). These results suggest that OBP-702 induces increased apoptosis when compared with Ad-p53 or OBP-301 in human osteosarcoma cells.

Figure 2.

OBP-702 induces increased apoptosis when compared with OBP-301 or Ad-p53. A, OBP-301–resistant SaOS-2 and MNNG/HOS cells were infected with OBP-702 or Ad-p53 at the indicated MOIs for 72 hours. Cell lysates were subjected to Western blot analysis for the C-PARP and PARP. β-Actin was assayed as a loading control. B–D, SaOS-2 and MNNG/HOS cells were infected with OBP-702, OBP-301, or Ad-p53 at an MOI of 10 for 48 hours. Mock-infected cells were used as control. Caspase-3 activation was quantified using the flow cytometric analysis. Representative flow cytometric data are shown in B. The mean percentage of SaOS-2 cells and MNNG/HOS cells that express active caspase-3 was calculated on the basis of 3 independent experiments (C). The cell-cycle state was analyzed by flow cytometry in SaOS-2 cells after staining with propidium iodide. Representative cell-cycle data are shown (D). The percentage of sub-G1 population was expressed in each graph. Bars, SD. Statistical significance was determined using Student t test. *, P < 0.05.

Figure 2.

OBP-702 induces increased apoptosis when compared with OBP-301 or Ad-p53. A, OBP-301–resistant SaOS-2 and MNNG/HOS cells were infected with OBP-702 or Ad-p53 at the indicated MOIs for 72 hours. Cell lysates were subjected to Western blot analysis for the C-PARP and PARP. β-Actin was assayed as a loading control. B–D, SaOS-2 and MNNG/HOS cells were infected with OBP-702, OBP-301, or Ad-p53 at an MOI of 10 for 48 hours. Mock-infected cells were used as control. Caspase-3 activation was quantified using the flow cytometric analysis. Representative flow cytometric data are shown in B. The mean percentage of SaOS-2 cells and MNNG/HOS cells that express active caspase-3 was calculated on the basis of 3 independent experiments (C). The cell-cycle state was analyzed by flow cytometry in SaOS-2 cells after staining with propidium iodide. Representative cell-cycle data are shown (D). The percentage of sub-G1 population was expressed in each graph. Bars, SD. Statistical significance was determined using Student t test. *, P < 0.05.

Close modal

p53 induction in human osteosarcoma cells infected with OBP-702

To investigate the molecular mechanism of OBP-702–induced apoptosis in human osteosarcoma cells, we evaluated p53 expression after OBP-702 infection in SaOS-2 (p53-null) and MNNG/HOS (p53-mutant) cells in which endogenous p53 expression level was confirmed by Western blot analysis (Supplementary Fig. S3). OBP-702 efficiently induced p53 expression in SaOS-2 and MNNG/HOS cells (Fig. 3A). The level of p53 expression was higher in OBP-702–treated cells than in Ad-p53–treated cells (Fig. 3A). Despite of OBP-702–induced high p53 expression, p53 downstream target p21 protein was induced only in Ad-p53–treated cells.

Figure 3.

OBP-702 induces p53 overexpression with E1A-mediated p21 suppression via miR-93 and miR-106b activation. A, expression of the p53 and p21 proteins in SaOS-2 and MNNG/HOS cells infected with OBP-702 or Ad-p53 at the indicated MOIs for 72 hours was assessed using Western blot analysis. B, expression of the E2F1 and viral E1A proteins in SaOS-2 and MNNG/HOS cells infected with OBP-702 at the indicated MOIs for 72 hours was assessed using Western blot analysis. C, expression of miR-93 and miR-106b was assayed using qRT-PCR in SaOS-2 cells infected with OBP-702 at the indicated MOIs for 72 hours on 3 independent experiments. The values of miR-93 and miR-106b at 0 MOI were set at 1, and the relative levels of miR-93 and miR-106b at the indicated MOIs were plotted as fold induction. Bars, SD. Statistical significance was determined by Student t test. *, P < 0.05. D, SaOS-2 cells were transfected with 10 nmol/L miR-93, miR-106, or control miRNA 24 hours before Ad-p53 infection at an MOI of 100. At 48 hours after Ad-p53 infection, the expression levels of p53, p21, PARP, and C-PARP were examined by Western blot analysis. β-Actin was assayed as a loading control. By using ImageJ software, the expression level of C-PARP protein was calculated relative to its expression in the control miR–treated cells, whose expression level was designated as 1.0.

Figure 3.

