Review Article

极光激酶在化学敏感、化学耐药癌症中的致癌信号传导的最新进展:针对极光激酶的新药物化学方法

卷 31, 期 23, 2024

发表于: 07 July, 2023

页: [3502 - 3528] 页: 27

弟呕挨: 10.2174/0929867330666230503124408

价格: $65

conference banner
摘要

极光激酶家族(AKI)由丝氨酸-苏氨酸蛋白激酶组成,参与细胞周期和有丝分裂的调节。这些激酶是调节遗传相关数据的粘附性所必需的。该家族成员可分为极光激酶A (Ark-A)、极光激酶B (Ark-B)和极光激酶C (Ark-C),由高度保守的苏氨酸蛋白激酶组成。这些激酶可以调节细胞分裂过程中的纺锤体组装、检查点通路和细胞质分裂。本综述的主要目的是探讨极光激酶在化学敏感/化学耐药癌症中的致癌信号传导的最新进展,并探讨针对这些激酶的各种药物化学方法。我们检索了Pubmed、Scopus、NLM、Pubchem和Relemed,以获得与极光激酶的最新信号作用和药物化学方法相关的信息,并讨论了每种极光激酶及其下游信号级联在几种化学敏感/化学耐药癌症进展中的最新作用;随后,我们讨论了天然产物(金黄紫堇碱, 紫堇灵,橙皮苷,杰多霉素,非瑟酮)和合成的药物化学分子作为极光激酶抑制剂(AKIs)。几种天然产物在化疗致敏和化疗耐药癌症中的疗效被解释为AKIs。例如,新型三唑类分子已被用于治疗胃癌,而氰吡啶类分子被用于治疗结直肠癌,三氟乙酸衍生物可用于治疗食管癌。此外,喹诺酮肼衍生物可用于针对乳腺癌和宫颈癌。相比之下,吲哚衍生物可优选用于口腔癌,而硫代氨基脲-吲哚可用于治疗前列腺癌,正如早期针对癌细胞的研究所报道的那样。此外,这些化学衍生物可以通过临床前研究作为AKIs进行检测。此外,通过这些药物化学底物在实验室中利用硅和合成途径合成新型AKIs可能有助于开发针对化疗耐药癌症的前瞻性新型AKIs。这项研究有助于肿瘤学家、化学家和药物化学家探索新的化学片段合成,以特异性靶向几种耐药癌细胞类型的极光激酶肽序列。

关键词: 极光激酶,下游信号传导,癌症,化学致敏,化学耐药,天然产物,药物化学。

[1]
Health enews-A news service from Advocate Aurora Health. Available from: https://www.ahchealthenews.com/(Accessed on: 2021 Jul 8).
[2]
Koh, H.M.; Jang, B.G.; Hyun, C.L.; Kim, Y.S.; Hyun, J.W.; Chang, W.Y.; Maeng, Y.H. Aurora Kinase A is a prognostic marker in colorectal adenocarcinoma. J. Pathol. Transl. Med., 2017, 51(1), 32-39.
[http://dx.doi.org/10.4132/jptm.2016.10.17] [PMID: 28013532]
[3]
Katsha, A.; Belkhiri, A.; Goff, L.; El-Rifai, W. Aurora kinase A in gastrointestinal cancers: Time to target. Mol. Cancer, 2015, 14(1), 106.
[http://dx.doi.org/10.1186/s12943-015-0375-4] [PMID: 25987188]
[4]
Goos, J.A.C.M.; Coupe, V.M.H.; Diosdado, B.; Delis-Van Diemen, P.M.; Karga, C.; Beliën, J.A.M.; Carvalho, B.; van den Tol, M.P.; Verheul, H.M.W.; Geldof, A.A.; Meijer, G.A.; Hoekstra, O.S.; Fijneman, R.J.A. Aurora kinase A (AURKA) expression in colorectal cancer liver metastasis is associated with poor prognosis. Br. J. Cancer, 2013, 109(9), 2445-2452.
