Oncolytic virotherapy is a promising alternative to conventional treatment, yet systemic delivery of these viruses to tumors remains a major challenge. In this regard, mesenchymal stem cells (MSC) with well-established tumor-homing property could serve as a promising systemic delivery tool. We showed that MSCs could be effectively infected by hepatocellular carcinoma (HCC)-targeted oncolytic adenovirus (HCC-oAd) through modification of the virus' fiber domain and that the virus replicated efficiently in the cell carrier. HCC-targeting oAd loaded in MSCs (HCC-oAd/MSC) effectively lysed HCC cells in vitro under both normoxic and hypoxic conditions as a result of the hypoxia responsiveness of HCC-oAd. Importantly, systemically administered HCC-oAd/MSC, which were initially infected with a low viral dose, homed to HCC tumors and resulted in a high level of virion accumulation in the tumors, ultimately leading to potent tumor growth inhibition. Furthermore, viral dose reduction and tumor localization of HCC-oAd/MSC prevented the induction of hepatotoxicity by attenuating HCC-oAd hepatic accumulation. Taken together, these results demonstrate that MSC-mediated systemic delivery of oAd is a promising strategy for achieving synergistic antitumor efficacy with improved safety profiles.

Significance:

Mesenchymal stem cells enable delivery of an oncolytic adenovirus specifically to the tumor without posing any risk associated with systemic administration of naked virions to the host.

Liver cancer is the third most common tumor type and the second leading cause of death in Asia. The most common form of liver cancer, hepatocellular carcinoma (HCC), lacks effective treatment options (1, 2). Only a small fraction of patients with HCC can undergo surgical resection and tumor recurrence occurs in more than 50% of these patients (3). Chemotherapeutics, such as sorafenib, elicit limited therapeutic effect against advanced HCC (4, 5). Therefore, there is an urgent need for new HCC therapy.

Mesenchymal stem cells (MSC) have emerged as a promising systemic delivery tool for cancer therapeutics due to their tumor-homing abilities (6, 7). Furthermore, MSCs exhibit relatively low immunogenicity (8) and do not stimulate lymphocyte proliferation, thus avoiding immune rejection (9). Despite these beneficial attributes, MSCs cannot control tumor growth (10). To address this problem, MSCs can be genetically engineered to express therapeutic genes (11, 12), loaded with drugs (13), or be infected with oncolytic viruses (14, 15).

Oncolytic adenovirus (oAd), which exhibits cancer-specific replication, viral production, and cytopathic effects, is a promising therapeutic cargo to enhance the overall therapeutic efficacy of MSC (16, 17). When oAd is utilized as the therapeutic cargo, MSCs are eventually lysed by oAd replication, thus preventing any adverse side effects that are associated with prolonged survival of stem cells in vivo. Importantly, MSC-mediated systemic delivery of oAd can improve intratumoral accumulation of the virus, which is not feasible with naked virion. In specific, the viral capsid, which is easily recognized by immune system as pathogen, can trigger adverse inflammatory responses and swift clearance of oAd in bloodstream, thus resulting in poor intratumoral accumulation (18, 19).

HCC is known for its active Wnt signaling, which contributes to the rapid and uncontrolled proliferation of tumor cells (20). An activated Wnt signaling pathway promotes the epithelial-to-mesenchymal transition (EMT), which contributes to the metastatic and drug-resistant phenotypes of cancer (21). Thus, to enhance the therapeutic efficacy of oAd against HCC, we inserted a sequence encoding a Wnt-inhibiting decoy receptor (WNTi) into the previously reported oAd that targets alpha-fetoprotein (AFP)-positive HCCs (22), generating a WNTi-expressing and HCC-targeting oAd (HCC-oAd-WNTi).

In this report, HCC-oAd-WNTi was loaded into bone marrow–derived human MSCs (HCC-oAd-WNTi/MSC) and evaluated whether this delivery method would reduce the liver toxicity of systemically administered oAd while improving its accumulation in tumor tissues. We illustrate that systemically administered HCC-oAd-WNTi/MSC efficiently targets orthotopic HCC tumors, thus enhancing the tumor accumulation and antitumor efficacy of oAd. Furthermore, HCC-oAd-WNTi/MSC overcomes well-known limitations of systemically administered oAd, such as strong immunogenicity, short blood circulation time, nonspecific liver sequestration, and hepatotoxicity in vivo.

Cells

Cell lines HEK293, A549, Hep3B, and BJ were purchased from the ATCC. Cells were cultured in DMEM (Gibco BRL) supplemented with 10% FBS (Gibco BRL), penicillin (100 IU/mL), and gentamicin (20 μg/mL). All cell lines were maintained at 37°C in a humidified incubator with an atmosphere containing 5% CO2. Cryopreserved human MSCs, which were isolated from bone marrow samples after aspiration from healthy adult male donors, were provided by Pharmicell Co., Ltd. MSCs were cultivated in low-glucose DMEM.

Construction of Ads

To construct a firefly luciferase–expressing HCC-targeting oAd vector, a firefly luciferase or WNTi expression cassettes were ligated into an Ad E3 shuttle vector (pSP72ΔE3; refs. 23, 24). These E3 shuttle vectors went through homologous recombination with linearized HCC-oAd (Ha2bm-d19-k35; ref. 22) to generate Ha2bm-d19-k35/Luc (HCC-oAd-Luc) or Ha2bm-d19-k35/WNTi (HCC-oAd-WNTi), respectively. Upon generation of viral particles (VP) in HEK293 cells by transfection, HCC-oAd-Luc and HCC-oAd-WNTi were propagated in A549 cells and purified by CsCl gradient centrifugation. The numbers of VPs were calculated from optical density measurements at 260 nm (OD260), where an absorbance of 1 (OD260 = 1) was equivalent to 1.1 × 1012 VP/mL. While infectious titers were determined by the plaque formation assay, VP: infectious viral particle (PFU) ratio was estimated to be 100:1, as described previously (25), for the experiments in this study. The genomic DNA from newly generated viruses were sequenced using PCR, then analyzed with SnapGene Software (GSL Biotech LLC) to confirm genomic integrity of final construct. The purified viruses were stored at −80°C until use.

Transduction evaluation

To optimize our transduction results, MSCs were transduced with serotype 5 Ad (Ad5) containing 5 different types of fibers: (i) a wild-type fiber (dE1/GFP), (ii) a fiber knob domain of Ad35 (dE1-k35/GFP), (iii) an RGD motif in the HI loop of the fiber (dE1-RGD/GFP), (iv) a full-length fiber shaft of Ad5 and the Ad3 fiber knob (dE1-k3L/GFP), and (v) a truncated fiber shaft of Ad5 and the Ad3 fiber knob (dE1-k3S/GFP). MSCs were then incubated for 48 hours at 37°C. GFP expression was observed with a fluorescence microscope (Olympus IX81; Olympus Optical). To assess luciferase expression by HCC-oAd-Luc, MSCs were infected with HCC-oAd-Luc at multiplicities of infection (MOI) of 50, 100, or 200. At 48 hours postinfection, luciferase signals were analyzed using the IVIS Imaging System (Xenogen Corp). The bioluminescence signal intensity was obtained as photons acquired per second (p/s) from the region of interest (26).

Optimization of oAd loading into MSCs

To assess the optimal dose of HCC-targeting oAd (HCC-oAd-Luc) for loading into MSCs, the viability of MSCs (5 × 104 cells/well in 24-well plates) was evaluated after infection with HCC-oAd-Luc at MOIs ranging from 0.5 to 50. At 2 or 5 days postinfection, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT; Sigma-Aldrich) was added to each well, after which, the plates were incubated at 37°C for 4 hours. The supernatant was then removed, and the precipitate was dissolved in DMSO. The absorbance at 540 nm was measured on a microplate reader. The viability of untreated MSCs was considered to be 100%.

