Antitumor effects of anlotinib in thyroid cancer

Xianhui Ruan, Xianle Shi, Qiman Dong, Yang Yu, Xiukun Hou, Xinhao Song, Xi Wei, Lingyi Chen, Ming Gao, Xianhui Ruan, Xianle Shi, Qiman Dong, Yang Yu, Xiukun Hou, Xinhao Song, Xi Wei, Lingyi Chen, Ming Gao

Abstract

There is no effective treatment for patients with poorly differentiated papillary thyroid cancer or anaplastic thyroid cancer (ATC). Anlotinib, a multi-kinase inhibitor, has already shown antitumor effects in various types of carcinoma in a phase I clinical trial. In this study, we aimed to better understand the effect and efficacy of anlotinib against thyroid carcinoma cells in vitro and in vivo. We found that anlotinib inhibits the cell viability of papillary thyroid cancer and ATC cell lines, likely due to abnormal spindle assembly, G2/M arrest, and activation of TP53 upon anlotinib treatment. Moreover, anlotinib suppresses the migration of thyroid cancer cells in vitro and the growth of xenograft thyroid tumors in mice. Our data demonstrate that anlotinib has significant anticancer activity in thyroid cancer, and potentially offers an effective therapeutic strategy for patients of advanced thyroid cancer type.

Keywords: anlotinib; apoptosis; migration; multi-kinase inhibitor; thyroid cancer.

Figures

Figure 1
Figure 1
Anlotinib inhibits thyroid cancer cells proliferation in vitro. (A) Thyroid cancer cells BCPAP and 8505C were treated with anlotinib (0, 1, 5 and 10 µM) for 24 h, and then photographed with inverted contrast microscopy (100× magnification). Scale bars: 50 µm. (B) Representative images of colony formation assays using BCPAP and 8505C cells treated with (0, 1, 5 and 10 µM) for 8 days. (C and D) Quantification of the colony formation assays in BCPAP and 8505C cells. Data are shown as mean ± s.d. (n ≥ 3).
Figure 2
Figure 2
Anlotinib induces G2/M phase arrest in thyroid cancer cells. (A and C) BCPAP cells (A) and 8505C cells (C) were treated with DMSO or 10 µM anlotinib for 24 h and stained with PI. Cell cycle distribution was assessed with flow cytometry and quantified by Modifit software. (B and D) The percentage of cells in sub-G1, G1, S and G2/M phase were calculated and plotted. Data are shown as mean ± s.d. (n ≥ 3).
Figure 3
Figure 3
Anlotinib leads to abnormal spindle formation in thyroid cancer cells. (A and C) BCPAP cells (A) and 8505C cells (C) were treated with anlotinib (0, 1, 5 and 10 µM) for 24 h. The morphological changes of nuclei were examined using Hoechst 33342 staining. Scale bars: 100 µm. The blowup images from the red boxes show nuclear condensation and structural changes after exposure to anlotinib. (B and D) Effect of anlotinib on mitotic spindle formation in thyroid cancer cells. Cells were treated with or without 10 µM anlotinib for 24 h and subjected to immunofluorescence staining for TUBULIN. The right plots show the percentage of cells with abnormal spindle in the metaphase cells treated with 0, 1, 5 and 10 µM anlotinib. Data are shown as mean ± s.d. (n ≥ 3).
Figure 4
Figure 4
Anlotinib induces apoptosis in thyroid cancer cells. (A and D) BCPAP (A) and 8505C (D) cells were treated with DMSO and 10 µM anlotinib for 36 h. Cells were stained with Annexin V-FITC/PI and analyzed by flow cytometry. The low right (LR) quadrant of the histograms indicates the early apoptotic cells, and the upper right quadrant indicates the late apoptotic cells. (B and E) Quantification results of experiments described in A and D. Data are shown as mean ± s.d. (n ≥ 3). (C and F) BCPAP (C) and 8505C (F) cells were treated with anlotinib (0, 1, 5, 10 and 15 µM) for 36 h. Cell lysates were prepared and subjected to Western blot to detect the expression of TP53, cleaved caspase 3 (CL-caspase 3) and cleaved PARP (CL-PARP).
Figure 5
Figure 5
Anlotinib induces apoptosis partly through activating the TP53 pathway. (A and B) siRNA target TP53 and control RNA were transfected into thyroid cancer cells. Twenty-four hours after transfection, the cells were treated with or without 10 µM anlotinib for 24 h. Cell lysates were prepared and subjected to Western blot. (C and D) Thyroid cancer cells described in (A and B) were stained with Annexin V-FITC/PI and analyzed with flow cytometry. Data are shown as mean ± s.d. (n ≥ 3).
Figure 6
Figure 6
The effects of anlotinib on the migration of thyroid cancer cells. (A and B) Representative images of wound-healing assays using BCPAP (A) and 8505C (B) cells in the presence of 0, 1, 5 and 10 µM anlotinib. Scale bars: 25 µm. The plots on the right show the quantification of wound healing assays. Data are shown as mean ± s.d. (n ≥ 3). (C and D) The expression and cellular distribution of F-actin in BCPAP (C) and 8505C cells (D). Cells were treated with DMSO or 10 µM anlotinib for 24 h, and then stained with rhodamine-phalloidin (red) for F-actin. Hoechst 33342 was applied to counter-stain the nucleus (blue). Scale bars: 100 µm.
Figure 7
Figure 7
Anlotinib suppresses tumor growth in vivo. (A) Images of dissected tumors from nude mice injected with K1 cells of DMSO treated group (n = 5) and anlotinib treated group (n = 5). (B) Tumor weights of control and anlotinib-treated groups. (C) Tumor volumes with or without anlotinib treatment. (D) HE staining, Ki67, TP53 and CL-caspase 3 IHC staining of the tumors with or without anlotinib treatment. Scale bars: 100 µm.

