Antitumor effect of FP3 in a breast cancer xenograft model

  • Authors:
    • Huanrong Lan
    • Lingzhi Zheng
    • Ketao Jin
    • Lisong Teng
  • View Affiliations

  • Published online on: October 26, 2012     https://doi.org/10.3892/etm.2012.773
  • Pages: 85-88
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

FP3 is a novel vascular endothelial growth factor (VEGF) blocker proposed to have antiangiogenic properties. Previous studies revealed that FP3 is a new promising agent for treating human choroidal neovascularization (CNV)‑associated age‑associated macular degeneration (AMD) and has an inhibitory effect on VEGF‑mediated proliferation and migration of human umbilical vein endothelial cells and VEGF‑mediated vessel sprouting of the rat aortic ring in vitro. Previous studies have also revealed that FP3 has antitumor effects and antiangiogenic effects in a non‑small cell lung cancer cell line (A549), as well as in patient‑derived tumor tissue xenograft models of gastric cancer and colon carcinoma with lymphatic and hepatic metastases in nude mice. In the present study, the antitumor effect of FP3 in an MDA‑MB‑231 breast cancer xenograft model was investigated. Treatment with FP3 for 3 weeks significantly suppressed xenograft growth and this inhibition was associated with a significant decrease in angiogenesis and direct inhibition of tumor cells. The results of the present study indicate that FP3 inhibits breast cancer tumor growth via the indirect inhibition of angiogenesis as well as a direct effect on tumor cells.

Introduction

Angiogenesis is the process of new blood vessel formation. It is an important process in the growth of malignant tumors as solid tumors must develop an angiogenic phenotype which promotes the establishment of an expanding vascular network for the delivery of oxygen and other nutrients (1). The predominant regulator of tumor angiogenesis is vascular endothelial growth factor (VEGF) (2,3) which is the only angiogenic factor known to be present throughout the entire tumor lifecycle (2,4). VEGF promotes endothelial cell proliferation, migration and survival in support of tumor angiogenesis. In addition, VEGF is a potent stimulator of vessel permeability, having originally been recognized for its function as a vascular permeability factor (5,6). Due to its fundamental role in tumor angiogenesis, VEGF serves as a logical target for antiangiogenic cancer therapy. Breast cancer is the most common neoplasm in women (7). Tumor angiogenesis is essential for the growth and spread of breast cancer cells. There are at least 6 different angiogenic growth factors associated with tumor angiogenesis in breast cancer. The major mediator of tumor angiogenesis is VEGF (8). VEGF expression is increased in a number of tumor types, including breast cancer (9). Overexpression of VEGF is associated with a poor prognosis for patients with breast cancer (10). In addition to its prognostic role, VEGF is also a validated target in the treatment of this disease. Recently, various antiangiogenic agents have shown efficacy in the treatment of breast cancer (11,12).

FP3 (also known as KH902 or KH903) is an engineered protein which contains the extracellular domain 2 of VEGF receptor 1 (Flt-1) and extracellular domains 3 and 4 of VEGF receptor 2 (Flk-1, KDR) fused to the Fc region of human immunoglobulin G1 (11,13). Previous studies revealed that FP3 has promise as a local antiangiogenic treatment for human choroidal neovascularization (CNV)-associated age-associated macular degeneration (AMD) (11,1416). In subsequent studies, it has been demonstrated that FP3 has an inhibitory effect on the VEGF-mediated proliferation and migration of human umbilical vein endothelial cells and VEGF-mediated vessel sprouting of the rat aortic ring in vitro(13). Previous studies also revealed that FP3 has antitumor effects and anti-angiogenic effects in a non-small cell lung cancer cell line (A549) (13) and patient-derived tumor tissue xenograft models of gastric cancer (17), as well as in colon carcinoma with lymphatic and hepatic metastases in nude mice (18).

It is unknown whether FP3 has an antitumor effect in breast cancer and what the mechanism behind the potential effect of FP3 would be. For this purpose, the present study was designed to evaluate the potential antitumor effects of FP3 in a breast cancer cell line subcutaneous xenograft model in nude mice and assess the antiangiogenenic effects of FP3 in this model.