[http://dx.doi.org/10.1038/bjc.2013.608] [PMID: 24104968]
[5]
Kivinummi, K.; Urbanucci, A.; Leinonen, K.; Tammela, T.L.J.; Annala, M.; Isaacs, W.B.; Bova, G.S.; Nykter, M.; Visakorpi, T. The expression of AURKA is androgen regulated in castration-resistant prostate cancer. Sci. Rep., 2017, 7(1), 17978.
[http://dx.doi.org/10.1038/s41598-017-18210-3] [PMID: 29269934]
[6]
Lo Iacono, M.; Monica, V.; Saviozzi, S.; Ceppi, P.; Bracco, E.; Papotti, M.; Scagliotti, G.V. Aurora Kinase A expression is associated with lung cancer histological-subtypes and with tumor de-differentiation. J. Transl. Med., 2011, 9(1), 100.
[http://dx.doi.org/10.1186/1479-5876-9-100] [PMID: 21718475]
[7]
D’Assoro, A.B.; Haddad, T.; Galanis, E. Aurora-A Kinase as a promising therapeutic target in cancer. Front. Oncol., 2016, 5(1), 295.
[http://dx.doi.org/10.3389/fonc.2015.00295] [PMID: 26779440]
[8]
Baldini, E; D’Armiento, M; Ulisse, S. A new aurora in anaplastic thyroid cancer therapy. Int J Endocrinol., 2014, 2014, 816430.
[http://dx.doi.org/10.1155/2014/816430]
[9]
Tang, A.; Gao, K.; Chu, L.; Zhang, R.; Yang, J.; Zheng, J. Aurora kinases: Novel therapy targets in cancers. Oncotarget, 2017, 8(14), 23937-23954.
[http://dx.doi.org/10.18632/oncotarget.14893]
[10]
Goldenson, B.; Crispino, J.D. The aurora kinases in cell cycle and leukemia. Oncogene, 2015, 34(5), 537-545.
[http://dx.doi.org/10.1038/onc.2014.14] [PMID: 24632603]
[11]
Mobley, A; Zhang, S; Bondaruk, J; Wang, Y; Majewski, T; Caraway, NP Aurora Kinase A is a biomarker for bladder cancer detection and contributes to its aggressive behavior. Sci Rep., 2017, 7, 40714.
[http://dx.doi.org/10.1038/srep40714]
[12]
Eterno, V.; Zambelli, A.; Villani, L.; Tuscano, A.; Manera, S.; Spitaleri, A.; Pavesi, L.; Amato, A. AURKA controls self-renewal of breast cancer-initiating cells promoting wnt3a stabilization through suppression of miR-128. Sci. Rep., 2016, 6(1), 28436.
[http://dx.doi.org/10.1038/srep28436] [PMID: 27341528]
[13]
Mignogna, C.; Staropoli, N.; Botta, C.; De Marco, C.; Rizzuto, A.; Morelli, M.; Di Cello, A.; Franco, R.; Camastra, C.; Presta, I.; Malara, N.; Salvino, A.; Tassone, P.; Tagliaferri, P.; Barni, T.; Donato, G.; Di Vito, A. Aurora Kinase A expression predicts platinum-resistance and adverse outcome in high-grade serous ovarian carcinoma patients. J. Ovarian Res., 2016, 9(1), 31.
[http://dx.doi.org/10.1186/s13048-016-0238-7] [PMID: 27209210]
[14]
Katsha, A.; Arras, J.; Soutto, M.; Belkhiri, A.; El-Rifai, W. AURKA regulates JAK2-STAT3 activity in human gastric and esophageal cancers. Mol. Oncol., 2014, 8(8), 1419-1428.
[http://dx.doi.org/10.1016/j.molonc.2014.05.012] [PMID: 24953013]
[15]
Zhang, H.; Bao, J.; Zhao, S.; Huo, Z.; Li, B. MicroRNA-490-3p suppresses hepatocellular carcinoma cell proliferation and migration by targeting the aurora kinase A gene (AURKA). Arch. Med. Sci., 2020, 16(2), 395-406.