Viral production of HCC-targeting oAd in MSCs

To assess the production of HCC-targeting oAd in MSCs, MSCs were seeded in 24-well plates and infected with HCC-oAd-Luc at MOIs ranging from 0.5 to 50. After 2 or 5 days of incubation, the cell pellets and supernatants were collected and freeze-thawed 3 times to harvest the virions. qRT-PCR (Q-PCR; TaqMan PCR detection; Applied Biosystems) was used to assess the number of viral genomes in each sample as described previously (27). Data were processed using the SDS 19.1 Software Package (Applied Biosystems). The results are representative of three independent experiments.

To quantify HCC-oAd-WNTi released from the MSC as a result of virus' lytic property and intracellular replication of the virus, 2 × 104 MSC were infected with 5 MOI of HCC-oAd-WNTi to generate HCC-oAd-WNTi/MSC. The viral genome copy number in culture supernatant and cell lysates were analyzed by qPCR as described above at 6 hours to 6 days postinfection.

Characterization of the HCC-oAd-WNTi

Hep3B cells were seeded on 100-mm plates containing DMEM supplemented with 5% FBS and infected with 1 MOI of H101, HCC-oAd, or HCC-oAd-WNTi. Untreated group served as a negative control. At 48 hours postinfection, cell lysates were obtained. Western blotting was performed with cell lysates in similar manner as described previously (28), and following primary antibodies were used: Wnt3a (Abcam), β-catenin (Cell Signaling Technology), epithelial cadherin (E-cadherin; Cell Signaling Technology), or phospho-MAPK/ERK (p-MEK; Cell Signaling Technology).

HCC and MSC killing effect of HCC-oAd-WNTi

To evaluate the effectiveness of HCC-oAd-WNTi for HCC killing, the viability of Hep3B cells was evaluated after infection with H101, HCC-oAd, or HCC-oAd-WNTi at MOIs ranging from 1 to 50. To assess the responsiveness of HCC-oAd-WNTi to hypoxia, the viability of Hep3B cells was evaluated under normoxic or hypoxic conditions after infection with 2 MOIs of HCC-oAd-WNTi. The cell viability in both set of experiments was determined at 48 hours postinfection using MTT as described above. Untreated cancer cells were considered to be 100% viable. The IC50 was determined by analysis of a dose–response curve in GraphPad Prism.

For the assessment of time-dependent change in MSC viability following infection with HCC-oAd-WNTi, MSCs (seeded on 12-well plate at 2 × 104 cells/well) were infected with HCC-oAd-WNTi at an MOI of 5. At 6 hours to 6 days postinfection, cell viability was assessed by the MTT assay as described previously. The results are representative of three independent experiments performed in triplicates.

Cancer-specific killing effect of HCC-oAd-WNTi/MSC

To evaluate the effectiveness of HCC-oAd-WNTi/MSC for killing cancer cells, Hep3B, A549, or BJ cells (seeded on 24-well plate at 2 × 104 cells/well) were treated with MSC, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC for 1, 3, or 5 days either under normoxic or hypoxic conditions. For the preparation of MSC and HCC-oAd-WNTi/MSC treatments, MSC (seeded on 24-well plate at 2 × 104 cells/well) were either treated with PBS or 5 MOI of HCC-oAd-WNTi for 2 days, respectively. Subsequently, MSC and HCC-oAd-WNTi/MSC went through several washing cycles with ice-cold PBS, then cells were detached from the plate, centrifuged, and treated to Hep3B, A549, or BJ cells. At 1, 3, or 5 days posttreatment, MTT assay was performed as described previously.

Antitumor efficacy of HCC-oAd-WNTi/MSC and bioluminescence imaging

To generate orthotopic HCC tumors, 1 × 106 Hep3B cells, which stably express the firefly luciferase gene, were injected into the left liver lobe of athymic nude mice. On day 7 after the cell injection, bioluminescence imaging was performed to confirm tumor establishment. Mice were anesthetized in a chamber filled with 2% isoflurane in oxygen and received d-luciferin (150 mg/kg; Caliper) by intraperitoneal injection. Orthotopic HCC tumor–harboring mice were randomized into four treatment groups; 200 μL of PBS, MSCs (1 × 106 cells), HCC-oAd-WNTi (5 × 108 VPs), or HCC-oAd-WNTi/MSC (1 × 106 cells infected with 5 × 108 VPs for 18 hours) was intravenously injected twice into each mouse, at 9 and 13 days postimplantation (n = 6/group). HCC-oAd-WNTi/MSC was prepared in similar manner as those described above to remove excess HCC-oAd-WNTi. Optical imaging was conducted every week following the first imaging with an IVIS SPECTRUM Instrument (Xenogen Corp.). Image signals were quantitatively analyzed with IGOR-PRO Living Image Software (Xenogen Corp). The in vivo bioluminescence signal intensity was obtained from a body region of interest (29). The livers from mice in each experimental group were harvested at 35 days posttumor implantation and incubated in PBS supplemented with d-luciferin (Caliper). Both optical and luminescent images were obtained using an IVIS Imaging System (Xenogen Corp). All procedures of animal research were provided in accordance with the Laboratory Animals Welfare Act, the Guide for the Care and Use of Laboratory Animals. This animal care and use protocol was reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) in College of Medicine, Hanyang University of Korea (Seoul, Korea).

Histologic and IHC analysis

Liver tissues were harvested from mice at 3 days after the last intravenous injection of PBS, MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC. The harvested liver tissues were fixed in 10% formalin, processed for paraffin embedding, and then cut into 5-μm sections. The sections were stained with hematoxylin and eosin (H&E), and then examined by light microscopy. The tumor area in the liver sections was also immunostained with proliferating cell nuclear antigen (PCNA)-specific antibody (DAKO) or CD90-specific antibody (Abcam).

Pharmacokinetic profile

To assess the rate of Ad clearance from the blood, qPCR was performed with whole-blood samples from oAd-treated mice as described previously (27). Blood was collected from retro-orbital plexus of mice at 5 minutes, 10 minutes, 30 minutes, 1 hours, 6 hours, 24 hours, and 48 hours postsystemic injection with 5 × 108 VPs of naked HCC-oAd-WNTi or HCC-oAd-WNTi/MSC (n = 3).

Biodistribution profile

Hep3B tumor–bearing mice were systemically treated twice in similar manner as above. The lung, heart, stomach, spleen, liver, tumor tissues, and blood were harvested from mice in each group at 24 hours after the second injection, then DNA extraction and qPCR were performed as described previously (27).

Hepatotoxicity

To assess the in vivo toxicity of each formulation, mice were intravenously injected twice with 200 μL of PBS alone (negative control) or MSCs (1 × 106 cells), HCC-oAd-WNTi (5 × 108 VPs), or HCC-oAd-WNTi/MSC (1 × 106 cells infected with 5 × 108 VPs for 18 hours) on days 9 and 13 (n = 3). The serum levels of alanine transaminase (ALT) and aspartate aminotransferase (AST) were measured at 3 days postinjection (30).

Statistical analysis

Data were expressed as the mean ± SD. Statistical significance was determined by the two-tailed Student t test or one-way ANOVA test (SPSS 13.0 Software; SPSS). P values less than 0.05 were considered as statistically significant.

Efficient transduction of HCC-targeting oAd into MSCs

To optimize the efficiency of Ad transduction into MSCs, we tested the transduction efficacy of GFP-expressing and replication-incompetent human serotype 5 Ads with 5 different types of fibers (dE1/GFP, dE1-k35/GFP, dE1-RGD/GFP, dE1-k3L/GFP, and dE1-k3S/GFP). As shown in Supplementary Fig. S1, the transduction efficacy of dE1/GFP and dE1-RGD/GFP was limited at all MOI in MSCs, whereas dE1-k35/GFP, dE1-k3L/GFP, and dE1-k3S/GFP led to higher GFP expression levels in a dose-dependent manner. Of note, dE1-k35/GFP showed the highest transduction efficacy in MSCs among the tested Ads with different fibers, suggesting that replacement of the fiber knob with that of Ad serotype 35 leads to optimal transduction of Ad into MSCs. On the basis of these results, we generated an oAd with an Ad type 35 knob; the oAd replicates under the control of the AFP-positive HCC-specific Ha2bm promoter and expresses luciferase (HCC-oAd-Luc; Fig. 1A). To evaluate the infection efficacy of HCC-oAd-Luc, MSCs were transduced with HCC-oAd-Luc at various MOIs. At 48 hours after infection, the luciferase signal from MSCs increased in a MOI-dependent manner (Fig. 1B). These results demonstrate that an HCC-targeting oAd can efficiently internalize into MSCs and express its transgene.