OBP-702 induces p53 overexpression with E1A-mediated p21 suppression via miR-93 and miR-106b activation. A, expression of the p53 and p21 proteins in SaOS-2 and MNNG/HOS cells infected with OBP-702 or Ad-p53 at the indicated MOIs for 72 hours was assessed using Western blot analysis. B, expression of the E2F1 and viral E1A proteins in SaOS-2 and MNNG/HOS cells infected with OBP-702 at the indicated MOIs for 72 hours was assessed using Western blot analysis. C, expression of miR-93 and miR-106b was assayed using qRT-PCR in SaOS-2 cells infected with OBP-702 at the indicated MOIs for 72 hours on 3 independent experiments. The values of miR-93 and miR-106b at 0 MOI were set at 1, and the relative levels of miR-93 and miR-106b at the indicated MOIs were plotted as fold induction. Bars, SD. Statistical significance was determined by Student t test. *, P < 0.05. D, SaOS-2 cells were transfected with 10 nmol/L miR-93, miR-106, or control miRNA 24 hours before Ad-p53 infection at an MOI of 100. At 48 hours after Ad-p53 infection, the expression levels of p53, p21, PARP, and C-PARP were examined by Western blot analysis. β-Actin was assayed as a loading control. By using ImageJ software, the expression level of C-PARP protein was calculated relative to its expression in the control miR–treated cells, whose expression level was designated as 1.0.

Close modal

To investigate the effect of exogenous p53 overexpression in virus replication, we next compared the replication abilities of OBP-702 and OBP-301 in p53-null SaOS-2 cells by measuring the relative amounts of E1A copy numbers. The E1A copy number of OBP-702 was similar to that of OBP-301 in SaOS-2 cells (Supplementary Fig. S4). These results indicate that OBP-702 efficiently induces exogenous p53 expression without affecting p21 expression and virus replication in human osteosarcoma cells.

OBP-702–mediated upregulation of miR-93 and miR-106b suppresses p21 expression

Adenoviral E1A protein has been shown to activate E2F1 expression (28), which is a multifunctional transcription factor that regulates diverse cell fates through induction of many target genes, including small noncoding miRNAs (29). Recently, E2F1-inducible miR-93 and miR-106b have been shown to suppress p21 expression in human cancer cells (30). Therefore, we sought to investigate whether OBP-702 induces expressions of E2F1 and E2F1-regulated miRNAs (miR-93 and miR-106b). OBP-702 infection activated E2F1 expression along with E1A accumulation in SaOS-2 and MNNG/HOS cells (Fig. 3B). The expression levels of miR-93 and miR-106b were increased in association with E2F1 activation in OBP-702–infected SaOS-2 and MNNG/HOS cells (Fig. 3C). In contrast, E1A-deleted Ad-p53 infection did not increase expressions of E2F1 and E2F1-regulated miR-93 and miR-106b (data not shown). Next, we assessed whether upregulation of miR-93 and miR-106b efficiently suppresses p21 expression induced by Ad-p53–mediated p53 overexpression. Ad-p53 infection at MOIs of 10 and 100 efficiently induced p21 expression at 48 hours after infection in SaOS-2 cells (Supplementary Fig. S5). When SaOS-2 cells were infected with Ad-p53 at an MOI of 100 for 48 hours, pretransfection with miR-93, miR-106b, or both efficiently suppressed Ad-p53–induced p21 expression (Fig. 3D). Interestingly, both miR-93- and miR-106b–transfected SaOS-2 cells showed the 1.5-fold increased expression of cleaved PARP (C-PARP) in consistency with remarkable p21 downregulation when compared with those transfected with control miR. However, the expression level of C-PARP was not increased in the miR-93- or miR-106b–transfected SaOS-2 cells, although transfection with miR-93 or miR-106b moderately decreased p21 expression. These results suggest that OBP-702 suppresses p21 expression through E1A-dependent upregulation of both E2F1-inducible miR-93 and miR-106b and contributes to induction of apoptosis.

Increased induction of autophagy by OBP-702 when compared with OBP-301

Recently, we showed that oncolytic adenovirus OBP-301 mainly induces programmed cell death in association with autophagy rather than apoptosis in human tumor cells (31). Therefore, we next investigated whether OBP-702 induces more profound autophagy than does OBP-301. Western blot analysis revealed that OBP-702 infection showed increased autophagy, which was confirmed by conversion of LC3-I to LC3-II (increased ratio of LC3-II/LC3-I) and p62 downregulation, when compared with OBP-301 in MNNG/HOS cells (Fig. 4A). Moreover, the expression level of the p53-induced modulator of autophagy, DRAM (32), was decreased after OBP-301 infection, but its expression was maintained after OBP-702 infection (Fig. 4A). As p53-mediated p21 overexpression is known to inhibit both apoptosis and autophagy (17, 18), we further evaluated whether miR-mediated p21 suppression is involved in the enhancement of p53-mediated autophagy induction. Ad-p53–induced autophagy was enhanced by miR-93- and miR-106b–mediated p21 suppression (Fig. 4B). These results suggest that OBP-702 induces more profound autophagy than does OBP-301 and that this effect occurs via p53-mediated DRAM activation and miR-mediated p21 suppression.

Figure 4.

OBP-702 induces increased autophagy when compared with OBP-301. A, MNNG/HOS cells were infected with OBP-702 or OBP-301 at the indicated MOIs for 72 hours. Cell lysates were subjected to Western blot analysis for LC3, p62, and DRAM. B, SaOS-2 cells were transfected with 10 nmol/L miR-93, miR-106, or control miRNA 24 hours before Ad-p53 infection. At 48 hours after Ad-p53 infection at an MOI of 100, the expression levels of LC3, p62, and DRAM were examined by Western blot analysis. β-Actin was assayed as a loading control. By using ImageJ software, the ratio of LC3-II/LC3-I expressions was calculated relative to its expression in the mock-infected cells, whose expression level was designated as 1.0.