[http://dx.doi.org/10.5114/aoms.2019.91351] [PMID: 32190151]
[16]
Furukawa, T.; Kanai, N.; Shiwaku, H.O.; Soga, N.; Uehara, A.; Horii, A. AURKA is one of the downstream targets of MAPK1/ERK2 in pancreatic cancer. Oncogene, 2006, 25(35), 4831-4839.
[http://dx.doi.org/10.1038/sj.onc.1209494] [PMID: 16532023]
[17]
Pokuri V.K.; Opyrchal M.; Boland P.M. Aurora Kinase A and gastrointestinal malignancies. Int. J. Cancer Res. Mol. Mech., 2015, 1(3), 1-7.
[18]
Yamada, M; Hirotsune, S; Wynshaw-Boris, A The essential role of LIS1, NDEL1 and Aurora-A in polarity formation and microtubule organization during neurogensis. Cell Adh Migr., 2010, 4(2), 180.
[19]
Wang, F.; Wang, L.; Fisher, L.A.; Li, C.; Wang, W.; Peng, A. Phosphatase 1 nuclear targeting subunit (PNUTS) regulates aurora kinases and mitotic progression. Mol. Cancer Res., 2019, 17(1), 10-19.
[http://dx.doi.org/10.1158/1541-7786.MCR-17-0670] [PMID: 30190438]
[20]
Huang, Y.; Li, T.; Ems-McClung, S.C.; Walczak, C.E.; Prigent, C.; Zhu, X.; Zhang, X.; Zheng, Y. Aurora A activation in mitosis promoted by BuGZ. J. Cell Biol., 2018, 217(1), 107-116.
[http://dx.doi.org/10.1083/jcb.201706103] [PMID: 29074706]
[21]
Liu, L.; Guo, C.; Dammann, R.; Tommasi, S.; Pfeifer, G.P. RASSF1A interacts with and activates the mitotic kinase Aurora-A. Oncogenes, 2008, 27(47), 6175-6186.
[http://dx.doi.org/10.1038/onc.2008.220]
[22]
Satinover, D.L.; Leach, C.A.; Stukenberg, P.T.; Brautigan, D.L. Activation of Aurora-A kinase by protein phosphatase inhibitor-2, a bifunctional signaling protein. Proc. Natl. Acad. Sci. USA, 2004, 101(23), 8625-8630.
[http://dx.doi.org/10.1073/pnas.0402966101] [PMID: 15173575]
[23]
Zhao, Z.; Lim, J.P.; Ng, Y.W.; Lim, L.; Manser, E. The GIT-associated kinase PAK targets to the centrosome and regulates Aurora-A. Mol. Cell, 2005, 20(2), 237-249.
[http://dx.doi.org/10.1016/j.molcel.2005.08.035] [PMID: 16246726]
[24]
Shao, S.; Wang, Y.; Jin, S.; Song, Y.; Wang, X.; Fan, W.; Zhao, Z.; Fu, M.; Tong, T.; Dong, L.; Fan, F.; Xu, N.; Zhan, Q. Gadd45a interacts with aurora-A and inhibits its kinase activity. J. Biol. Chem., 2006, 281(39), 28943-28950.
[http://dx.doi.org/10.1074/jbc.M600235200] [PMID: 16772293]
[25]
Interaction and feedback regulation between STK15/BTAK/Aurora-A kinase and protein phosphatase 1 through mitotic cell division cycle. Early Detection Research Network, Available from: https://edrn.nci.nih.gov/data-and-resources/publications/11551964-interaction-and-feedback-regulation (Accessed on: 2022 Aug 22).
[26]
Sarkissian, M.; Mendez, R.; Richter, J.D. Progesterone and insulin stimulation of CPEB-dependent polyadenylation is regulated by Aurora A and glycogen synthase kinase-3. Genes Dev, 2004, 18(1), 48.
[27]
Johnson, E.O.; Chang, K.H.; de Pablo, Y.; Ghosh, S.; Mehta, R.; Badve, S.; Shah, K. PHLDA1 is a crucial negative regulator and effector of Aurora A kinase in breast cancer. J. Cell Sci., 2011, 124(16), 2711-2722.
