Terminal prostate cancer is refractory to conventional anticancer treatments because of frequent overexpression of antiapoptotic proteins Bcl-2 and/or Bcl-xL. Adenovirus-mediated delivery of melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24), a secreted cytokine having cancer-selective apoptosis-inducing properties, profoundly inhibits prostate cancer cell growth. However, forced overexpression of Bcl-2 or Bcl-xL renders prostate cancer cells resistant to Ad.mda-7. We constructed a conditionally replication-competent adenovirus in which expression of the adenoviral E1A gene, necessary for replication, is driven by the cancer-specific promoter of progression elevated gene-3 (PEG-3) and which simultaneously expresses mda-7/IL-24 in the E3 region of the adenovirus (Ad.PEG-E1A-mda-7), a cancer terminator virus (CTV). This CTV generates large quantities of MDA-7/IL-24 as a function of adenovirus replication uniquely in cancer cells. Infection of Ad.PEG-E1A-mda-7 (CTV) in normal prostate epithelial cells and parental and Bcl-2– or Bcl-xL–overexpressing prostate cancer cells confirmed cancer cell–selective adenoviral replication, mda-7/IL-24 expression, growth inhibition, and apoptosis induction. Injecting Ad.PEG-E1A-mda-7 (CTV) into xenografts derived from DU-145-Bcl-xL cells in athymic nude mice completely eradicated not only primary tumors but also distant tumors (established in the opposite flank), thereby implementing a cure. These provocative findings advocate potential therapeutic applications of this novel virus for advanced prostate cancer patients with metastatic disease. [Cancer Res 2007;67(11):5434–42]

Prostate cancer represents one of the most important health problems in industrialized countries (1, 2). It is the most common cancer and the second leading cause of cancer-related deaths in men in the United States. In 2006, the estimated new prostate cancer cases were 234,460, of which 27,350 men were estimated to die predominantly from metastatic prostate cancer in the United States. Therapeutic options vary according to the stage of the disease at the time of presentation and diagnosis. Patients with localized disease may be treated with surgery or radiation, whereas the treatment for patients with metastatic disease is purely palliative (1, 2). Hormonal treatment with antiandrogens is the standard therapy for stage IV prostate cancer, but patients ultimately become nonresponsive to androgen ablation (1, 2). Current therapy options for patients with hormone-refractory prostate cancer include radiotherapy and cytotoxic chemotherapeutic agents, such as mitoxantrone, estramustine, and taxanes (13). Despite a palliative benefit, none of these approaches engender a beneficial effect on the overall survival of patients. Consequently, no consistently effective therapy exists for these patients mandating the development of novel, more efficacious, and innovative treatment approaches, especially those targeting metastasis.

Overexpression of antiapoptotic proteins Bcl-2 and Bcl-xL is a frequent occurrence in prostate cancer development and progression (46). Bcl-2 immunostaining increases with Gleason grade of prostate cancer and Bcl-2 expression is a predictor of poor prognosis in prostate cancer patients (710). Moreover, Bcl-2 expression is augmented following androgen ablation and correlates with progression of prostate carcinomas from androgen dependence to androgen independence (1113). Bcl-xL overexpression associates with resistance to drug-induced apoptosis in prostate cancer cells (1416). Forced overexpression of Bcl-2 has been shown to desensitize prostate cancer cells to apoptotic stimuli (17, 18). Accordingly, strategies that overcome apoptosis resistance afforded by either Bcl-2 or Bcl-xL overexpression would clearly have important therapeutic implications in treating patients with prostate cancer.

Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24) was originally identified as a gene associated with terminal differentiation and irreversible growth suppression of metastatic human melanoma cells (19, 20). mda-7 belongs to the IL-10 family of cytokines that include IL-19, IL-20, IL-22, mda-7/IL-24, and IL-26 (21, 22). A unique property of mda-7/IL-24, when delivered by an adenoviral expression system (Ad.mda-7), is selective induction of growth suppression and apoptosis in a broad spectrum of human cancers, including prostate carcinoma, without exerting any deleterious effects to their normal counterparts (reviewed in refs. 2326). In addition to its direct apoptosis-inducing properties, Ad.mda-7 also shows antiangiogenic, radiosensitizing, immunostimulatory, and potent “bystander” antitumor activity (2732). A phase I clinical trial evaluating Ad.mda-7 (INGN 241) activity by intratumoral injection in patients with advanced solid tumors was done, and the results indicate that mda-7/IL-24 is safe and could induce as much as 70% apoptosis in tumors following a single injection of recombinant virus, and multiple injections promote an objective clinical response (23, 33, 34). These exciting results provide direct support for using mda-7/IL-24 in potentially developing an effective gene-based therapy for cancer.

In multiple cancer subtypes, Ad.mda-7 infection reduces the levels of antiapoptotic proteins, including Bcl-2 and/or Bcl-xL, and enhances expression of proapoptotic proteins, including Bax and/or Bak, thus shifting the balance towards an apoptotic phenotype and tumor cell death (2426, 35). Although parental prostate cancer cells are highly susceptible to Ad.mda-7–induced apoptosis, stable overexpression of Bcl-2 and Bcl-xL renders prostate cancer cells resistant to the apoptotic effects of Ad.mda-7 (36). A differential protective effect of Bcl-2 and Bcl-xL is evident in human prostate cancer cells. Although Bcl-2 is primarily responsible for making LNCaP prostate cancer cells resistant to Ad.mda-7, it is Bcl-xL that promotes this resistance in DU-145 and PC-3 cells (36).