Figure 4.

OBP-702 induces increased autophagy when compared with OBP-301. A, MNNG/HOS cells were infected with OBP-702 or OBP-301 at the indicated MOIs for 72 hours. Cell lysates were subjected to Western blot analysis for LC3, p62, and DRAM. B, SaOS-2 cells were transfected with 10 nmol/L miR-93, miR-106, or control miRNA 24 hours before Ad-p53 infection. At 48 hours after Ad-p53 infection at an MOI of 100, the expression levels of LC3, p62, and DRAM were examined by Western blot analysis. β-Actin was assayed as a loading control. By using ImageJ software, the ratio of LC3-II/LC3-I expressions was calculated relative to its expression in the mock-infected cells, whose expression level was designated as 1.0.

Close modal

Enhanced antitumor effect of OBP-702 in an orthotopic xenograft tumor model

Finally, to assess the in vivo antitumor effect of OBP-702, we used an orthotopic MNNG/HOS tumor xenograft model. OBP-702, OBP-301, Ad-p53, or PBS were intratumorally injected for 3 cycles every 2 days. OBP-702 administration significantly suppressed tumor growth when compared with OBP-301, Ad-p53, or PBS in an orthotopic MNNG/HOS tumor model (Fig. 5A and B). 3D-CT examination revealed that OBP-702–treated tumors had less bone destruction than did OBP-301- or Ad-p53–treated tumors (Fig. 5C). On histopathologic analysis, there were large necrotic areas in OBP-702–treated tumors but not in OBP-301- or Ad-p53–treated tumors (Fig. 5D). Moreover, the expression of the cell proliferation marker, Ki67, was also decreased, especially in OBP-702–treated tumor cells (Supplementary Fig. S6). These results suggest that OBP-702 eliminates tumor tissues more efficiently when compared with OBP-301 or Ad-p53.

Figure 5.

Antitumor effect of OBP-702 in an orthotopic MNNG/HOS osteosarcoma xenograft model. A, athymic nude mice were inoculated intratibially with MNNG/HOS cells (5 × 106 cells/site). Twenty-one days after inoculation (designated as day 0), Ad-p53, OBP-301, or OBP-702 were injected into the tumor with 1 × 108 PFUs on days 0, 2, and 4 (black arrows). PBS was used as a control. Three mice were used for each group. Each tumor volume was assessed by CT examination. Tumor growth was expressed as mean tumor volume ± SD. Statistical significance was determined by Student t test. *, P < 0.05. B, macroscopic appearance of MNNG/HOS tumors in nude mice on days 0 and 28 after treatment with PBS, Ad-p53, OBP-301, or OBP-702. Tumor masses are outlined by a dotted line. C, 3D-CT images of MNNG/HOS tumors. The tumor volumes were calculated by the image viewer (INTAGE Realia) based on 3D-CT images of tumors after trimming. The white arrowheads indicate the osteolytic areas within tumor tissues treated with PBS, Ad-p53, or OBP-301. Left side images are low magnification and right side images are high magnification of the area outlined by a white square. D, histologic analysis of the MNNG/HOS tumors. Tumor tissues were obtained on day 28 after first treatment with PBS, Ad-p53, OBP-301, or OBP-702. Paraffin-embedded sections of MNNG/HOS tumors were stained with hematoxylin and eosin solutions. There were large necrotic areas in MNNG/HOS tumors treated with OBP-702. Left side images are low magnification and right side images are high magnification of the area outlined by a white square. Left scale bars, 500 μm. Right scale bars, 100 μm.

Figure 5.

Antitumor effect of OBP-702 in an orthotopic MNNG/HOS osteosarcoma xenograft model. A, athymic nude mice were inoculated intratibially with MNNG/HOS cells (5 × 106 cells/site). Twenty-one days after inoculation (designated as day 0), Ad-p53, OBP-301, or OBP-702 were injected into the tumor with 1 × 108 PFUs on days 0, 2, and 4 (black arrows). PBS was used as a control. Three mice were used for each group. Each tumor volume was assessed by CT examination. Tumor growth was expressed as mean tumor volume ± SD. Statistical significance was determined by Student t test. *, P < 0.05. B, macroscopic appearance of MNNG/HOS tumors in nude mice on days 0 and 28 after treatment with PBS, Ad-p53, OBP-301, or OBP-702. Tumor masses are outlined by a dotted line. C, 3D-CT images of MNNG/HOS tumors. The tumor volumes were calculated by the image viewer (INTAGE Realia) based on 3D-CT images of tumors after trimming. The white arrowheads indicate the osteolytic areas within tumor tissues treated with PBS, Ad-p53, or OBP-301. Left side images are low magnification and right side images are high magnification of the area outlined by a white square. D, histologic analysis of the MNNG/HOS tumors. Tumor tissues were obtained on day 28 after first treatment with PBS, Ad-p53, OBP-301, or OBP-702. Paraffin-embedded sections of MNNG/HOS tumors were stained with hematoxylin and eosin solutions. There were large necrotic areas in MNNG/HOS tumors treated with OBP-702. Left side images are low magnification and right side images are high magnification of the area outlined by a white square. Left scale bars, 500 μm. Right scale bars, 100 μm.