[http://dx.doi.org/10.1242/jcs.084970] [PMID: 21807936]
[28]
Cheng, A.; Zhang, P.; Wang, B.; Yang, D.; Duan, X.; Jiang, Y.; Xu, T.; Jiang, Y.; Shi, J.; Ding, C.; Wu, G.; Sang, Z.; Wu, Q.; Wang, H.; Wu, M.; Zhang, Z.; Pan, X.; Pan, Y.; Gao, P.; Zhang, H.; Zhou, C.; Guo, J.; Yang, Z. Aurora-A mediated phosphorylation of LDHB promotes glycolysis and tumor progression by relieving the substrate-inhibition effect. Nat. Commun., 2019, 10(1), 5566.
[http://dx.doi.org/10.1038/s41467-019-13485-8] [PMID: 31804482]
[29]
Mandati, V.; Maestro, L.; Del; Dingli, F.; Lombard, B.; Loew, D.; Molinie, N. Phosphorylation of Merlin by Aurora A kinase appears necessary for mitotic progression. J. Biol. Chem., 2019, 294(35), 12992.
[30]
Wang-Bishop, L.; Chen, Z.; Gomaa, A.; Lockhart, A.C.; Salaria, S.; Wang, J.; Lewis, K.B.; Ecsedy, J.; Washington, K.; Beauchamp, R.D.; El-Rifai, W. Inhibition of AURKA reduces proliferation and survival of gastrointestinal cancer cells with activated KRAS by preventing activation of RPS6KB1. Gastroenterology, 2019, 156(3), 662-675.e7.
[http://dx.doi.org/10.1053/j.gastro.2018.10.030] [PMID: 30342037]
[31]
Shi, Y.; Solomon, L.R.; Pereda-Lopez, A.; Giranda, V.L.; Luo, Y.; Johnson, E.F.; Shoemaker, A.R.; Leverson, J.; Liu, X. Ubiquitin-specific cysteine protease 2a (USP2a) regulates the stability of Aurora-A. J. Biol. Chem., 2011, 286(45), 38960-38968.
[http://dx.doi.org/10.1074/jbc.M111.231498] [PMID: 21890637]
[32]
Zhao, J.W.; Wu, Z.H.; Guo, J.W.; Huang, M.J.; You, Y.Z.; Liu, H.M. Synthesis and anti-gastric cancer activity evaluation of novel triazole nucleobase analogues containing steroidal/coumarin/quinoline moieties. Eur J Med Chem., 2019, 181, 111520.
[33]
Xu, L.; Shi, L.; Qiu, S.; Chen, S.; Lin, M.; Xiang, Y.; Zhao, C.; Zhu, J.; Shen, L.; Zuo, Z. Design, synthesis, and evaluation of cyanopyridines as anti-colorectal cancer agents via inhibiting STAT3 pathway. Drug Des. Devel. Ther., 2019, 13, 3369-3381.
[http://dx.doi.org/10.2147/DDDT.S217800] [PMID: 31576111]
[34]
Ke, Y.; Hu, T.X.; Huo, J.F.; Yan, J.K.; Wang, J.Y.; Yang, R.H.; Xie, H.; Liu, Y.; Wang, N.; Zheng, Z.J.; Sun, Y.X.; Wang, C.; Du, J.; Liu, H.M. Synthesis and in vitro biological evaluation of novel derivatives of Flexicaulin A condensation with amino acid trifluoroacetate. Eur. J. Med. Chem., 2019, 182, 111645.
[http://dx.doi.org/10.1016/j.ejmech.2019.111645] [PMID: 31494472]
[35]
Bingul, M.; Tan, O.; Gardner, C.; Sutton, S.; Arndt, G.; Marshall, G.; Cheung, B.; Kumar, N.; Black, D. Synthesis, characterization and anti-cancer activity of hydrazide derivatives incorporating a quinoline moiety. Molecules, 2016, 21(7), 916.
