Conditionally replicating adenoviruses (CRAd) have been under extensive investigations as anticancer agents. Previously, we found that ZD55, an adenovirus serotype 5-based CRAd, infected and killed the leukemia cells expressing coxsackie adenovirus receptor (CAR). However, majority of leukemic cells lack CAR expression on their cell surface, resulting in resistance to CRAd infection. In this study, we showed that SG235, a novel fiber chimeric CRAd that has Ad35 tropism, permitted CAR-independent cell entry, and this in turn produced selective cytopathic effects in a variety of human leukemic cells in vitro and in vivo. Moreover, SG235 expressing exogenous tumor necrosis factor-related apoptosis-inducing ligand (SG235-TRAIL) effectively induced apoptosis of leukemic cells via the activation of extrinsic and intrinsic apoptotic pathway and elicited a superior antileukemia activity compared with SG235. In addition, normal hematopoietic progenitors were resistant to the inhibitory activity of SG235 and SG235-TRAIL. Our data suggest that these novel oncolytic agents may serve as useful tools for the treatment of leukemia. [Mol Cancer Ther 2009;8(5):1387–97]

Novel therapeutic approaches for cancer treatment are urgently needed because conventional treatment regimens are frequently acutely toxic, nonselective, or ineffective. Oncolytic virotherapy provides a new platform to treat cancer as biotherapeutic agents that lack cross-resistance with currently available treatments (1, 2) and represents a novel approach for gene therapy (3, 4). Results from preclinical studies with several viral types are promising and some oncolytic viruses have entered or even completed clinical trials (5, 6); however, little of the previously used therapeutic viruses is able to infect hematopoietic malignant cells. Recently, oncolytic measles viruses have been shown to be active in preclinical in vitro and animal models of multiple myeloma (79) and lymphoma (1012). These data have raised the possibility of clinical application of virotherapy for leukemia.

Conditionally replicating adenoviruses (CRAd) have attracted considerable interest owing to their several attributes including lytic replication, high stability, efficient genome transfer, and low pathogenicity (13). The most studied CRAd in cancer therapy is adenovirus serotype 5 (Ad5), which requires expression of coxsackie adenovirus receptor (CAR) on target cells for successful transduction (1416). Unfortunately, hematopoietic and leukemia cells express few CARs, which essentially renders the tumor cell resistant to Ad5 infection (14, 15, 17). To enhance tumor cell infectivity, concerted efforts have been made to modify fiber knob domain of Ad5. In this regard, recent reports showed that a nonreplicating adenovirus independent of CAR by substitution of a chimeric Ad5/35 fiber improved infectious efficiency and antitumor gene therapy effects of the Ad5 tropism in leukemic cells (18, 19). In this study, we constructed a novel fiber chimeric Ad5 oncolytic vector (SG235) that has Ad35 tropism generated through the substitution of the Ad5 fiber protein by the Ad35 knob domain. Then, SG235 was armed to express exogenous apoptosis-inducing gene tumor necrosis factor-related apoptosis-inducing ligand (SG235-TRAIL). The infectivity and cytotoxic effects of SG235 and SG235-TRAIL on leukemic cell lines and primary blasts obtained from leukemia patients were determined, and the potent antitumor activity in human leukemia xenograft mouse model was evaluated.

Primary Leukemia Samples, Cell Lines, and Reagents

Bone marrow aspirates from the patients with acute myeloid leukemia (AML; n = 70), acute promyelocytic leukemia (n = 8), B-cell acute lymphocytic leukemia (n = 37), and T-cell acute lymphocytic leukemia (n = 7) were obtained after institutional review board approval and informed patient consent. The percentage of infiltrating blasts in the bone marrow was >85%. Mononuclear cells were prepared as described (3). To examine the apoptosis of primary leukemic cells induced by CRAds, we established a short time culture of patient's blasts as reported previously (20). Briefly, mononuclear cells from another 6 patients with AML were allowed to settle in MEM (Life Technologies) for 17 min at 37°C to eliminate cell sticking. The nonsticking cells were cultured in MEM with 15% heat-inactivated FCS (Hyclone Laboratories). Viability was better than 96% after 72 h as determined by trypan blue dye exclusion.

Human T-cell leukemia lines Molt-4 and Jurkat, chronic myelogenous leukemia line K562, AML line HL-60, myeloma line RPMI 8226, and lymphoma line L428 were purchased from the American Type Culture Collection. B-cell leukemia line Mutz-1 was provided by Dr. Z Hu, and AML line Kasumi was kindly provided by Prof. S Chen. Cells were cultured in RPMI 1640 (Hyclone) supplemented with 10% FCS (Hyclone) and 1% l-glutamine (Life Technologies). Human embryonic kidney cell line (Microbix Biosystem) and cervical cancer cell line HeLa (Shanghai Cell Collection) were maintained in MEM supplemented with FCS. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide was purchased from Sigma, and caspase inhibitors z-DEVD-FMK and z-IETD-FMK were from Bio Vision.

Construction of Recombinant Adenovirus Vectors

Vectors ZD55, pZD55-TRAIL, and pZD55-enhanced green fluorescence protein (GFP) were constructed as described previously (21, 22). Plasmid DC311-TRAIL was constructed as follows: the TRAIL expression cassette of pZD55-TRAIL was cloned into shuttle plasmid (pDC311) by cutting with BglII (New England Biolabs) and religating to generate pDC311-TRAIL. The resulting shuttle vector (pDC311-TRAIL) or pZD55-TRAIL was cotransfected with a serotype 35 adenovirus vector (pPE35) into 293 cells to generate the Ad-TRAIL and SG235-TRAIL CRAd, respectively, through homologous recombination.

Cell Viability Assay

Leukemic cells were plated on 96-well plates at 1 × 104 per well 1 day before virus infection. Cells were then infected with viruses at the indicated multiplicity of infection (MOI) for 48 h. PBS was used as a control. The cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and trypan blue dye exclusion as described previously (23).