Figure 1.

Construction and transduction efficacy of oAd replicating under the control of the HCC-specific Ha2bm promoter (HCC-oAd-Luc). A, Schematic HCC-oAd-Luc structure. This oAd replicates under the control of the hypoxic response element and a modified AFP promoter. The expression cassette for luciferase was inserted into the E3 region of the Ad genome. B, Efficiency of HCC-oAd-Luc transduction into MSCs. MSCs were treated with HCC-oAd-Luc at MOIs of 50 to 200. At 48 hours postinfection, luciferase signals were assessed by an IVIS imaging system (original magnification, ×1). The bioluminescence signal intensity was obtained as photons acquired per second (p/s) from regions of interest. The experiment was assessed at least three times and the data shown are representative of experiments performed in triplicate.

Figure 1.

Construction and transduction efficacy of oAd replicating under the control of the HCC-specific Ha2bm promoter (HCC-oAd-Luc). A, Schematic HCC-oAd-Luc structure. This oAd replicates under the control of the hypoxic response element and a modified AFP promoter. The expression cassette for luciferase was inserted into the E3 region of the Ad genome. B, Efficiency of HCC-oAd-Luc transduction into MSCs. MSCs were treated with HCC-oAd-Luc at MOIs of 50 to 200. At 48 hours postinfection, luciferase signals were assessed by an IVIS imaging system (original magnification, ×1). The bioluminescence signal intensity was obtained as photons acquired per second (p/s) from regions of interest. The experiment was assessed at least three times and the data shown are representative of experiments performed in triplicate.

Close modal

Dose optimization for oAd loading into MSCs

Optimization of the oAd dose for loading into cell carriers is critical for preventing premature cytolysis. Thus, the optimal oAd loading dose for MSCs was assessed. As shown in Fig. 2A, MSC viability was significantly lower at 5 versus 2 days postinfection for all MOIs (P < 0.001). These results suggest that infecting MSCs with HCC-oAd-Luc for 5 days is suboptimal because the majority of the MSCs were lysed by the virus.

Figure 2.

MSC killing effect of HCC-targeting oAd and its viral production in MSCs. A, MSC killing effect of HCC-targeting oAd. MSCs were infected with HCC-oAd-Luc at MOIs ranging from 0.5 to 50. At 2 or 5 days postinfection, cell viability was assessed by the MTT assay. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. B, Viral production of HCC-oAd-Luc in MSCs. MSCs were infected with HCC-oAd-Luc at MOIs ranging from 0.5 to 50. At 2 or 5 days postinfection, the total viral yield produced in MSCs was quantified by qPCR. Dashed line represents the detection limit of qPCR. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. ***, P < 0.001.

Figure 2.

MSC killing effect of HCC-targeting oAd and its viral production in MSCs. A, MSC killing effect of HCC-targeting oAd. MSCs were infected with HCC-oAd-Luc at MOIs ranging from 0.5 to 50. At 2 or 5 days postinfection, cell viability was assessed by the MTT assay. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. B, Viral production of HCC-oAd-Luc in MSCs. MSCs were infected with HCC-oAd-Luc at MOIs ranging from 0.5 to 50. At 2 or 5 days postinfection, the total viral yield produced in MSCs was quantified by qPCR. Dashed line represents the detection limit of qPCR. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. ***, P < 0.001.

Close modal

For further optimization, we assessed the level of viral replication in MSCs (Fig. 2B). A 10-fold increase in the viral dose, from 0.5 to 5 MOI, resulted in a 5,000-fold higher level of viral production at day 2 postinfection, whereas a same fold increase in the viral dose, from 5 to 50 MOI, only led to a minimal increase (1.8-fold) in viral production, showing that increasing the viral dose above 5 MOI yields a diminishing return on viral production. Because the viral production level plateaued at MOIs higher than 5 and a significant dose-dependent reduction in MSC viability was observed at the same viral dose range, we chose day 2 of infection and an MOI of 5 as the optimal conditions, which balance viral replication and MSC viability, to generate oAd-loaded MSCs in our subsequent experiments.

Characterization of HCC-oAd-WNTi

HCC cells demonstrate rapid and uncontrolled growth driven by the highly activated Wnt pathway (20). Therefore, we previously developed an oAd that expresses WNTi and demonstrated that WNTi expression can effectively inhibit the overactive Wnt signaling pathway in cancer (31). To continue this line of work, in our current study we designed and generated a HCC-specific oAd that expresses WNTi by utilizing a enhancer region–modified AFP promoter (Ha2bm; ref. 22) to restrict oAd replication to AFP-positive HCCs, ultimately generating HCC-oAd-WNTi (Fig. 3A).

Figure 3.

Construction and characterization of HCC-oAd-WNTi. A, Schematic HCC-oAd and HCC-oAd-WNTi structures. These oAds replicate under the control of the hypoxic response element and a modified AFP promoter. The expression cassette for WNTi was inserted into the E3 region of the Ad genome. B, Wnt signaling pathway–related gene expression. Hep3B cells were treated with HCC-oAd or HCC-oAd-WNTi at a MOI of 1. At 48 hours postinfection, Wnt3a, β-catenin, p-MEK, and E-cadherin expression levels were assessed by Western blot. C, HCC killing effect of HCC-oAd-WNTi. Hep3B cells were infected with H101, HCC-oAd, or HCC-oAd-WNTi at MOIs ranging from 1 to 50. At 48 hours postinfection, cell viability was assessed by the MTT assay. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. ***, P < 0.001, HCC-oAd versus HCC-oAd-WNTi. D, HCC killing effect of HCC-oAd-WNTi under hypoxic conditions. Hep3B cells were infected with H101, HCC-oAd, or HCC-oAd-WNTi at an MOI of 2 under normoxic or hypoxic conditions. At 48 hours postinfection, cell viability was assessed by the MTT assay. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. *, P < 0.05; ***, P < 0.001.

Figure 3.

Construction and characterization of HCC-oAd-WNTi. A, Schematic HCC-oAd and HCC-oAd-WNTi structures. These oAds replicate under the control of the hypoxic response element and a modified AFP promoter. The expression cassette for WNTi was inserted into the E3 region of the Ad genome. B, Wnt signaling pathway–related gene expression. Hep3B cells were treated with HCC-oAd or HCC-oAd-WNTi at a MOI of 1. At 48 hours postinfection, Wnt3a, β-catenin, p-MEK, and E-cadherin expression levels were assessed by Western blot. C, HCC killing effect of HCC-oAd-WNTi. Hep3B cells were infected with H101, HCC-oAd, or HCC-oAd-WNTi at MOIs ranging from 1 to 50. At 48 hours postinfection, cell viability was assessed by the MTT assay. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. ***, P < 0.001, HCC-oAd versus HCC-oAd-WNTi. D, HCC killing effect of HCC-oAd-WNTi under hypoxic conditions. Hep3B cells were infected with H101, HCC-oAd, or HCC-oAd-WNTi at an MOI of 2 under normoxic or hypoxic conditions. At 48 hours postinfection, cell viability was assessed by the MTT assay. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. *, P < 0.05; ***, P < 0.001.

Close modal

To assess whether the newly generated HCC-oAd-WNTi could suppress the Wnt signaling pathway, we investigated the expression levels of Wnt and various associated downstream factors in HCC-oAd- or HCC-oAd-WNTi–infected Hep3B cells via Western blotting. As shown in Fig. 3B, Hep3B cells treated with HCC-oAd-WNTi exhibited a significantly lower Wnt expression level than did cells infected with control oAd (HCC-oAd) or the negative control group (untreated). Furthermore, HCC-oAd-WNTi treatment significantly attenuated the expression levels of various downstream factors, such as β-catenin and p-MEK that are known to promote rapid cancer cell proliferation and EMT. In addition, the expression level of a mesenchymal marker, E-cadherin, was markedly higher in HCC-oAd-WNTi–infected Hep3B cells than in untreated or HCC-oAd–infected cells. Together, these data show that HCC-oAd-WNTi effectively inhibited the Wnt signaling pathway and decreased levels of an EMT-related protein in HCC cells.