[http://dx.doi.org/10.3390/molecules21070916] [PMID: 27428941]
[36]
Li, X.; Ding, J.; Li, N.; Liu, W.; Ding, F.; Zheng, H.; Ning, Y.; Wang, H.; Liu, R.; Ren, S. Synthesis and biological evaluation of celastrol derivatives as anti-ovarian cancer stem cell agents. Eur. J. Med. Chem., 2019, 179, 667-679.
[http://dx.doi.org/10.1016/j.ejmech.2019.06.086] [PMID: 31279299]
[37]
Xu, Y.; Zhang, X.J.; Li, W.B.; Wang, X.R.; Wang, S.; Qiao, X.P.; Chen, S.W. Design, synthesis and biological evaluation of indole-2-one derivatives as potent BRD4 inhibitors. Eur. J. Med. Chem., 2020, 208, 112780.
[http://dx.doi.org/10.1016/j.ejmech.2020.112780] [PMID: 32883643]
[38]
Li, J.; Zheng, T.; Jin, Y.; Xu, J.; Yu, J.; Lv, Y. Synthesis, molecular docking and biological evaluation of quinolone derivatives as novel anticancer agents. Chem. Pharm. Bull., 2018, 66(1), 55-60.
[http://dx.doi.org/10.1248/cpb.c17-00035] [PMID: 29118308]
[39]
He, Z.X.; Huo, J.L.; Gong, Y.P.; An, Q.; Zhang, X.; Qiao, H.; Yang, F.F.; Zhang, X.H.; Jiao, L.M.; Liu, H.M.; Ma, L.Y.; Zhao, W. Design, synthesis and biological evaluation of novel thiosemicarbazone-indole derivatives targeting prostate cancer cells. Eur. J. Med. Chem., 2021, 210, 112970.
[http://dx.doi.org/10.1016/j.ejmech.2020.112970] [PMID: 33153765]
[40]
Yang, N.; Wang, C.; Wang, Z.; Zona, S.; Lin, S-X.; Wang, X.; Yan, M.; Zheng, F-M.; Li, S-S.; Xu, B.; Bella, L.; Yong, J-S.; Lam, E.W-F.; Liu, Q. FOXM1 recruits nuclear Aurora kinase A to participate in a positive feedback loop essential for the self-renewal of breast cancer stem cells. Oncogene, 2017, 36(24), 3428-3440.
[http://dx.doi.org/10.1038/onc.2016.490] [PMID: 28114286]
[41]
Tang, J.; Yang, L.; Li, Y.; Ning, X.; Chaulagain, A.; Wang, T.; Wang, D. ARID3A promotes the development of colorectal cancer by upregulating AURKA. Carcinogenesis, 2021, 42(4), 578-586.
[http://dx.doi.org/10.1093/carcin/bgaa118] [PMID: 33165575]
[42]
Long, Q.; An, X.; Chen, M.; Wang, N.; Sui, S.; Li, Y. PUF60/AURKA axis contributes to tumor progression and malignant phenotypes in bladder cancer. Front Oncol., 2020, 10, 568015.
[43]
Tanaka, M.; Ueda, A.; Kanamori, H.; Ideguchi, H.; Yang, J.; Kitajima, S.; Ishigatsubo, Y. Cell-cycle-dependent regulation of human aurora A transcription is mediated by periodic repression of E4TF1. J. Biol. Chem., 2002, 277(12), 10719-10726.
[http://dx.doi.org/10.1074/jbc.M108252200] [PMID: 11790771]
[44]
Udayakumar, T.S.; Belakavadi, M.; Choi, K.H.; Pandey, P.K.; Fondell, J.D. Regulation of Aurora-A kinase gene expression via GABP recruitment of TRAP220/MED1. J. Biol. Chem., 2006, 281(21), 14691-14699.
