117
Views
3
CrossRef citations to date
0
Altmetric
Original Research

OSU-03012 Disrupts Akt Signaling and Prevents Endometrial Carcinoma Progression in vitro and in vivo

, , , , , , & show all
Pages 1797-1810 | Published online: 30 Apr 2021

References

  • RahibL, SmithB, AizenbergR, RosenzweigA, FleshmanJ, MatrisianL. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74(11):2913–2921. doi:10.1158/0008-5472.CAN-14-015524840647
  • MakkerV, TaylorM, AghajanianC, et al. Lenvatinib plus pembrolizumab in patients with advanced endometrial cancer. J Clin Oncol. 2020;38(26):2981–2992. doi:10.1200/JCO.19.0262732167863
  • FlemingG. Systemic chemotherapy for uterine carcinoma: metastatic and adjuvant. J Clin Oncol. 2007;25(20):2983–2990. doi:10.1200/JCO.2007.10.843117617530
  • BashoR, GilcreaseM, MurthyR, et al. Targeting the PI3K/AKT/mTOR pathway for the treatment of mesenchymal triple-negative breast cancer: evidence from a phase 1 trial of mTOR inhibition in combination with liposomal doxorubicin and bevacizumab. JAMA Oncol. 2017;3(4):509–515. doi:10.1001/jamaoncol.2016.528127893038
  • ShiN, YuH, ChenT. Inhibition of esophageal cancer growth through the suppression of PI3K/AKT/mTOR signaling pathway. Onco Targets Ther. 2019;12:7637–7647. doi:10.2147/OTT.S20545731571914
  • RoncolatoF, LindemannK, WillsonM, MartynJ, MileshkinL. PI3K/AKT/mTOR inhibitors for advanced or recurrent endometrial cancer. Cochrane Database Syst Rev. 2019;10. doi:10.1002/14651858.CD012160.pub2
  • EdiriweeraM, TennekoonK, SamarakoonS. Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: biological and therapeutic significance. Semin Cancer Biol. 2019;59:147–160. doi:10.1016/j.semcancer.2019.05.01231128298
  • QianD, XiaoX, ByunJ, et al. PI3K/Akt/mTOR signaling and plasma membrane proteins are implicated in responsiveness to adjuvant dendritic cell vaccination for metastatic colorectal cancer. Clin Cancer Res. 2017;23(2):399–406. doi:10.1158/1078-0432.CCR-16-062327435399
  • JohnsenJ, SegerströmL, OrregoA, et al. Inhibitors of mammalian target of rapamycin downregulate MYCN protein expression and inhibit neuroblastoma growth in vitro and in vivo. Oncogene. 2008;27(20):2910–2922. doi:10.1038/sj.onc.121093818026138
  • OpelD, PorembaC, SimonT, DebatinK, FuldaS. Activation of Akt predicts poor outcome in neuroblastoma. Cancer Res. 2007;67(2):735–745. doi:10.1158/0008-5472.CAN-06-220117234785
  • KandothC, SchultzN, CherniackA, et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67–73. doi:10.1038/nature1211323636398
  • SalvesenH, CarterS, MannelqvistM, et al. Integrated genomic profiling of endometrial carcinoma associates aggressive tumors with indicators of PI3 kinase activation. Proc Natl Acad Sci U S A. 2009;106(12):4834–4839. doi:10.1073/pnas.080651410619261849
  • BansalN, YendluriV, WenhamR. The molecular biology of endometrial cancers and the implications for pathogenesis, classification, and targeted therapies. Cancer Control. 2009;16(1):8–13. doi:10.1177/10732748090160010219078924
  • LiuZ, HongZ, QuP. Proteomic analysis of human endometrial tissues reveals the roles of PI3K/AKT/mTOR pathway and tumor angiogenesis molecules in the pathogenesis of endometrial cancer. Biomed Res Int. 2020;2020:5273969.32908897