The present study explored the efficacy of a conditionally replication-competent adenovirus (CRCA)–based cancer gene therapy approach for eradicating resistant prostate cancer cells. We constructed a CRCA in which replication is driven by a minimal active region of the promoter of progression elevated gene-3 (PEG-3), which functions selectively in diverse cancer cells with limited activity in normal cells, generating a cancer terminator virus (CTV; refs. 37, 38). PEG-3 was cloned as an up-regulated transcript from a transformation progression rodent cancer model, and attractively, the activity of its promoter (PEG-Prom) was found to be significantly and often markedly higher not only in rodent but also in human cancer cells of diverse origin when compared with normal cells (3941). The cancer cell specificity of the PEG-Prom is governed by two transcription factors (activator protein-1 and polyoma enhancing activator-3) that are expressed at elevated levels, either singly or in combination, in virtually all types of cancers (37, 38, 40, 41). Employing the PEG-Prom to drive GFP or luciferase via a replication-incompetent adenovirus confirmed prominent cancer cell–specific transgene expression in human prostate and breast cancer cells as well as in malignant glioma cells (41). These observations prompted us to investigate the use of the PEG-Prom to drive expression of the E1A gene, necessary for adenovirus replication, to create cancer cell–specific CRCAs (37, 38). One engineered CRCA simultaneously expresses mda-7/IL-24 in the E3 region (Ad.PEG-E1A-mda-7; CTV), thereby mediating robust production of this cytokine as a function of adenoviral replication (37). We now show that Ad.PEG-E1A-mda-7 (CTV) effectively circumvents Bcl-2– and Bcl-xL–dependent resistance of prostate cancer cells and eradicates both primary and distant tumors in a nude mouse xenograft model. These observations establish that Ad.PEG-E1A-mda-7 (CTV) might prove to be an efficient tool for elimination of terminal metastatic and therapy-refractory prostate cancers in patients.

Cell lines, culture conditions, and viability assays. DU-145, PC-3, and LNCaP prostate cancer cells were obtained from the American Type Culture Collection and cultured as described (36). The generation and characterization of DU-145-Bcl-xL, PC-3-Bcl-xL, and LNCaP-Bcl-2 have been described previously (36). P69 cells are normal human prostate epithelial cells immortalized by SV40 T/t antigen and are cultured as described (32). Cell viability was determined by standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays (35).

Construction of a CRCA. To construct the CRCA Ad.PEG-E1A-mda-7, a CTV, the AdenoQuick cloning system from OD 260, Inc. was employed (37, 38). This system uses two shuttle vectors (pE1.2 and pE3.1) in which the transgene cassettes, PEG-Prom driving E1A and cytomegalovirus (CMV) promoter driving mda-7/IL-24, were inserted, respectively, before being transferred into a large adenoviral plasmid (pAd). Adenoviral amplification, purification, titration, and infection were done as described (37, 38). Similar strategies were used to generate Ad.CMV-E1A-mda-7. Ad.CMV-mda-7 and Ad.PEG-mda-7 were constructed as previously described (37).

Annexin V binding assay. Annexin V binding assays were done as described (36).

Preparation of whole-cell lysates and Western blot analyses. Preparation of whole-cell lysates and Western blot analyses was done as described (37). The primary antibodies used were anti-E1A (1:1,000; mouse monoclonal; Upstate Biotechnology), anti-MDA-7 (1:2,000; rabbit polyclonal), and anti-EF1α (1:1,000; mouse monoclonal; Upstate Biotechnology).

Human prostate cancer xenografts in athymic nude mice. DU-145-Bcl-xL cells (2 × 106) were injected s.c. in 100 μL of PBS in both flanks of male athymic nude mice (NCRnu/nu, 4 weeks old, ∼20 g body weight; refs. 37, 38). After establishment of visible tumors of ∼75 mm3, requiring ∼4 to 5 days, intratumoral injections of different adenoviruses were given only to the tumors on the left flank at a dose of 1 × 108 plaque-forming units in 100 μL. No injection was given to the right-sided tumors. The injections were given thrice a week for the first week and then twice a week for two more weeks for a total of seven injections. A minimum of five animals was used per experimental point. Tumor volume was measured twice weekly with a caliper and calculated using the formula: π/6 × larger diameter × (smaller diameter)2. At the end of the experiment, the animals were sacrificed, and the tumors were removed and weighed.

Immunofluorescence analysis. Tumors were harvested from the animals, fixed in formalin, and embedded in paraffin. The sections were deparaffinized and were permeabilized with 0.1% Triton X-100 in PBS for 30 min. Sections were then blocked for 1 h at room temperature with 2% goat serum and 1% bovine serum albumin in PBS and incubated with anti-E1A antibody (1:100) or anti-MDA-7 antibody (1:100) overnight at 4°C. Sections were then rinsed in PBS and incubated with Alexa 488–conjugated anti-mouse or anti-rabbit IgG (Molecular Probes), respectively, for 1 h at room temperature. The sections were mounted in VectaShield fluorescence mounting medium containing 4,6-diamidino-2-phenylindole (Vector Laboratories). For CD31 staining, FITC-conjugated rat anti-mouse CD31 monoclonal antibody (BD PharMingen) was used. A confocal laser scanning microscope analyzed the images.

Statistical analysis. Statistical analysis was done using one-way ANOVA, followed by Fisher's protected least significant difference analysis. P < 0.05 was considered significant.