Western Blotting

Western blot analysis was done as described previously (23), with 40 mg protein loaded on 12% SDS-PAGE. The primary antibodies used were as follows: caspase-9, caspase-8, caspase-3, poly(ADP-ribose) polymerase, Bcl-1, Bid, Bcl-xL, and Bax antibodies were purchased from Cell Signaling Technology. Bim and Mcl-1 were obtained from Epitomics. GAPDH and actin were used as housekeeping protein control and provided by Kangchen. After incubation with secondary antibodies (KPL), blots were revealed by enhanced chemiluminescence procedures according to the manufacturer's recommendation.

Flow Cytometric Analysis

FITC-conjugated anti-human CD46 (BD Pharmingen) and phycoerythrin-conjugated anti-human DR4, DR5, DcR1, and DcR2 (eBioscience) antibodies were used. Stained cells were analyzed on a FACSCalibur flow cytometer (Becton Dickinson).

Apoptosis was assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining (Roche) and phycoerythrin-conjugated Annexin V apoptosis detection kit (Becton Dickinson) following the manufacturer's instructions. For mitochondrial membrane potential evaluation, cells were stained with 5 μg/mL rhodamine 123 (Invitrogen) at 37°C for 30 min and then measured at 530 nm using flow cytometry.

ELISA for TRAIL

An ELISA for levels of TRAIL in leukemic cell supernatant was done using a Quantikine kit as per the manufacturer's instructions (R&D Systems).

Colony-Forming Cell Assay

Leukemic colony formation assay was done as described elsewhere (24, 25). Briefly, blasts were collected from the peripheral blood or bone marrow of untreated AML patients. Cells were cultured in MEM for 20 min at 37°C in plastic flasks, and nonsticking cells were collected and resuspended in serum-free MEM with or without viruses at MOI of 50 for 4 h and plated at 2 × 105/mL in methylcellulose, growth medium, and 10% phytohemagglutinin-leukocyte conditioned medium for incubation at 37°C for 6 days. Colonies containing in excess of 20 cells were counted under an inverted microscope (Olympus). For normal hematopoietic progenitor assays, bone marrow cells (2 × 105/mL) were exposed to viruses (50 MOI) in serum-free medium for 4 h and cultured in 0.8% methylcellulose and 20% FCS/Iscove's modified Dulbecco's medium supplemented with granulocyte-macrophage colony-stimulating factor (Sigma). The cultures in triplicates were maintained for 6 days with humidity and 5% CO2, after which colonies were enumerated.

Animal Studies

The local animal care and use committee approved this study. Cell implantation and virus treatment were done as described (3). Briefly, 3- to 4-week-old female severe combined immunodeficient mice (Shanghai Experimental Animal Center of the Chinese Academy of Sciences) were injected with 1 × 107 Kasumi cells in 100 μL s.c. into the hind flanks. Tumor volume was measured and calculated as π/6 length × width2. When the tumors reached a volume of ∼120 mm3, animals were randomly assigned to treatment groups and received three intratumoral injections of 1.7 × 108 plaque-forming units viruses diluted in a volume of 100 μL PBS. The untreated control received PBS. On the day 5 after treatments, one mouse of each group was humanely killed, and tumors were harvested and then processed for TUNEL assay using In situ Cell Death Detection Kit (Roche).

Statistical Analysis

Differences among the treatment groups were assessed by ANOVA using GraphPad Prism 4. P < 0.05 was considered significant.

Infection and Replication of Chimeric Oncolytic Adenoviruses in Leukemia Cells

First, we analyzed transduction efficiency of SG235 in a panel of leukemia cell lines. Infection of Kasumi cells, which were shown to be CAR negative (3), with SG235-GFP at MOIs of 5, 25, and 50 resulted in a dose- and time-dependent shift in fluorescence (Fig. 1A). Flow cytometric analysis showed that SG235 infected all tested hematopoietic malignant cell lines (Fig. 1B). To assess the replication ability of SG235, we compared the viral production of SG235 with that of ZD55, a 5 type oncolytic adenovirus. The total viral particles in the culture medium and cell fraction were determined by performing plaque assay on 293 cells. After 48 h of virus infection, the viral yield increased significantly for SG235. In contrast, ZD55 replicated in CAR-positive Mutz-1 cells but not in CAR-negative Kasumi cells (Fig. 1C). These results confirm that SG235 infects and replicates effectively in a variety of leukemic cells through a CAR-independent mechanism.

Figure 1.

Infection and replication of chimeric oncolytic adenoviruses in leukemia cell lines. A, Kasumi cells were infected with SG235-GFP at 5, 25, and 50 MOIs for the indicated times and then visualized under a green filter microscope. B, a panel of hematopoietic malignant cell lines treated with SG235-GFP at MOI of 50 for 48 h was subjected to flow cytometric analysis. C, Kasumi and Mutz-1 cells were infected with ZD55 and SG235 vectors at MOI of 50, respectively. After 48 h, medium and cells were harvested and subjected to three freeze-thaw cycles. The collected supernatants were tested for virus production by a plaque assay on 293 cells. Mean of three independent experiments.

Figure 1.

Infection and replication of chimeric oncolytic adenoviruses in leukemia cell lines. A, Kasumi cells were infected with SG235-GFP at 5, 25, and 50 MOIs for the indicated times and then visualized under a green filter microscope. B, a panel of hematopoietic malignant cell lines treated with SG235-GFP at MOI of 50 for 48 h was subjected to flow cytometric analysis. C, Kasumi and Mutz-1 cells were infected with ZD55 and SG235 vectors at MOI of 50, respectively. After 48 h, medium and cells were harvested and subjected to three freeze-thaw cycles. The collected supernatants were tested for virus production by a plaque assay on 293 cells. Mean of three independent experiments.

