Therapeutic potential of bone marrow-derived mesenchymal stem cells producing pigment epithelium-derived factor in lung carcinoma

  • Authors:
    • Qiaoling Chen
    • Ping Cheng
    • Tao Yin
    • Hong He
    • Li Yang
    • Yuquan Wei
    • Xiancheng Chen
  • View Affiliations

  • Published online on: June 6, 2012     https://doi.org/10.3892/ijmm.2012.1015
  • Pages: 527-534
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Specific and efficient gene delivery to target cells and the subsequent expression of the RNA and protein is crucial to the success of gene-based therapy for cancer. Mesenchymal stem cells (MSCs) represent novel and efficient tools for delivery of therapeutic agents to tumor cells. In this study, we evaluated the potential of bone marrow-derived mesenchymal stem cells, genetically modified to express pigment epithelium-derived factor (PEDF) for the treatment of Lewis lung carcinoma (LLC). MSCs derived from murine bone marrow were efficiently engineered to express human PEDF by adenoviral transduction, and the expression and bioactivity of the transgenic protein from engineered MSCs were confirmed in vitro. Animal experiments showed that the systemic administration of MSCs treated with PEDF dramatically reduced the growth of LLC tumors and significantly prolonged survival. Immunohistochemistry analysis of the tumors from MSC-PEDF-treated animals indicated an increase in apoptosis and a decrease in microvessel density. ELISA showed that the group of MSCs treated with PEDF had relatively higher expression levels of PEDF in tumor tissue and lower levels in serum compared with the free Ad-PEDF group. These results suggest that MSCs have potential use as effective delivery of vehicles for therapeutic genes in the treatment of LLC.

Introduction

Lung cancer is one of the most common malignant tumors in humans and has high morbidity and mortality. Worldwide, over 1.3 million people are diagnosed each year with lung cancer, with over 1.1 million deaths (1,2). Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, accounting for 85% of cases. The prognosis of patients with NSCLC is extremely poor because most patients are diagnosed in an advanced stage with local or distant metastases. Surgical treatment is reserved only for patients with early stage disease. Radiotherapy and chemotherapy are not specific to cancer cells and produce severe side effects, including bone marrow suppression, renal impairment and second cancer occurrence (3,4). Therefore, development of new therapeutic strategies is an urgent need for the treatment of NSCLC.

Angiogenesis, the outgrowth of new blood vessels from pre-existing ones, plays an essential role in tumor growth and metastasis (5). A positive association between tumor angiogenesis and tumor aggressiveness in carcinoma of the breast, lung, prostate, malignant melanoma and other solid tumors has been confirmed by many clinical studies (6). Thus, depriving a tumor of its vascular supply with anti-angiogenic agents is a promising approach against tumors (7). Pigment epithelium-derived factor (PEDF), a 50 kDa secreted glycoprotein that belongs to the non-inhibitory serpin group, is a potent endogenous inhibitor of angiogenesis (8,9). PEDF exerts its antitumor effects via antiangiogenesis, tumor differentiation, and direct tumor suppression by apoptosis (8). Much focus on PEDF as a promising therapeutic target in cancer has stemmed from its potent antiangiogenic activity, which has shown to be more effective than any other known endogenous angiogenic inhibitor, including angiostatin, thrombospondin-1, and endostatin (10). It has been demonstrated that PEDF can inhibit the proliferation and migration of endothelial cells and can induce apoptosis (8). A number of studies showed inverse correlation between PEDF levels, grade and metastatic potential of prostate cancer (11), pancreatic cancer (12), glioma (13), cervical carcinoma (14) as well as lung carcinoma (15).