PEG-Prom targets selective expression of Ad E1A and MDA-7/IL-24 in prostate cancer cells. Apart from Ad.PEG-E1A-mda-7 (CTV), a series of additional adenoviruses were created, such as Ad.CMV-E1A-mda-7, in which viral replication is controlled by the CMV promoter and which also expresses mda-7/IL-24, and Ad.CMV-E1A and Ad.PEG-E1A, in which viral replication is controlled by the CMV promoter or the PEG-Prom, respectively, to compare and contrast their relative efficacy. We also employed Ad.CMV-mda-7 and Ad.PEG-mda-7, replication-incompetent adenoviruses in which the CMV or the PEG promoter drives mda-7/IL-24 expression, respectively. A replication-incompetent empty adenovirus, Ad.vec was used as a control. Experiments were done in three prostate cancer cell lines, androgen-nonresponsive DU-145 and PC-3 cells, and androgen-responsive LNCaP cells and their Ad.mda-7-resistant variants (i.e., DU-145-Bcl-xL, PC-3-Bcl-xL, and LNCaP-Bcl-2). These Bcl-2– or Bcl-xL–overexpressing clones and their resistance to Ad.mda-7 have been extensively characterized in previous studies (36). As a control, P69, a normal prostate epithelial cells immortalized by SV40 T/t antigen, was employed. The replication-incompetent adenoviruses were infected at a multiplicity of infection of 5,000 viral particles per cell, whereas the replication-competent adenoviruses were infected at 1,000 viral particles per cell. The functionality of these constructs was ascertained following adenovirus infection by monitoring protein levels of MDA-7/IL-24 and E1A, a marker for adenoviral replication, by Western blot analysis after appropriate viral infection (Fig. 1). Western blot analysis detects multiple E1A gene products ranging from 36 to 50 kDa and multiple glycosylated forms of MDA-7/IL-24 protein ranging from 21 to 28 kDa (37, 38).

Figure 1.

PEG-Prom promotes adenovirus replication and transgene expression selectively in prostate cancer cells. The indicated cells were uninfected (control) or infected with the indicated adenovirus (as described at the top) at a multiplicity of infection of 1,000 viral particles per cell for replication-competent adenovirus and 5,000 viral particles per cell for replication-incompetent adenovirus for 48 h. The expressions of E1A, MDA-7/IL-24, and EF1-α proteins were analyzed by Western blot analyses.

Figure 1.

PEG-Prom promotes adenovirus replication and transgene expression selectively in prostate cancer cells. The indicated cells were uninfected (control) or infected with the indicated adenovirus (as described at the top) at a multiplicity of infection of 1,000 viral particles per cell for replication-competent adenovirus and 5,000 viral particles per cell for replication-incompetent adenovirus for 48 h. The expressions of E1A, MDA-7/IL-24, and EF1-α proteins were analyzed by Western blot analyses.

Close modal

Infection of normal immortal human P69 prostate epithelial cells with Ad.CMV-E1A or Ad.CMV-E1A-mda-7, but not Ad.PEG-E1A or Ad.PEG-E1A-mda-7 (CTV), resulted in production of E1A proteins, whereas in prostate carcinoma cells, infection with all four replication-competent adenoviruses generated E1A proteins (Fig. 1). No E1A proteins were detected in any cell line following infection with replication-incompetent adenoviruses. In P69 cells, infection with Ad.CMV-E1A-mda-7 and Ad.CMV-mda-7 resulted in MDA-7/IL-24 production, whereas infection with Ad.PEG-mda-7 or Ad.PEG-E1A-mda-7 (CTV) resulted in barely detectable levels of MDA-7/IL-24 production (Fig. 1). In prostate cancer cells, infection with Ad.CMV-mda-7, Ad.PEG-mda-7, Ad.CMV-E1A-mda-7, or Ad.PEG-E1A-mda-7 (CTV) generated significant MDA-7/IL-24 production. No MDA-7/IL-24 protein production could be detected in control uninfected cells or following infection with Ad.vec, Ad.CMV-E1A, or Ad.PEG-E1A. These findings document that the PEG-Prom facilitates cancer cell–selective replication of adenovirus and mda-7/IL-24 expression.

Ad.PEG-E1A-mda-7 (CTV) selectively kills prostate cancer cells, without harming normal prostate cells. The effects of the engineered adenoviruses on cell viability and apoptosis were evaluated in the various prostate cell lines. Cells were infected with replication-competent adenoviruses at 10, 100, and 1,000 viral particles per cell and with replication-incompetent adenoviruses at 1,000, 2,000, and 5,000 viral particles per cell. In P69 cells, infection with only Ad.CMV-E1A or Ad.CMV-E1A-mda-7, but not with Ad.PEG-E1A, Ad.CMV-mda-7, Ad.PEG-mda-7, or Ad.PEG-E1A-mda-7 (CTV), induced profound growth inhibition (Fig. 2). In contrast, in all prostate cancer cells, both parental and mda-7/IL-24–resistant, Ad.CMV-E1A-mda-7, Ad.PEG-E1A-mda-7 (CTV), Ad.CMV-E1A, and Ad.PEG-E1A infection resulted in significant growth inhibition. Infection with Ad.CMV-mda-7 and Ad.PEG-mda-7 inhibited growth of parental DU-145, PC-3, and LNCaP cells but not their resistant counterparts. It should be noted that the level of growth inhibition observed with 5,000 viral particles per cell of Ad.CMV-mda-7 and Ad.PEG-mda-7 was equivalent to that observed with only 100 viral particles per cell of replication-competent adenovirus, indicating that the replication-competent adenoviruses are much more potent than the replication-incompetent adenoviruses in cell growth inhibition. These findings indicate that the PEG-Prom allows adenovirus replication specifically in cancer cells, protecting normal cells from growth inhibition because of adenovirus replication. The observation that mda-7/IL-24 exerted no direct growth inhibitory effect on normal cells confirms the cancer cell selectivity of this therapeutic approach. Most importantly, Ad.PEG-E1A-mda-7 was able to overcome the resistance of Bcl-2– and Bcl-xL–overexpressing clones to mda-7/IL-24, indicating its potential therapeutic application in prostate cancer patients frequently showing Bcl-2 and Bcl-xL overexpression.