Close modal

Chimeric Oncolytic Adenovirus SG235 Induces Apoptosis of Leukemia Cell Lines

Next, a panel of leukemic cell lines was examined for apoptosis by Annexin V staining and fluorescence-activated cell sorting analysis. Infection with SG235 at a MOI 50 resulted in apoptosis of all tested leukemic cells (data not shown). The apoptosis induced by SG235 was verified by TUNEL assay in Kasumi cells. The number of cells with TUNEL positivity increased in a dose-dependent manner after treatment with SG235 (Fig. 2A). To determine the underlying mechanism by which SG235 induces apoptosis of leukemia cell, the activation of caspases was examined by Western blot analysis. After infection with SG235 at the time interval indicated, pro-caspase-9 levels decreased and the cleavage of caspase-9, caspase-3, and poly(ADP-ribose) polymerase was observed (Fig. 2B). These data showed that infection with SG235 triggers activation of caspase cascade. Similar results were obtained from human HeLa cell line (Supplementary Fig. S1),5

5Supplementary material for this article is available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

suggesting the broad activity of SG235 against tumor cells.

Figure 2.

Oncolytic adenovirus SG235 triggers apoptosis of leukemia cells. A, Kasumi cells were treated with or without SG235 at MOIs of 5, 10, and 20 for 96 h and then detected using TUNEL assays. B, Kasumi cells were infected with SG235. Cells were harvested at various time points and analyzed for the activation of caspases by Western blotting. GAPDH was used as a loading control.

Figure 2.

Oncolytic adenovirus SG235 triggers apoptosis of leukemia cells. A, Kasumi cells were treated with or without SG235 at MOIs of 5, 10, and 20 for 96 h and then detected using TUNEL assays. B, Kasumi cells were infected with SG235. Cells were harvested at various time points and analyzed for the activation of caspases by Western blotting. GAPDH was used as a loading control.

Close modal

SG235-TRAIL Exhibits Enhanced Cytotoxicity and Induction of Apoptosis via Activation of Caspase

To determine whether TRAIL expression could improve the antitumor activity of fiber chimeric CRAd, cell proliferation was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Both SG235 and SG235-TRAIL significantly inhibited growth of leukemic cells in a dose-dependent manner compared with no viral treatment (P < 0.001). Moreover, SG235-TRAIL achieved stronger cytotoxicity than did SG235 (Fig. 3A). Similarly, when cells were exposed to SG235-TRAIL, a significant increase in apoptosis was observed in Kasumi cells (Fig. 3B) and Mutz-1 cells (data not shown). The activation of caspase-8 and the caspase-8 substrate Bid was observed in the cells treated with SG235-TRAIL for 48 h but not in the control cells. Dysfunction of mitochondria, up-regulation of Bax, and activation of caspase-9 and caspase-3 were detected as well. In contrast, SG235 only showed a slight activation of caspase-9 and caspase-3 but not caspase-8 (Fig. 3C and D). Specific inhibitors of caspases were used to further confirm involvement of the extrinsic apoptotic pathway. Either z-DEVD-FMK (caspase-3 inhibitor) or z-IETD-FMK (caspase-8 inhibitor) provided a partial protection against cytotoxicity of SG235-TRAIL. Moreover, the inhibitory effect of caspase-3 inhibitor appeared to be stronger than that of caspase-8 inhibitor (Fig. 3E). We further analyzed activation of caspase-9 and caspase-3 in a panel of cell lines including HL-60 (AML), K562 (chronic myelogenous leukemia), RPMI 8226 (myeloma), and Jurkat (T-cell leukemia). The activation of caspases was detected in all tested cell lines (Supplementary Fig. S1).5 Together, these data are consistent with the previous observation that both membrane and mitochondrial pathways were activated on TRAIL exposure in K562 cells (25), indicating that increased cell killing by SG235-TRAIL is mainly dependent on TRAIL-mediated apoptosis.

Figure 3.

SG235-TRAIL induces apoptosis via activation of both membrane and mitochondrial pathways and results in enhanced cytotoxicity. A, leukemic cells were treated with SG235 and SG235-TRAIL at the indicated viral particles for 48 h. Ad-TRAIL was used as the vector control. Cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Mean ± SD of three independent assays. B, Kasumi cells were infected with the indicated vectors at MOI of 50. On day 2 after the treatment, apoptosis was assessed by flow cytometric analysis. C, mitochondrial membrane potential was evaluated by rhodamine 123 staining in Kasumi cells treated with SG235-TRAIL at 50 MOI for 48 h. D, cell extracts prepared from Kasumi cells untreated or exposed to the viruses for 48 h were examined with Western blotting using antibodies that recognize the indicated polypeptide. E, Kasumi cells were treated with SG235-TRAIL in the presence or absence of z-DEVD-FMK (50 μmol/L) or z-IETD-FMK (50 μmol/L) for 3 d and then examined for cell apoptosis by flow cytometric analysis. *, P < 0.001 versus SG235-TRAIL alone; #, P < 0.01 versus SG235-TRAIL alone.

Figure 3.

SG235-TRAIL induces apoptosis via activation of both membrane and mitochondrial pathways and results in enhanced cytotoxicity. A, leukemic cells were treated with SG235 and SG235-TRAIL at the indicated viral particles for 48 h. Ad-TRAIL was used as the vector control. Cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Mean ± SD of three independent assays. B, Kasumi cells were infected with the indicated vectors at MOI of 50. On day 2 after the treatment, apoptosis was assessed by flow cytometric analysis. C, mitochondrial membrane potential was evaluated by rhodamine 123 staining in Kasumi cells treated with SG235-TRAIL at 50 MOI for 48 h. D, cell extracts prepared from Kasumi cells untreated or exposed to the viruses for 48 h were examined with Western blotting using antibodies that recognize the indicated polypeptide. E, Kasumi cells were treated with SG235-TRAIL in the presence or absence of z-DEVD-FMK (50 μmol/L) or z-IETD-FMK (50 μmol/L) for 3 d and then examined for cell apoptosis by flow cytometric analysis. *, P < 0.001 versus SG235-TRAIL alone; #, P < 0.01 versus SG235-TRAIL alone.