To assess whether expression of WNTi by the HCC-targeting oAd would enhance the cell-killing effect of oAd in HCC cells, AFP-positive HCC cells were infected with a clinically approved oAd (H101), the cognate control HCC-oAd, or HCC-oAd-WNTi at various MOIs. As shown in Fig. 3C, HCC-oAd-WNTi elicited a dose-dependent and significantly greater cancer cell–killing effect than H101 or HCC-oAd at all MOIs (P < 0.001), suggesting that WNTi expression enhances the cancer cell–killing effect of HCC-oAd. The IC50 values for the control HCC-oAd and HCC-oAd-WNTi were 10.3 and 1.6 MOI, respectively, showing that the expression of WNTi augments the anticancer effect of oAd. We had previously shown that the Ha2bm promoter–driven viral replication of HCC-oAd overcomes hypoxia-induced repression of Ad replication (22). Thus, we assessed whether HCC-oAd-WNTi elicits more potent cancer cell–killing effect under hypoxic conditions than normoxia. As shown in Fig. 3D, HCC-oAd-WNTi elicited greater cancer cell–killing efficacy than HCC-oAd under both hypoxia and normoxia. Furthermore, HCC-oAd-WNTi showed 1.2-fold greater cancer cell–killing efficacy under hypoxia than normoxia (P < 0.001). These findings suggest that HCC-oAd-WNTi elicits a potent cell-killing effect in AFP-positive HCCs and overcomes hypoxia-mediated downregulation of viral replication (22).

Potent cancer-specific killing effect of HCC-oAd-WNTi/MSC

To evaluate the cancer-specific killing effect of HCC-oAd-WNTi/MSC (infected with 5 MOI of oAd), we cocultured a cancer or normal cell line [HCC (Hep3B), non-HCC cancer (A549), or normal (BJ)] with HCC-oAd-WNTi/MSC at a 1:1 MSC-to-cell ratio. As shown in Fig. 4, similar level of viability was observed for the untreated and MSC-treated groups under both normoxic and hypoxic conditions, suggesting that MSCs alone did not affect the cell growth rate. In contrast, Hep3B cells treated with HCC-oAd-WNTi or HCC-oAd-WNTi/MSC exhibited significantly reduced viability in comparison with untreated or MSC-treated cells, suggesting that both naked HCC-oAd-WNTi and the virus loaded in MSCs can kill AFP-positive HCC cells. At day 1 posttreatment, HCC-oAd-WNTi- and HCC-oAd-WNTi/MSC–treated Hep3B cells exhibited a similar level of viability to that of the untreated group, emphasizing that the reduction in cancer cell viability was due to replication-mediated cytopathic effects in the oAd-incorporated groups. In marked contrast, HCC-oAd-WNTi/MSC treatment elicited a significantly more potent HCC killing effect than HCC-oAd-WNTi on day 3 or 5 of treatment (P < 0.05, P < 0.01), showing that HCC-oAd-WNTi replication in MSCs enhances the cytopathic effect. These results are in agreement with results shown in Fig. 2B and S2. In AFP-negative A549 lung cancer cells, similar results were observed only under hypoxic conditions (P < 0.01), confirming the HRE-mediated enhancement of Ad replication (22, 26). These results suggest that HCC-oAd-WNTi/MSC can elicit cancer-specific cell-killing effect in response to hypoxia, which is a hallmark of solid tumors (32), regardless of cellular AFP level; the expanded cancer targeting by hypoxia-responsiveness of Ha2bm promoter would be beneficial in clinical setting where heterogeneity of tumors leads to large phenotypical variance in tumors, yet hypoxia is conserved in all types of solid tumors, including 30% of HCC patient cases with AFP-negative HCC tumors (22, 33). Furthermore, both HCC-oAd-WNTi and HCC-oAd-WNTi/MSC elicited minimal killing effects in the normal BJ cell line, showing that the oncolytic effects of HCC-oAd-WNTi/MSC occur with high specificity toward cancer cells.

Figure 4.

Cancer-specific killing effect of HCC-oAd-WNTi/MSC. Hep3B, A549, and BJ cells were infected with MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC (MSCs infected with 5 MOI of virus for 2 days) at a 1:1 cancer cell-to-MSC ratio under normoxic or hypoxic conditions. At 1, 3, or 5 days postinfection, cell viability was assessed by the MTT assay. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 4.

Cancer-specific killing effect of HCC-oAd-WNTi/MSC. Hep3B, A549, and BJ cells were infected with MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC (MSCs infected with 5 MOI of virus for 2 days) at a 1:1 cancer cell-to-MSC ratio under normoxic or hypoxic conditions. At 1, 3, or 5 days postinfection, cell viability was assessed by the MTT assay. The data are representative of three independent experiments performed in triplicate. Bars, mean ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

Potent therapeutic efficacy of systemically delivered HCC-oAd-WNTi/MSC in an orthotopic HCC tumor model

Orthotopic tumor models are emerging as important cancer research models due to their clinical relevance (34). Solid tumors inaccessible by a needle, such as lung, liver, and pancreatic cancers, cannot be treated by intratumoral administration, and thus require systemic treatment. To evaluate the therapeutic efficacy of systemically administered oAd-loaded MSCs, luciferase-expressing orthotopic Hep3B tumors were treated with PBS, MSCs, HCC-oAd-WNTi (5 × 108 VP), or HCC-oAd-WNTi/MSC (1 × 106 MSC infected with 5 × 108 VP of oAd; 5 MOI) via tail-vein injection. Infection of MSC with 5 MOI of HCC-oAd-WNTi for 18 hours (HCC-oAd-WNTi/MSC) had negligible impact on MSC viability, because more than 95% of MSCs were viable with these conditions (Supplementary Fig. S3). In addition, HCC-oAd-WNTi/MSC prepared by these conditions was determined to possess intracellular VP count that is equivalent to infection with 0.08 MOI (Supplementary Fig. S2), which would translate to 8.0 × 106 VP being delivered by MSC at time of administration. These results indicate that 62.5-fold lower VP of HCC-oAd-WNTi/MSC were administered compared with HCC-oAd-WNTi. As shown in Fig. 5A and B, the systemic administration of HCC-oAd-WNTi/MSC resulted in markedly higher antitumor activity than either HCC-oAd-WNTi or MSCs alone at 35 days postimplantation (P < 0.01), showing 44.7-, 24.0-, or 8.1-fold greater therapeutic efficacy than PBS, MSCs, or HCC-oAd-WNTi, respectively.

Figure 5.

Antitumor effect of HCC-oAd-WNTi/MSC in an orthotopic Hep3B tumor model. A and B, Luciferase-expressing Hep3B cells were injected directly into the left lobe of the liver in mice to generate tumors. For the treatment of tumors, PBS, MSCs (1 × 106 cells), HCC-oAd-WNTi (5 × 108 VPs), or HCC-oAd-WNTi/MSC (1 × 106 cells infected with 5 × 108 VPs) were intravenously injected at 9 and 13 days after the tumor cell injection. Tumor growth was analyzed every week by optical and bioluminescence imaging. Image signals were quantitatively analyzed with IGOR-PRO Living Image software. C and D, At 35 days following the initial treatment with PBS, MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC, livers of mice from each experimental group were harvested and photographed by bioluminescence imaging. Image signals were quantitatively analyzed with IGOR-PRO Living Image software. E, Tumor weights were measured at the end of experiment at 35 days posttreatment, after ex vivo imaging. All tumors were separated from the liver and weighed with a conventional scale. *, P < 0.05; **, P < 0.01.

Figure 5.