Figure 2.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) selectively kills prostate cancer cells. The indicated cells were uninfected (control) or infected with the indicated adenovirus (as described at the top) at the indicated multiplicity of infection (viral particles per cell). Cell viability was analyzed by standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay after 2, 4, and 6 d of infection. Columns, mean; bars, SD.

Figure 2.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) selectively kills prostate cancer cells. The indicated cells were uninfected (control) or infected with the indicated adenovirus (as described at the top) at the indicated multiplicity of infection (viral particles per cell). Cell viability was analyzed by standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay after 2, 4, and 6 d of infection. Columns, mean; bars, SD.

Close modal

Ad.PEG-E1A-mda-7 (CTV) selectively induces apoptosis in prostate cancer cells. To investigate the mechanism of growth inhibition, Annexin V staining assays, which permit differentiation between apoptotic and necrotic cells, were done (Fig. 3). Infection with only Ad.CMV-E1A and Ad.CMV-E1A-mda-7 (CTV) elevated the percentage of early-apoptotic and late-apoptotic (necrotic) P69 cells. However, all of the adenoviruses, except for Ad.vec, resulted in significant apoptosis in DU-145, PC-3, and LNCaP parental prostate cancer cells. Infection with the replication-competent adenoviruses resulted predominantly in necrosis as manifested by an increase in late apoptotic cells, whereas infection with Ad.CMV-mda-7 and Ad.PEG-mda-7 resulted in predominantly apoptosis as evidenced by an increase in early-apoptotic cells. Although Ad.CMV-mda-7 and Ad.PEG-mda-7 had no apoptotic effect on DU-145-Bcl-xL, PC-3-Bcl-xL, and LNCaP-Bcl-2 cells, Ad.CMV-E1A-mda-7 and Ad.PEG-E1A-mda-7 (CTV) could override this resistance and induce significant apoptosis and necrosis in these cells. It is important to mention that whereas the replication-incompetent adenoviruses were used at 5,000 viral particles per cell, replication-competent adenoviruses were used at 1,000 viral particles per cell.

Figure 3.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) selectively induces apoptosis in prostate cancer cells. The indicated cells were treated as in Fig. 1. Annexin V staining was analyzed by flow cytometry 48 h after infection.

Figure 3.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) selectively induces apoptosis in prostate cancer cells. The indicated cells were treated as in Fig. 1. Annexin V staining was analyzed by flow cytometry 48 h after infection.

Close modal

Ad.PEG-E1A-mda-7 (CTV) eradicates both primary and distant Bcl-XL–overexpressing prostate tumors in nude mice. To expand on the in vitro studies, in vivo assays were done using nude mice containing established DU-145-Bcl-xL s.c. xenografts on both right and left flanks. After palpable tumors of ∼75 mm3 developed, in ∼4 to 5 days, seven intratumoral injections with different adenoviruses, thrice per week for the first week and twice per week for an additional 2 weeks, were administered to the tumors on the left flank at a dose of 1 × 1010 viral particles in 100 μL. No injections were given to the right-sided tumors. The experiment was terminated after 6 weeks with injections of Ad.CMV-E1A-mda-7 or Ad.PEG-E1A-mda-7 (CTV) because tumors on both sides showed regression after only three injections, and with seven injections they were completely eradicated (Fig. 4). Further studies are required to determine if a single or double injection with the CTV elicits any discernible antitumor activity in this animal model. Additionally, both control and Ad.vec–infected DU-145-Bcl-xL tumor xenografts reached tumor volumes of ∼2,000 mm3 requiring them to be sacrificed. Although Ad.CMV-E1A or Ad.PEG-E1A inhibited the growth of tumors on the left flank, they had some inhibitory effect on tumors on the right side, which was not statistically significant. Ad.CMV-mda-7 or Ad.PEG-mda-7 also displayed marginal effects on the growth of both left- and right-sided tumors, which correlates with the in vitro findings of resistance of DU-145-Bcl-xL cells to mda-7/IL-24. The observation that intratumoral injection of Ad.PEG-E1A-mda-7 (CTV) completely eradicated the primary and the distant tumor (comparable with a metastasis) provides confidence that this strategy may prove amenable for successfully treating aggressive cancers.

Figure 4.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) eradicates primary and distant tumors. S.c. tumor xenografts from DU-145-Bcl-xL cells were established in athymic nude mice in both right and left flanks, and only tumors on the left side were injected with PBS (control) or with the indicated adenovirus for 3 wks (total of seven injections). A, measurement of tumor volume. Points, mean (with a minimum of five mice in each group); bars, SD. B, photograph of the animals of each representative group. White arrow, tumor. C, left, photograph of the tumor at the end of the study; right, measurement of tumor weight at the end of the study. Columns, mean (with at least five mice in each group); bars, SD.

Figure 4.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) eradicates primary and distant tumors. S.c. tumor xenografts from DU-145-Bcl-xL cells were established in athymic nude mice in both right and left flanks, and only tumors on the left side were injected with PBS (control) or with the indicated adenovirus for 3 wks (total of seven injections). A, measurement of tumor volume. Points, mean (with a minimum of five mice in each group); bars, SD. B, photograph of the animals of each representative group. White arrow, tumor. C, left, photograph of the tumor at the end of the study; right, measurement of tumor weight at the end of the study. Columns, mean (with at least five mice in each group); bars, SD.