Close modal

Effects of SG235-TRAIL on the Expression of TRAIL Receptors and Bcl-2 Family Proteins

TRAIL induces apoptosis in a wide variety of tumor cells by interacting with death signaling receptors (DR4 and DR5). The decoy receptors (DcR1 and DcR2) compete with death receptors for TRAIL engagement (26). We thus examined the expression of TRAIL receptors on leukemic cell lines. The expression of DR4 and DR5 was undetectable on Kasumi, Mutz-1, and K562 cells, which is consistent with a previous report showing similar phenomena in leukemic cell lines and primary blasts (27). A weak expression of DcR1 and DcR2 at the protein level was observed (Fig. 4A). DR4 and DR5 expression was not up-regulated by SG235-TRAIL (data not shown). We further determined whether SG235-TRAIL virus secreted TRAIL into the medium of infected cells using an ELISA. Figure 4B showed that TRAIL could be detected in the culture supernatants of Kasumi and K562 cells treated with SG235-TRAIL. In contrast, the medium of cells infected with Ad-TRAIL or SG235 showed that TRAIL levels were below the detectable range of the assay (<31.2 pg/mL).

Figure 4.

TRAIL receptor expressions in leukemic cell lines and effect of the CRAds on Bcl-2 family protein expression. A, cell surface expression levels of TRAIL death and decoy receptors in K562, Kasumi, and Mutz-1 were assessed by flow cytometric analysis. Shadowed histograms, cells stained with anti-TRAIL receptor antibodies; unshadowed histograms, control cells stained with control goat antibodies. Representative of three separate experiments. B, TRAIL levels in the medium of cells treated with the viruses at a MOI of 50 for the indicated time were measured by ELISA. Mean ± SD from three independent experiments. #, P > 0.001 versus PBS control; *, P < 0.001 versus Ad-TRAIL. C, Kasumi cells were infected with Ad-TRAIL, SG235, or SG235-TRAIL at MOI of 50 for 48 h. Cell lysates were probed with antibodies to the indicated polypeptides. Actin served as a loading control.

Figure 4.

TRAIL receptor expressions in leukemic cell lines and effect of the CRAds on Bcl-2 family protein expression. A, cell surface expression levels of TRAIL death and decoy receptors in K562, Kasumi, and Mutz-1 were assessed by flow cytometric analysis. Shadowed histograms, cells stained with anti-TRAIL receptor antibodies; unshadowed histograms, control cells stained with control goat antibodies. Representative of three separate experiments. B, TRAIL levels in the medium of cells treated with the viruses at a MOI of 50 for the indicated time were measured by ELISA. Mean ± SD from three independent experiments. #, P > 0.001 versus PBS control; *, P < 0.001 versus Ad-TRAIL. C, Kasumi cells were infected with Ad-TRAIL, SG235, or SG235-TRAIL at MOI of 50 for 48 h. Cell lysates were probed with antibodies to the indicated polypeptides. Actin served as a loading control.

Close modal

Because proapoptotic Bcl-2 family members are involved in tumor cell resistance to TRAIL (28, 29), we investigated the behavior of Bcl-2 family member proteins in leukemic cells infected with the viruses. Cells treated with SG235 and SG235-TRAIL showed a decreased expression of Bcl-2 and Mcl-1, suggesting an inhibitory effect of the CRAds on Bcl-2 and Mcl-1 proteins. The expression levels of Bim, Bak, and Bcl-xL were not affected by SG235 or SG235-TRAIL infection (Fig. 4C). These observations were further confirmed by quantitative real-time PCR analysis (data not shown).

Expression of CD46 and Selective Cytotoxic Effect of Chimeric CRAds in Primary Leukemic Cells

Because Ad35 uses CD46 as a high-affinity primary attachment receptor (15, 30), the expression level of CD46 in primary leukemic cells from 122 patients was determined by flow cytometry. Most of the tested primary blasts expressed CD46 (data not shown). As determined, 60 of 70 cases with AML were found to be CD46+ (≥30% of reactive cells), and 7 of 8 acute promyelocytic leukemia patient samples were CD46+. In B-cell acute lymphocytic leukemia, 24 of 37 patient samples were CD46+, and in T-cell acute lymphocytic leukemia, 5 of 7 patients were CD46+.

To examine infectivity and cytotoxicity effects of SG235 on primary leukemic cells, the blasts from 6 AML patients were infected with SG235-GFP at MOI 50 for 48 h, and GFP-positive cells were checked by a fluorescence microscope or counted by fluorescence-activated cell sorting. Results showed that SG235 effectively infected primary AML cells and achieved 25.68% of GFP-positive cells on average (Fig. 5A). Furthermore, leukemic cells treated with Ad-TRAIL, SG235, and SG235-TRAIL vector showed average of 9.2%, 14.9%, and 30.5% Annexin V-positive/propidium iodide-negative cells, respectively (Fig. 5B). The increased apoptosis induced by SG235-TRAIL was also confirmed through analyzing the cleaved forms of caspase-3 and poly(ADP-ribose) polymerase (Fig. 5C). Next, we examined the ability of infectious and cytopathic effect of the CRAds on primary leukemic cells by performing the colony formation assay. AML cells from 2 patients and incubated for 5 days in methylcellulose culture with SG235-GFP showed bright green fluorescence dots (Fig. 6A). As expected, SG235 significantly inhibited leukemic colony formation in all tested 6 AML specimens (SG235 versus untreated control; P < 0.01). On infection by SG235-TRAIL, almost no colonies were recovered in any cases, suggesting enhanced cytopathic effects induced by SG235-TRAIL. In contrast, Ad-TRAIL slightly inhibited clonogenic survival of primary AML samples.

Figure 5.

Infection and induction of apoptosis of CRAds in primary leukemia cells. A, leukemic blasts from six patients with AML [a, AML with maturation (M2 subtype); b, acute erythroleukemia (M6); c, M2; d, acute monoblastic leukemia (M5); e, M5; f, AML without maturation (M1)] were cultured with SG235-GFP at 50 MOI for 48 h, visualized under a green filter microscope, and analyzed by flow cytometry. g, average of six patients. B, leukemic cells were treated with the indicated viruses at MOI of 50 for 48 h, stained with FITC-conjugated Annexin V and propidium iodide, and subjected to flow cytometric analysis. Representative of results from six patients. C, after treatment with the viruses at 50 MOI for 48 h, primary blasts were collected. Whole-cell extracts were prepared and immunoblotted for the detection of activation of caspases.