Marrow-derived stromal cells (MSCs), also known as mesenchymal stem cells or nonhematopoietic progenitor cells, are precursors that have the potential to differentiate along osteogenic, adipogenic and chondrogenic lineages when placed in the appropriate environments. This differentiation plasticity makes MSCs the ultimate candidates for future utilization in cell therapy and tissue regeneration (16,17). Furthermore, MSCs expressing transgenes maintained long-term expression (up to 6 months) in vivo because of their hypoimmunogenic properties and production of immunosuppressive molecules (18,19). The tumor microenvironment is known to provide a preferential niche for MSCs homing and survival. As MSCs have the capacity to home to the tumor microenvironment, they may be promising tools for the selective delivery of antineoplastic drugs to tumors (20,21).

In this study, we developed a novel strategy of tumor gene therapy in which MSCs loaded with recombinant adenoviruses expressing PEDF could effectively suppress tumor growth and prolong the lifespan through inhibiting angiogenesis in a mouse model, indicating that MSCs may be employed as an effective carrier for tumor gene therapy.

Materials and methods

Cell lines and culture

Lewis lung carcinoma (LL/2) and human embryonic kidney 293 cell lines (purchased from American Type Culture Collection, ATCC, Rockville, MD) were maintained in Dulbecco’s modified Eagle’s medium (Gibco-BRL, Grand Island, NY) supplemented with 10% heat-inactivated fetal bovine serum (Gibco, Auckland, NZ), 2 mM L-glutamine and 100 μg/ml of Amikacin. Human umbilical vein endothelial cells (HUVECs) were isolated from human umbilical cord veins as previously described (22), and grown in EBM-2 medium with SingleQuots (Lonza) containing VEGF and other growth factors. HUVECs at passages 2 to 6 were used for all experiments.

Isolation and culture of marrow MSCs

Female C57BL/6 mice 4- to 6-week-old were sacrificed, femurs and tibias were isolated, and whole bone marrow was retrieved by flushing these bones with low-glucose DMEM (Gibco), supplemented with 10% FBS and 50 U/ml penicillin, 50 μg/ml streptomycin. All of these bone marrow cells were then cultured at 37°C in a 5% CO2 and a 95% humidified atmosphere. Two days later, the nonadherent hematopoietic cells were discarded and the adherent MSCs were preserved for further expansion. The medium was changed twice/week and the MSCs were cultured for 14-days before first passage. Cells of 4–5 passages were used in the experiments (23). MSCs have been proved to express several cell lineage-specific antigens (24). Therefore, the plate-adhering cells were further sorted by flow cytometry using CD34, CD44, CD45, CD73, CD90 and CD105 (BD Biosciences) for phenotype characteristics of MSCs. Isotypic control analyses were conducted in parallel. Flow cytometric analysis was performed on a BD Biosciences FACSCalibur flow cytometer, and data were analyzed with the CellQuest Pro software (BD Immunocytometry Systems).

Adenoviral transduction of MSCs

The adenoviruses were created using the AdEasy system. The viruses were amplified in HEK293 cells and purified on CsCl gradients according to standard methods (25). When the confluence of proliferating MSCs reached or surpassed ∼90%, the cells were transduced with recombinant adenovirus at a multiplicity of infection (MOI) of 1,500 for 4 h. MSCs were also infected with adenovirus-LacZ (Ad-LacZ) at an MOI of 1,500 as a control.

Verification of expressed PEDF from MSCs-PEDF in vitro

Western blot analysis was performed as described previously (26). Briefly, MSCs were infected with adenoviruses for 4 h and then the virus-containing medium was removed and additionally incubated in low-serum medium (low-glucose DMEM containing 2% FBS). After a further 48 h of incubation, the conditioned media was collected. The secreted proteins in the supernatants of the culture were precipitated by TCA-DOC/acetone and the western blotting assay was performed using a standard method with a mouse anti-human PEDF monoclonal antibody (R&D Systems, Boston, MA, USA) and a biotinylated secondary antibody, and the bands were detected using an enhanced chemiluminescence detection system (Pierce, Rockford, IL, USA). The concentration of the PEDF secreted in the culture supernatants was detected using a sandwich enzyme-linked immunosorbent assay (ELISA) kit for the human PEDF protein (Groundwork Biotechnology Diagnosticate Ltd., San Diego, CA, USA) following the manufacturer’s protocol.