Antitumor effect of HCC-oAd-WNTi/MSC in an orthotopic Hep3B tumor model. A and B, Luciferase-expressing Hep3B cells were injected directly into the left lobe of the liver in mice to generate tumors. For the treatment of tumors, PBS, MSCs (1 × 106 cells), HCC-oAd-WNTi (5 × 108 VPs), or HCC-oAd-WNTi/MSC (1 × 106 cells infected with 5 × 108 VPs) were intravenously injected at 9 and 13 days after the tumor cell injection. Tumor growth was analyzed every week by optical and bioluminescence imaging. Image signals were quantitatively analyzed with IGOR-PRO Living Image software. C and D, At 35 days following the initial treatment with PBS, MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC, livers of mice from each experimental group were harvested and photographed by bioluminescence imaging. Image signals were quantitatively analyzed with IGOR-PRO Living Image software. E, Tumor weights were measured at the end of experiment at 35 days posttreatment, after ex vivo imaging. All tumors were separated from the liver and weighed with a conventional scale. *, P < 0.05; **, P < 0.01.

Close modal

Although HCC-oAd-WNTi did not induce tumor regression in this report due to extremely low viral dose deflating the overall antitumor effect to a suboptimal level, systemic administration of HCC-specific oAd replicating under the control of Ha2bm promoter at higher dose would likely achieve more potent tumor growth inhibition. Indeed, a systemic administration of HCC-oAd at 2.5 × 1010 VP has been reported to elicit potent antitumor effect against orthotopic HCC tumor (22). Ex vivo imaging of the livers further supports these data; PBS-, MSC-, or HCC-oAd-WNTi–treated livers showed a relatively larger tumor burden, whereas livers from HCC-oAd-WNTi/MSC–treated mice had a significantly lower tumor burden (Fig. 5C and D). Similar results were observed when the tumor burden was assessed by end-point tumor weight measurement (Fig. 5E). Of note, the HCC-oAd-WNTi/MSC was estimated to harbor 8.0 × 106 VP of oAd based on the results from Supplementary Fig. S2, which meant that the injected dose was 2,500-fold lower than the oAd dose (∼2 × 1010 VPs) frequently utilized for systemic treatment of tumor-bearing mice (27). These results suggest that loading of oAd into MSCs augments the therapeutic efficacy of systemically administered virus while minimizing any potential dose-related side effects.

To further investigate the therapeutic effect of HCC-oAd-WNTi/MSC, liver tissues were harvested at 3 days after the last treatment (with PBS, MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC). As shown in Fig. 6A, H&E staining revealed large tumor areas in both PBS- and MSC-treated liver tissues. In contrast, HCC-oAd-WNTi/MSC–treated liver tissues showed no observable tumor cells. In support of the H&E staining results, HCC-oAd-WNTi/MSC treatment led to a markedly lower level of proliferating cells compared with PBS, MSC, or HCC-oAd-WNTi treatments (Fig. 6B), indicating that HCC-oAd-WNTi/MSC induces a potent antitumor effect by inhibiting HCC cell proliferation. Next, we assessed how the oAd affects MSC viability in the tumor tissues by staining for CD90, a marker of human MSCs. As shown in Fig. 6C, the tumor tissues from the mice treated with MSCs were CD90+, whereas no MSCs were observed in the liver tissues treated with HCC-oAd-WNTi/MSC.

Figure 6.

Histologic and IHC changes in orthotopic Hep3B tumors treated with HCC-oAd-WNTi/MSC. A, Representative sections were stained with H&E. Original magnification, ×50 and ×400. B, IHC staining of PCNA was performed on tumor sections from each group of mice. C, MSC localization was assessed by IHC using an CD90–specific antibody.

Figure 6.

Histologic and IHC changes in orthotopic Hep3B tumors treated with HCC-oAd-WNTi/MSC. A, Representative sections were stained with H&E. Original magnification, ×50 and ×400. B, IHC staining of PCNA was performed on tumor sections from each group of mice. C, MSC localization was assessed by IHC using an CD90–specific antibody.

Close modal

Improved pharmacokinetic profile of HCC-oAd-WNTi/MSC

For the efficient treatment of HCC tumors by systemic injection, the blood retention time of Ad must be prolonged (35). Therefore, we assessed whether MSC prolongs the blood retention time of HCC-oAd-WNTi. As shown in Fig. 7A, naked HCC-oAd-WNTi was rapidly cleared from the blood. In contrast, HCC-oAd-WNTi/MSC was retained at 17- and 3,200-fold higher levels in the blood compared with naked HCC-oAd-WNTi at 6 hours and 24 hours postinjection (P < 0.001 for HCC-oAd-WNTi vs. HCC-oAd-WNTi/MSC at 6 hours: 2.4 × 103 vs. 4.0 × 104 VP and 24 hours: 1.8 × 101 vs. 5.9 × 104 VP), respectively, suggesting that MSCs efficiently extend the oAd blood circulation time.

Figure 7.

Pharmacokinetic profile, biodistribution, and safety profile of HCC-oAd-WNTi/MSC. A, Pharmacokinetic profile of HCC-oAd-WNTi/MSC. HCC-oAd-WNTi or HCC-oAd-WNTi/MSC was injected intravenously into mice. Total viral DNA was extracted from blood samples (obtained 5 minutes, 10 minutes, 30 minutes, 1 hours, 6 hours, 24 hours, and 48 hours after injection). The Ad genome copy number was determined by qRT-PCR. The data are representative of three independent experiments performed in triplicate. B, Biodistribution of HCC-oAd-WNTi and HCC-oAd-WNTi/MSC. HCC-oAd-WNTi or HCC-oAd-WNTi/MSC was systemically injected into the tail vein twice, on days 9 and 13 posttumor implantation. The lung, heart, stomach, spleen, liver, tumor, and blood were harvested at 24 hours after the second injection. qRT-PCR was performed to detect Ad genomes. The data are representative of three independent experiments performed in triplicate. Data were normalized by subtracting experimental group values from the corresponding values from the PBS-treated group and are presented as mean ± SD. C, Liver toxicity test. Serum ALT and AST levels were measured at 3 days after systemic administration of PBS, MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC. Nontumor-bearing normal mice were used as a control. The data are representative of three independent experiments performed in triplicate. Data represent mean ± SD *, P < 0.05; ***, P < 0.001.

Figure 7.

Pharmacokinetic profile, biodistribution, and safety profile of HCC-oAd-WNTi/MSC. A, Pharmacokinetic profile of HCC-oAd-WNTi/MSC. HCC-oAd-WNTi or HCC-oAd-WNTi/MSC was injected intravenously into mice. Total viral DNA was extracted from blood samples (obtained 5 minutes, 10 minutes, 30 minutes, 1 hours, 6 hours, 24 hours, and 48 hours after injection). The Ad genome copy number was determined by qRT-PCR. The data are representative of three independent experiments performed in triplicate. B, Biodistribution of HCC-oAd-WNTi and HCC-oAd-WNTi/MSC. HCC-oAd-WNTi or HCC-oAd-WNTi/MSC was systemically injected into the tail vein twice, on days 9 and 13 posttumor implantation. The lung, heart, stomach, spleen, liver, tumor, and blood were harvested at 24 hours after the second injection. qRT-PCR was performed to detect Ad genomes. The data are representative of three independent experiments performed in triplicate. Data were normalized by subtracting experimental group values from the corresponding values from the PBS-treated group and are presented as mean ± SD. C, Liver toxicity test. Serum ALT and AST levels were measured at 3 days after systemic administration of PBS, MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC. Nontumor-bearing normal mice were used as a control. The data are representative of three independent experiments performed in triplicate. Data represent mean ± SD *, P < 0.05; ***, P < 0.001.