Materials and methods

Materials

The MDA-MB-231 human breast cancer cells were purchased from American Type Culture Collection (ATCC, Manassas, VA, USA). RPMI-1640 medium, fetal bovine serum (FBS), penicillin and streptomycin were purchased from Gibco (Grand Island, NY, USA). Methanol, acetic acid, crystal violet and hydrogen peroxide were purchased from Sigma (St. Louis, MO, USA). The antibody against platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31; rat monoclonal, clone MEC 13.3) was purchased from BD Pharmingen (San Diego, CA, USA). Fluorescent (Cy3-conjugated) secondary antibody (goat anti-rat) was purchased from Jackson ImmunoResearch (West Grove, PA, USA). Bevacizumab (Avastin) was purchased from Roche, Inc. (Roche, Nutley, NJ, USA). FP3 was provided as a gift by Kanghong Biotechnology Inc. (Kanghong, Chengdu, China).

Animals

Female BALB/c nude mice (4–6 weeks old) purchased from Slaccas Laboratory Animal Co. (Shanghai, China) were housed in a barrier facility and acclimated to 12-h light-dark cycles for ≥3 days prior to use. The use of experimental animals adhered to the ‘Principles of Laboratory Animal Care’ (NIH publication #85-23, revised in 1985). All experiments were approved by the Institutional Animal Care and Use Committee of Zhejiang University (approval ID: SYXK(ZHE)2005-0072).

Cell culture

MDA-MB-231, a breast cancer cell line, was maintained in RPMI-1640 medium supplemented with 10% FBS, 200 IU/ml penicillin and 200 μg/ml streptomycin. The culture medium was replaced every other day. After reaching confluence, the cells were subcultured.

In vitro MDA-MB-231 cell clonogenic assay

MDA-MB-231 cells were cultured in RPMI-1640 with 10% FBS. Cells in the exponential growth phase were trypsinized, washed and counted. The cells were seeded in triplicate at a density of 2,500 cells per 100-mm dish containing 10 ml complete medium. Cells were treated 2 h after seeding with FP3 (20, 50 or 100 μg/ml) or bevacizumab (50 μg/ml). The control plates received medium only. After 10 days of incubation at 37°C, the medium was drained and colonies were rinsed, fixed with a mixture of methanol and acetic acid (10:1) and stained with 1% crystal violet. The colonies containing >50 cells were counted.

Mice tumor xenografts

For inoculation, ∼1.0×107 MDA-MB-231 cells in 0.2 ml serum-free RPMI-1640 were injected subcutaneously into the right flank of 32 female athymic nude mice. The mice developed visible tumors within 3 weeks of the inoculation. The tumors were allowed to grow to >60 mm3 prior to imaging. Subsequently, the mice were divided into 4 treatment groups containing 8 mice each and the treatments were initiated. An 8-mouse group was used as an implantation and normal saline (NS)-treated negative control.

Mouse drug treatments and tumor growth regression assay

Following the tumor cell implantation and within 2 weeks of inoculation, the mice received 200 μl vehicle (NS), FP3 (2, 6 or 18 mg/kg) or bevacizumab (Avastin; 6 mg/kg) intravenously. The mice were treated with the drugs from 21 days after implantation at the indicated doses twice per week (n=8 for each dose) for 3 weeks. The animals were then sacrificed and the tumors were measured ex vivo with calipers (tumor volume = length × width2 /2).