Tube formation assay

The tube formation assay was conducted as described previously (27) to evaluate the antiangiogenic activity of PEDF produced by MSCs-PEDF. Each well of a 96-well plate was coated with 50 μl Matrigel (BD Biosciences). After polymerization at 37°C for 30 min, HUVECs (2×104/well) were suspended in 200 μl of the conditioned media derived from MSCs, MSCs-LacZ, MSCs-PEDF respectively and seeded onto the Matrigel. Six hours later, cells were viewed and photographed under an inverted microscope with a digital camera. The tubes of five fields were counted and the data were averaged.

Transwell invasion assay in vitro

To determine whether PEDF secreted by MSCs-PEDF can inhibit HUVEC invasion, a Transwell invasion assay was performed as previously described (28). Briefly, the filter of the Transwell chamber (Millipore) was coated with 50 μl Matrigel (BD Biosciences). After Matrigel polymerization, HUVEC (2×104/well) were suspended in 200 μl of the conditioned media derived from MSCs, MSCs-LacZ, MSCs-PEDF respectively and loaded in the upper chamber. The lower well of the Transwell plate was filled with 600 μl of EBM-2 medium containing various growth factors. Cells were allowed to migrate for 24 h at 37°C. Non-migrated cells were scraped with a cotton swab, and migrated cells were fixed with 100% methanol and stained with 0.05% crystal violet. The number of cells that had migrated to the lower side of the filter was quantified by manual counting under a light microscope with five fields (x100). All assays were conducted in triplicate.

Alginate encapsulation assay

An alginate-encapsulated tumor cell assay was conducted as previously described (26). Alginate beads were formed with about 1×105 LLC cells per bead and implanted s.c. into both dorsal sides of C57BL/6 mice. Then the mice were injected with PBS, MSCs-LacZ, MSCs-PEDF or free Ad-PEDF via the tail vein on Day 2 for once. Twelve days after the beads were implanted, the mice were injected intravenously with a dose at 100 mg/kg FITC-dextran solution (Sigma) in 100 μl. Alginate beads were removed and photographed with a digital camera within 20 min after FITC-dextran administration. The uptake of FITC-dextran was quantified against a calibration curve of FITC-dextran.

PEDF-expressing MSCs reduce subcutaneous tumor growth in vivo

Female syngeneic C57BL/6 mice 6–8 weeks of age were purchased from the West China Experimental Animal Center of Sichuan University (Sichuan, China) and maintained in a dedicated pathogen-free environment. All animal procedures were approved by the Institute’s Animal Care and Use Committee. Mice were implanted s.c. in the right flank with LLC cells at 5×105/mouse. When tumor diameters reached 3 mm, mice were randomly divided into four groups and i.v. injected with PBS, 108 plaque forming units (PFU) of Ad-PEDF, 5×105 MSCs-LacZ or 5×105 MSCs-PEDF at 4 days intervals for two times. Tumor growth and survival rate were monitored every 3 days by caliper. Tumor volumes were determined at various time points using the formula: width2 × length × 0.52. Cohorts of mice (n=8) from each group were sacrificed at Day-30 to determine the effect of therapy and remaining mice (n=10) were monitored for long-term survival. The duration of survival was recorded when the mouse died or had to be sacrificed secondary to tumor diameter >20 mm, tumor ulceration or bleeding. The difference in survival was determined by a log-rank test.

Immunofluorescence staining of tumor vessels

To determine the degree of tumor-induced angiogenesis, frozen sections were prepared from the tumor tissue 30 days after inoculation. The procedure of immunofluorescence staining was done as described previously (29). Briefly, sections were probed with a monoclonal anti-CD31 antibody (BD Biosciences) and nuclei were stained with DAPI. The sections were viewed at low magnification (x40) to identify the most vascular-rich area (hot spot) and 5 non-overlapping fields were selected. Microvessel counting was performed at a high-power field (×200) under fluorescence microscopy and data were expressed as the mean number of CD31-positive vessels per field from three sections in each tumor.

