98
Views
2
CrossRef citations to date
0
Altmetric
Original Research

Characterisation of Levonorgestrel-Resistant Endometrial Cancer Cells

, , & ORCID Icon
Pages 7871-7884 | Published online: 14 Oct 2021

References

  • Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–E386. doi:10.1002/ijc.2921025220842
  • Duska LR, Garrett A, Rueda BR, Haas J, Chang Y, Fuller AF. Endometrial cancer in women 40 years old or younger. Gynecol Oncol. 2001;83(2):388–393. doi:10.1006/gyno.2001.643411606102
  • Gottwald L, Pluta P, Piekarski J, et al. Long-term survival of endometrioid endometrial cancer patients. Arch Med Sci. 2010;6(6):937–944. doi:10.5114/aoms.2010.1930522427770
  • Koh W-J, Abu-Rustum NR, Bean S, et al. Uterine neoplasms, version 1.2018, NCCN clinical practice guidelines in oncology. J Natl Compr Cancer Netw. 2018;16(2):170–199. doi:10.6004/jnccn.2018.0006
  • Pellerin GP, Finan MA. Endometrial cancer in women 45 years of age or younger: a clinicopathological analysis. Am J Obstet Gynecol. 2005;193(5):1640–1644. doi:10.1016/j.ajog.2005.05.00316260203
  • McMahon MD, Scott DM, Saks E, Tower A, Raker CA, Matteson KA. Impact of obesity on outcomes of hysterectomy. J Minim Invasive Gynecol. 2014;21(2):259–265. doi:10.1016/j.jmig.2013.08.70724012923
  • Acharya S, Esthappan J, Badiyan S, et al. Medically inoperable endometrial cancer in patients with a high body mass index (BMI): patterns of failure after 3-D image-based high dose rate (HDR) brachytherapy. Radiother Oncol. 2016;118(1):167–172. doi:10.1016/j.radonc.2015.12.01926743834
  • Setiawan VW, Yang HP, Pike MC, et al. Type I and II endometrial cancers: have they different risk factors? J Clin Oncol. 2013;31(20):2607–2618. doi:10.1200/JCO.2012.48.259623733771
  • Christensen K, Doblhammer G, Rau R, Vaupel JW. Ageing populations: the challenges ahead. Lancet. 2009;374(9696):1196–1208. doi:10.1016/S0140-6736(09)61460-419801098
  • Wilson C, Tobin S, Young R. The exploding worldwide cancer burden. Int J Gynecol Cancer. 2004;14(1):1–11.
  • Mountzios G, Pectasides D, Bournakis E, et al. Developments in the systemic treatment of endometrial cancer. Crit Rev Oncol Hematol. 2011;79(3):278–292. doi:10.1016/j.critrevonc.2010.07.01320833559
  • Beatty MN, Blumenthal PD. The levonorgestrel-releasing intrauterine system: safety, efficacy, and patient acceptability. Ther Clin Risk Manag. 2009;5(3):561–574.19707273
  • Pal N, Broaddus RR, Urbauer DL, et al. Treatment of low-risk endometrial cancer and complex atypical hyperplasia with the levonorgestrel-releasing intrauterine device. Obstet Gynecol. 2018;131(1):109–116. doi:10.1097/AOG.000000000000239029215513
  • Behrouzi R, Ryan NAJ, Barr CE, et al. Baseline serum HE4 but not tissue HE4 expression predicts response to the levonorgestrel-releasing intrauterine system in atypical hyperplasia and early stage endometrial cancer. Cancers. 2020;12(2):276. doi:10.3390/cancers12020276
  • Westin SN, Fellman B, Sun CC, et al. Prospective Phase II trial of levonorgestrel intrauterine device: nonsurgical approach for complex atypical hyperplasia and early-stage endometrial cancer. Am J Obstet Gynecol. 2020;224(2):191–e1.32805208
  • Janda M, Robledo KP, Gebski V, et al. Complete pathological response following levonorgestrel intrauterine device in clinically stage 1 endometrial adenocarcinoma: results of a randomized clinical trial. Gynecol Oncol. 2021;161(1):143–151. doi:10.1016/j.ygyno.2021.01.02933762086
  • Gallos I, Alazzam M, Clark T, et al. Management of endometrial hyperplasia. Green-top guideline no. 67. In: RCOG/BSGE Joint Guideline. 2016. London: Royal College of Obstetricians and Gynaecologists;2016.
