|
|
 |
|
ORIGINAL ARTICLE |
|
Year : 2019 | Volume
: 12
| Issue : 14 | Page : 54-58 |
|
Anti-tumor activity of a recombinant endoglin-MIP3α Fc-fusion protein in mice with hepatocellular carcinoma
Zhi-Hui He, Si-Ru Liu, Xin-Lai Wu, Yong-Hao Huang, Yan Chen, Yi Deng, Pan-Pan Xie, Yan-Da Lu
Tumor Institute, the First Affiliated Hospital of Hainan Medical University, Haikou 571199, P.R. China
Date of Submission | 10-Sep-2019 |
Date of Decision | 27-Oct-2019 |
Date of Acceptance | 08-Nov-2019 |
Date of Web Publication | 03-Dec-2019 |
Correspondence Address: Yan-Da Lu Tumor Institute, the First Affiliated Hospital of Hainan Medical University, Haikou 571199 P.R. China
 Source of Support: None, Conflict of Interest: None
DOI: 10.4103/1995-7645.271980
Objective: To investigate the effects of a recombinant endoglin-macrophage inflammatory protein 3α Fc-fusion protein (EM) vaccine on tumor angiogenesis and growth in mice with H22 hepatocellular carcinoma. Methods: An in vivo hepatoma mouse model was established. Seven days after subcutaneous inoculation of H22 tumor cells, mice were randomly divided into four groups: EM, endoglin Fc-fusion protein, macrophage inflammatory protein 3α Fc-fusion protein, and normal saline groups. Tumor volume and survival rate of mice were studied at 3-day intervals. Microvessel density of the tumors and tumor cell proliferation were detected by immunohistochemistry, and tumor cell apoptosis was detected by TdT-mediated biotinylated-dUTP nick-end label staining. The number of CD11c and CD86 positive dendritic cells were detected by flow cytometry. Results: Compared with the other groups, the tumor volume became smaller, and the survival time was longer in the EM-treated group. Besides, microvessel density and cell proliferation index were significantly lower, while the tumor cell apoptosis index was significantly higher in the EM-treated group. Besides the number of CD11c and CD86 positive dendritic cells in EM- treated mice was larger than that in other groups. Conclusions: EM Fc-fusion protein could effectively inhibit tumor growth through inhibiting endoglin-related tumor angiogenesis and cell proliferation, promoting tumor cell apoptosis, and could induce a certain degree of antitumor immune responses.
Keywords: Endoglin, MIP3α, Fc-fusion protein, Angiogenesis, Hepatocellular carcinoma
How to cite this article: He ZH, Liu SR, Wu XL, Huang YH, Chen Y, Deng Y, Xie PP, Lu YD. Anti-tumor activity of a recombinant endoglin-MIP3α Fc-fusion protein in mice with hepatocellular carcinoma. Asian Pac J Trop Med 2019;12, Suppl S2:54-8 |
How to cite this URL: He ZH, Liu SR, Wu XL, Huang YH, Chen Y, Deng Y, Xie PP, Lu YD. Anti-tumor activity of a recombinant endoglin-MIP3α Fc-fusion protein in mice with hepatocellular carcinoma. Asian Pac J Trop Med [serial online] 2019 [cited 2023 Jun 2];12, Suppl S2:54-8. Available from: https://www.apjtm.org/text.asp?2019/12/14/54/271980 |
Zhi-hui He#, Si-ru Liu#, Xin-lai Wu
These authors contributed equally.
1. Introduction | |  |
Hepatocellular carcinoma is the most common type of liver cancer, causing approximately 1 million deaths annually[1]. In 2012, approximately 50% of new cancer cases occurred in Asia, mostly in China[2]. The primary aetiological factors in these regions are chronic infections by hepatitis viruses and dietary exposure to aflatoxins, a group of mycotoxins that are natural contaminants of the staple diet[3]. Thus, it is essential to seek multidisciplinary approaches for the treatment of hepatocellular carcinoma.