  • UegakiK, KanamoriY, KigawaJ, et al. PTEN-positive and phosphorylated-Akt-negative expression is a predictor of survival for patients with advanced endometrial carcinoma. Oncol Rep. 2005;14(2):389–392.16012720
  • Megino-LuqueC, MoiolaC, Molins-EscuderC, et al. Small-molecule inhibitors (SMIs) as an effective therapeutic strategy for endometrial cancer. Cancers. 2020;12(10):2751. doi:10.3390/cancers12102751
  • ZhuJ, HuangJ, TsengP, et al. From the cyclooxygenase-2 inhibitor celecoxib to a novel class of 3-phosphoinositide-dependent protein kinase-1 inhibitors. Cancer Res. 2004;64(12):4309–4318. doi:10.1158/0008-5472.CAN-03-406315205346
  • JohnsonA, SmithL, ZhuJ, et al. A novel celecoxib derivative, OSU03012, induces cytotoxicity in primary CLL cells and transformed B-cell lymphoma cell line via a caspase- and Bcl-2-independent mechanism. Blood. 2005;105(6):2504–2509. doi:10.1182/blood-2004-05-195715454489
  • MurataM, NaraharaS, KawanoT, et al. Design and function of engineered protein nanocages as a drug delivery system for targeting pancreatic cancer cells via neuropilin-1. Mol Pharm. 2015;12(5):1422–1430. doi:10.1021/mp500712925811429
  • LiuJ, QinC, LvW, ZhaoQ, QinC. OSU-03012, a non-Cox inhibiting celecoxib derivative, induces apoptosis of human esophageal carcinoma cells through a p53/Bax/cytochrome c/caspase-9-dependent pathway. Anticancer Drugs. 2013;24(7):690–698. doi:10.1097/CAD.0b013e328362469f23652278
  • WestN, Garcia-VargasA, ChalfantC, ParkM. OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role for Nck1 but not Nck2. BMC Cancer. 2013;13:256. doi:10.1186/1471-2407-13-25623706161
  • GaoM, YehP, LuY, et al. OSU-03012, a novel celecoxib derivative, induces reactive oxygen species-related autophagy in hepatocellular carcinoma. Cancer Res. 2008;68(22):9348–9357. doi:10.1158/0008-5472.CAN-08-164219010909
  • KucabJ, LeeC, ChenC, et al. Celecoxib analogues disrupt Akt signaling, which is commonly activated in primary breast tumours. Breast Cancer Res. 2005;7(5):R796–807. doi:10.1186/bcr129416168126
  • MateoJ, De BonoJS, RamanathanRK, et al. A first-in-human phase I trial of AR-12, a PDK-1 inhibitor, in patients with advanced solid tumors. 2013.
  • Muñoz-AlonsoM, AcostaJ, RichardC, DelgadoM, SedivyJ, LeónJ. p21Cip1 and p27Kip1 induce distinct cell cycle effects and differentiation programs in myeloid leukemia cells. J Biol Chem. 2005;280(18):18120–18129. doi:10.1074/jbc.M50075820015746092
  • KalkavanH, GreenD. MOMP, cell suicide as a BCL-2 family business. Cell Death Differ. 2018;25(1):46–55. doi:10.1038/cdd.2017.17929053143
  • NagarajN, AnilakumarK, SinghO. Diallyl disulfide causes caspase-dependent apoptosis in human cancer cells through a Bax-triggered mitochondrial pathway. J Nutr Biochem. 2010;21(5):405–412. doi:10.1016/j.jnutbio.2009.01.01519423321
  • JiangX, WangX. Cytochrome C-mediated apoptosis. Annu Rev Biochem. 2004;73:87–106. doi:10.1146/annurev.biochem.73.011303.07370615189137
  • WangY, KulpS, WangD, et al. Targeting endoplasmic reticulum stress and Akt with OSU-03012 and gefitinib or erlotinib to overcome resistance to epidermal growth factor receptor inhibitors. Cancer Res. 2008;68(8):2820–2830. doi:10.1158/0008-5472.CAN-07-133618413750
  • WengS, KashidaY, KulpS, et al. Sensitizing estrogen receptor-negative breast cancer cells to tamoxifen with OSU-03012, a novel celecoxib-derived phosphoinositide-dependent protein kinase-1/Akt signaling inhibitor. Mol Cancer Ther. 2008;7(4):800–808. doi:10.1158/1535-7163.MCT-07-043418413793