Figure 5.

Infection and induction of apoptosis of CRAds in primary leukemia cells. A, leukemic blasts from six patients with AML [a, AML with maturation (M2 subtype); b, acute erythroleukemia (M6); c, M2; d, acute monoblastic leukemia (M5); e, M5; f, AML without maturation (M1)] were cultured with SG235-GFP at 50 MOI for 48 h, visualized under a green filter microscope, and analyzed by flow cytometry. g, average of six patients. B, leukemic cells were treated with the indicated viruses at MOI of 50 for 48 h, stained with FITC-conjugated Annexin V and propidium iodide, and subjected to flow cytometric analysis. Representative of results from six patients. C, after treatment with the viruses at 50 MOI for 48 h, primary blasts were collected. Whole-cell extracts were prepared and immunoblotted for the detection of activation of caspases.

Close modal
Figure 6.

Effects of chimeric oncolytic adenoviruses on formation of blast cell colony and normal hematopoietic progenitor. A, blast cells from two patients with AML were infected with SG235-GFP and then cultured in methylcellulose medium supplemented with phytohemagglutinin-leukocyte conditioned medium. After 6 d, colonies were visualized by regular light (top) or fluorescence (bottom). B, leukemic cells of patients (n = 6) were treated with Ad-TRAIL, SG235, or SG235-TRAIL at MOI of 50, respectively, and their long-term survival was evaluated by colony formation assays. Mean ± SE. *, P < 0.05; **, P < 0.01; #, P < 0.01 from control. C, mononuclear cells obtained from bone marrow of three healthy volunteers were cultured in methylcellulose medium supplemented with recombinant human granulocyte-macrophage colony-stimulating factor for 6 d, at which time colonies were visualized and scored with an inverted microscope. #, P < 0.01 from control; *, P < 0.05 from Ad-TRAIL.

Figure 6.

Effects of chimeric oncolytic adenoviruses on formation of blast cell colony and normal hematopoietic progenitor. A, blast cells from two patients with AML were infected with SG235-GFP and then cultured in methylcellulose medium supplemented with phytohemagglutinin-leukocyte conditioned medium. After 6 d, colonies were visualized by regular light (top) or fluorescence (bottom). B, leukemic cells of patients (n = 6) were treated with Ad-TRAIL, SG235, or SG235-TRAIL at MOI of 50, respectively, and their long-term survival was evaluated by colony formation assays. Mean ± SE. *, P < 0.05; **, P < 0.01; #, P < 0.01 from control. C, mononuclear cells obtained from bone marrow of three healthy volunteers were cultured in methylcellulose medium supplemented with recombinant human granulocyte-macrophage colony-stimulating factor for 6 d, at which time colonies were visualized and scored with an inverted microscope. #, P < 0.01 from control; *, P < 0.05 from Ad-TRAIL.

Close modal

To assess the cytotoxicity of CRAds to normal hematopoietic progenitor cells, CFU-GM assays were done by plating appropriate numbers of mononuclear cells obtained from normal bone marrow samples in methylcellulose medium supplemented with recombinant human granulocyte-macrophage colony-stimulating factor. The data presented in Fig. 6C revealed that the number of normal myeloid progenitor cells (CFU-GM) was not significantly affected by Ad-TRAIL and SG235. Treatment with SG235-TRAIL resulted in 17% reduction of the colony number. These results indicate that the cytopathic effect of SG235 and SG235-TRAIL on normal hematopoietic progenitors is minimal.

In vivo Antitumor Activity of the Chimeric Oncolytic Adenoviruses

We subsequently investigated the relative antileukemia efficacy of SG235 in vivo. Treatment of CAR-negative Kasumi xenograft-bearing severe combined immunodeficient mice with direct intratumor injection of 5 × 108 plaque-forming units SG235 achieved significant tumor growth inhibition compared with the PBS control (P < 0.001). In contrast, treatment with ZD55 almost had no effect on the tumor growth (Fig. 7A). We next assessed the therapeutic efficacy of SG235 versus SG235-TRAIL against leukemic cells in vivo (Fig. 7B). SG235-TRAIL was more effective than SG235 in tumor growth inhibition (SG235-TRAIL versus SG235; P < 0.001 at the conclusion of the treatment). The antitumor therapeutic effect of nonreplicative Ad-TRAIL was not observed. Furthermore, one animal from each treatment group was euthanized 5 days after treatment, and tumors were harvested for histochemical examination. Results of in situ TUNEL assay showed a marked increase of apoptotic cells within the tumor treated with SG235-TRAIL compared with the treatment of SG235 (Fig. 7C).

Figure 7.

Antitumor activity of chimeric oncolytic adenoviruses in s.c. Kasumi xenografts in animal model. A, tumor-bearing mice (n = 7) were treated with either PBS or 5 × 108 plaque-forming units of the indicated viruses by intratumor injection. *, P > 0.05 from PBS control; #, P < 0.001 from control. B, animals with Kasumi tumors were intratumorally injected with 5 × 108 plaque-forming units of different vectors. The size of tumor was monitored and tumor volume was calculated. Points, mean of tumor volume (n = 7); bars, SD. *, P < 0.001 versus SG235-treated group. C, apoptotic cell death is induced by treatment with SG235 and SG235-TRAIL in vivo; apoptotic nuclei are stained in blue. TUNEL pictures were taken at ×50 original magnification.

Figure 7.

Antitumor activity of chimeric oncolytic adenoviruses in s.c. Kasumi xenografts in animal model. A, tumor-bearing mice (n = 7) were treated with either PBS or 5 × 108 plaque-forming units of the indicated viruses by intratumor injection. *, P > 0.05 from PBS control; #, P < 0.001 from control. B, animals with Kasumi tumors were intratumorally injected with 5 × 108 plaque-forming units of different vectors. The size of tumor was monitored and tumor volume was calculated. Points, mean of tumor volume (n = 7); bars, SD. *, P < 0.001 versus SG235-treated group. C, apoptotic cell death is induced by treatment with SG235 and SG235-TRAIL in vivo; apoptotic nuclei are stained in blue. TUNEL pictures were taken at ×50 original magnification.