Close modal

Ad.PEG-E1A-mda-7 (CTV) replicates at distant tumor sites and generates MDA-7/IL-24 protein. Because Ad.CMV-E1A-mda-7 or Ad.PEG-E1A-mda-7 (CTV) eradicated both the left-sided injected and right-sided uninjected tumors, we analyzed the replication efficiency and transgene delivery by these adenoviruses (Fig. 5). Tumors were harvested from the animals after three injections and formalin-fixed, paraffin embedded sections were stained for E1A and MDA-7/IL-24 and analyzed by a confocal laser scanning microscopy. The replication-competent adenoviruses, and not the replication-incompetent adenoviruses, could effectively replicate in the left-sided tumors as evidenced by robust staining for E1A protein (Fig. 5). Interestingly, staining for E1A could also be detected in the right-sided tumors, albeit at a much lower level than their left-sided counterparts, indicating that the adenovirus could enter into the circulation and replicate in the right-sided tumor cells. Staining for MDA-7/IL-24 supported these findings (Fig. 5). Although all of the mda-7/IL-24 expressing adenoviruses effectively generated MDA-7/IL-24 protein in the left-sided tumors, only Ad.CMV-E1A-mda-7 and Ad.PEG-E1A-mda-7 (CTV), and not Ad.CMV-mda-7 and Ad.PEG-mda-7, generated MDA-7/IL-24 protein in the right-sided tumors. These data indicate that the combination of adenovirus replication and robust MDA-7/IL-24 generation resulted in a significant response that could effectively eliminate both the primary and distant tumor.

Figure 5.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) replicates at distant sites and generates MDA-7/IL-24 protein. Tumors were harvested after three injections of adenovirus. Formalin-fixed, paraffin-embedded sections were immunostained for E1A and MDA-7/IL-24 as described in Materials and Methods.

Figure 5.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) replicates at distant sites and generates MDA-7/IL-24 protein. Tumors were harvested after three injections of adenovirus. Formalin-fixed, paraffin-embedded sections were immunostained for E1A and MDA-7/IL-24 as described in Materials and Methods.

Close modal

Ad.PEG-E1A.mda-7 (CTV) inhibits angiogenesis. MDA-7/IL-24, as a secreted cytokine, is known to inhibit angiogenesis (34). Based on this consideration, we analyzed the microvessel density in the tumor by staining for CD31 (Fig. 6). After three injections, Ad.CMV-E1A and Ad.PEG-E1A showed marginal effects on angiogenesis inhibition on the left-sided tumors with no effect on the right-sided uninjected tumors. Ad.CMV-mda-7 and Ad.PEG-mda-7 significantly inhibited angiogenesis in both left- and right-sided tumors. Because these two adenoviruses did not affect on the growth of the tumors, it might be inferred that inhibition of angiogenesis alone may not be sufficient to inhibit the growth of the tumors, and inhibition of growth of the cancer cells themselves is mandatory to provoke an enduring antitumor effect. Ad.CMV-E1A-mda-7 and Ad.PEG-E1A-mda-7 (CTV) profoundly inhibited angiogenesis in both left- and right-sided tumors, indicating that the MDA-7/IL-24 protein generated exhibits its complete spectrum of biological activities (Fig. 6). The summation of these findings strongly implies that Ad.PEG-E1A-mda-7 (CTV) might be an effective therapeutic for efficiently eliminating aggressive and resistant prostate cancers.

Figure 6.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) inhibits angiogenesis. Tumors were harvested after three injections of adenovirus. Formalin-fixed, paraffin-embedded sections were immunostained for CD31 A, confocal images of the tumors stained for CD31. B, quantification of microvessel density in the tumors. Columns, mean; bars, SD.

Figure 6.

PEG-Prom–driven CRCA (Ad.PEG-E1A-mda-7; CTV) inhibits angiogenesis. Tumors were harvested after three injections of adenovirus. Formalin-fixed, paraffin-embedded sections were immunostained for CD31 A, confocal images of the tumors stained for CD31. B, quantification of microvessel density in the tumors. Columns, mean; bars, SD.

Close modal

Genetic therapies for prostate cancer represent promising strategies for the treatment of this neoplasm. The prostate gland is not vital for survival and is accessible by ultrasound; potential therapeutic genes can be injected directly into the primary tumor; tissue-specific promoters exist that can target therapeutic gene expression or viral replication uniquely in the prostate gland; and disease progression can be monitored by measuring prostate-specific antigen (42). Moreover, because prostate cancer is commonly a relatively slow-growing disease, it may be necessary to use repeated gene therapy applications, with single or multiple genes, over the life span of the patient. In these contexts, the use of replication defective adenoviruses to administer therapeutic and cytotoxic genes and CRCA to selectively induce cytolysis in prostate tumor cells represent viable treatment options.

Cancer is a complicated process involving abnormalities in multiple genetic and signal transduction pathways. As such, effective eradication of cancer requires a combinatorial treatment approach. CRCAs are being evaluated for treatment of prostate cancers (4345). Although administration of a CRCA alone in phase I/II clinical trials showed their safety, very limited objective clinical responses have been attained using this strategy (4345). Additionally, most currently employed CRCAs are based on the ONYX-015 backbone, which is dependent on the p53 status of the cancer cells, thus limiting their universal applicability (46). Our approach is unique and superior to other viral therapy approaches for several reasons. First, we employ the PEG-Prom that functions in all types of cancer cells, irrespective of their p53 or Rb status, with very limited to no activity in normal cells (38, 40, 41). Consequently, the PEG-Prom may provide a generic tool for ensuring cancer selectivity. Second, our approach involves the unique tumor suppressor molecule mda-7/IL-24 that not only has cancer-selective apoptosis-inducing properties but also displays a plethora of indirect antitumor “bystander” activities (2326). The finding that Ad.PEG-E1A-mda-7 (CTV) eradicated not only primary injected tumors, but also distant non-injected tumors derived from a resistant prostate cancer cell line in a nude mouse xenograft model support the anticancer potency of this cancer therapeutic virus. This powerful genetic tool is now ready for further evaluation in animal models, such as Syrian hamsters that facilitate adenovirus replication and therefore analysis of toxicity (47) and transgenic mouse models that recapitulate prostate cancer development, and eventually in phase I/II clinical trials.