References

    1. Baudin E, Schlumberger M. 2007. New therapeutic approaches for metastatic thyroid carcinoma. Lancet Oncology 8 148–156. (10.1016/s1470-2045(07)70034-7)
    1. Chen W, Zheng R, Baade P D, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. 2016. Cancer statistics in China 2015. CA: A Cancer Journal for Clinicians 66 115–132. (10.3322/caac.21338)
    1. Cho BY, Choi HS, Park YJ, Lim JA, Ahn HY, Lee EK, Kim KW, Yi KH, Chung JK, Youn YK, et al. 2013. Changes in the clinicopathological characteristics and outcomes of thyroid cancer in Korea over the past four decades. Thyroid 23 797–804. (10.1089/thy.2012.0329)
    1. Covell LL, Ganti AK. 2015. Treatment of advanced thyroid cancer: role of molecularly targeted therapies. Targeted Oncology 10 311–324. (10.1007/s11523-014-0331-z)
    1. Chipuk JE, Maurer U, Green DR, Schuler M. 2003. Pharmacologic activation of p53 elicits Bax-dependent apoptosis in the absence of transcription, Cancer Cell 4 371–381. (10.1016/S1535-6108(03)00272-1)
    1. Enewold L, Zhu K, Ron E, Marrogi AJ, Stojadinovic A, Peoples GE, Devesa SS. 2009. Rising thyroid cancer incidence in the United States by demographic and tumor characteristics, 1980–2005. Cancer Epidemiology, Biomarkers and Prevention 18 784–791. (10.1158/1055-9965.EPI-08-0960)
    1. Espinosa AV, Porchia L, Ringel MD. 2007. Targeting BRAF in thyroid cancer. British Journal of Cancer 96 16–20. (10.1038/sj.bjc.6603520)
    1. Fagin JA, Mitsiades N. 2008. Molecular pathology of thyroid cancer: diagnostic and clinical implications. Best Practice and Research Clinical Endocrinology and Metabolism 22 955–969. (10.1016/j.beem.2008.09.017)
    1. Girotti MR, Marais R. 2013. Deja Vu: EGF receptors drive resistance to BRAF inhibitors. Cancer Discovery 3 487–490. (10.1158/-13-0131)
    1. Green DR, Kroemer G. 2009. Cytoplasmic functions of the tumour suppressor p53. Nature 458 1127–1130. (10.1038/nature07986)
    1. Haupt Y, Rowan S, Shaulian E, Vousden KH, Oren M. 1995. Iduction of apoptosis in Hela cells by trans-activation-deficient p53. Genes and Development 9 2170–2183. (10.1101/gad.9.17.2170)
    1. Kilfoy BA, Zheng T, Holford TR, Han X, Ward MH, Sjodin A, Zhang Y, Bai Y, Zhu C, Guo GL, et al. 2009. International patterns and trends in thyroid cancer incidence, 1973–2002. Cancer Causes Control 20 525–531. (10.1007/s10552-008-9260-4)
    1. Kondo T, Ezzat S, Asa SL. 2006. Pathogenetic mechanisms in thyroid follicular-cell neoplasia. Nature Reviews Cancer 6 292–306. (10.1038/nrc1836)
    1. Li C, Lee KC, Schneider EB, Zeiger MA. 2012. BRAF V600E mutation and its association with clinicopathological features of papillary thyroid cancer: a meta-analysis. Journal of Clinical Endocrinology and Metabolism 97 4559–4570. (10.1210/jc.2012-2104)
    1. Liu C, Chen T, Liu Z. 2016. Associations between BRAF(V600E) and prognostic factors and poor outcomes in papillary thyroid carcinoma: a meta-analysis. World Journal of Surgical Oncology 14 241 (10.1186/s12957-016-0979-1)
    1. Lorusso L, Pieruzzi L, Biagini A, Sabini E, Valerio L, Giani C, Passannanti P, Pontillo-Contillo B, Battaglia V, Mazzeo S, et al. 2016. Lenvatinib and other tyrosine kinase inhibitors for the treatment of radioiodine refractory, advanced, and progressive thyroid cancer. OncoTargets Therapy 9 6467–6477. (10.2147/OTT.S84625)