Close modal

We previously reported that telomerase-specific replication-competent oncolytic adenovirus OBP-301 has strong antitumor activity in a variety of human epithelial and nonepithelial malignant cells (12, 14, 27). However, some human osteosarcoma cells were resistant to the cytopathic activity of OBP-301 (14). In this study, we showed that a novel p53-expressing oncolytic adenovirus, OBP-702, had increased in vitro and in vivo antitumor effects than did OBP-301 in human osteosarcoma cells (Fig. 1 and 5). OBP-702 induced increased apoptosis in association with p53 upregulation and p21 downregulation when compared with replication-deficient Ad-p53 (Fig. 2 and 3A). E1A-dependent upregulation of miR-93 and miR-106b was involved in OBP-702–mediated suppression of p21 expression (Fig. 3). Moreover, p53-mediated DRAM activation with p21 suppression enhanced oncolytic adenovirus-mediated autophagy induction (Fig. 4). Recent studies suggest that transgene-expressing armed oncolytic adenoviruses are a promising antitumor strategy for induction of oncolytic and transgene-induced cell death (33). Although p53 overexpression has been shown to enhance antitumor activity of oncolytic adenoviruses (34), the molecular mechanisms by which p53 mediates enhancement of the antitumor effect remain unclear. Recently, we reported that OBP-702 induces profound apoptosis through p53-dependent BAX upregulation and E1A-dependent p21 and MDM2 downregulation in epithelial malignant cells (26). Thus, oncolytic adenovirus-mediated p53 overexpression likely induces dual apoptotic and autophagic cell death pathways through p53-dependent BAX/DRAM activation and E1A-dependent p21/MDM2 suppression with E2F1-inducible miR-93/106b upregulation (Fig. 6).

Figure 6.

Outline of OBP-702–mediated induction of dual programmed cell death pathways. OBP-702 infection induces apoptosis and autophagy, leading to cell death, through p53-dependent BAX/DRAM upregulation and E1A-dependent p21 downregulation via E2F1-inducible miR-93/106b activation.

Figure 6.

Outline of OBP-702–mediated induction of dual programmed cell death pathways. OBP-702 infection induces apoptosis and autophagy, leading to cell death, through p53-dependent BAX/DRAM upregulation and E1A-dependent p21 downregulation via E2F1-inducible miR-93/106b activation.

Close modal

OBP-702 efficiently suppressed the cell viability of both OBP-301–sensitive and -resistant osteosarcoma cells (Fig. 1). We previously reported that OBP-301–resistant SaOS-2 cells have no hTERT mRNA expression (Table 1), suggesting that SaOS-2 cells maintain telomere length through alternative lengthening of telomeres (ALT). As hTERT gene promoter is used for tumor-specific replication of OBP-301, ALT-type human osteosarcoma cells such as SaOS-2 cells may be resistant to OBP-301. However, ALT-type SaOS-2 cells showed similar sensitivity to OBP-702 as well as non–ALT-type MNNG/HOS cells (Fig. 1 and Table 1). These results suggest that p53 overexpression overcomes resistance to OBP-301 in ALT-type SaOS-2 cells. As the replication rate of OBP-702 was almost similar that of OBP-301 in ALT-type SaOS-2 cells (Supplementary Fig. S2), p53-induced cell death pathway would suppress the cell viability of ALT-type human osteosarcoma cells.

OBP-702–mediated p53 overexpression induced 2 types of programmed cell deaths (i.e., apoptosis and autophagy), thereby contributing to the enhancement of the antitumor effect of OBP-301 in human osteosarcoma cells (Fig. 2 and 4). As p53 downstream target p21 functions as a suppressor of apoptosis and autophagy (17, 18), p21 suppression may be a critical factor to induce dual programmed cell death pathways in response p53 overexpression. Suppression of p21 expression by genetic deletion or artificial p21 target microRNA has been shown to enhance the Ad-p53–induced apoptosis (18, 35). Inactivation of p21 by adenoviral E1A has been shown to enhance apoptosis in chemotherapeutic drug-treated human colon cancer cells that overexpress p53 (36). Genetic deletion of p21 has been also shown to induce autophagy in mouse embryonic fibroblasts treated with C(2)-ceramide or γ-irradiation (17). In contrast, p21 overexpression inhibited the Ad-p53–mediated apoptosis induction (18). Thus, E1A-mediated p21 downregulation would enhance p53-induced apoptosis and autophagy in OBP-702–infected cells.