TUNEL assay for in situ apoptosis

To determine the degree of apoptosis, terminal dUTP nick-end labeling (TUNEL) staining was performed using an in situ cell death detection kit (Promega) following the manufacturer’s protocol. Tumor species were prepared as described above. Three tumors from each group were analyzed. The number of apoptotic cells was quantified in 5 randomly selected fields at ×200 magnifications using a fluorescence microscope.

ELISA for expressed PEDF in serum and tumor tissue

Serum samples and fresh tumor tissues were harvested and detected using ELISA (Groundwork Biotechnology Diagnosticate, Ltd.) in triplicate for the tendencies of PEDF levels at 7, 14 and 21 days after i.v. administration of treatment. Non-necrotic fresh tumor tissues were cut into small pieces, lysed in RIPA buffer on ice, passed through a sieve (BellCo Glass), and evaluated for the local production of PEDF as ng/mg.

Statistical analysis

All values are presented as means ± SEM (standard error of the mean). SPSS 17.0 was used for statistical analysis. Significance was evaluated using one-way ANOVA, and P<0.05 was considered significant differences. The survival rates were compared by means of the log-rank test.

Results

MSCs isolation, genetic modification and confirmation of PEDF expression in vitro

The adherent cells have a typical spindle-like morphology (Fig. 1B). Analysis of immunophenotype showed that isolated BM-derived cells shared classical immunophenotype of MSCs, including positivity for CD44, CD73, CD90, CD105, but negativity for CD34, CD45 (Fig. 1A). Thus, as the data showed, we successfully obtained abundant MSCs from mouse bone marrow.

After BMSCs were cultured to reach 90% confluence and incubated with adenoviruses at a MOI of 1,500 for 4 h, the secreted PEDF in the supernatants of conditioned media was verified by western blotting. As shown in Fig. 1C, a specific 50 kDa band was observed only in the supernatants from Ad-PEDF-transduced MSCs, which indicated that PEDF was appropriately synthesized and processed. PEDF levels in the collected media (n=3) were determined utilizing a sandwich enzyme-linked immunosorbent assay (ELISA) kit for the human PEDF protein (Groundwork Biotechnology Diagnosticate Ltd.). It showed that PEDF was produced in vitro in great amounts for 48 h, with a concentration as high as 78.8±4.8 ng/ml (Fig. 1D).

Bioactivity of PEDF produced by PEDF-transduced MSCs in vitro

HUVECs when seeded on Matrigel become elongated and form capillary-like structures mimicking the in vivo neoangiogenesis process (30). We used this assay to examine the antiangiogenic activity of PEDF expressed by MSCs-PEDF. As shown in Fig. 2A, HUVECs plated on the surface of Matrigel formed capillary-like structures in the MSCs and MSCs-LacZ groups after 6 h, however, treatment with the conditioned media (CM) from MSCs-PEDF dramatically blocked the tube formation (P<0.01). We also conducted Transwell invasion assays to evaluate the ability of HUVECs to pass through the Matrigel and membrane barrier of the Transwell in the presence of the CM derived from MSCs, MSCs-LacZ and MSCs-PEDF. Data showed that the CM from the MSCs-PEDF significantly inhibited the invasion properties of endothelial cells (Fig. 2B) (P<0.01).

Therapeutic effect of MSCs-PEDF on LLC cell growth in vivo

To learn if the PEDF secreted in vivo by PEDF gene-modified MSCs can engender a beneficial effect against LLC cells in vivo, we implanted isogenic C57BL/6 mice with LLC cells s.c. and treated with PBS, 108 PFU of Ad-PEDF, 5×105 MSCs-LacZ or 5×105 MSCs-PEDF at 4 days intervals for two times i.v. Cohorts of mice (n=8) from each group were sacrificed at Day-30 to determine the effect of therapy and remaining mice (n=10) were monitored for long-term survival. As shown in Fig. 3A, the tumor volume in the MSCs-PEDF group was dramatically smaller compared with that of the PBS group (1038.8±139.0 vs. 2897.7±274.9 mm3, ANOVA; P<0.01). By contrast, the growth rate of tumors was not affected by administration of free Ad-PEDF (2622.8±359.1 mm3) and MSCs transduced with Ad-LacZ (3019.2±360.9 mm3). These data suggest that intravenous administration of MSC-PEDF cells inhibits the growth of tumors, whereas systemically injected free Ad-PEDF or MSC-LacZ cells does not.