A series of preclinical and clinical studies have indicated that endoglin is one of the most important angiogenic growth factors for tumor angiogenesis, and it is overexpressed and upregulated in tumor-associated angiogenic vasculature relative to normal tissue vasculature[4],[5],[6],[7]. Therefore, it is conceivable that using endoglin as a target of tumor immunotherapy strategy could inhibit endoglin- related angiogenesis and further suppress tumor growth. Our previous work also found that targeted endoglin immunotherapy of tumors showed an effective and synergistic anti-tumor activity[8],[9],[10],[11]. Macrophage inflammatory protein 3α (MIP3α) is the most important specific chemokine of dendritic cells (DCs)[12]. By attracting immature DCs which express the DC-unique receptor CCR6, MIP3α chemoattracts pathogenic infected tissues, enabling DCs to contact and present antigens[12],[13],[14],[15]. Our previous study found that recombinant MIP3α adenovirus used as a vaccine effectively attracted DCs and induced antitumor immunities[16]. Therefore, if we try to increase the level of MIP3α in tumor tissues, we can promote the chemotaxis of DCs to tumor tissues and improve antitumor immunity. The purpose of our study was to evaluate the antitumor efficacy of the endoglin-MIP3α (EM) Fc-fusion protein in mice with H22 liver cancer.
2. Materials and methods | |  |
2.1. Proteins preparation
In this study, the lyophilized recombinant Fc-fusion proteins of endoglin-MIP3α, endoglin, and MIP3α were prepared as described previously[9].
2.2. Animals model and treatment
To established the H22 hepatoma model, forty six to eight-week- old female and specific pathogen-free (SPF) BALA/c mice were randomly divided into four groups of ten mice each and injected subcutaneously with 2×106 corresponding tumor cells in the right flank. The temperature in the animal facility was maintained at 20-26 °C, with a relative humidity between 40% and 70%. Environmental conditions in the laboratory animal center were in line with the national standard for SPF-grade facilities. The EM, endoglin and MIP3α Fc-fusion protein were injected intratumorally once a week for 4 wk with a dose of 10 μg (100 μL) per mouse, respectively. Additional untreated control animals were injected with 100 μL normal saline. Tumor growth was evaluated every 3 days, and tumor volume was estimated using the formula for an ellipsoid (0.5×length×width×height, cm). This study followed the animal protocols of the College’s Animal Care and Use Committee and was ethically approved by the First Affiliated Hospital of Hainan Medical University Committee (code of ethics: ZDYF2017165).
2.3. Immunohistochemistry
For microvessel density and cell proliferation analyses, frozen sections were fixed in acetone, incubated, and stained with antibodies reactive to either CD31 or proliferating cell nuclear antigen (BD Pharmingen, USA) respectively. Moreover, sections of tissue were fixed with 1% paraformaldehyde in phosphate buffer solution (PBS) and stained for apoptosis analysis using TdT-mediated biotinylated- dUTP nick-end label (TUNEL) assay (In Situ Cell Death Detection Kit; Roche, UK) as previously described[9].
2.4. Flow cytometry
Lymphocytes were isolated from the tumor using a lymphocyte separation medium. The cells were collected by centrifugation and then were washed twice using PBS. The suspension was removed by centrifugation. The cells were resuspended in 90 μL PBS, then mixed with antibodies (CD11c: 0.25 μg/test; CD86: 0.125 μg/test) and incubated with antibodies at 4 °C for 30 min. After the addition of 400 μL PBS, FACS data collection was performed on a FACScan (Becton Dickinson) using CellQuest Pro software.
2.5. Statistical analysis
To compare data at individual time points, ANOVA and an unpaired Student’s t-test were used. Survival curves were constructed according to the Kaplan-Meier method. P<0.05 was considered statistically significant.
3. Results | |  |
3.1. Inhibition of tumor growth and extended survival time
The tumor volume was smaller and the survival time was significantly longer in the EM-treated group than that in other groups (P<0.001, [Figure 1]). | Figure 1: Induction of anti-tumor activities in the H22 hepatoma model. A: Tumor volume, B: Survival rate. NS: normal saline group, EM: endoglin-MIP3α Fc-fusion protein group, Endoglin-Fc: endoglin Fc-fusion protein group, MIP3α-Fc: MIP3α Fc-fusion protein group.