  • Barr CE, Crosbie EJ. Mirena coil is a suitable treatment of early-stage endometrial cancer in obese women. BJOG. 2020;127(8):1001.32350976
  • Dore M, Filoche S, Danielson K, Henry C. Efficacy of the LNG-IUS for treatment of endometrial hyperplasia and early stage endometrial cancer: can biomarkers predict response? Gynecol Oncol Rep. 2021;36:100732. doi:10.1016/j.gore.2021.10073233718563
  • Janzen DM, Rosales MA, Paik DY, et al. Progesterone receptor signaling in the microenvironment of endometrial cancer influences its response to hormonal therapy. Cancer Res. 2013;73(15):4697–4710. doi:10.1158/0008-5472.CAN-13-093023744837
  • Reyes HD, Carlson MJ, Devor EJ, et al. Downregulation of FOXO1 mRNA levels predicts treatment failure in patients with endometrial pathology conservatively managed with progestin-containing intrauterine devices. Gynecol Oncol. 2016;140(1):152–160. doi:10.1016/j.ygyno.2015.10.02326524723
  • Li W, Wang S, Qiu C, et al. Comprehensive bioinformatics analysis of acquired progesterone resistance in endometrial cancer cell line. J Transl Med. 2019;17(1):58. doi:10.1186/s12967-019-1814-630813939
  • Barros FSV, Brosens JJ, Brighton PJ. Isolation and primary culture of various cell types from whole human endometrial biopsies. Bio-Protocol. 2016;6(22):e2028. doi:10.21769/BioProtoc.2028
  • Skok K, Maver U, Gradišnik L, Kozar N, Takač I, Arko D. Endometrial cancer and its cell lines. Mol Biol Rep. 2020;47(2):1399–1411. doi:10.1007/s11033-019-05226-331848918
  • Qu W, Zhao Y, Wang X, et al. Culture characters, genetic background, estrogen/progesterone receptor expression, and tumorigenic activities of frequently used sixteen endometrial cancer cell lines. Clinica Chimica Acta. 2019;489:225–232. doi:10.1016/j.cca.2018.08.013
  • McDermott M, Eustace AJ, Busschots S, et al. In vitro development of chemotherapy and targeted therapy drug-resistant cancer cell lines: a practical guide with case studies. Front Oncol. 2014;4:40. doi:10.3389/fonc.2014.0004024639951
  • Henry C, Llamosas E, Knipprath-Meszaros A, et al. Targeting the ROR1 and ROR2 receptors in epithelial ovarian cancer inhibits cell migration and invasion. Oncotarget. 2015;6(37):40310–40326. doi:10.18632/oncotarget.564326515598
  • Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9(7):671–675. doi:10.1038/nmeth.208922930834
  • Gebäck T, Schulz MMP, Koumoutsakos P, Detmar M. TScratch: a novel and simple software tool for automated analysis of monolayer wound healing assays. BioTechniques. 2009;46(4):265–274. doi:10.2144/00011308319450233
  • Vinci M, Box C, Eccles SA. Three-dimensional (3D) tumor spheroid invasion assay. J Vis Exp. 2015;99:e52686.