Moreover, MSCs producing PEDF was found to significantly increase the survival of mice. All animals in the control groups died by Day 42, whereas 40% of mice treated with MSC-PEDF survived beyond that time (P<0.01). There was no significant increase in survival of mice treated with MSC transduced with Ad-LacZ or free Ad-PEDF (Fig. 3B).

Inhibition of angiogenesis in vivo

Immunofluorescence anti-CD31 staining was performed to evaluate the consequence of anti-angiogenesis therapy. The most highly vascularized area of each tumor section was identified on low power and five high-powered fields were counted in this area of greatest vessel density. The tumor tissue from MSCs-PEDF-treated mice showed apparently decreased microvessel density compared with the other three control groups (Fig. 4A). The inhibition of angiogenesis was confirmed in the alginate-encapsulated tumor cell assay. The treated tumor exhibited relatively little vascularity (Fig. 4B). Vascularization of beads over 12 days can then be measured by uptake of FITC-dextran into beads. Vascularization of alginate beads was apparently reduced, and FITC-dextran uptake was decreased in MSCs-PEDF-treated mice compared with that in controls (ANOVA; P<0.01).

Induction of apoptosis in vivo

The TUNEL assay was performed to detect tumor cell apoptosis to further investigate the role of MSCs-PEDF treatment in tumors in vivo. As shown in Fig. 5, MSCs-PEDF-treated tumors showed significantly more apoptotic cells (with green nuclei) than tumors from the PBS, Ad-PEDF or MSCs-LacZ-treated groups. The apoptosis index was also significantly higher in the MSCs-PEDF-treated group compared with the controls (ANOVA; P<0.01). The inhibition of angiogenesis and the consequent induction of apoptosis underlie the mechanism of tumor volume decrease and the lifespan prolongation in tumor-bearing mice.

ELISA for expressed PEDF in serum and tumor tissue

The systemic and local (serum and intratumoral) expression tendencies of PEDF at indicated points by ELISA are shown in Fig. 6. In the MSCs-PEDF group, the serum levels of PEDF were ∼38 ng/ml on Day 7, and declined to ∼26 ng/ml after that; meanwhile, the intratumoral levels of PEDF rose from ∼168 ng/mg (7 days) to ∼253 ng/mg (14 days), and then slightly declined to ∼238 ng/mg in the 21 days, ∼7-fold vs. the intratumoral level in the free Ad-PEDF group (32 ng/mg). In free Ad-PEDF group, the levels of PEDF, diminished quickly from ∼60 to 25 ng/ml in serum and from ∼50 to ∼32 ng/mg intratumorally. Other controls including PBS, MSCs-LacZ hardly led to any elevation of serous or intratumoral levels of PEDF.

Discussion

In this study, we demonstrated that systemically delivered MSCs loaded with the PEDF gene could preferentially home to tumor loci and induce antiangiogenesis as well as apoptosis of cancer cells, resulting in the inhibition of tumor growth and prolongation of lifespan in the mouse Lewis lung carcinoma (LLC) model.