    1. Machens A, Holzhausen H-J, Dralle H. 2005. The prognostic value of primary tumor size in papillary and follicular thyroid carcinoma. Cancer 103 2269–2273. (10.1002/cncr.21055)
    1. McFarland D, Misiukiewicz K. 2014. Sorafenib in radioactive iodine-refractory well-differentiated metastatic thyroid cancer. OncoTargets and Therapy 7 1291 (10.2147/ott.s49430)
    1. Murugan AK, Dong J, Xie J, Xing M. 2009. MEK1 mutations, but not ERK2 mutations, occur in melanomas and colon carcinomas, but none in thyroid carcinomas. Cell Cycle 8 2122–2124. (10.4161/cc.8.13.8710)
    1. Messina RL, Sanfilippo M, Vella V, Pandini G, Vigneri P, Nicolosi ML, Giani F, Vigneri R, Frasca F. 2012. Reactivation of p53 mutants by prima-1 (corrected) in thyroid cancer cells. International Journal of Cancer 130 2259–2270. (10.1002/ijc.26228)
    1. Nagaiah G, Hossain A, Mooney CJ, Parmentier J, Remick SC. 2011. Anaplastic thyroid cancer: a review of epidemiology, pathogenesis, and treatment. Journal of Oncology 2011 542358 (10.1155/2011/542358)
    1. Rehman A, Chahal MS, Tang X, Bruce JE, Pommier Y, Daoud SS. 2005. Proteomic identification of heat shock protein 90 as a candidate target for p53 mutation reactivation by PRIMA-1 in breast cancer cells. Breast Cancer Research 7 R765–R774. (10.1186/bcr1290)
    1. Sherman SI. 2009. Advances in chemotherapy of differentiated epithelial and medullary thyroid cancers. Journal of Clinical Endocrinology and Metabolism 94 1493–1499. (10.1210/jc.2008-0923)
    1. Smallridge RC, Marlow LA, Copland JA. 2009. Anaplastic thyroid cancer: molecular pathogenesis and emerging therapies. Endocrine-Related Cancer 16 17–44. (10.1677/ERC-08-0154)
    1. Smallridge RC, Ain KB, Asa SL, Bible KC, Brierley JD, Burman KD, Kebebew E, Lee NY, Nikiforov YE, Rosenthal MS, et al. 2012. American Thyroid Association guidelines for management of patients with anaplastic thyroid cancer. Thyroid 22 1104–1139. (10.1089/thy.2012.0302)
    1. Sun Y, Niu W, Du F, Du C, Li S, Wang J, Li L, Wang F, Hao Y, Li C, et al. 2016. Safety, pharmacokinetics, and antitumor properties of anlotinib, an oral multi-target tyrosine kinase inhibitor, in patients with advanced refractory solid tumors. Journal of Hematology and Oncology 9 105 (10.1186/s13045-016-0332-8)
    1. Saiselet M, Floor S, Tarabichi M, Dom G, Hebrant A, van Staveren WC, Maenhaut C. 2012. Thyroid cancer cell lines: an overview. Frontiers in Endocrinology 3 133 (10.3389/fendo.2012.00133)
    1. Wang C, Chen J, Cao W, Sun L, Sun H, Liu Y. 2016. Aurora-B and HDAC synergistically regulate survival and proliferation of lymphoma cell via AKT, mTOR and Notch pathways. European Journal of Pharmacology 779 1–7. (10.1016/j.ejphar.2015.11.049)
    1. Xing M. 2013. Molecular pathogenesis and mechanisms of thyroid cancer. Nature Reviews Cancer 13 184–199. (10.1038/nrc3431)
    1. Xing M, Alzahrani AS, Carson KA, Viola D, Elisei R, Bendlova B, Yip L, Mian C, Vianello F, Tuttle RM, et al. 2013. Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer. JAMA 309 1493 (10.1001/jama.2013.3190)
    1. Zhang L, Liu L, He X, Shen Y, Liu X, Wei J, Yu F, Tian J. 2016. CHIP promotes thyroid cancer proliferation via activation of the MAPK and AKT pathways. Biochemical and Biophysical Research Communications 477 356–362. (10.1016/j.bbrc.2016.06.101)

Source: PubMed

3
Tilaa