Click here to view |
3.2. Suppression of angiogenesis
The microvessel density staining with an antibody reactive to CD31 was lower in the EM-treated mice than in controls [14.9±1.9 vs. 35.4±1.8 (endoglin) vs. 42.4±2.8 (MIP3α) and 46.8±3.7 (NS)] (P<0.001, [Figure 2]). | Figure 2: Inhibition of tumor angiogenesis by immunohistochemical analysis. Endoglin-MIP3α Fc-fusion protein group (A), endoglin Fc-fusion protein group (B), MIP3α Fc-fusion protein group (C), and normal saline group (D). Vessel density (E) was determined in H22 tumor sections stained with an antibody reactive to CD31 (**P<0.01,***P<0.001).
Click here to view |
3.3. Suppression of cell proliferation
Tumor cell proliferation index detected by immunohistochemistry [Figure 3]A, [Figure 3]B, [Figure 3]C, [Figure 3]D was significantly higher in the EM-treated group than that in endoglin, MIP3α, and NS group (9.4 ± 1.6 vs. 30.8 ± 4.6, 24.7 ± 3.1, 42.1 ± 3.9, P<0.001) [Figure 3]E. | Figure 3: Inhibition of tumor cell proliferation by immunohistochemical analysis. Endoglin-MIP3α Fc-fusion protein group (A), endoglin Fc-fusion protein group (B), MIP3α Fc-fusion protein group (C), and normal saline group (D). Tumor cell proliferation (E) was determined in H22 tumor sections stained with an antibody reactive to proliferating cell nuclear antigen (**P<0.01,***P<0.001).
Click here to view |
3.4. Induction of tumor cell apoptosis
Tumor cell apoptosis index [Figure 4] was significantly higher in the EM-treated group [(15.2 ± 3.2)%] than that in endoglin group [(6.8±1.3)%], MIP3α group [(2.7±0.9)%], and NS group [(1.4 ± 0.7)%] (P<0.001 for all). | Figure 4: Induction of tumor cell apoptosis. Section of frozen H22 model tumor tissues was stained for apoptosis by TUNEL. Endoglin-MIP3α Fc-fusion protein group (A), endoglin Fc-fusion protein group (B), MIP3α Fc-fusion protein group (C), and normal saline group (D)(***P<0.001, NS P>0.05).
Click here to view |
3.5. Induction of anti-tumor immunity
Flow cytometry result showed that the number of CD11c and CD86 positive dendritic cells was significantly larger in the EM-treated group [(37.9 ± 3.5)%] than that in endoglin group [(7.3 ± 1.2)%], MIP3α group [(34.2 ± 1.5)%] , and NS group [(1.2 ± 0.3)%] (P<0.001).
4. Discussion | |  |
Angiogenesis is critical to the growth and metastasis of tumors, and endoglin is one of the most critical angiogenic growth factors for tumor angiogenesis[4],[5],[6],[7]. Since endoglin is a crucial regulatory molecule of angiogenesis in the solid tumor as described previously[7],[8],[9],[10], it is viable to use anti-cancer angiogenesis therapy to block the TGF-ß/endoglin signal pathway. However, due to inadequate DC targeting, the inadequate anti-tumor activity of endoglin alone, and inhibition of tumor microenvironment, it is necessary to seek effective treatment strategies. MIP3α is the most important specific chemokine of DCs and could effectively attract DCs and induce antitumor immunities[12],[13],[14],[15],[16]. Therefore, this research is to explore the anti-cancer activities of a recombinant EM Fc- fusion protein in mice with H22 hepatocellular carcinoma.
The results of this study suggest that the treatment of the EM Fc- fusion protein could effectively inhibit tumor growth, not only by inducing more effective anti-angiogenesis, but also by promoting apoptosis and suppression of cell proliferation in tumor tissues. Moreover, the number of CD11c and CD86 positive DCs was more significantly increased in the EM-treated group as compared with other groups, suggesting that the Fc fusion protein brings about better chemotactic recruitment of DCs.
Our findings demonstrate that the endoglin-MIP3α Fc-fusion protein treatment may be used as a useful and new approach to treat solid tumors. In addition, we also find that endoglin-MIP3α Fc- fusion protein induces a certain degree of the anti-tumor immune response, the specific mechanism remains to be further explored.
Conflict of interest statement
The authors declare that there is no conflict of interest.
Founding project
The work was supported by grants from the Finance Science and Technology Projects of Hainan Province (grant No.ZDYF2019148 and ZDYF2017165).
Authors’ contribution
Y.D.L. and Z.H.H. conceived and coordinated the study. S.R.L., X.L.W., Y.H.H., Y.C. and P.P.X. carried out the experiments. X.L.W. and Y.D. collected and analysed the data. Both Z.H.H. and Y.D.L. authors contributed to the final version of the manuscript.