Immunohistochemical staining

Formalin-fixed, paraffin-embedded 5-μm thick tumor sections were analyzed by immunohistochemical analysis according to the previously described method (18). The sections were baked, deparaffinized in xylene and rehydrated. Antigen retrieval was performed with citrate buffer (pH 6.0) in a microwave at 98°C for 5 min. Following a PBS wash, endogenous peroxidases were quenched in 3% hydrogen peroxide for 20 min. The slides were then blocked with 1% normal goat serum for 20 min at room temperature and incubated with rat anti-mouse PECAM-1 (CD31) polyclonal antibody at a 1:100 dilution. The slides were incubated overnight at 4°C. The following day, the slides were washed several times with PBS and the specimens were incubated for 1 h at room temperature with fluorescent (Cy3-conjugated) secondary antibody (goat anti-rat) diluted (1:200) in PBS. Specimens were rinsed again with PBS and mounted in Vectashield (Vector Laboratories, Burlingame, CA, USA). The tissue sections were examined and digitally photographed using a Zeiss Axiophot fluorescence microscope (Carl Zeiss, Thornwood, NY, USA) equipped with single, dual and triple fluorescence filters and a low-light, externally cooled, three-chip charge-coupled device (CCD) camera (480×640 pixel RGB-color images, CoolCam; SciMeasure Analytical Systems, Atlanta, GA, USA) and saved as TIFF files.

Statistical analysis

Data are presented as the mean ± SEM and were analyzed using SPSS 16.0 software (SPSS, Inc., Chicago, IL, USA). Differences among the means of the groups were determined using one-way ANOVA. P<0.05 was considered to indicate a statistically significant difference.

Results

FP3 significantly inhibits MDA-MB-231 cell proliferation in vitro and blocks tumor growth in vivo

The antiproliferative effect of FP3 on MDA-MB-231 cells in vitro was evaluated. The results demonstrated that FP3 directly inhibited the tumor cells. FP3 reduced MDA-MB-231 cell colony formation by >77%. (Fig. 1)

To begin to evaluate FP3 as an anticancer therapeutic agent for breast cancer and to compare it with other effective agents targeting the VEGF pathway, its ability to block the growth of a breast cancer cell line, MDA-MB-231, in a mouse subcutaneous tumor model was evaluated. Following implantation, tumor cells were allowed to grow for 3 weeks and formed large retroperitoneal tumors >60 mm3. Injections of FP3 (2, 6 and 18 mg/kg body weight), bevacizumab (6 mg/kg body weight) or NS were then administered intravenously, biweekly for 3 weeks, after which the animals were sacrificed and the tumors excised and measured. FP3 significantly inhibited the growth of the tumor xenografts (Fig. 2).

FP3 results in decreased vasculature of tumors

To evaluate the effects of FP3 on tumor-associated angiogenesis, the tumor xenografts were sectioned and immune-stained with an anti-body to PECAM-1 so that the vasculature was visualized. This analysis revealed that the higher doses of FP3 almost completely blocked tumor-associated angiogenesis, with the stunted tumors being largely avascular (Fig. 3D and E). The lowest dose of FP3 (2 mg/kg) was not as effective at inhibiting tumor growth compared to a higher dose (6 or 18 mg/kg). However, the lowest dose of FP3 (2 mg/kg) appeared to be as effective as the higher doses (6 or 18 mg/kg) at blocking tumor-associated angiogenesis (Fig. 3C–E). In contrast to the FP3-treated tumors, the control tumors in the vehicle-treated mice were not only much larger but also had a high vascular density (Fig. 3A).

Discussion

Tumor vessels are considered to be dynamic in terms of the formation of new vessels or angiogenesis. Tumors acquire their vasculature by endothelial cell sprouting, co-option of pre-existing vessels, intussusceptive microvascular growth, postnatal vasculogenesis, glomeruloid angiogenesis or vasculogenic mimicry. It should be emphasized that in the majority of cases, these mechanisms are interlinked, participating simultaneously in physiological and pathological angiogenesis (11). VEGF promotes certain or all of these processes, rendering VEGF a rational target for antiangiogenic drug development (19). Since anti-VEGF approaches act by blocking tumor-associated angiogenesis, which appears to be widely required by numerous different types of tumors, these approaches may prove to be generally useful against a wide variety of cancer types (20).

VEGF expression is increased in a number of tumor types, including breast cancer (9). VEGF-A is highly expressed in numerous tumors of the lung, brain and gastrointestinal and urogenital tracts, as well as in situ and invasive breast cancer (21). There is a positive correlation between VEGF levels and poor clinical outcomes, including patient survival (10). Anti-VEGF treatment inhibits the growth of human breast tumor xenografts in animals (22).