Close modal

This study found that SG235, a new fiber chimeric CRAd, could effectively infect a variety of malignant hematopoietic cell lines regardless of their CAR expression status. Leukemia cells treated with SG235 showed a significant growth inhibition and apoptosis evidenced by Annexin V staining and TUNEL assay. Importantly, in addition to an effect of SG235 on commercially available permanent tumor cell lines, a strong cytotoxicity effect on primary leukemic cells was also observed, whereas almost no effect on normal hematopoietic progenitors was detected. Because chimeric Ad5 vectors possessing Ad35 fibers use CD46, often abundant on tumor cells compared with normal cells (31), as a primary attachment receptor, we analyzed CD46 expression on bone marrow samples from the patients with AML and acute lymphocytic leukemia by flow cytometry. CD46 was expressed in a majority of tested primary blasts. These findings, along with our data that leukemic cells are sensitive to SG235, suggest that this new fiber chimeric CRAd is an attractive vector for treating hematopoietic tumors including leukemia.

TRAIL, a member of the tumor necrosis factor superfamily, has garnered considerable attention as a novel anticancer agent, as it appears to selectively induce apoptosis of cancer cells but not of normal cells in various tissue types (32). Human recombinant TRAIL is reported to induce apoptosis of multiple myeloma (33, 34) and myeloid leukemia cells (35, 36) and elicit potent antitumor activity and synergistic effect with chemotherapy (37, 38). However, some leukemic cell lines and fresh blasts are unresponsive to TRAIL treatment (39, 40). In the present study, we have shown for the first time that SG235-TRAIL, a fiber chimeric CRAd expressing TRAIL, exhibits enhanced oncolytic potency in a variety of leukemic cells compared with its parent vector SG235. Moreover, SG235-TRAIL is much stronger than SG235 in killing established leukemia xenografts, which is related to apoptosis induction. The data reported herein have confirmed the results of others and our previous observation in different cancer cell models (21, 41) and suggested that integrating TRAIL gene therapy into an oncolytic adenovirus overcomes the weaknesses of the TRAIL gene therapy and virotherapy used individually.

TRAIL activates the extrinsic apoptotic pathway in type I cells. In type II cells, the intrinsic mitochondrial pathway is recruited to amplify the apoptotic signal through cleavage of Bid by caspase-8 (39). Our study revealed that the intrinsic mitochondrial pathway is also involved in SG235-TRAIL-induced apoptosis of leukemic cells evidenced by the activation of caspase-8 and caspase-9, decreased Bid, and dysfunction of mitochondria. Moreover, Bcl-2 family proteins are critical for the determination of sensitivity to TRAIL (28, 29). Recent studies have shown that degradation of Mcl-1, a Bcl-2 family member, initiates mitochondrial apoptotic cascade (42). In this study, we observed down-regulated expression of Bcl-2 and Mcl-1 in SG235-treated or SG235-TRAIL-treated cells. This loss of Mcl-1 might contribute to the activated intrinsic pathway.

Several intracellular events blocking TRAIL-mediated apoptosis have been identified by researchers, such as lack of expression of death receptor on the cell surface and competitive binding of TRAIL by the decoy receptors (39). In this regard, we found that the DR4/DR5 expression was absent in three leukemic lines, whereas dimly positive DcR1/DcR2 expression was detected, which is in line with the recent data that DR4 expressed in ∼50% of AML cases and DR5 expressed only in a minority (∼10%) of the cases analyzed (40). Moreover, there was no any modification of DR4/DR5 and DcR1/DcR2 expression in SG235-TRAIL-treated cells. Thus, the reason why SG235-TRAIL induced a massive apoptosis in leukemic cells with defect of the DR4/DR5 expressions is still unclear; however, it was reported that the status of signaling or decoy TRAIL receptors, assessed through either immunoblotting analysis or flow cytometry, cannot serve to reliably predict the TRAIL sensitivity (or resistance) of multiple myeloma cells (33). Recently, Wu et al. reported that a nonreplicative Ad5/35-TRAIL is more efficient than exogenous TRAIL in triggering apoptosis of HL-60 leukemia cells (43). The exact reason for this discrepancy is unclear. It is important to note that the authors reported TRAIL-induced apoptosis of HL-60 cells only at very high doses of TRAIL (10-100 ng/mL) and did not exam secreted TRAIL in the medium of Ad5/35-TRAIL-infected cells. Data from other studies support the observation that high doses of recombinant TRAIL are needed for apoptosis of leukemic cells (35, 40). Our results showed that low concentrations of TRAIL were detected in the medium of SG235-TRAIL-treated cells, indicating that the mechanisms by which SG235-TRAIL induces apoptosis may differ from that of exogenous TRAIL. Because constitutive endocytosis and predominant intracellular location of some of the TRAIL receptors were reported previously (44, 45), further studies are needed to determine whether TRAIL produced by SG235-TRAIL ligates the death receptors within intracellular organelles and thus triggers apoptosis. Additionally, adenovirus E1A gene expression in tumor cells was reported to enhance killing by TRAIL (46), which may contribute to enhanced apoptosis induced by SG235-TRAIL.

In conclusion, we showed that the fiber-modified oncolytic adenovirus SG235 permits CAR-independent cell entry and induces selective cytopathic effects in human leukemic cells in vitro and in vivo. Furthermore, SG235-TRAIL treatment could elicit much stronger cytotoxic effects through induction of apoptosis than did SG235. These findings suggest that the current gene-virotherapeutic strategy could be a promising approach for treating leukemia.

No potential conflicts of interest were disclosed.