Close modal

Biodistribution and safety profiles of HCC-oAd-WNTi/MSC in an orthotopic HCC tumor model

Naked Ad is nonspecifically sequestered in the liver following intravenous injection due to interactions with Kupffer cells and coagulation factors (27), resulting in hepatotoxicity and a limited antitumor effect. To examine whether HCC-oAd-WNTi/MSC can circumvent hepatic sequestration while enhancing intratumoral oAd accumulation, biodistribution profiles of systemically administered MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC were analyzed in tumor-bearing mice. As shown in Fig. 7B, the liver uptake of HCC-oAd-WNTi/MSC was significantly less than that of naked HCC-oAd-WNTi (P < 0.001). Importantly, HCC-oAd-WNTi/MSC showed 4,824-fold higher intratumoral accumulation than naked HCC-oAd-WNTi (P < 0.001). These results show that MSCs can protect their oAd cargo effectively during systemic circulation and curtail native hepatic oAd tropism, leading to efficient intratumoral accumulation of the virus. In addition, these data suggest that MSC-mediated intratumoral oAd localization, even at a low viral dose, can lead to a potent antitumor effect as the HCC-oAd-WNTi can effectively replicate within tumor tissues. Consequently, the tumor-to-liver ratio of HCC-oAd-WNTi/MSC was 6,481-fold greater than that of naked HCC-oAd-WNTi, showing that MSC-based delivery of oncolytic Ad can overcome the hurdles of systemic administration to improve the therapeutic efficacy and safety profile of the virus.

Next, hepatotoxicity was analyzed by assessing the serum level of ALT and AST after the systemic administration of PBS, MSCs, HCC-oAd-WNTi, or HCC-oAd-WNTi/MSC in tumor-bearing mice, as well as in normal mice without tumor. As shown in Fig. 7C, mice treated with HCC-oAd-WNTi showed the highest AST levels (1.6-fold higher than in the PBS-treated group; P < 0.05). Conversely, we did not observe a significant increase in AST levels in mice treated with HCC-oAd-WNTi/MSC relative to levels in the normal mice group, showing that MSC-mediated tumor-specific oAd delivery prevents the hepatic damage that is traditionally associated with systemically administered Ad as well as with an HCC tumor burden (22, 36).

MSCs can infiltrate into tumor tissues and release therapeutic cargos, such as conventional chemotherapeutics or oncolytic viruses (37, 38). Although several types of drugs can be loaded into MSCs, efficient loading of Ad into MSCs is hampered by a low level of CAR expression on the MSC surface (39). This problem was addressed in this report by replacement of the endogenous Ad serotype 5 fiber knob with an Ad serotype 35 fiber knob to enhance Ad transduction into MSCs (Supplementary Fig. S1). Here, we employed other genetic modification strategies to further improve the therapeutic efficacy of oAd against HCC. We previously developed HCC-specific oAd (HCC-oAd), which utilizes modified AFP hypoxia–responsive and modified AFP enhancer regions to enhance transcription of the Ad E1A gene; this modification overcomes hypoxia-mediated downregulation of Ad replication in tumors. Building on this previous work, we have now inserted a sequence encoding WNTi, a soluble Wnt decoy receptor, into HCC-oAd (generating HCC-oAd-WNTi) to inhibit the hyperactive Wnt signaling pathway, a problem found in 50% of clinical HCCs that is associated with a poor patient survival rate (31, 40). The addition of WNTi as a therapeutic gene allowed for effective suppression of various Wnt signaling–related factors and improved the HCC killing potency of HCC-oAd-WNTi (Fig. 3). The disruption of the overactive Wnt signaling pathway in HCC is of critical importance as only few Wnt-inhibitory chemical drugs have reached phase I clinical trials to date, even though the Wnt/β-catenin pathway is frequently upregulated and implicated in drug resistance, tumor progression, and metastasis of HCC (41). Furthermore, oAd-mediated therapeutic gene expression has been shown to be highly localized to tumor tissues due to tumor-specific replication and gene expression by the virus (28), making oAd better suited for HCC therapy than chemical compounds with insufficient cancer specificity, given that Wnt/β-catenin signaling is critical for the maintenance of diverse liver functions in normal hepatic tissues (42).

One of the innovative aspects of using MSCs as a carrier for oncolytic viruses is that these cells could function as a biological factory to enable viral replication, suggesting that it may be possible for a small quantity of virus to be initially loaded into MSCs to deliver a sufficient viral dose to tumor tissues (15, 43). Even though replication of oncolytic virus in MSCs has been reported by our current study and others before (44, 45), the detailed mechanism is not clear. One plausible explanation is that Ha2bm promoter is predicted to possess several transcription factor–binding domains for C-JUN, FOXP3, and C/EBP-β, which are expressed in the MSC and upregulated during determination of MSC fate (46, 47), and these attributes may allow replication of HCC-oAd-WNTi in MSC through Ha2bm promoter activation.

Although viral replication within MSCs is a promising attribute, excessive viral replication may induce premature MSC lysis, which would decrease the overall efficacy of the system. Given this background, we carefully optimized the viral dose for MSC infection to balance the increase in total viral yield with the reduction in MSC viability (Fig. 2), showing that too high of an initial viral dose critically attenuates overall MSC viability with minimal improvement in total viral yield. Profiling of MCS survival and viral release also demonstrate that MSCs infected with HCC-oAd-WNTi (5 MOI; 18 hours or 2 days of infection; in vivo or in vitro preparation condition of HCC-oAd-WNTi/MSC, respectively) are healthy and viable enough to produce progeny viruses (Supplementary Figs. S3 and S2). In particular, plenty of viral progenies were released from the MSCs due to cytolytic property of oAd. These results are in line with a previously reported Ad production protocol in which a low initial dose of Ad (2 MOI) was utilized to optimize viral production in a virus producer cell line, suggesting that an excessive viral dose may lead to poor viral production and yield due to an asymmetrical compensation in cell viability (48).

Notably, the conditions for optimal viral replication and MSC viability (Fig. 2) enabled HCC-oAd-WNTi/MSC to elicit a more potent HCC killing effect than that elicited by naked HCC-oAd-WNTi (Figs. 3 and 4), showing that, over time, viral replication in MSCs may increase the final viral titer. The difference between the anticancer effects of naked HCC-oAd-WNTi and HCC-oAd-WNTi/MSC was even greater in vivo: systemic treatment with a substandard therapeutic dose of HCC-oAd-WNTi/MSC led to complete regression in a subpopulation of treated mice (Fig. 5), supporting in vitro findings (Figs. 2 and 4). In general, the systemic administration of various oAds for the treatment of orthotopic HCC tumors has required more than 1 × 1010 VPs to elicit a notable antitumor effect (22, 26), and to best of our knowledge there are no available reports of systemically administered oAd inducing complete HCC regression at a 40-fold lower viral dose than the conventionally reported dose of 2 × 1010 VPs. Such dose reduction is critical for the clinical development of new therapeutics, because it can greatly improve the safety profile.

The tumor-homing and cargo-protective properties of MSCs acting as a cell carrier are well known (15, 49). In present, our findings show that cargo-protective properties of MSC enabled the virions loaded in HCC-oAd-WNTi/MSC to efficiently accumulate in tumor tissues, in part, through enhanced virion retention in the bloodstream (Fig. 7A and B, respectively). In agreement with this finding, others have reported that several types of systemic carriers can improve the intratumoral accumulation of systemically administered oAd by prolonging the blood circulation of virions (27, 50). Carrier MSCs also attenuated hepatic sequestration of oAd and prevented hepatic damage (Fig. 7B and C), a promising result given that the hepatic tropism and inflammatory attributes of Ad are major concerns associated with the systemic delivery of Ad in clinical trials. These findings show that MSCs functioning as a systemic carrier of oAd can overcome conventional shortcomings associated with systemic oAd delivery, such as nonspecific hepatic sequestration, hepatotoxicity, and poor blood circulation (29, 30).

Finally, one of the major concerns about all stem cell–based therapy in clinical trials is that stem cells may uncontrollably differentiate into undesirable lineages, potentially leading to tumorigenesis and inflammation; thus, the FDA requires long periods of patient follow-up during and after clinical trials (51). Our findings provide strong evidence that this potential risk can be addressed by utilizing the cytolytic effects of oAd, because CD90+ MSCs were not detected in liver tissues from mice treated with HCC-oAd-WNTi/MSC at 3 days after administration, whereas MSCs remained in the liver tissues from mice treated with MSCs alone (Fig. 6C). Our in vitro findings shown in Fig. 2 also support this claim because active oAd replication in MSCs led to cell lysis, thus abolishing any potential risk that may arise due to engraftment of therapeutic MSCs into tumor tissues.