FP3 is a humanized fusion protein which combines ligand binding elements taken from the extracellular domains of VEGF receptors 1 and 2 and the Fc portion of IgG1 and is designed to bind to all forms of VEGF-A (13). In order to further substantiate the antitumor and anti-angiogenesis effects of FP3 in breast cancer, an MDA-MB-231 subcutaneous xenograft model in nude mice was used. The results of the study showed that, in the MDA-MB-231 human breast cancer xenograft model, FP3 effectively inhibited tumor growth (Fig. 2). Treatment with FP3 also resulted in stunted and almost completely avascular tumors (Fig. 3). The antitumor activity of FP3 is most likely mediated by the inhibition of angiogenesis since the microvessel density values in FP3-treated tumors were significantly decreased. The fact that FP3 resembled the well-defined angiogesis inhibitor bevacizumab with regard to tumor growth and microvessel density measurements, is an additional indication of the antiangiogenic activity of FP3.

Whether FP3 has a direct cell-killing or inhibitory effect in MDA-MB-231 cells in vitro was investigated using a clonogenic assay. FP3 was identified to have a direct cell-killing effect on MDA-MB-231 cells (Fig. 1). These results indicated that the inhibitory effect of FP3 on the MDA-MB-231 tumor xenograft model growth may partially result from the inhibition of the tumor cells.

The results of the present study reveal that FP3 has an excellent antitumor effect against breast cancer xenografts and indicate that it may have potential as an effective antiangiogenic agent in the treatment of breast cancer.

Acknowledgements

This study was supported by the State Key Basic Research and Development Program of China (973 Program, Grant No. 2009CB521704), National High-tech Research & Development Program of China (863 Program, Grant No. 2006AA02A245), National Natural Science Foundation of China (Grant No. 81000894), Zhejiang Provincial Natural Science Foundation of China (Grant No. Y12H160045), Zhejiang Provincial Science and Technology Projects (Grant No. 2009C13021, 2011C23087), Zhejiang Provincial Medical and Healthy Science and Technology Projects (Grant No. 2011KYB137), Science Research Fund of Taizhou (Grant No. A102KY09), Science Research Fund of Shaoxing (Grant No. 2011D10013) and Science Research Fund of Zhuji (Grant No. 2011CC7874).

References

1 

Folkman J: Fundamental concepts of the angiogenic process. Curr Mol Med. 3:643–651. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Hicklin DJ and Ellis LM: Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 23:1011–1027. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Ferrara N, Gerber HP and LeCouter J: The biology of VEGF and its receptors. Nat Med. 9:669–676. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Ferrara N: Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev. 25:581–611. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS and Dvorak HF: Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science. 219:983–985. 1983. View Article : Google Scholar

6 

Shibuya M: Structure and function of VEGF/VEGF-receptor system involved in angiogenesis. Cell Struct Funct. 26:25–35. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

8 

Cook KM and Figg WD: Angiogenesis inhibitors: current strategies and future prospects. CA Cancer J Clin. 60:222–243. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Brown LF, Berse B, Jackman RW, Tognazzi K, Guidi AJ, Dvorak HF, Senger DR, Connolly JL and Schnitt SJ: Expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in breast cancer. Hum Pathol. 26:86–91. 1995. View Article : Google Scholar : PubMed/NCBI

10 

Linderholm B, Grankvist K, Wilking N, Johansson M, Tavelin B and Henriksson R: Correlation of vascular endothelial growth factor content with recurrences, survival, and first relapse site in primary node-positive breast carcinoma after adjuvant treatment. J Clin Oncol. 18:1423–1431. 2000.