E1A-dependent E2F1 activation and subsequent upregulation of E2F1-inducible miRNAs efficiently suppressed p21 expression, leading to the enhancement of p53-induced apoptosis and autophagy, in OBP-702–infected osteosarcoma cells (Figs. 2–4). Recent studies suggest that the cross-talk between p53 and E2F1 play a role in the regulation of diverse cell fates (37). For example, co-expression of p53 and E2F1 contributes to induction of apoptosis (38, 39). We previously showed that E2F1 enhanced Ad-p53–mediated apoptosis through p14ARF-dependent MDM2 downregulation (39) and that OBP-702 infection showed E1A-dependent MDM2 downregulation in association with apoptosis (26). Recently, E2F1 has been shown to suppress MDM2 expression by suppressing the promoter activity (40) or by inducing upregulation of miR-25/32, which targets MDM2 (41). Furthermore, E2F1-inducible miR-93/106b enhanced Ad-p53–induced apoptosis and autophagy via p21 suppression (Figs. 3D and 4B). Therefore, the cooperation between the MDM2/p53/p21 pathway and the E2F1/miRNA pathway may be involved in the induction of apoptotic and autophagic cell death in response to OBP-702.

OBP-702–mediated p53 overexpression enhanced autophagy that was induced by oncolytic adenovirus in human osteosarcoma cells. OBP-702 infection induced increased expression of DRAM and decreased expression of p62 when compared with OBP-301 (Fig. 4), suggesting that OBP-702–mediated p53 overexpression enhances autophagy through DRAM activation. We recently reported that OBP-301 induces autophagy through E1A-dependent activation of E2F1/miR-7 pathway and subsequent suppression of EGF receptor (EGFR; ref. 31). Restoration of p53 expression enhances the sensitivity to EGFR inhibitors in human cancer cells (42). Moreover, EGFR downregulation by transfection of specific antisense oligonucleotide promotes the differentiation status of human osteosarcoma U2OS cells (43). Thus, OBP-702 may induce differentiation as well as cell death through autophagy activation by DRAM upregulation and EGFR downregulation in human osteosarcoma cells.

The 3D-CT imaging system was a useful method to assess both tumor volume and bone destruction status in MNNG/HOS tumors. OBP-702–treated tumors were smaller and had less bone destruction than PBS-, Ad-p53-, or OBP-301–treated tumors (Fig. 5A and C). Recent reports have suggested that zoledronic acid suppresses tumor growth as well as osteolytic components in human osteosarcoma xenograft tumor models (44, 45). These results suggest that combination therapy with OBP-702 and zoledronic acid may be more effective and more protective against bone destruction in human osteosarcomas. Further study using a 3D-CT imaging system may provide important information about bone destruction status in osteosarcomas treated with OBP-702 and zoledronic acid.

Adenovirus-mediated p53 gene therapy exerts an antitumor effect in human osteosarcoma cells (46). However, the antitumor activity of replication-deficient Ad-p53 is limited in some human osteosarcoma cells (47). Ad-p53–mediated p53 overexpression increases the sensitivity of human osteosarcoma cells to the chemotherapeutic drugs, cisplatin and doxorubicin (48). A synergistic antitumor effect between doxorubicin and roscovitine was also associated with autophagy induction in human osteosarcoma U2OS cells (49). As OBP-702 induced more profound apoptosis and autophagy than did OBP-301 or Ad-p53 (Fig. 2 and 4), combination therapy with OBP-702 and chemotherapeutic agents may be more effective than monotherapy with OBP-702. Moreover, a recent report has shown that p53-armed replication-competent oncolytic adenovirus is a safe antitumor agent in rodents and non-human primates (50). However, for clinical application of OBP-702, it must be necessary to establish the systemic delivery method and confirm the host biologic contributions in patients with cancer. Although there are some unsolved issues, the combination of p53-armed oncolytic adenovirus and chemotherapy may provide us a promising antitumor strategy against human osteosarcoma cells.

In conclusion, we clearly showed that the p53-expressing oncolytic adenovirus OBP-702 has a much stronger antitumor effect than does OBP-301. Oncolytic adenovirus-mediated p53 gene transduction may induce dual apoptotic and autophagic cell death pathways through p53-dependent activation of cell death inducers and E1A-dependent suppression of cell death inhibitors, resulting in the enhancement of antitumor effect.

Y. Urata is President & CEO of Oncolys BioPharma, Inc., the manufacturer of OBP-301 (Telomelysin). H. Tazawa and T. Fujiwara are consultants of Oncolys BioPharma, Inc. No potential conflicts of interest were disclosed by the other authors.

Conception and design: J. Hasei, F. Uno, S. Kagawa, T. Ozaki, T. Fujiwara

Development of methodology: J. Hasei, F. Uno, S. Kagawa

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J. Hasei, H. Tazawa, S. Osaki, Y. Yamakawa, A. Yoshida, T. Onishi, T. Ozaki

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J. Hasei, H. Tazawa, T. Fujiwara

Writing, review, and/or revision of the manuscript: J. Hasei, H. Tazawa, T. Kunisada, Y. Urata, T. Fujiwara

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): H. Tazawa, T. Kunisada, Y. Hashimoto, S. Kagawa, Y. Urata, T. Ozaki

Study supervision: T. Sasaki, T. Kunisada, F. Uno, Y. Urata, T. Fujiwara

The authors thank Dr. Satoru Kyo (Kanazawa University) for providing the HOS and SaOS-2 cells and Tomoko Sueishi for her excellent technical support.

