小分子酪氨酸激酶抑制剂安罗替尼对鼻咽癌细胞的抗肿瘤活性及其机制
作者:
作者单位:

1.湖南省人民医院 检验科,湖南 长沙,410000;中山大学附属第五医院 2.肿瘤中心;2.临床检验科, 广东 珠海,519000

作者简介:

谢婉莹,女,硕士,检验师,研究方向:肿瘤靶向免疫治疗。

通讯作者:

刘志刚,男,博士,主任医师,研究方向:肿瘤学

中图分类号:

R739.63

基金项目:

★北京市医卫健康公益基金(F3040C)。


Anti-tumor activity and mechanism of the small molecular tyrosine kinase inhibitor anlotinib in nasopharyngeal cancer cells
Author:
Affiliation:

1.Department of Clinical Laboratory, the People's Hospital of Hunan Province, Changsha, Hunan, 410000, China;2.The Cancer Center;3.Department of Clinical Laboratory, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China

  • 摘要
  • | |
  • 访问统计
  • |
  • 参考文献
  • |
  • 相似文献
  • | | |
  • 文章评论
    摘要:

    目的 探索小分子酪氨酸激酶抑制剂安罗替尼对鼻咽癌细胞的抗肿瘤作用及其潜在机制,为临床应用安罗替尼治疗鼻咽癌提供可靠的实验证据。方法 通过CCK-8实验和克隆形成实验检测不同剂量梯度安罗替尼对鼻咽癌细胞6-10B和SUNE-1的细胞活性、增殖能力的影响;通过Transwell和划痕实验分析安罗替尼对鼻咽癌细胞侵袭及迁移能力的影响;利用流式细胞术检测安罗替尼对鼻咽癌细胞周期及凋亡的影响;采用Western blotting验证安罗替尼处理后鼻咽癌细胞凋亡相关蛋白的表达变化。结果 在鼻咽癌细胞中加入安罗替尼处理后,6-10B实验组和SUNE-1实验组细胞存活分数降低,细胞集落形成数量明显减少,且呈显著的剂量依赖性,在药物浓度为2.0 μmol·L-1时可达到约50%的增殖及克隆抑制率(P<0.01)。Western blotting分析发现,安罗替尼可能是由Erk通路介导线粒体通路激活,促进鼻咽癌细胞凋亡,且以药物浓度达到2.0 μmol·L-1时作用最为明显。凋亡检测结果证实,2.0 μmol·L-1安罗替尼处理后的实验组较对照组具有更高的细胞凋亡率,且以早期凋亡为主(P<0.001)。此外,2.0 μmol·L-1安罗替尼处理后的鼻咽癌细胞发生明显的周期分布改变,6-10B实验组和SUNE-1实验组均发生G2/M期阻滞,且6-10B实验组作用较为显著(P=0.0004),而SUNE-1实验组的变化无统计学意义(P=0.0723)。Transwell和划痕实验结果表明,实验组侵袭和迁移能力较对照组相比显著下降(P<0.05)。结论 安罗替尼可能通过Erk通路介导鼻咽癌细胞6-10B和SUNE-1的凋亡,并抑制其增殖,有望成为治疗鼻咽癌的新型靶向药物。

    Abstract:

