157
Views
1
CrossRef citations to date
0
Altmetric
Original Research

Triptolide is a Promising Therapeutic Approach in Treating Thyroid Cancer Based on in silico and in vitro Experiment

, , , , , , & show all
Pages 4275-4287 | Published online: 11 Oct 2021

References

  • CabanillasME, McFaddenDG, DuranteC. Thyroid cancer. Lancet. 2016;388(10061):2783–2795. doi:10.1016/S0140-6736(16)30172-627240885
  • SungH, FerlayJ, SiegelRL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249. doi:10.3322/caac.2166033538338
  • AsaSL. The current histologic classification of thyroid cancer. Endocrinol Metab Clin North Am. 2019;48(1):1–22. doi:10.1016/j.ecl.2018.10.00130717895
  • GraniG, LamartinaL, DuranteC, et al. Follicular thyroid cancer and Hürthle cell carcinoma: challenges in diagnosis, treatment, and clinical management. Lancet Diabetes Endocrinol. 2018;6(6):500–514. doi:10.1016/S2213-8587(17)30325-X29102432
  • SchmidbauerB, MenhartK, HellwigD, et al. Differentiated thyroid cancer-treatment: state of the art. Int J Mol Sci. 2017;18(6):E1292. doi:10.3390/ijms1806129228629126
  • NoelP, Von HoffDD, SalujaAK, et al. Triptolide and its derivatives as cancer therapies. Trends Pharmacol Sci. 2019;40(5):327–341. doi:10.1016/j.tips.2019.03.00230975442
  • JiangW, ChenM, XiaoC, et al. Triptolide suppresses growth of breast cancer by targeting HMGB1 in vitro and in vivo. Biol Pharm Bull. 2019;42(6):892–899. doi:10.1248/bpb.b18-0081830956264
  • SunYY, XiaoL, WangD, et al. Triptolide inhibits viability and induces apoptosis in liver cancer cells through activation of the tumor suppressor gene p53. Int J Oncol. 2017;50(3):847–852. doi:10.3892/ijo.2017.385028098861
  • RenoTA, KimJY, RazDJ. Triptolide inhibits lung cancer cell migration, invasion, and metastasis. Ann Thorac Surg. 2015;100(5):1817–1824. doi:10.1016/j.athoracsur.2015.05.07426298168
  • CalòPG, MedasF, ConzoG, et al. Intraoperative neuromonitoring in thyroid surgery: is the two-staged thyroidectomy justified? Int J Surg. 2017;41(Suppl 1):S13–S20. doi:10.1016/j.ijsu.2017.02.00128506407
  • DocimoG, ToloneS, RuggieroR, et al. Total thyroidectomy without prophylactic central neck dissection combined with routine oral calcium and vitamin D supplements: is it a good option to achieve a low recurrence rate avoiding hypocalcemia? A retrospective study. Minerva Chir. 2013;68(3):321–328.23774098
  • ConzoG, DocimoG, MaurielloC, et al. The current status of lymph node dissection in the treatment of papillary thyroid cancer. A literature review. Clin Ter. 2013;164(4):e343–e346.24045534
  • ChenSR, DaiY, ZhaoJ, et al. A mechanistic overview of triptolide and celastrol, natural products from Tripterygium wilfordii Hook F. Front Pharmacol. 2018;9:104. doi:10.3389/fphar.2018.0010429491837
  • YuanK, LiX, LuQ, et al. Application and mechanisms of triptolide in the treatment of inflammatory diseases-a review. Front Pharmacol. 2019;10:1469. doi:10.3389/fphar.2019.0146931866868
  • HouW, LiuB, XuH. Triptolide: medicinal chemistry, chemical biology and clinical progress. Eur J Med Chem. 2019;176:378–392. doi:10.1016/j.ejmech.2019.05.03231121546
  • ZhuW, OuY, LiY, et al. A small-molecule triptolide suppresses angiogenesis and invasion of human anaplastic thyroid carcinoma cells via down-regulation of the nuclear factor-κB pathway. Mol Pharmacol. 2009;75(4):812–819. doi:10.1124/mol.108.05260519158360
  • ZhuW, HeS, LiY, et al. Anti-angiogenic activity of triptolide in anaplastic thyroid carcinoma is mediated by targeting vascular endothelial and tumor cells. Vascul Pharmacol. 2010;52(1–2):46–54. doi:10.1016/j.vph.2009.10.00619854299
  • CalcinottoA, KohliJ, ZagatoE, et al. Cellular senescence: aging, cancer, and injury. Physiol Rev. 2019;99(2):1047–1078. doi:10.1152/physrev.00020.201830648461
  • IcardP, FournelL, WuZ, et al. Interconnection between metabolism and cell cycle in cancer. Trends Biochem Sci. 2019;44(6):490–501. doi:10.1016/j.tibs.2018.12.00730655165
  • TchakarskaG, SolaB. The double dealing of cyclin D1. Cell Cycle. 2020;19(2):163–178. doi:10.1080/15384101.2019.170690331885322
  • OttoT, PiotrP. Cell cycle proteins as promising targets in cancer therapy. Nat Rev Cancer. 2017;17(2):93–115. doi:10.1038/nrc.2016.13828127048
  • HydbringP, MalumbresM, SicinskiP. Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat Rev Mol Cell Biol. 2016;17(5):280–292. doi:10.1038/nrm.2016.2727033256
  • MalumbresM, BarbacidM. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9(3):153–166. doi:10.1038/nrc260219238148
  • TaniguchiK, MichaelM. NF-κB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018;18(5):309–324.29379212
  • EluardB, ThieblemontC, BaudV. NF-κB in the new era of cancer therapy. Trends Cancer. 2020;6(8):677–687. doi:10.1016/j.trecan.2020.04.00332409139
  • BraicuC, BuseM, BusuiocC, et al. A comprehensive review on MAPK: a promising therapeutic target in cancer. Cancers. 2019;11(10):1618. doi:10.3390/cancers11101618
  • CarneiroBA, El-DeiryWS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 2020;17(7):395–417.32203277
  • WarrenCFA, Wong-BrownMW, BowdenNA. BCL-2 family isoforms in apoptosis and cancer. Cell Death Dis. 2019;10(3):177. doi:10.1038/s41419-019-1407-630792387
  • AubreyB, KellyG, JanicA, et al. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018;25(1):104–113.29149101
  • HafnerA, BulykML, JambhekarA, et al. The multiple mechanisms that regulate p53 activity and cell fate. Nat Rev Mol Cell Biol. 2019;20(4):199–210. doi:10.1038/s41580-019-0110-x30824861