1
Kirn
D
,
Martuza
RL
,
Zwiebel
J
. 
Replication-selective virotherapy for cancer: biological principles, risk management and future directions
.
Nat Med
2001
;
7
:
781
7
.
2
Hawkins
LK
,
Lemoine
NR
,
Kirn
D
. 
Oncolytic biotherapy: a novel therapeutic platform
.
Lancet Oncol
2002
;
3
:
17
26
.
3
Qian
W
,
Liu
J
,
Tong
Y
, et al
. 
Enhanced antitumor activity by a selective conditionally replicating adenovirus combining with MDA-7/interleukin-24 for B-lymphoblastic leukemia via induction of apoptosis
.
Leukemia
2008
;
22
:
361
9
.
4
Fisher
PB
. 
Is mda/IL24 a “magic bullet” for cancer?
Cancer Res
2005
;
65
:
10128
38
.
5
Martuza
RL
,
Malick
A
,
Markert
JM
,
Ruffner
KL
,
Coen
DM
. 
Experimental therapy of human glioma by means of a genetically engineered virus mutant
.
Science
1991
;
252
:
854
6
.
6
Everts
B
,
van der Poel
HG
. 
Replication-selective oncolytic viruses in the treatment of cancer
.
Cancer Gene Ther
2005
;
12
:
141
61
.
7
Peng
KW
,
Ahmann
GJ
,
Pham
L
,
Greipp
PR
,
Cattaneo
R
,
Russell
SJ
. 
Systemic therapy of myeloma xenografts by an attenuated measles virus
.
Blood
2001
;
98
:
2002
7
.
8
Ong
HT
,
Timm
MM
,
Greipp
PR
, et al
. 
Oncolytic measles virus targets high CD46 expression on multiple myeloma cells
.
Exp Hematol
2006
;
46
:
713
20
.
9
Peng
KW
,
Donovan
KA
,
Schneider
U
,
Cattaneo
R
,
Lust
JA
,
Russell
SJ
. 
Oncolytic measles viruses displaying a single-chain antibody against CD38, a myeloma cell marker
.
Blood
2003
;
101
:
2557
62
.
10
Grote
D
,
Russell
SJ
,
Cornu
TI
,
Cattaneo
R
,
Poland
GA
,
Fielding
AK
. 
Live attenuated measles virus induces regression of human lymphoma xenografts in immunodeficient mice
.
Blood
2001
;
97
:
3746
54
.
11
Kunzi
V
,
Oberholzer
PA
,
Heinzerling
L
,
Dummer
R
,
Nai
HY
. 
Recombinant measles virus induces cytolysis of cutaneous T-cell lymphoma in vitro and in vivo
.
J Invest Dermatol
2006
;
126
:
2525
32
.
12
Heinzerling
L
,
Kunzi
V
,
Oberholzer
PA
,
Kundig
T
,
Naim
H
,
Dummer
R
. 
Oncolytic measles virus in cutaneous T-cell lymphomas mounts antitumor immune responses in vivo and targets interferon-resistant tumor cells
.
Blood
2005
;
106
:
2287
94
.
13
Rivera
AA
,
Davydova
J
,
Schierer
S
, et al
. 
Combining high selectivity of replication with fiber chimerism for effective adenoviral oncolysis of CAR-negative melanoma cells
.
Gene Ther
2004
;
11
:
1694
702
.
14
Mentel
R
,
Dopping
G
,
Wegner
U
,
Seidel
W
,
Liebermann
H
,
Dohner
L
. 
Adenovirus-receptor interaction with human lymphocytes
.
J Med Virol
1997
;
51
:
252
7
.
15
Rebel
VI
,
Hartnett
S
,
Denham
J
,
Chan
M
,
Finberg
R
,
Sieff
CA
. 
Maturation and lineage-specific expression of the coxsackie and adenovirus receptor in hematopoietic cells
.
Stem Cells
2000
;
18
:
176
82
.
16
Wang
Y
,
Xue
S
,
Hallden
G
, et al
. 
Virus-associated RNA I-deleted adenovirus, a potential oncolytic agent targeting EBV-associated tumors
.
Cancer Res
2005
;
65
:
1523
31
.
17
Medina
D
,
Sheay
W
,
Goodell
L
, et al
. 
Adenovirus mediated cytotoxicity of chronic lymphocytic leukemia cells
.
Blood
1999
;
94
:
3499
508
.
18
Yotnda
P
,
Onishi
H
,
Heslop
HE
, et al
. 
Efficient infection of primitive hematopoietic stem cells by modified adenovirus
.
Gene Ther
2001
;
8
:
930
7
.
19
Yotnda
P
,
Zompeta
C
,
Heslop
HE
,
Andreeff
M
,
Brenner
MK
,
Marini
F
. 
Comparison of the efficiency of transduction of leukemic cells by fiber-modified adenoviruses
.
Hum Gene Ther
2004
;
15
:
1229
42
.
20
Sauter
C
,
Baumberger
U
,
Ekenbark
S
,
Lindenmann
J
. 
Replication of an avian myxovirus in primary cultures of human leukemic cells
.
Cancer Res
1973
;
33
:
3002
7
.
21
Pei
Z
,
Chu
L
,
Zou
W
, et al
. 
An oncolytic adenoviral vector of Smac increases antitumor activity of TRAIL against HCC in human cells and in mice
.
Hepatology
2004
;
39
:
1371
81
.
22
Zhang
Z
,
Zou
W
,
Luo
C
, et al
. 
An armed oncolytic adenovirus system, ZD55-gene, demonstrating potent antitumoral efficacy
.
Cell Res
2003
;
13
:
481
9
.
23
Qian
W
,
Liu
J
,
Jin
J
,
Ni
W
,
Xu
W
. 
Arsenic trioxide induces not only apoptosis but also autophagic cell death in leukemia cell lines via up-regulation of Beclin-1
.
Leukemia Res
2007
;
31
:
329
39
.
24
Minden
MD
,
Till
JE
,
McCulloch
EA
. 
Proliferative state of blast cell progenitors in acute myeloblastic leukemia (AML)
.
Blood
1978
;
52
:
592
600
.
25
Rosato
RR
,
Almenara
JA
,
Dai
Y
,
Grant
S
. 