    Objective To explore the anti-tumor effects of anlotinib, a small molecular tyrosine kinase inhibitor, on nasopharyngeal carcinoma (NPC) cells and its potential mechanism, and to provide reliable experimental evidence for the clinical treatment of NPC with anlotinib.Methods The responses of serial dose gradients of anlotinib on the cell viability and proliferation of 6-10B and SUNE-1 cell lines were detected by cell proliferation viability assay and colony formation assay. Then, the downstream pathway proteins in cell lines after anlotinib treatment were visualized by Western blotting. The effects of promoting apoptosis and inducing cell cycle arrest by anlotinib were confirmed through flow cytometry. The capacity of anlotinib in inhibiting migration and invasion of 6-10B and SUNE-1 cell lines was verified by transwell assay and wound healing assay.Results After treatment of anlotinib, cell viability and proliferation were inhibited in nasopharyngeal carcinoma 6-10B and SUNE-1 cell lines in a dose-dependent way, and the inhibition rate was up to 50% when then concentration of anlotinib was 2 μmol·L-1 (P<0.01). Western blotting results showed that anlotinib might induce the cell apoptosis by activating the mitochondrial pathway and potentially the Erk pathway, with the highest apoptosis rate when the anlotinib concentration up to 2 μmol·L-1. The results of flow cytometry showed that 2 μmol·L-1 anlotinib induced remarkable cell apoptosis, with early apoptosis predominantly (P<0.001). What's more, the nasopharyngeal carcinoma cells had obvious changes in cycle distribution after treated with 2 μmol·L-1 anlotinib. Both 6-10B and SUNE-1 cells had a G2/M arrest, and there was a significant difference in 6-10B group (P=0.0004), but no statistical differences in SUNE-1 group (P=0.0723). By transwell assay and wound healing assay, the significant suppression of invasion and migration in 6-10B and SUNE-1 was definitude (P<0.05).Conclusion This study indicated that anlotinib promoted the apoptosis and inhibited the proliferation of nasopharyngeal carcinoma cells 6-10B and SUNE-1 through the Erk pathway. It could be a potential target drug for nasopharyngeal carcinoma.