The question is how does Ad.PEG-E1A-mda-7 (CTV) induce complete elimination of right-sided non-injected tumors. Our immunofluorescence studies show that Ad.PEG-E1A-mda-7 (CTV) is able to enter into the circulation and replicate and generate MDA-7/IL-24 protein in distant tumors. This observation is exciting because it indicates that the universality of PEG-Prom will allow adenovirus replication and transgene expression, wherever there is any tumor tissue in the animal. Additionally, we have observed that Ad.PEG-E1A-mda-7 (CTV) also effectively inhibited angiogenesis in right-sided tumors. Although Ad.CMV-mda-7 or Ad.PEG-mda-7 also inhibited angiogenesis in both left- and right-sided tumors, these adenoviruses had very limited effect on tumor growth owing to the inherent resistance of DU-145-Bcl-xL cells to mda-7/IL-24. Accordingly, it might be inferred that the combination of adenovirus replication with generation of robust amounts of MDA-7/IL-24, that can override resistance and evoke a profound “bystander effect,” is essential for promoting a clinically significant effect, especially in the case of metastatic, end-stage, and therapy-resistant cancers.

As a secreted cytokine, MDA-7/IL-24 induces IL-6, IFN-γ, tumor necrosis factor-α, IL-1β, IL-12, and granulocyte macrophage colony-stimulating factor, potent immunomodulatory agents, from peripheral blood mononuclear cells (30, 31). These secondary cytokines induced by MDA-7/IL-24 might activate antigen-presenting cells to present tumor antigens, thereby triggering an antitumor immune response. Studies in a phase I clinical trial involving intratumoral injection of Ad.mda-7 (INGN 241) suggest that these in vitro effects are recapitulated in the context of patients, supporting the immune-modulating properties of this cytokine (32, 33). Experiments done in syngeneic mouse tumor models also support this hypothesis (48). Although current studies were done in athymic nude mice that are immunocompromised, these mice still have a spleen and a liver and display potent natural killer cell activity (49). Therefore, in addition to inhibition of angiogenesis, augmentation of an antitumor immune response might also be a potential mechanism of the “bystander effect” of Ad.PEG-E1A-mda-7 (CTV). This function of mda-7/IL-24 is especially significant in a patient with an intact immune system where the effect of Ad.PEG-E1A-mda-7 (CTV) might be further augmented. In this context, the balance between clearance of adenovirus by the immune system and the modulation of the immune system by mda-7/IL-24 will likely represent major determinants in the antitumor potency of Ad.PEG-E1A-mda-7 (CTV) in patients. The robust activity of this adenovirus suggests a need for only limited administration, which in principle will preclude the activation of the immune system promoting viral clearance. Additionally, the observation that neutralizing anti-adenovirus antibodies do not inhibit replicating adenoviruses indicates that Ad.PEG-E1A-mda-7 (CTV) might engender a profound effect in inducing complete eradication of primary and metastatic cancers (50).

In summary, the present studies support the proposition that Ad.PEG-E1A-mda-7 (CTV) might provide a nontoxic effective alternative or complement to conventional adjuvant treatment modalities for patients with prostate cancer. Based on our previous combinatorial studies in cell culture and in animal models (reviewed in refs. 2426), a combination of Ad.PEG-E1A-mda-7 (CTV) with localized low-dose radiotherapy might promote an even more profound effect, potentially further enhancing patient survival and possibly inducing a cure. The feasibility of intratumoral injection of Ad.PEG-E1A-mda-7 (CTV) into prostate cancers, so that it is shielded from immune system-mediated clearance while evoking a potent antitumor response, makes this genetic vector an ideal therapeutic for prostate cancer. Current studies are ongoing for stringent evaluation of this CTV in immunocompetent animal models for eventual translation into clinical trials in patients with localized and metastatic prostate cancer.

Note: P.B. Fisher is the Michael and Stella Chernow Urological Cancer Research Scientist and a Samuel Waxman Cancer Research Foundation Investigator.

Grant support: NIH grants P01 CA104177, R01 CA35675, and R01 CA097318; Samuel Waxman Cancer Research Foundation; and Chernow Endowment (P.B. Fisher).