  • Henry C, Quadir A, Hawkins NJ, et al. Expression of the novel Wnt receptor ROR2 is increased in breast cancer and may regulate both β-catenin dependent and independent Wnt signalling. J Cancer Res Clin Oncol. 2015;141(2):243–254. doi:10.1007/s00432-014-1824-y25209439
  • Henry C, Hacker N, Ford C. Silencing ROR1 and ROR2 inhibits invasion and adhesion in an organotypic model of ovarian cancer metastasis. Oncotarget. 2017;8(68):112727–112738. doi:10.18632/oncotarget.2255929348860
  • Chandrashekar DS, Bashel B, Balasubramanya SAH, et al. UALCAN: a portal for facilitating tumour subgroup gene expression and survival analyses. Neoplasia. 2017;19(8):659. doi:10.1016/j.neo.2017.05.00228739011
  • Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15(3):178–196.24556840
  • Feitelson MA, Arzumanyan A, Kulathinal RJ, et al. Sustained proliferation in cancer: mechanisms and novel therapeutic targets. Semin Cancer Biol. 2015;35:S25–S54.25892662
  • Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29(34):4741–4751. doi:10.1038/onc.2010.21520531305
  • Ørbo A, Arnes M, Lyså LM, Borgfelt C, Straume B. HE4 is a novel tissue marker for therapy response and progestin resistance in medium-and low-risk endometrial hyperplasia. Br J Cancer. 2016;115(6):725–730. doi:10.1038/bjc.2016.24727537387
  • Bedard PL, Hansen AR, Ratain MJ, Siu LL. Tumour heterogeneity in the clinic. Nature. 2013;501(7467):355–364. doi:10.1038/nature1262724048068
  • Stanta G, Bonin S. Overview on clinical relevance of intra-tumor heterogeneity. Front Med. 2018;5(85). doi:10.3389/fmed.2018.00085
  • Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338–345. doi:10.1038/nature1262524048066
  • Ghaleb AM, Yang VW. Krüppel-like factor 4 (KLF4): what we currently know. Gene. 2017;611:27–37. doi:10.1016/j.gene.2017.02.02528237823
  • Lung HL, Kan R, Chau WY, et al. The anti-tumor function of the IKK inhibitor PS1145 and high levels of p65 and KLF4 are associated with the drug resistance in nasopharyngeal carcinoma cells. Sci Rep. 2019;12064(9):1.
  • Jiang Z, Zhang Y, Chen X, Wu P, Chen D. Long non-coding RNA LINC00673 silencing inhibits proliferation and drug resistance of prostate cancer cells via decreasing KLF4 promoter methylation. J Cell Mol Med. 2020;24(2):1878–1892. doi:10.1111/jcmm.1488331881124
  • Deng X, Kong F, Li S, et al. A KLF4/PiHL/EZH2/HMGA2 regulatory axis and its function in promoting oxaliplatin-resistance of colorectal cancer. Cell Death Dis. 2021;485(12):1–3.