Collectively, we demonstrate that (i) HCC-targeted oAd can be efficiently loaded into MSCs through modification of the viral capsid, (ii) oAd loaded into MSCs can elicit potent cancer-specific killing effects through active viral replication in the MSC carrier, (iii) MSC tumor–homing tropism improves tumor-specific oAd accumulation, (iv) the cargo-protective attributes of MSCs prolong and enhance virion circulation in the blood, and (v) oAd-loaded MSC improves the safety profile of both oAd and MSCs by decreasing oAd hepatic sequestration and hepatotoxicity while promoting elimination of MSCs through viral replication. These findings show that MSCs loaded with HCC-targeting oAd can serve as a “stealth carrier” to preferentially deliver the virus to the tumor while attenuating any potential risk that may arise from systemic administration of naked virions in the clinical environment.

J. Hong is a researcher at GeneMedicine. C.-O. Yun is the CEO at GeneMedicine. H.C. Shin and H Lee are researchers at Phamicell. H.S. Kim is the CEO at Pharmicell Co. Ltd. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A.-R. Yoon, H. Lee, C.-O. Yun

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.-R. Yoon, Y. Li, H.C. Shin

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.-R. Yoon, J. Hong, Y. Li

Writing, review, and/or revision of the manuscript: A.-R. Yoon, J. Hong, C.-O. Yun

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.-R. Yoon, Y. Li, H. Lee, H.S. Kim, C.-O. Yun

Study supervision: C.-O. Yun

This work was supported by grants from the National Research Foundation of Korea (2016M3A9B5942352 to C.-O. Yun; 2016R1C1B2015558 to A.-R. Yoon) and Korea Drug Development Fund (KDDF) funded by MSIP, MOTIE, and MOHW (KDDF-201611-05, Republic of Korea, to A.-R. Yoon).