This study was supported by grants-in-aid from the Ministry of Education, Science, and Culture, Japan (T. Fujiwara) and grants from the Ministry of Health and Welfare, Japan (T. Fujiwara), and in part by the National Cancer Center Research and Development Fund (23-A-10) (T. Ozaki).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ottaviani
G
,
Jaffe
N
. 
The etiology of osteosarcoma
.
Cancer Treat Res
2009
;
152
:
15
32
.
2.
Damron
TA
,
Ward
WG
,
Stewart
A
. 
Osteosarcoma, chondrosarcoma, and Ewing's sarcoma: National Cancer Data Base Report
.
Clin Orthop Relat Res
2007
;
459
:
40
7
.
3.
Lewis
IJ
,
Nooij
MA
,
Whelan
J
,
Sydes
MR
,
Grimer
R
,
Hogendoorn
PC
, et al
Improvement in histologic response but not survival in osteosarcoma patients treated with intensified chemotherapy: a randomized phase III trial of the European Osteosarcoma Intergroup
.
J Natl Cancer Inst
2007
;
99
:
112
28
.
4.
Bacci
G
,
Longhi
A
,
Versari
M
,
Mercuri
M
,
Briccoli
A
,
Picci
P
. 
Prognostic factors for osteosarcoma of the extremity treated with neoadjuvant chemotherapy: 15-year experience in 789 patients treated at a single institution
.
Cancer
2006
;
106
:
1154
61
.
5.
Bielack
SS
,
Kempf-Bielack
B
,
Delling
G
,
Exner
GU
,
Flege
S
,
Helmke
K
, et al
Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols
.
J Clin Oncol
2002
;
20
:
776
90
.
6.
Siegel
R
,
Naishadham
D
,
Jemal
A
. 
Cancer statistics, 2012
.
CA Cancer J Clin
2012
;
62
:
10
29
.
7.
Buseman
CM
,
Wright
WE
,
Shay
JW
. 
Is telomerase a viable target in cancer?
Mutat Res
2012
;
730
:
90
7
.
8.
Artandi
SE
,
DePinho
RA
. 
Telomeres and telomerase in cancer
.
Carcinogenesis
2010
;
31
:
9
18
.
9.
Umehara
N
,
Ozaki
T
,
Sugihara
S
,
Kunisada
T
,
Morimoto
Y
,
Kawai
A
, et al
Influence of telomerase activity on bone and soft tissue tumors
.
J Cancer Res Clin Oncol
2004
;
130
:
411
6
.
10.
Aogi
K
,
Woodman
A
,
Urquidi
V
,
Mangham
DC
,
Tarin
D
,
Goodison
S
. 
Telomerase activity in soft-tissue and bone sarcomas
.
Clin Cancer Res
2000
;
6
:
4776
81
.
11.
Nakayama
J
,
Tahara
H
,
Tahara
E
,
Saito
M
,
Ito
K
,
Nakamura
H
, et al
Telomerase activation by hTRT in human normal fibroblasts and hepatocellular carcinomas
.
Nat Genet
1998
;
18
:
65
8
.
12.
Kawashima
T
,
Kagawa
S
,
Kobayashi
N
,
Shirakiya
Y
,
Umeoka
T
,
Teraishi
F
, et al
Telomerase-specific replication-selective virotherapy for human cancer
.
Clin Cancer Res
2004
;
10
:
285
92
.
13.
Nemunaitis
J
,
Tong
AW
,
Nemunaitis
M
,
Senzer
N
,
Phadke
AP
,
Bedell
C
, et al
A phase I study of telomerase-specific replication competent oncolytic adenovirus (telomelysin) for various solid tumors
.
Mol Ther
2010
;
18
:
429
34
.
14.
Sasaki
T
,
Tazawa
H
,
Hasei
J
,
Kunisada
T
,
Yoshida
A
,
Hashimoto
Y
, et al
Preclinical evaluation of telomerase-specific oncolytic virotherapy for human bone and soft tissue sarcomas
.
Clin Cancer Res
2011
;
17
:
1828
38
.
15.
Li
G
,
Kawashima
H
,
Ogose
A
,
Ariizumi
T
,
Xu
Y
,
Hotta
T
, et al
Efficient virotherapy for osteosarcoma by telomerase-specific oncolytic adenovirus
.
J Cancer Res Clin Oncol
2011
;
137
:
1037
51
.
16.
Vousden
KH
,
Prives
C
. 
Blinded by the light: the growing complexity of p53
.
Cell
2009
;
137
:
413
31
.
17.
Fujiwara
K
,
Daido
S
,
Yamamoto
A
,
Kobayashi
R
,
Yokoyama
T
,
Aoki
H
, et al
Pivotal role of the cyclin-dependent kinase inhibitor p21WAF1/CIP1 in apoptosis and autophagy