    参考文献
    [1] CHEN Y P, CHAN A T C, LE Q T, et al. Nasopharyngeal carcinoma [J]. Lancet, 2019, 394(10192): 64-80. DOI: 10.1016/s0140-6736(19)30956-0.
    [2] FARRELL P J. Epstein-Barr virus and cancer [J]. Annu Rev Pathol, 2019, 14: 29-53. DOI: 10.1146/annurev-pathmechdis-012418-013023.
    [3] SUNG H, FERLAY J, SIEGEL R L, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries [J]. CA A Cancer J Clin, 2021, 71(3): 209-249. DOI: 10.3322/caac.21660.
    [4] LEE A W M, NG W T, CHAN L L K, et al. Evolution of treatment for nasopharyngeal cancer - success and setback in the intensity-modulated radiotherapy era [J]. Radiother Oncol, 2014, 110(3): 377-384. DOI: 10.1016/j.radonc.2014.02.003.
    [5] TSENG M, HO F, LEONG Y H, et al. Emerging radiotherapy technologies and trends in nasopharyngeal cancer [J]. Cancer Commun (Lond), 2020, 40(9): 395-405. DOI: 10.1002/cac2.12082.
    [6] LIN B, SONG X, YANG D, et al. Anlotinib inhibits angiogenesis via suppressing the activation of VEGFR2, PDGFRβ and FGFR1 [J]. Gene, 2018, 654: 77-86. DOI: 10.1016/j.gene.2018.02.026.
    [7] XIE C, WAN X, QUAN H, et al. Preclinical characterization of anlotinib, a highly potent and selective vascular endothelial growth factor receptor-2 inhibitor [J]. Cancer Sci, 2018, 109(4): 1207-1219. DOI: 10.1111/cas.13536.
    [8] SUN Y K, NIU W, DU F, et al. Safety, pharmacokinetics, and antitumor properties of anlotinib, an oral multi-target tyrosine kinase inhibitor, in patients with advanced refractory solid tumors [J]. J Hematol Oncol, 2016, 9(1): 105. DOI: 10.1186/s13045-016-0332-8.
    [9] WANG G, SUN M, JIANG Y, et al. Anlotinib, a novel small molecular tyrosine kinase inhibitor, suppresses growth and metastasis via dual blockade of VEGFR2 and MET in osteosarcoma [J]. Int J Cancer, 2019, 145(4): 979-993. DOI: 10.1002/ijc.32180.
    [10] DENG Z M, LIAO W, WEI W, et al. Anlotinib as a promising inhibitor on tumor growth of oral squamous cell carcinoma through cell apoptosis and mitotic catastrophe [J]. Cancer Cell Int, 2021, 21(1): 37. DOI: 10.1186/s12935-020-01721-x.
    [11] HAN B H, LI K, ZHAO Y Z, et al. Anlotinib as a third-line therapy in patients with refractory advanced non-small-cell lung cancer: a multicentre, randomised phase II trial (ALTER0302) [J]. Br J Cancer, 2018, 118(5): 654-661. DOI: 10.1038/bjc.2017.478.
    [12] ZHANG Y, CHEN L, HU G Q, et al. Gemcitabine and cisplatin induction chemotherapy in nasopharyngeal carcinoma [J]. N Engl J Med, 2019, 381(12): 1124-1135. DOI: 10.1056/nejmoa1905287.
    [13] ZHANG L, HUANG Y, HONG S, et al. Gemcitabine plus cisplatin versus fluorouracil plus cisplatin in recurrent or metastatic nasopharyngeal carcinoma: a multicentre, randomised, open-label, phase 3 trial [J]. Lancet, 2016, 388(10054): 1883-1892. DOI: 10.1016/s0140-6736(16)31388-5.
    [14] CHEN X, LIANG W, WAN N, et al. Cost-effectiveness analysis of gemcitabine plus cisplatin versus fluorouracil plus cisplatin for first-line treatment of recurrent or metastatic nasopharyngeal carcinoma [J]. Oral Oncol, 2019, 94: 80-85. DOI: 10.1016/j.oraloncology.2019.04.022.
    [15] HUI E P, MA B B Y, LOONG H H F, et al. Efficacy, safety, and pharmacokinetics of axitinib in nasopharyngeal carcinoma: a preclinical and phase II correlative study [J]. Clin Cancer Res, 2018, 24(5): 1030-1037. DOI: 10.1158/1078-0432.CCR-17-1667.
    [16] PàEZ-RIBES M, ALLEN E, HUDOCK J, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis [J]. Cancer Cell, 2009, 15(3): 220-231. DOI: 10.1016/j.ccr.2009.01.027.
    [17] GAVET O, PINES J. Progressive activation of CyclinB1-Cdk1 coordinates entry to mitosis [J]. Dev Cell, 2010, 18(4): 533-543. DOI: 10.1016/j.devcel.2010.02.013.
    [18] HARLEY M E, ALLAN L A, SANDERSON H S, et al. Phosphorylation of Mcl-1 by CDK1–cyclin B1 initiates its Cdc20-dependent destruction during mitotic arrest [J]. EMBO J, 2010, 29(14): 2407-2420. DOI: 10.1038/emboj.2010.112.
    [19] OLA M S, NAWAZ M, AHSAN H. Role of Bcl-2 family proteins and caspases in the regulation of apoptosis [J]. Mol Cell Biochem, 2011, 351(1/2): 41-58. DOI: 10.1007/s11010-010-0709-x.
    [20] DECKER P, MULLER S. Modulating poly (ADP-ribose) polymerase activity: potential for the prevention and therapy of pathogenic situations involving DNA damage and oxidative stress [J]. Curr Pharm Biotechnol, 2002, 3(3): 275-283. DOI: 10.2174/1389201023378265.
    [21] AREDIA F, SCOVASSI A I. Poly(ADP-ribose): a signaling molecule in different paradigms of cell death [J]. Biochem Pharmacol, 2014, 92(1): 157-163. DOI: 10.1016/j.bcp.2014.06.021.
    引证文献
    网友评论
    网友评论
    分享到微博
    发 布
引用本文

谢婉莹,钟桂华,魏威,周玉玲,黎村艳,江冠民,刘志刚.小分子酪氨酸激酶抑制剂安罗替尼对鼻咽癌细胞的抗肿瘤活性及其机制[J].肿瘤药学,2022,(1):46-52 ( in Chinese)

复制
分享
文章指标
  • 点击次数:669
  • 下载次数: 753
  • HTML阅读次数: 595
  • 引用次数: 0
历史
  • 收稿日期:2021-06-10
  • 在线发布日期: 2022-03-15
我要投稿 杂志简介 杂志简介 二维码
TOP
×
《肿瘤药学》
《肿瘤药学》编辑部打假维权声明