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.
Bosch
FX
,
Ribes
J
,
Diaz
M
,
Cleries
R
. 
Primary liver cancer: worldwide incidence and trends
.
Gastroenterology
2004
;
127
:
S5
S16
.
2.
Llovet
JM
,
Burroughs
A
,
Bruix
J
. 
Hepatocellular carcinoma
.
Lancet
2003
;
362
:
1907
17
.
3.
Tabrizian
P
,
Jibara
G
,
Shrager
B
,
Schwartz
M
,
Roayaie
S
. 
Recurrence of hepatocellular cancer after resection: patterns, treatments, and prognosis
.
Ann Surg
2015
;
261
:
947
55
.
4.
Llovet
JM
,
Ricci
S
,
Mazzaferro
V
,
Hilgard
P
,
Gane
E
,
Blanc
JF
, et al
Sorafenib in advanced hepatocellular carcinoma
.
N Engl J Med
2008
;
359
:
378
90
.
5.
Bruix
J
,
Raoul
JL
,
Sherman
M
,
Mazzaferro
V
,
Bolondi
L
,
Craxi
A
, et al
Efficacy and safety of sorafenib in patients with advanced hepatocellular carcinoma: subanalyses of a phase III trial
.
J Hepatol
2012
;
57
:
821
9
.
6.
Spaeth
E
,
Klopp
A
,
Dembinski
J
,
Andreeff
M
,
Marini
F
. 
Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells
.
Gene Ther
2008
;
15
:
730
8
.
7.
Chamberlain
G
,
Fox
J
,
Ashton
B
,
Middleton
J
. 
Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing
.
Stem Cells
2007
;
25
:
2739
49
.
8.
Devine
SM
,
Cobbs
C
,
Jennings
M
,
Bartholomew
A
,
Hoffman
R
. 
Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates
.
Blood
2003
;
101
:
2999
3001
.
9.
Moradian Tehrani
R
,
Verdi
J
,
Noureddini
M
,
Salehi
R
,
Salarinia
R
,
Mosalaei
M
, et al
Mesenchymal stem cells: a new platform for targeting suicide genes in cancer
.
J Cell Physiol
2018
;
233
:
3831
45
.
10.
Sage
EK
,
Thakrar
RM
,
Janes
SM
. 
Genetically modified mesenchymal stromal cells in cancer therapy
.
Cytotherapy
2016
;
18
:
1435
45
.
11.
Chen
XC
,
Wang
R
,
Zhao
X
,
Wei
YQ
,
Hu
M
,
Wang
YS
, et al
Prophylaxis against carcinogenesis in three kinds of unestablished tumor models via IL12-gene-engineered MSCs
.
Carcinogenesis
2006
;
27
:
2434
41
.
12.
Matsumoto
R
,
Omura
T
,
Yoshiyama
M
,
Hayashi
T
,
Inamoto
S
,
Koh
KR
, et al
Vascular endothelial growth factor-expressing mesenchymal stem cell transplantation for the treatment of acute myocardial infarction
.
Arterioscler Thromb Vasc Biol
2005
;
25
:
1168
73
.
13.
Duchi
S
,
Sotgiu
G
,
Lucarelli
E
,
Ballestri
M
,
Dozza
B
,
Santi
S
, et al
Mesenchymal stem cells as delivery vehicle of porphyrin loaded nanoparticles: effective photoinduced in vitro killing of osteosarcoma
.
J Control Release
2013
;
168
:
225
37
.
14.
Ahmed
AU
,
Rolle
CE
,
Tyler
MA
,
Han
Y
,
Sengupta
S
,
Wainwright
DA
, et al
Bone marrow mesenchymal stem cells loaded with an oncolytic adenovirus suppress the anti-adenoviral immune response in the cotton rat model
.
Mol Ther
2010
;
18
:
1846
56
.
15.
Komarova
S
,
Kawakami
Y
,
Stoff-Khalili
MA
,
Curiel
DT
,
Pereboeva
L
. 
Mesenchymal progenitor cells as cellular vehicles for delivery of oncolytic adenoviruses
.
Mol Cancer Ther
2006
;
5
:
755
66
.
16.
Bischoff
JR
,
Kirn
DH
,
Williams
A
,
Heise
C
,
Horn
S
,
Muna
M
, et al
An adenovirus mutant that replicates selectively in p53-deficient human tumor cells
.
Science
1996
;
274
:
373
6
.
17.
Kirn
D
. 
Replication-selective oncolytic adenoviruses: virotherapy aimed at genetic targets in cancer
.
Oncogene
2000
;
19
:
6660
9
.
18.
Su
C
,
Peng
L
,
Sham
J
,
Wang
X
,
Zhang
Q
,
Chua
D
, et al
Immune gene-viral therapy with triplex efficacy mediated by oncolytic adenovirus carrying an interferon-gamma gene yields efficient antitumor activity in immunodeficient and immunocompetent mice
.
Mol Ther
2006
;
13
:
918
27
.
19.
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
.
20.
Jia
Q
,
Bu
Y
,
Wang
Z
,
Chen
B
,
Zhang
Q
,
Yu
S
, et al
Maintenance of stemness is associated with the interation of LRP6 and heparin-binding protein CCN2 autocrined by hepatocellular carcinoma
.
J Exp Clin Cancer Res
2017
;
36
:
117
.
21.
Thompson
EW
,
Haviv
I
. 
The social aspects of EMT-MET plasticity
.
Nat Med
2011
;
17
:
1048
9
.
22.
Yoon
AR
,
Hong
J
,
Kim
M
,
Yun
CO
. 
Hepatocellular carcinoma-targeting oncolytic adenovirus overcomes hypoxic tumor microenvironment and effectively disperses through both central and peripheral tumor regions
.
Sci Rep
2018
;
8
:
2233
.
23.
Choi
IK
,
Lee
YS
,
Yoo
JY
,
Yoon
AR
,
Kim
H
,
Kim
DS
, et al
Effect of decorin on overcoming the extracellular matrix barrier for oncolytic virotherapy
.
Gene Ther
2010
;
17
:
190
201
.
24.
Lee
JS
,
Hur
MW
,
Lee
SK
,
Choi
WI
,
Kwon
YG
,
Yun
CO
. 
A novel sLRP6E1E2 inhibits canonical Wnt signaling, epithelial-to-mesenchymal transition, and induces mitochondria-dependent apoptosis in lung cancer
.
PLoS One
2012
;
7
:
e36520
.
25.
Yoon
AR
,
Kim
JH
,
Lee
YS
,
Kim
H
,
Yoo
JY
,
Sohn
JH
, et al
Markedly enhanced cytolysis by E1B-19kD-deleted oncolytic adenovirus in combination with cisplatin
.
Hum Gene Ther
2006
;
17
:
379
90
.
26.
Kwon
OJ
,
Kim
PH
,
Huyn
S
,
Wu
L
,
Kim
M
,
Yun
CO
. 
A hypoxia- and {alpha}-fetoprotein-dependent oncolytic adenovirus exhibits specific killing of hepatocellular carcinomas
.
Clin Cancer Res
2010
;
16
:
6071
82
.
27.
Yoon
AR
,
Kasala
D
,
Li
Y
,
Hong
J
,
Lee
W
,
Jung
SJ
, et al
Antitumor effect and safety profile of systemically delivered oncolytic adenovirus complexed with EGFR-targeted PAMAM-based dendrimer in orthotopic lung tumor model
.
J Control Release
2016
;
231
:
2
16
.
28.
Yoon
AR
,
Hong
J
,
Yun
CO
. 
Adenovirus-mediated decorin expression induces cancer cell death through activation of p53 and mitochondrial apoptosis
.
Oncotarget
2017
;
8
:
76666
85
.
29.
Kwon
OJ
,
Kang
E
,
Choi
JW
,
Kim
SW
,
Yun
CO
. 
Therapeutic targeting of chitosan-PEG-folate-complexed oncolytic adenovirus for active and systemic cancer gene therapy
.
J Control Release
2013
;
169
:
257
65
.
30.
Kim
PH
,
Sohn
JH
,
Choi
JW
,
Jung
Y
,
Kim
SW
,
Haam
S
, et al
Active targeting and safety profile of PEG-modified adenovirus conjugated with herceptin
.
Biomaterials
2011
;
32
:
2314
26
.
31.
Lachenmayer
A
,
Alsinet
C
,
Savic
R
,
Cabellos
L
,
Toffanin
S
,
Hoshida
Y
, et al
Wnt-pathway activation in two molecular classes of hepatocellular carcinoma and experimental modulation by sorafenib
.
Clin Cancer Res
2012
;
18
:
4997
5007
.
32.
Graham
K
,
Unger
E
. 
Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment
.
Int J Nanomed
2018
;
13
:
6049
58
.
33.
Colombo
M
. 
Screening for cancer in viral hepatitis
.
Clin Liver Dis
2001
;
5
:
109
22
.
34.
Fei
XF
,
Zhang
QB
,
Dong
J
,
Diao
Y
,
Wang
ZM
,
Li
RJ
, et al
Development of clinically relevant orthotopic xenograft mouse model of metastatic lung cancer and glioblastoma through surgical tumor tissues injection with trocar
.
J Exp Clin Cancer Res
2010
;
29
:
84
.
35.
Carlisle
R
,
Choi
J
,
Bazan-Peregrino
M
,
Laga
R
,
Subr
V
,
Kostka
L
, et al
Enhanced tumor uptake and penetration of virotherapy using polymer stealthing and focused ultrasound
.
J Natl Cancer Inst
2013
;
105
:
1701
10
.
36.
Lopez
JB
,
Balasegaram
M
,
Thambyrajah
V
,
Timor
J
. 
The value of liver function tests in hepatocellular carcinoma
.
Malays J Pathol
1996
;
18
:
95
9
.
37.
Sasportas
LS
,
Kasmieh
R
,
Wakimoto
H
,
Hingtgen
S
,
van de Water
JA
,
Mohapatra
G
, et al
Assessment of therapeutic efficacy and fate of engineered human mesenchymal stem cells for cancer therapy
.
Proc Natl Acad Sci U S A
2009
;
106
:
4822
7
.
38.
Yong
RL
,
Shinojima
N
,
Fueyo
J
,
Gumin
J
,
Vecil
GG
,
Marini
FC
, et al
Human bone marrow-derived mesenchymal stem cells for intravascular delivery of oncolytic adenovirus Delta24-RGD to human gliomas
.
Cancer Res
2009
;
69
:
8932
40
.
39.
Knaan-Shanzer
S
,
van de Watering
MJ
,
van der Velde
I
,
Goncalves
MA
,
Valerio
D
,
de Vries
AA
. 
Endowing human adenovirus serotype 5 vectors with fiber domains of species B greatly enhances gene transfer into human mesenchymal stem cells
.
Stem Cells
2005
;
23
:
1598
607
.
40.
Giles
RH
,
van Es
JH
,
Clevers
H
. 
Caught up in a Wnt storm: Wnt signaling in cancer
.
Biochim Biophys Acta
2003
;
1653
:
1
24
.
41.
Vilchez
V
,
Turcios
L
,
Marti
F
,
Gedaly
R
. 
Targeting Wnt/β-catenin pathway in hepatocellular carcinoma treatment
.
World J Gastroenterol
2016
;
22
:
823
32
.
42.
Nejak-Bowen
KN
,
Monga
SP
. 
Beta-catenin signaling, liver regeneration and hepatocellular cancer: sorting the good from the bad
.
Semin Cancer Biol
2011
;
21
:
44
58
.
43.
Shayakhmetov
DM
,
Lieber
A
. 
Dependence of adenovirus infectivity on length of the fiber shaft domain
.
J Virol
2000
;
74
:
10274
86
.
44.
Alfano
AL
,
Nicola Candia
A
,
Cuneo
N
,
Guttlein
LN
,
Soderini
A
,
Rotondaro
C
, et al
Oncolytic adenovirus-loaded menstrual blood stem cells overcome the blockade of viral activity exerted by ovarian cancer ascites
.
Mol Ther Oncolytics
2017
;
6
:
31
44
.
45.
Dembinski
JL
,
Spaeth
EL
,
Fueyo
J
,
Gomez-Manzano
C
,
Studeny
M
,
Andreeff
M
, et al
Reduction of nontarget infection and systemic toxicity by targeted delivery of conditionally replicating viruses transported in mesenchymal stem cells
.
Cancer Gene Ther
2010
;
17
:
289
97
.
46.
Sundin
M
,
D'Arcy
P
,
Johansson
CC
,
Barrett
AJ
,
Lonnies
H
,
Sundberg
B
, et al
Multipotent mesenchymal stromal cells express FoxP3: a marker for the immunosuppressive capacity?
J Immunother
2011
;
34
:
336
42
.
47.
Rangatia
J
,
Vangala
RK
,
Treiber
N
,
Zhang
P
,
Radomska
H
,
Tenen
DG
, et al
Downregulation of c-Jun expression by transcription factor C/EBPalpha is critical for granulocytic lineage commitment
.
Mol Cell Biol
2002
;
22
:
8681
94
.
48.
Jager
L
,
Hausl
MA
,
Rauschhuber
C
,
Wolf
NM
,
Kay
MA
,
Ehrhardt
A
. 
A rapid protocol for construction and production of high-capacity adenoviral vectors
.
Nat Protoc
2009
;
4
:
547
64
.
49.
Stuckey
DW
,
Shah
K
. 
Stem cell-based therapies for cancer treatment: separating hope from hype
.
Nat Rev Cancer
2014
;
14
:
683
91
.
50.
Na
Y
,
Choi
JW
,
Kasala
D
,
Hong
J
,
Oh
E
,
Li
Y
, et al
Potent antitumor effect of neurotensin receptor-targeted oncolytic adenovirus co-expressing decorin and Wnt antagonist in an orthotopic pancreatic tumor model
.
J Control Release
2015
;
220
:
766
82
.
51.
Charo
RA
,
Sipp
D
. 
Rejuvenating regenerative medicine regulation
.
N Engl J Med
2018
;
378
:
504
5
.