The choice of PEDF over other cytokines for antitumor effects on LLC in our studies is based on its potent antiangiogenic activity, which has shown to be more effective than any other known endogenous angiogenic inhibitor, including angiostatin, thrombospondin-1, and endostatin. Moreover, PEDF can also exert its antitumor activity via tumor differentiation and direct tumor suppression by apoptosis. It has been reported that PEDF can inhibit several types of tumors including lung carcinomas (8). Purified recombinant PEDF and PEDF-based viral vectors have been investigated in a lot of studies. Nevertheless, a suboptimal half-life in plasma reduces their possible therapeutic effects (14,25). To overcome this shortcoming, a novel and more effective vehicle is needed. In this study, we demonstrated that the engineered MSCs can secret high-level PEDF and the conditioned media derived from these engineered MSCs was capable of inhibiting tube formation and migration of human umbilical vein endothelial cells in vitro.

MSCs are attractive as a cellular vehicle for delivery of antitumoral agents in tumor gene therapy, since they possess several unique biological features. First, the poorer immunogenicity of MSCs enables them to survive for hundreds of days in the host, which can offer a relatively long effect for chronic disease (31). Second, the capacity of MSCs homing to malignant tissues can offer a higher concentration of antineoplastic drugs in the local tumor sites and lower side effect on other healthy organs (21,31). Last, MSCs are easy to harvest, isolate, expand and to gene modify in vitro, which means the offer convenience and cost effectiveness in future preclinical or clinical applications.

It has been shown that the migratory ability of MSCs was not affected by viral transduction in vitro and in vivo (21,32,33). As a relatively perfect candidate of cellular vehicles used in our study, MSCs could protect PEDF to escape from being destroyed due to entering the circulatory system and being accompanied by potential adverse effects owing to interfering spontaneously with other systems and thereby improving its pharmacokinetics. MSCs were capable of migrating to the tumor sites after intravenous infusion; by contrast, free Ad-PEDF lacks this attribute (32,33). In our study, consecutive administration of free Ad-PEDF could only partly retard the tumor growth to some extent in the midway stage; yet, with potential systemic side effects to the hosts. This also has no therapeutic effect in the ultimate stage. This phenomenon could be partly attributed to the temporary high serum yet low intratumor levels of PEDF as a result of nonselective expression and subsequent elimination of free Ad-PEDF.

Our results suggest that the antitumor activity of MSC-PEDF cells is associated not with the serum concentration of PEDF but rather with the engraftment of MSCs in tumors and the local effects of the PEDF they produce on malignant cells. Therefore, selective engraftment of MSC in tumors is an important issue for further clinical development of this method. Despite the antitumor effects of MSCs-PEDF observed, there was an absence of higher systemic levels of the transgenic protein, suggesting the local effects of MSCs-produced PEDF in the tumor microenvironment. These findings originally suggest that i.v. infused MSCs can circulate over the lung vascular bed and migrate into s.c. growing tumors, supporting the concept that MSCs have the ability of homing into tumor sites. Therefore, therapy with MSCs-PEDF cells could be used to increase PEDF expression in tumors and surrounding tissues and to control the growth of malignant cells. In fact, the potential clinical application of MSCs-PEDF cells could be broad, because many types of malignancies are sensitive to the antiproliferative or proapoptotic effects of PEDF in vitro. The data indicate that systemic administration of MSCs-PEDF led to a pronounced growth inhibition of established subcutaneous LLC and prolonged survival of mice.

In conclusion, our investigation reveals the potential of genetically modified MSC expressing PEDF for Lewis lung cancer. This may prefigure an integrated cellular and molecular therapeutic weapon targeted against advanced human malignancies. However, the same approach could be explored in the delivery of other agents with antitumor activities.

Acknowledgements

The authors thank members of the State Key Laboratory of Biotherapy for helpful discussions. This study was supported by the National Natural Science Foundation (30973507) and the National 973 Basic Research Program of China (2010CB529900 and 2010CB529906).

References

1. 

J FerlayP AutierM BoniolM HeanueM ColombetP BoyleEstimates of the cancer incidence and mortality in Europe in 2006Ann Oncol18581592200710.1093/annonc/mdl49817287242

2. 

DM ParkinF BrayJ FerlayP PisaniGlobal cancer statistics, 2002CA Cancer J Clin5574108200510.3322/canjclin.55.2.74

3. 