11 

Teng LS, Jin KT, He KF, Wang HH, Cao J and Yu DC: Advances in combination of antiangiogenic agents targeting VEGF-binding and conventional chemotherapy and radiation for cancer treatment. J Chin Med Assoc. 73:281–288. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Teng LS, Jin KT, He KF, Zhang J, Wang HH and Cao J: Clinical applications of VEGF-trap (aflibercept) in cancer treatment. J Chin Med Assoc. 73:449–456. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Jin K, He K, Teng F, Li G, Wang H, Han N, Xu Z, Cao J, Wu J, Yu D and Teng L: FP3: a novel VEGF blocker with antiangiogenic effects in vitro and antitumour effects in vivo. Clin Transl Oncol. 13:878–884. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Zhang M, Zhang J, Yan M, Li H, Yang C and Yu D: Recombinant anti-vascular endothelial growth factor fusion protein efficiently suppresses choridal neovascularization in monkeys. Mol Vis. 14:37–49. 2008.

15 

Zhang M, Yu D, Yang C, Xia Q, Li W, Liu B and Li H: The pharmacology study of a new recombinant human VEGF receptor-fc fusion protein on experimental choroidal neovascularization. Pharm Res. 26:204–210. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Zhang M, Zhang J, Yan M, Luo D, Zhu W, Kaiser PK and Yu DC; KH902 Phase 1 Study Group: A phase 1 study of KH902, a vascular endothelial growth factor receptor decoy, for exudative age-related macular degeneration. Ophthalmology. 118:672–678. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Jin K, He K, Han N, Li G, Wang H, Xu Z, Jiang H, Zhang J and Teng L: Establishment of a PDTT xenograft model of gastric carcinoma and its application in personalized therapeutic regimen selection. Hepatogastroenterology. 58:1814–1822. 2011.PubMed/NCBI

18 

Jin K, Li G, Cui B, Zhang J, Lan H, Han N, Xie B, Cao F, He K, Wang H, Xu Z, Teng L and Zhu T: Assessment of a novel VEGF targeted agent using patient-derived tumor tissue xenograft models of colon carcinoma with lymphatic and hepatic metastases. PLoS One. 6:e283842011. View Article : Google Scholar : PubMed/NCBI

19 

Huang J, Frischer JS, New T, Kim ES, Serur A, Lee A, Kadenhe-Chiwishe A, Pollyea DA, Yokoi A, Holash J, Yancopoulos GD, Kandel JJ and Yamashiro DJ: TNP-470 promotes initial vascular sprouting in xenograft tumors. Mol Cancer Ther. 3:335–343. 2004.PubMed/NCBI

20 

Holash J, Davis S, Papadopoulos N, Croll SD, Ho L, Russell M, Boland P, Leidich R, Hylton D, Burova E, Ioffe E, Huang T, Radziejewski C, Bailey K, Fandl JP, Daly T, Wiegand SJ, Yancopoulos GD and Rudge JS: VEGF-Trap: a VEGF blocker with potent antitumor effects. Proc Natl Acad Sci USA. 99:11393–11398. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Roskoski R Jr: Vascular endothelial growth factor (VEGF) signaling in tumor progression. Crit Rev Oncol Hematol. 62:179–213. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Borgström P, Gold DP, Hillan KJ and Ferrara N: Importance of VEGF for breast cancer angiogenesis in vivo: implications from intravital microscopy of combination treatments with an anti-VEGF neutralizing monoclonal antibody and doxorubicin. Anticancer Res. 19:4203–4214. 1999.PubMed/NCBI

Related Articles

Journal Cover

January 2013
Volume 5 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lan H, Zheng L, Jin K and Teng L: Antitumor effect of FP3 in a breast cancer xenograft model. Exp Ther Med 5: 85-88, 2013
APA
Lan, H., Zheng, L., Jin, K., & Teng, L. (2013). Antitumor effect of FP3 in a breast cancer xenograft model. Experimental and Therapeutic Medicine, 5, 85-88. https://doi.org/10.3892/etm.2012.773
MLA
Lan, H., Zheng, L., Jin, K., Teng, L."Antitumor effect of FP3 in a breast cancer xenograft model". Experimental and Therapeutic Medicine 5.1 (2013): 85-88.
Chicago
Lan, H., Zheng, L., Jin, K., Teng, L."Antitumor effect of FP3 in a breast cancer xenograft model". Experimental and Therapeutic Medicine 5, no. 1 (2013): 85-88. https://doi.org/10.3892/etm.2012.773