.
J Biol Chem
2008
;
283
:
388
97
.
18.
Gorospe
M
,
Cirielli
C
,
Wang
X
,
Seth
P
,
Capogrossi
MC
,
Holbrook
NJ
. 
p21(Waf1/Cip1) protects against p53-mediated apoptosis of human melanoma cells
.
Oncogene
1997
;
14
:
929
35
.
19.
Blagosklonny
MV
,
el-Deiry
WS
. 
In vitro evaluation of a p53-expressing adenovirus as an anti-cancer drug
.
Int J Cancer
1996
;
67
:
386
92
.
20.
Zeng
Y
,
Prabhu
N
,
Meng
R
,
Eldeiry
W
. 
Adenovirus-mediated p53 gene therapy in nasopharyngeal cancer
.
Int J Oncol
1997
;
11
:
221
6
.
21.
Clayman
GL
,
el-Naggar
AK
,
Lippman
SM
,
Henderson
YC
,
Frederick
M
,
Merritt
JA
, et al
Adenovirus-mediated p53 gene transfer in patients with advanced recurrent head and neck squamous cell carcinoma
.
J Clin Oncol
1998
;
16
:
2221
32
.
22.
Swisher
SG
,
Roth
JA
,
Nemunaitis
J
,
Lawrence
DD
,
Kemp
BL
,
Carrasco
CH
, et al
Adenovirus-mediated p53 gene transfer in advanced non-small-cell lung cancer
.
J Natl Cancer Inst
1999
;
91
:
763
71
.
23.
Shimada
H
,
Matsubara
H
,
Shiratori
T
,
Shimizu
T
,
Miyazaki
S
,
Okazumi
S
, et al
Phase I/II adenoviral p53 gene therapy for chemoradiation resistant advanced esophageal squamous cell carcinoma
.
Cancer Sci
2006
;
97
:
554
61
.
24.
Fujiwara
T
,
Tanaka
N
,
Kanazawa
S
,
Ohtani
S
,
Saijo
Y
,
Nukiwa
T
, et al
Multicenter phase I study of repeated intratumoral delivery of adenoviral p53 in patients with advanced non-small-cell lung cancer
.
J Clin Oncol
2006
;
24
:
1689
99
.
25.
Sakai
R
,
Kagawa
S
,
Yamasaki
Y
,
Kojima
T
,
Uno
F
,
Hashimoto
Y
, et al
Preclinical evaluation of differentially targeting dual virotherapy for human solid cancer
.
Mol Cancer Ther
2010
;
9
:
1884
93
.
26.
Yamasaki
Y
,
Tazawa
H
,
Hashimoto
Y
,
Kojima
T
,
Kuroda
S
,
Yano
S
, et al
A novel apoptotic mechanism of genetically engineered adenovirus-mediated tumour-specific p53 overexpression through E1A-dependent p21 and MDM2 suppression
.
Eur J Cancer
2012
;
48
:
2282
91
.
27.
Hashimoto
Y
,
Watanabe
Y
,
Shirakiya
Y
,
Uno
F
,
Kagawa
S
,
Kawamura
H
, et al
Establishment of biological and pharmacokinetic assays of telomerase-specific replication-selective adenovirus
.
Cancer Sci
2008
;
99
:
385
90
.
28.
Bagchi
S
,
Raychaudhuri
P
,
Nevins
JR
. 
Adenovirus E1A proteins can dissociate heteromeric complexes involving the E2F transcription factor: a novel mechanism for E1A trans-activation
.
Cell
1990
;
62
:
659
69
.
29.
Emmrich
S
,
Putzer
BM
. 
Checks and balances: E2F-microRNA crosstalk in cancer control
.
Cell Cycle
2010
;
9
:
2555
67
.
30.
Petrocca
F
,
Vecchione
A
,
Croce
CM
. 
Emerging role of miR-106b-25/miR-17-92 clusters in the control of transforming growth factor beta signaling
.
Cancer Res
2008
;
68
:
8191
4
.
31.
Tazawa
H
,
Yano
S
,
Yoshida
R
,
Yamasaki
Y
,
Sasaki
T
,
Hashimoto
Y
, et al
Genetically engineered oncolytic adenovirus induces autophagic cell death through an E2F1-microRNA-7-epidermal growth factor receptor axis
.
Int J Cancer
2012
;
131
:
2939
50
.
32.
Crighton
D
,
Wilkinson
S
,
O'Prey
J
,
Syed
N
,
Smith
P
,
Harrison
PR
, et al
DRAM, a p53-induced modulator of autophagy, is critical for apoptosis
.
Cell
2006
;
126
:
121
34
.
33.
Liu
TC
,
Galanis
E
,
Kirn
D
. 
Clinical trial results with oncolytic virotherapy: a century of promise, a decade of progress
.
Nat Clin Pract Oncol
2007
;
4
:
101
17
.
34.