DS EttingerW AkerleyG BeplerNon-small cell lung cancerJ Natl Compr Cancer Netw8740801201020679538

4. 

A JemalR SiegelE WardY HaoJ XuMJ ThunCancer statistics, 2009CA Cancer J Clin59225249200910.3322/caac.20006

5. 

J FolkmanY ShingAngiogenesisJ Biol Chem26710931109341992

6. 

VW van HinsberghA CollenP KoolwijkAngiogenesis and anti-angiogenesis: perspectives for the treatment of solid tumorsAnn Oncol10Suppl 4S60S63199910436787

7. 

CR DassTM TranPF ChoongAngiogenesis inhibitors and the need for anti-angiogenic therapeuticsJ Dent Res86927936200710.1177/15440591070860100517890668

8. 

ET EkCR DassPF ChoongPigment epithelium-derived factor: a multimodal tumor inhibitorMol Cancer Ther516411646200610.1158/1535-7163.MCT-06-010716891449

9. 

FR SteeleGJ ChaderLV JohnsonJ Tombran-TinkPigment epithelium-derived factor: neurotrophic activity and identification as a member of the serine protease inhibitor gene familyProc Natl Acad Sci USA9015261530199310.1073/pnas.90.4.15268434014

10. 

DW DawsonOV VolpertP GillisPigment epithelium-derived factor: a potent inhibitor of angiogenesisScience285245248199910.1126/science.285.5425.24510398599

11. 

JA DollVM StellmachNP BouckPigment epithelium-derived factor regulates the vasculature and mass of the prostate and pancreasNat Med9774780200310.1038/nm87012740569

12. 

R HaseM MiyamotoH UeharaPigment epithelium-derived factor gene therapy inhibits human pancreatic cancer in miceClin Cancer Res1187378744200510.1158/1078-0432.CCR-05-132316361561

13. 

M GuanHF YamB SuLoss of pigment epithelium derived factor expression in glioma progressionJ Clin Pathol56277282200310.1136/jcp.56.4.27712663639

14. 

J YangS ChenX HuangGrowth suppression of cervical carcinoma by pigment epithelium-derived factor via anti-angiogenesisCancer Biol Ther9967974201010.4161/cbt.9.12.1163520364117

15. 

L WangV SchmitzA Perez-MediavillaI IzalJ PrietoC QianSuppression of angiogenesis and tumor growth by adenoviral-mediated gene transfer of pigment epithelium-derived factorMol Ther87279200310.1016/S1525-0016(03)00128-X12842430

16. 

BD RoordaA ter ElstWA KampsES de BontBone marrow-derived cells and tumor growth: contribution of bone marrow-derived cells to tumor micro-environments with special focus on mesenchymal stem cellsCrit Rev Oncol Hematol69187198200910.1016/j.critrevonc.2008.06.00418675551

17. 

MF PittengerAM MackaySC BeckMultilineage potential of adult human mesenchymal stem cellsScience284143147199910.1126/science.284.5411.14310102814

18. 

N EliopoulosA Al-KhaldiM CrosatoK LachapelleJ GalipeauA neovascularized organoid derived from retrovirally engineered bone marrow stroma leads to prolonged in vivo systemic delivery of erythropoietin in nonmyeloablated, immunocompetent miceGene Ther10478489200310.1038/sj.gt.3301919

19. 

SH SeoKS KimSH ParkThe effects of mesenchymal stem cells injected via different routes on modified IL-12-mediated antitumor activityGene Ther18488495201110.1038/gt.2010.17021228885

20. 

U GalderisiA GiordanoMG PaggiThe bad and the good of mesenchymal stem cells in cancer: Boosters of tumor growth and vehicles for targeted delivery of anticancer agentsWorld J Stem Cells2512201010.4252/wjsc.v2.i1.521607110

21. 

M StudenyFC MariniRE ChamplinC ZompettaIJ FidlerM AndreeffBone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumorsCancer Res6236033608200212097260

22. 