References | |  |
1. | Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in China: Good or bad news from the 2018 global cancer statistics? Cancer Commun (Lond) 2019; 39(1): 22. |
2. | Ge S, Huang D. Systemic therapies for hepatocellular carcinoma. Drug Discov Ther 2015; 9: 352-362. |
3. | Szymañska K, Lesi OA, Kirk GD, Sam O, Taniere P, Scoazec JY, et al. Ser-249TP53 mutation in tumour and plasma DNA of hepatocellular carcinoma patients from a high incidence area in the Gambia, West Africa. Int J Cancer 2004; 110(3): 374-379. |
4. | Ferrara N, Alitalo K. Clinical application of angiogenic growth factors and their inhibitors. Nature Med 1999; 5: 1359-1364. |
5. | Burrows FJ, Derbyshire EJ, Tazzari PL, Amlot P, Gazdar AF, King SW, et al. Up-regulation of endoglin on vascular endothelial cells in human solid tumors: Implications for diagnosis and therapy. Clin Cancer Res1995; 1(12): 1623-1634. |
6. | Bodey B, Bodey B Jr, Siegel SE, Kaiser HE. Over-expression of endoglin (CD105): A marker of breast carcinoma-induced neo-vascularization. Anticancer Res 1998; 18(5A): 3621-3628. |
7. | Takahashi N, Haba A, Matsuno F, Seon BK. Antiangiogenic therapy of established tumors in human skin/severe combined immunodeficiency mouse chimeras by anti-endoglin (CD105) monoclonal antibodies, and synergy between anti-endoglin antibody and cyclophosphamide. Cancer Res 2001; 61(21): 7846-7854. |
8. | Li YN, Huang FY, Mei WL, Dai HF, Guo JL, Tan GH, et al. Toxicarioside A, isolated from tropical Antiaris toxicaria, blocks endoglin/TGF-β signaling in a bone marrow stromal cell line. Asian Pac J Trop Med 2012; 5(2): 91-97. |
9. | Huang FY, Li L, Liu Q,Li YN, Bai RZ, Huang YH, et al. Bacterial surface display of endoglin by antigen 43 induces antitumor effectiveness via bypassing immunotolerance and inhibition of angiogenesis. Int J Cancer 2014; 134(8): 1981-1990. |
10. | Huang FY, Mei WL, Li YN, Tan GH, Dai HF, Guo JL, et al. Toxicarioside A inhibits tumor growth and angiogenesis: Involvement of TGF-β/endoglin signaling. PLoS One 2012; 7(11): e50351. |
11. | Tan GH, Huang FY, Wang H, YH, Lin YY, Li YN. Immunotherapy of hepatoma with a monoclonal antibody against murine endoglin. World J Gastroenterol 2007; 13(17): 2479-2483. |
12. | Ghadjar P, Rubie C, Aebersold DM, Keilholz U. The chemokine CCL20 and its receptor CCR6 in human malignancy with focus on colorectal cancer. Int J Cancer 2009; 125(4): 741-745. |
13. | Samaniego R, Gutiérrez-González A, Gutiérrez-Seijo A, Sánchez- Gregorio S, García-Giménez J, Mercader E, et al. CCL20 expression by tumor-associated macrophages predicts progression of human primary cutaneous melanoma. Cancer Immunol Res 2018; 6(3): 267-275. |
14. | Lu E, Su J, Zhou Y, Zhang C, Wang Y. CCL20/CCR6 promotes cell proliferation and metastasis in laryngeal cancer by activating p38 pathway. Biomed Pharmacother 2017; 85: 486-492. |
15. | Zhang XP, Hu ZJ, Meng AH, Duan GC, Zhao QT, Yang J, et al. Role of CCL20/CCR6 and the ERK signaling pathway in lung adenocarcinoma. Oncol Lett 2017; 14(6): 8183-8189. |
16. | Huang FY, Huang FR, Chen B, Liu Q, Wang H, Zhou SL, et al. Microencapsulation of tumor lysates and live cell engineering with MIP- 3a as an effective vaccine. Biomaterials 2015; 53: 554-565. |
[Figure 1], [Figure 2], [Figure 3], [Figure 4]
|