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
Sternberg CN. Highlights of contemporary issues in the medical management of prostate cancer.
Crit Rev Oncol Hematol
2002
;
43
:
105
–21.
2
Di Lorenzo G, De Placido S. Hormone refractory prostate cancer (HRPC): present and future approaches of therapy.
Int J Immunopathol Pharmacol
2006
;
19
:
11
–34.
3
Dyrstad SW, Shah P, Rao K. Chemotherapy for prostate cancer.
Curr Pharm Des
2006
;
12
:
819
–37.
4
Colombel M, Symmans F, Gil S, et al. Detection of the apoptosis-suppressing oncoprotein bc1-2 in hormone-refractory human prostate cancers.
Am J Pathol
1993
;
143
:
390
–400.
5
Krajewska M, Krajewski S, Epstein JI, et al. Immunohistochemical analysis of bcl-2, bax, bcl-X, and mcl-1 expression in prostate cancers.
Am J Pathol
1996
;
148
:
1567
–76.
6
Krajewski S, Krajewska M, Shabaik A, et al. Immunohistochemical analysis of in vivo patterns of Bcl-X expression.
Cancer Res
1994
;
54
:
5501
–7.
7
Bauer JJ, Sesterhenn IA, Mostofi FK, McLeod DG, Srivastava S, Moul JW. Elevated levels of apoptosis regulator proteins p53 and bcl-2 are independent prognostic biomarkers in surgically treated clinically localized prostate cancer.
J Urol
1996
;
156
:
1511
–6.
8
Keshgegian AA, Johnston E, Cnaan A. Bcl-2 oncoprotein positivity and high MIB-1 (Ki-67) proliferative rate are independent predictive markers for recurrence in prostate carcinoma.
Am J Clin Pathol
1998
;
110
:
443
–9.
9
Sullivan GF, Amenta PS, Villanueva JD, Alvarez CJ, Yang JM, Hait WN. The expression of drug resistance gene products during the progression of human prostate cancer.
Clin Cancer Res
1998
;
4
:
1393
–403.
10
Bai XZ, Masters JR, O'Donoghue N, et al. Prognostic markers in clinically localised prostate cancer.
Int J Oncol
1999
;
14
:
785
–91.
11
McDonnell TJ, Troncoso P, Brisbay SM, et al. Expression of the protooncogene bcl-2 in the prostate and its association with emergence of androgen-independent prostate cancer.
Cancer Res
1992
;
52
:
6940
–4.
12
Beham AW, Sarkiss M, Brisbay S, Tu SM, von Eschenbach AC, McDonnell TJ. Molecular correlates of bcl-2-enhanced growth following androgen-ablation in prostate carcinoma cells in vivo.
Int J Mol Med
1998
;
1
:
953
–9.
13
Kajiwara T, Takeuchi T, Ueki T, et al. Effect of Bcl-2 overexpression in human prostate cancer cells in vitro and in vivo.
Int J Urol
1999
;
6
:
520
–5.
14
Lebedeva I, Rando R, Ojwang J, Cossum P, Stein CA. Bcl-xL in prostate cancer cells: effects of overexpression and down-regulation on chemosensitivity.
Cancer Res
2000
;
60
:
6052
–60.
15
Li X, Marani M, Mannucci R, et al. Overexpression of BCL-XL underlies the molecular basis for resistance to staurosporine-induced apoptosis in PC-3 cells.
Cancer Res
2001
;
61
:
1699
–706.
16
Liu QY, Stein CA. Taxol and estramustine-induced modulation of human prostate cancer cell apoptosis via alteration in bcl-xL and bak expression.
Clin Cancer Res
1997
;
3
:
2039
–46.
17
Kyprianou N, King ED, Bradbury D, Rhee JG. bcl-2 over-expression delays radiation-induced apoptosis without affecting the clonogenic survival of human prostate cancer cells.
Int J Cancer
1997
;
70
:
341
–8.
18
Tu SM, McConnel K, Marin MC, et al. Combination Adriamycin and suramin induces apoptosis in bcl-2 expressing prostate carcinoma cells.
Cancer Lett
1995
;
93
:
147
–55.
19
Jiang H, Lin JJ, Su ZZ, Goldstein NI, Fisher PB. Subtraction hybridization identifies a novel melanoma differentiation associated gene, mda-7, modulated during human melanoma differentiation, growth and progression.
Oncogene
1995
;
11
:
2477
–86.
20
Jiang H, Su ZZ, Lin JJ, Goldstein NI, Young CSH, Fisher PB. The melanoma differentiation associated gene mda-7 suppresses cancer cell growth.
Proc Natl Acad Sci U S A
1996
;
93
:
9160
–5.
21
Pestka S, Krause CD, Sarkar D, Walter MR, Shi Y, Fisher PB. Interleukin-10 and related cytokines and receptors.
Annu Rev Immunol
2004
;
22
:
929
–79.
22
Huang EY, Madireddi MT, Gopalkrishnan RV, et al. Genomic structure, chromosomal localization and expression profile of a novel melanoma differentiation associated (mda-7) gene with cancer specific growth suppressing and apoptosis inducing properties.
Oncogene
2001
;
20
:
7051
–63.
23
Fisher PB, Sarkar D, Lebedeva IV, et al. Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24): novel gene therapeutic for metastatic melanoma. Toxicol Appl Pharmacol. In press 2007.
24
Fisher PB. Is mda-7/IL-24 a “magic bullet” for cancer?
Cancer Res
2005
;
65
:
10128
–38.
25
Gupta P, Su Z-z, Lebedeva IV, et al. mda-7/IL-24: multifunctional cancer-specific apoptosis-inducing cytokine.
Pharmacol Ther
2006
;
111
:
596
–628.
26
Lebedeva IV, Sauane M, Gopalkrishnan RV, et al. mda-7/IL-24: exploiting cancer's Achilles' heel.
Mol Ther
2005
;
11
:
4
–18.
27
Ramesh R, Mhashilkar AM, Tanaka F, et al. Melanoma differentiation-associated gene 7/interleukin (IL)-24 is a novel ligand that regulates angiogenesis via the IL-22 receptor.
Cancer Res
2003
;
63
:
5105
–13.
28