Simultaneous activation of the intrinsic and extrinsic pathways by histone deacetylase (HDAC) inhibitors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) synergistically induces mitochondrial damage and apoptosis in human leukemia cells
.
Mol Cancer Ther
2003
;
2
:
1273
84
.
26
Srivastava
RK
. 
TRAIL/Apo-2L: mechanisms and clinical applications in cancer
.
Neoplasia
2001
;
3
:
535
46
.
27
Clodi
K
,
Wimmer
D
,
Li
Y
, et al
. 
Expression of tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors and sensitivity to TRAIL-induced apoptosis in primary B-cell acute lymphoblastic leukaemia cells
.
Br J Haematol
2000
;
111
:
580
6
.
28
Guo
BC
,
Xu
YH
. 
Bcl-2 over-expression and activation of protein kinase C suppress the TRAIL-induced apoptosis in Jurkat T cells
.
Cell Res
2001
;
11
:
101
6
.
29
Lamothe
B
,
Aggarwal
BB
. 
Ectopic expression of Bcl-2 and Bcl-xL inhibits apoptosis induced by TNF-related apoptosis-inducing ligand (TRAIL) through suppression of caspases-8, 7, and 3 and BID cleavage in human acute myelogenous leukemia cell line HL-60
.
J Interferon Cytokine Res
2002
;
22
:
269
79
.
30
Shayakhmetov
DM
,
Eberly
AM
,
Li
ZY
,
Lieber
A
. 
Deletion of penton RGD motifs affects the efficiency of both the internalization and the endosome escape of viral particles containing adenovirus serotype 5 or 35 fiber knobs
.
J Virol
2005
;
79
:
1053
61
.
31
Hara
T
,
Kojima
A
,
Fukuda
H
, et al
. 
Levels of complement regulatory proteins, CD35 (CR1), CD46 (MCP) and CD55 (DAF) in human haematological malignancies
.
Br J Haematol
1992
;
82
:
368
73
.
32
LeBlanc
HN
,
Ashkenazi
A
. 
Apo2L/TRAIL and its death and decoy receptors
.
Cell Death Differ
2003
;
10
:
66
75
.
33
Mitsiades
CS
,
Treon
SP
,
Mitsiades
N
, et al
. 
TRAIL/Apo2L ligand selectively induces apoptosis and overcomes drug resistance in multiple myeloma: therapeutic applications
.
Blood
2001
;
98
:
795
804
.
34
Chen
Q
,
Gong
B
,
Mahmoud-Ahmed
AS
, et al
. 
Apo2L/TRAIL and Bcl-2-related proteins regulate type I interferon-induced apoptosis in multiple myeloma
.
Blood
2001
;
98
:
2183
92
.
35
Plasilova
M
,
Zivny
J
,
Jelinek
J
, et al
. 
TRAIL (Apo2L) suppresses growth of primary human leukemia and myelodysplasia progenitors
.
Leukemia
2002
;
16
:
67
73
.
36
Uno
K
,
Inukai
T
,
Kayagaki
N
, et al
. 
TNF-related apoptosis-inducing ligand (TRAIL) frequently induces apoptosis in Philadelphia chromosome-positive leukemia cells
.
Blood
2003
;
101
:
3658
67
.
37
Ashkenazi
A
,
Pai
RC
,
Fong
S
, et al
. 
Safety and antitumor activity of recombinant soluble Apo2 ligand
.
J Clin Invest
1999
;
104
:
155
62
.
38
Jazirehi
AR
,
Ng
CP
,
Gan
XH
,
Schiller
G
,
Bonavida
B
. 
Adriamycin sensitizes the Adriamycin-resistant 8226/Dox40 human multiple myeloma cells to Apo2L/tumor necrosis factor-related apoptosis-inducing ligand-mediated (TRAIL) apoptosis
.
Clin Cancer Res
2001
;
7
:
3874
83
.
39
Cheng
J
,
Hylander
BL
,
Baer
MR
,
Chen
X
,
Repasky
EA
. 
Multiple mechanisms underlie resistance of leukemia cells to Apo2 ligand/TRAIL
.
Mol Cancer Ther
2006
;
5
:
1844
53
.
40
Riccioni
R
,
Pasquini
L
,
Mariani
G
, et al
. 
TRAIL decoy receptors mediate resistance of acute myeloid leukemia cells to TRAIL
.
Haematologica
2005
;
90
:
612
24
.
41
Dong
F
,
Wang
L
,
Davis
JJ
, et al
. 
Eliminating established tumor in nu/nu nude mice by a tumor necrosis factor-α-related apoptosis-inducing ligand-armed oncolytic adenovirus
.
Clin Cancer Res
2006
;
12
:
5224
30
.
42
Han
J
,
Goldstein
LA
,
Gastman
BR
,
Rabinowich
H
. 
Interrelated roles for Mcl-1 and BIM in regulation of TRAIL-mediated mitochondrial apoptosis
.
J Biol Chem
2006
;
281
:
10153
63
.
43
Wu
CH
,
Kao
CH
,
Safa
AR
. 
TRAIL recombinant adenovirus triggers robust apoptosis in multidrug-resistant HL-60/Vinc cells preferentially through death receptor DR5
.
Hum Gene Ther
2008
;
19
:
731
43
.
44
Zhang
Y
,
Zhang
B
. 
TRAIL resistance of breast cancer cells is associated with constitutive endocytosis of death receptors 4 and 5
.
Mol Cancer Res
2008
;
6
:
1861
71
.
45
Zhang
XD
,
Franco
AV
,
Nguyen
T
,
Gray
CP
,
Hersey
P
. 
Differential localization and regulation of death and decoy receptors for TNF-related apoptosis-inducing ligand (TRAIL) in human melanoma cells
.
J Immunol
2000
;
164
:
3961
70
.
46
Routes
JM
,
Ryan
S
,
Clase
A
, et al
. 
Adenovirus E1A oncogene expression in tumor cells enhances killing by TNF-related apoptosis-inducing ligand (TRAIL)
.
J Immunol
2000
;
165
:
4522
7
.

Competing Interests

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.

Supplementary data