  • Jia Y, Zhou J, Luo X, et al. KLF4 overcomes tamoxifen resistance by suppressing MAPK signaling pathway and predicts good prognosis in breast cancer. Cell Signal. 2018;42:165–175. doi:10.1016/j.cellsig.2017.09.02528988130
  • Lee SI, Kim DK, Seo EJ, et al. Role of Krüppel-like factor 4 in the maintenance of chemoresistance of anaplastic thyroid Cancer. Thyroid. 2017;27(11):1424–1432. doi:10.1089/thy.2016.041428920531
  • Schoenhals M, Kassambara A, Veyrune JL, et al. Krüppel-like factor 4 blocks tumor cell proliferation and promotes drug resistance in multiple myeloma. Haematologica. 2013;98(9):1442. doi:10.3324/haematol.2012.06694423585530
  • Shimizu Y, Takeuchi T, Mita S, et al. Krüppel-like factor 4 mediates anti-proliferative effects of progesterone with G0/G1 arrest in human endometrial epithelial cells. J Endocrinol Invest. 2010;33(10):745–750. doi:10.1007/BF0334668120479568
  • Ghaleb AM, Katz JP, Kaestner KH, et al. Krüppel-like factor 4 exhibits antiapoptotic activity following gamma-radiation-induced DNA damage. Oncogene. 2007;26(16):2365–2373. doi:10.1038/sj.onc.121002217016435
  • Rowland BD, Bernards R, Peeper DS. The KLF4 tumour suppressor is a transcriptio na l repressor of p53 that acts as a context-dependent oncogene. Nat Cell Biol. 2005;7(11):1074–1082. doi:10.1038/ncb131416244670
  • Eaton JL, Unno K, Caraveo M, et al. Increased AKT or MEK1/2 activity influences progesterone receptor levels and localization in endometriosis. J Clin Endocrinol Metab. 2013;98(12):E1871–E18. doi:10.1210/jc.2013-166124064688
  • Akesson E, Gallos ID, Ganesan R, et al. Prognostic significance of estrogen and progesterone receptor expression in LNG-IUS (Mirena®) treatment of endometrial hyperplasia: an immunohistochemical study. Acta Obstet Gynecol Scand. 2010;89(3):393–398. doi:10.3109/0001634090355600620199355
  • Fawzy M, Mosbah A, Zalata K, et al. Predictors of progestin therapy response in endometrial hyperplasia: an Immunohistochemical Study. Egypt J Fertil Steril. 2016;20(2):6–11. doi:10.21608/egyfs.2016.19528
  • Tang G, Liu D, Xiao G, et al. Transcriptional repression of FOXO1 by KLF4 contribute s to glioma progression. Oncotarget. 2016;7(49):81757–81767. doi:10.18632/oncotarget.1318427835585
  • Uhlén M, Fagerberg L, Hallstrom BM. Tissue-based map of the human proteome. Science. 2015;347(6220):1260419. PubMed: 25613900. doi:10.1126/science.126041925613900
  • Sato S, Maekawa R, Tamura I, et al. SATB2 and NGR1: potential upstream regulatory factors in uterine leiomyo mas. J Assist Reprod Genet. 2019;36(11):2385–2397. doi:10.1007/s10815-019-01582-y31728810
  • Moh M, Krings G, Ates D, Aysal A, Kim GE, Rabban JT. SATB2 expression distinguishes ovarian metastases of colorectal and appendiceal origin from primary ovarian tumors of mucinous or endometrioid type. Am J Surg Pathol. 2016;40(3):419–432. doi:10.1097/PAS.000000000000055326551622
  • Sletten ET, Smaglyukova N, Ørbo A, Sager G. Expression of nuclear progesterone receptors (nPRs), membrane progesterone receptors (mPRs) and progesterone receptor membrane components (PGRMCs) in the human endometrium after 6 months levonorgestrel low dose intrauterine therapy. J Steroid Biochem Mol Biol. 2020;202:105701. doi:10.1016/j.jsbmb.2020.10570132479983
  • Patel B, Elguero S, Thakore S, Dahoud W, Bedaiwy M, Mesiano S. Role of nuclear progesterone receptor isoforms in uterine pathophysiology. Hum Reprod Update. 2015;21(2):155–173. doi:10.1093/humupd/dmu05625406186
  • Mote PA, Johnston JF, Manninen T, Tuohimaa P, Clarke CL. Detection of progesterone receptor forms A and B by immunohistochemical analysis. J Clin Pathol. 2001;54(8):624–630. doi:10.1136/jcp.54.8.62411477119
  • Yang X, Kui L, Tang M, et al. High-throughput transcriptome profiling in drug and biomarker discovery. Front Genet. 2020;11:19. doi:10.3389/fgene.2020.0001932117438
  • Kapałczyńska M, Kolenda T, Przybyła W, et al. 2D and 3D cell cultures - a comparison of different types of cancer cell cultures. Arch Med Sci. 2018;14(4):910–919.30002710