Yacoub A, Mitchell C, Lebedeva IV, et al. mda-7 (IL-24) Inhibits growth and enhances radiosensitivity of glioma cells in vitro via JNK signaling.
Cancer Biol Ther
2003
;
2
:
347
–53.
29
Su Z-z, Lebedeva IV, Sarkar D, et al. Melanoma differentiation associated gene-7, mda-7/IL-24, selectively induces growth suppression, apoptosis and radiosensitization in malignant gliomas in a p53-independent manner.
Oncogene
2003
;
22
:
1164
–80.
30
Caudell EG, Mumm JB, Poindexter N, et al. The protein product of the tumor suppressor gene, melanoma differentiation-associated gene 7, exhibits immunostimulatory activity and is designated IL-24.
J Immunol
2002
;
168
:
6041
–6.
31
Poindexter NJ, Walch ET, Chada S, Grimm EA. Cytokine induction of interleukin-24 in human peripheral blood mononuclear cells.
J Leukoc Biol
2005
;
78
:
745
–52. Epub 2005 Jul 6.
32
Su Z-z, Emdad L, Sauane M, et al. Unique aspects of mda-7/IL-24 antitumor bystander activity: establishing a role for secretion of MDA-7/IL-24 protein by normal cells.
Oncogene
2005
;
24
:
7552
–66.
33
Cunningham CC, Chada S, Merritt JA, et al. Clinical and local biological effects of an intratumoral injection of mda-7 (IL24; INGN 241) in patients with advanced carcinoma: a phase I study.
Mol Ther
2005
;
11
:
149
–59.
34
Tong AW, Nemunaitis J, Su D, et al. Intratumoral injection of INGN 241, a nonreplicating adenovector expressing the melanoma-differentiation associated gene-7 (mda-7/IL24): biologic outcome in advanced cancer patients.
Mol Ther
2005
;
11
:
160
–72.
35
Lebedeva IV, Su Z-z, Chang Y, Kitada S, Reed JC, Fisher PB. The cancer growth suppressing gene mda-7 induces apoptosis selectively in human melanoma cells.
Oncogene
2002
;
21
:
708
–18.
36
Lebedeva IV, Sarkar D, Su Z-z, et al. Bcl-2 and Bcl-xL differentially protect human prostate cancer cells from induction of apoptosis by melanoma differentiation associated gene-7, mda-7/IL-24.
Oncogene
2003
;
22
:
8758
–73.
37
Sarkar D, Su Z-z, Vozhilla N, Park ES, Gupta P, Fisher PB. Dual cancer-specific targeting strategy cures primary and distant breast carcinomas in nude mice.
Proc Natl Acad Sci U S A
2005
;
102
:
14034
–9.
38
Sarkar D, Su Z-z, Vozhilla N, et al. Targeted virus replication plus immunotherapy eradicates primary and distant pancreatic tumors in nude mice.
Cancer Res
2005
;
65
:
9056
–63.
39
Su ZZ, Shi Y, Fisher PB. Subtraction hybridization identifies a progression elevated gene PEG-3 with sequence homology to a growth arrest and DNA damage inducible gene.
Proc Natl Acad Sci U S A
1997
;
94
:
9125
–30.
40
Su ZZ, Shi Y, Fisher PB. Cooperation between AP1 and PEA3 sites within the progression elevated gene-3 (PEG-3) promoter regulate basal and differential expression of PEG-3 during progression of the oncogenic phenotype in transformed rat embryo cells.
Oncogene
2000
;
19
:
3411
–21.
41
Su Z-z, Sarkar D, Emdad L, et al. Targeting gene expression selectively in cancer cells by using the progression-elevated gene-3 promoter.
Proc Natl Acad Sci U S A
2005
;
102
:
1059
–64.
42
Gopalkrishnan RV, Kang DC, Fisher PB. Molecular markers and determinants of prostate cancer metastasis.
J Cell Physiol
2001
;
189
:
245
–56.
43
Small EJ, Schellhammer PF, Higano CS, et al. A phase I trial of intravenous CG7870, a replication-selective, prostate-specific antigen-targeted oncolytic adenovirus, for the treatment of hormone-refractory, metastatic prostate cancer.
Mol Ther
2006
;
14
:
107
–17.
44
Freytag SO, Stricker H, Pegg J, et al. Phase I study of replication-competent adenovirus-mediated double-suicide gene therapy in combination with conventional-dose three-dimensional conformal radiation therapy for the treatment of newly diagnosed, intermediate- to high-risk prostate cancer.
Cancer Res
2003
;
63
:
7497
–506.
45
DeWeese TL, van der Poel H, Li S, et al. A phase I trial of CV706, a replication-competent, PSA selective oncolytic adenovirus, for the treatment of locally recurrent prostate cancer following radiation therapy.
Cancer Res
2001
;
61
:
7464
–72.
46
Heise C, Sampson-Johannes A, Williams A, McCormick F, Von Hoff DD, Kirn DH. ONYX-015, an E1B gene-attenuated adenovirus, causes tumor-specific cytolysis and antitumoral efficacy that can be augmented by standard chemotherapeutic agents.
Nat Med
1997
;
3
:
639
–45.
47
Thomas MA, Spencer JF, La Regina MC, et al. Syrian hamster as a permissive immunocompetent animal model for the study of oncolytic adenovirus vectors.
Cancer Res
2006
;
66
:
1270
–6.
48
Miyahara R, Banerjee S, Kawano K, et al. Melanoma differentiation-associated gene-7 (mda-7)/interleukin (IL)-24 induces anticancer immunity in a syngeneic murine model.
Cancer Gene Ther
2006
;
13
:
753
–61.
49
Smyth MJ, Hayakawa Y, Takeda K, Yagita H. New aspects of natural-killer-cell surveillance and therapy of cancer.
Nat Rev Cancer
2002
;
2
:
850
–61.
50
Tsai V, Johnson DE, Rahmnan A, et al. Impact of human neutralizing antibodies on antitumor efficacy of an oncolytic adenovirus in a murine model.
Clin Cancer Res
2004
;
10
:
7199
–206.