van Beusechem
VW
,
van den Doel
PB
,
Grill
J
,
Pinedo
HM
,
Gerritsen
WR
. 
Conditionally replicative adenovirus expressing p53 exhibits enhanced oncolytic potency
.
Cancer Res
2002
;
62
:
6165
71
.
35.
Idogawa
M
,
Sasaki
Y
,
Suzuki
H
,
Mita
H
,
Imai
K
,
Shinomura
Y
, et al
A single recombinant adenovirus expressing p53 and p21-targeting artificial microRNAs efficiently induces apoptosis in human cancer cells
.
Clin Cancer Res
2009
;
15
:
3725
32
.
36.
Chattopadhyay
D
,
Ghosh
MK
,
Mal
A
,
Harter
ML
. 
Inactivation of p21 by E1A leads to the induction of apoptosis in DNA-damaged cells
.
J Virol
2001
;
75
:
9844
56
.
37.
Polager
S
,
Ginsberg
D
. 
p53 and E2f: partners in life and death
.
Nat Rev Cancer
2009
;
9
:
738
48
.
38.
Wu
X
,
Levine
AJ
. 
p53 and E2F-1 cooperate to mediate apoptosis
.
Proc Natl Acad Sci U S A
1994
;
91
:
3602
6
.
39.
Itoshima
T
,
Fujiwara
T
,
Waku
T
,
Shao
J
,
Kataoka
M
,
Yarbrough
WG
, et al
Induction of apoptosis in human esophageal cancer cells by sequential transfer of the wild-type p53 and E2F-1 genes: involvement of p53 accumulation via ARF-mediated MDM2 down-regulation
.
Clin Cancer Res
2000
;
6
:
2851
9
.
40.
Tian
X
,
Chen
Y
,
Hu
W
,
Wu
M
. 
E2F1 inhibits MDM2 expression in a p53-dependent manner
.
Cell Signal
2011
;
23
:
193
200
.
41.
Suh
SS
,
Yoo
JY
,
Nuovo
GJ
,
Jeon
YJ
,
Kim
S
,
Lee
TJ
, et al
MicroRNAs/TP53 feedback circuitry in glioblastoma multiforme
.
Proc Natl Acad Sci U S A
2012
;
109
:
5316
21
.
42.
Huang
S
,
Benavente
S
,
Armstrong
EA
,
Li
C
,
Wheeler
DL
,
Harari
PM
. 
p53 modulates acquired resistance to EGFR inhibitors and radiation
.
Cancer Res
2011
;
71
:
7071
9
.
43.
Salvatori
L
,
Caporuscio
F
,
Coroniti
G
,
Starace
G
,
Frati
L
,
Russo
MA
, et al
Down-regulation of epidermal growth factor receptor induced by estrogens and phytoestrogens promotes the differentiation of U2OS human osteosarcoma cells
.
J Cell Physiol
2009
;
220
:
35
44
.
44.
Dass
CR
,
Choong
PF
. 
Zoledronic acid inhibits osteosarcoma growth in an orthotopic model
.
Mol Cancer Ther
2007
;
6
:
3263
70
.
45.
Labrinidis
A
,
Hay
S
,
Liapis
V
,
Ponomarev
V
,
Findlay
DM
,
Evdokiou
A
. 
Zoledronic acid inhibits both the osteolytic and osteoblastic components of osteosarcoma lesions in a mouse model
.
Clin Cancer Res
2009
;
15
:
3451
61
.
46.
Ternovoi
VV
,
Curiel
DT
,
Smith
BF
,
Siegal
GP
. 
Adenovirus-mediated p53 tumor suppressor gene therapy of osteosarcoma
.
Lab Invest
2006
;
86
:
748
66
.
47.
Hellwinkel
OJ
,
Muller
J
,
Pollmann
A
,
Kabisch
H
. 
Osteosarcoma cell lines display variable individual reactions on wildtype p53 and Rb tumour-suppressor transgenes
.
J Gene Med
2005
;
7
:
407
19
.
48.
Ganjavi
H
,
Gee
M
,
Narendran
A
,
Parkinson
N
,
Krishnamoorthy
M
,
Freedman
MH
, et al
Adenovirus-mediated p53 gene therapy in osteosarcoma cell lines: sensitization to cisplatin and doxorubicin
.
Cancer Gene Ther
2006
;
13
:
415
9
.
49.
Lambert
LA
,
Qiao
N
,
Hunt
KK
,
Lambert
DH
,
Mills
GB
,
Meijer
L
, et al
Autophagy: a novel mechanism of synergistic cytotoxicity between doxorubicin and roscovitine in a sarcoma model
.
Cancer Res
2008
;
68
:
7966
74
.
50.
Su
C
,
Cao
H
,
Tan
S
,
Huang
Y
,
Jia
X
,
Jiang
L
, et al
Toxicology profiles of a novel p53-armed replication-competent oncolytic adenovirus in rodents, felids, and nonhuman primates
.
Toxicol Sci
2008
;
106
:
242
50
.