  • SalvesenH, HaldorsenI, TrovikJ. Markers for individualised therapy in endometrial carcinoma. Lancet Oncol. 2012;13(8):e353–e361. doi:10.1016/S1470-2045(12)70213-922846840
  • CatasusL, GallardoA, CuatrecasasM, PratJ. Concomitant PI3K-AKT and p53 alterations in endometrial carcinomas are associated with poor prognosis. Mod Pathol. 2009;22(4):522–529. doi:10.1038/modpathol.2009.519234438
  • DedesK, WetterskogD, AshworthA, KayeS, Reis-FilhoJ. Emerging therapeutic targets in endometrial cancer. Nat Rev Clin Oncol. 2011;8(5):261–271. doi:10.1038/nrclinonc.2010.21621221135
  • PoreS, HahmE, KimS, et al. A novel sulforaphane-regulated gene network in suppression of breast cancer-induced osteolytic bone resorption. Mol Cancer Ther. 2020;19(2):420–431. doi:10.1158/1535-7163.MCT-19-061131784454
  • NoguchiK, KitanakaC, YamanaH, KokubuA, MochizukiT, KuchinoY. Regulation of c-Myc through phosphorylation at Ser-62 and Ser-71 by c-Jun N-terminal kinase. J Biol Chem. 1999;274(46):32580–32587. doi:10.1074/jbc.274.46.3258010551811
  • SalomonsG, BradyH, Verwijs-JanssenM, et al. The Bax alpha:Bcl-2 ratio modulates the response to dexamethasone in leukaemic cells and is highly variable in childhood acute leukaemia. Int J Cancer. 1997;71(6):959–965. doi:10.1002/(SICI)1097-0215(19970611)71:6<959::AID-IJC9>3.0.CO;2-X9185697
  • SusinS, LorenzoH, ZamzamiN, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature. 1999;397(6718):441–446. doi:10.1038/171359989411
  • Khosravi-FarR, EspostiM. Death receptor signals to mitochondria. Cancer Biol Ther. 2004;3(11):1051–1057. doi:10.4161/cbt.3.11.117315640619
  • ZouH, LiY, LiuX, WangX. An APAF-1.cytochrome c multimeric complex is a functional apoptosome that activates procaspase-9. J Biol Chem. 1999;274(17):11549–11556. doi:10.1074/jbc.274.17.1154910206961
  • KaleJ, OsterlundE, AndrewsD. BCL-2 family proteins: changing partners in the dance towards death. Cell Death Differ. 2018;25(1):65–80. doi:10.1038/cdd.2017.18629149100
  • RosséT, OlivierR, MonneyL, et al. Bcl-2 prolongs cell survival after Bax-induced release of cytochrome c. Nature. 1998;391(6666):496–499. doi:10.1038/351609461218
  • ThornberryN, LazebnikY. Caspases: enemies within. Science. 1998;281(5381):1312–1316. doi:10.1126/science.281.5381.13129721091
  • RomanoG. The role of the dysfunctional Akt-related pathway in cancer: establishment and maintenance of a malignant cell phenotype, resistance to therapy, and future strategies for drug development. Scientifica. 2013;2013:317186. doi:10.1155/2013/31718624381788
  • NitulescuG, Van De VenterM, NitulescuG, et al. The Akt pathway in oncology therapy and beyond (Review). Int J Oncol. 2018;53(6):2319–2331. doi:10.3892/ijo.2018.459730334567
  • HaD, LeeA. Insulin-like growth factor 1-receptor signaling stimulates GRP78 expression through the PI3K/AKT/mTOR/ATF4 axis. Cell Signal. 2020;75:109736. doi:10.1016/j.cellsig.2020.10973632805346
  • YangJ, NieJ, MaX, WeiY, PengY, WeiX. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer. 2019;18(1):26.30782187
  • Rodriguez-CupelloC, DamM, SeriniL, et al. The STRIPAK complex regulates response to chemotherapy through p21 and p27. Front Cell Dev Biol. 2020;8:146. doi:10.3389/fcell.2020.0014632258031