EA JaffeRL NachmanCG BeckerCR MinickCulture of human endothelial cells derived from umbilical veins. Identification by morphologic and immunologic criteriaJ Clin Invest5227452756197310.1172/JCI1074704355998

23. 

N EliopoulosM FrancoisMN BoivinD MartineauJ GalipeauNeo-organoid of marrow mesenchymal stromal cells secreting interleukin-12 for breast cancer therapyCancer Res6848104818200810.1158/0008-5472.CAN-08-016018559528

24. 

S BobisD JarochaM MajkaMesenchymal stem cells: characteristics and clinical applicationsFolia Histochem Cytobiol44215230200617219716

25. 

LP YangP ChengXC PengAntitumor effect of adenovirus-mediated gene transfer of pigment epithelium-derived factor on mouse B16–F10 melanomaJ Exp Clin Cancer Res2875200919500366

26. 

JY LiuYQ WeiL YangImmunotherapy of tumors with vaccine based on quail homologous vascular endothelial growth factor receptor-2Blood10218151823200310.1182/blood-2002-12-377212750177

27. 

X PangZ YiX ZhangAcetyl-11-keto-beta-boswellic acid inhibits prostate tumor growth by suppressing vascular endothelial growth factor receptor 2-mediated angiogenesisCancer Res6958935900200910.1158/0008-5472.CAN-09-075519567671

28. 

S ZhangZ CaoH TianSKLB1002, a novel potent inhibitor of VEGF receptor 2 signaling, inhibits angiogenesis and tumor growth in vivoClin Cancer Res1744394450201110.1158/1078-0432.CCR-10-310921622720

29. 

KA MohamedaliD KedarP SweeneyThe vascular-targeting fusion toxin VEGF121/rGel inhibits the growth of orthotopic human bladder carcinoma tumorsNeoplasia7912920200510.1593/neo.0529216242074

30. 

G TarabolettiR GiavazziModelling approaches for angiogenesisEur J Cancer40881889200410.1016/j.ejca.2004.01.00215120043

31. 

V FritzC JorgensenMesenchymal stem cells: an emerging tool for cancer targeting and therapyCurr Stem Cell Res Ther33242200810.2174/15748880878348946218220921

32. 

X ChenX LinJ ZhaoA tumor-selective biotherapy with prolonged impact on established metastases based on cytokine gene-engineered MSCsMol Ther16749756200810.1038/mt.2008.318362930

33. 

Y GaoA YaoW ZhangHuman mesenchymal stem cells overexpressing pigment epithelium-derived factor inhibit hepatocellular carcinoma in nude miceOncogene2927842794201010.1038/onc.2010.38

Related Articles

Journal Cover

September 2012
Volume 30 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Q, Cheng P, Yin T, He H, Yang L, Wei Y and Chen X: Therapeutic potential of bone marrow-derived mesenchymal stem cells producing pigment epithelium-derived factor in lung carcinoma. Int J Mol Med 30: 527-534, 2012
APA
Chen, Q., Cheng, P., Yin, T., He, H., Yang, L., Wei, Y., & Chen, X. (2012). Therapeutic potential of bone marrow-derived mesenchymal stem cells producing pigment epithelium-derived factor in lung carcinoma. International Journal of Molecular Medicine, 30, 527-534. https://doi.org/10.3892/ijmm.2012.1015
MLA
Chen, Q., Cheng, P., Yin, T., He, H., Yang, L., Wei, Y., Chen, X."Therapeutic potential of bone marrow-derived mesenchymal stem cells producing pigment epithelium-derived factor in lung carcinoma". International Journal of Molecular Medicine 30.3 (2012): 527-534.
Chicago
Chen, Q., Cheng, P., Yin, T., He, H., Yang, L., Wei, Y., Chen, X."Therapeutic potential of bone marrow-derived mesenchymal stem cells producing pigment epithelium-derived factor in lung carcinoma". International Journal of Molecular Medicine 30, no. 3 (2012): 527-534. https://doi.org/10.3892/ijmm.2012.1015