[http://dx.doi.org/10.1074/jbc.M600163200] [PMID: 16574658]
[45]
Hung, L.Y.; Tseng, J.T.; Lee, Y.C.; Xia, W.; Wang, Y.N.; Wu, M.L.; Chuang, Y.H.; Lai, C.H.; Chang, W.C. Nuclear epidermal growth factor receptor (EGFR) interacts with signal transducer and activator of transcription 5 (STAT5) in activating Aurora-A gene expression. Nucleic Acids Res., 2008, 36(13), 4337-4351.
[http://dx.doi.org/10.1093/nar/gkn417] [PMID: 18586824]
[46]
Chou, C.H.; Yang, N.K.; Liu, T.Y.; Tai, S.K.; Hsu, D.S.S.; Chen, Y.W. Chromosome instability modulated by BMI1-AURKA signaling drives progression in head and neck cancer. Cancer Res., 2013, 73(2), 953-66.
[http://dx.doi.org/10.1158/0008-5472.CAN-12-2397]
[47]
Lai, C.H.; Huang, Y.C.; Lee, J.C.; Tseng, J.T.C.; Chang, K.C.; Chen, Y.J.; Ding, N.J.; Huang, P.H.; Chang, W.C.; Lin, B.W.; Chen, R.Y.; Wang, Y.C.; Lai, Y.C.; Hung, L.Y. Translational upregulation of Aurora-A by hnRNP Q1 contributes to cell proliferation and tumorigenesis in colorectal cancer. Cell Death Dis., 2017, 8(1), e2555.
[http://dx.doi.org/10.1038/cddis.2016.479] [PMID: 28079881]
[48]
Ice, R.J.; Mclaughlin, S.L.; Livengood, R.H.; Culp, M.V.; Eddy, E.R.; Ivanov, A.V. NEDD9 depletion destabilizes Aurora A kinase and heightens the efficacy of Aurora A inhibitors: implications for treatment of metastatic solid tumors. Cancer Res., 2013, 73(10), 3168.
[http://dx.doi.org/10.1158/0008-5472.CAN-12-4008]
[49]
Eyers, P.A.; Erikson, E.; Chen, L.G.; Maller, J.L. A novel mechanism for activation of the protein kinase Aurora A. Curr. Biol., 2003, 13(8), 691-697.
[http://dx.doi.org/10.1016/S0960-9822(03)00166-0] [PMID: 12699628]
[50]
Bayliss, R.; Sardon, T.; Vernos, I.; Conti, E. Structural basis of Aurora-A activation by TPX2 at the mitotic spindle. Mol. Cell, 2003, 12(4), 851-862.
[http://dx.doi.org/10.1016/S1097-2765(03)00392-7] [PMID: 14580337]
[51]
Giubettini, M.; Asteriti, I.A.; Scrofani, J.; De Luca, M.; Lindon, C.; Lavia, P.; Guarguaglini, G. Control of Aurora-A stability through interaction with TPX2. J. Cell Sci., 2011, 124(1), 113-122.
[http://dx.doi.org/10.1242/jcs.075457] [PMID: 21147853]
[52]
Huang, Y.H.; Wu, C.C.; Chou, C.K.; Huang, C.Y.F. A translational regulator, PUM2, promotes both protein stability and kinase activity of Aurora-A. PLoS One, 2011, 6(5), e19718.
[http://dx.doi.org/10.1371/journal.pone.0019718] [PMID: 21589936]
[53]
Johnson, E.O.; Chang, K.H.; Ghosh, S.; Venkatesh, C.; Giger, K.; Low, P.S.; Shah, K. LIMK2 is a crucial regulator and effector of Aurora-A-kinase-mediated malignancy. J. Cell Sci., 2012, 125(5), 1204-1216.
[http://dx.doi.org/10.1242/jcs.092304] [PMID: 22492986]
[54]
Wang, J.; Nikhil, K.; Viccaro, K.; Chang, L.; Jacobsen, M.; Sandusky, G.; Shah, K. Aurora A-Twist1 axis promotes highly aggressive phenotypes in pancreatic carcinoma. J. Cell Sci., 2017, 130(6), jcs.196790.
[http://dx.doi.org/10.1242/jcs.196790] [PMID: 28167680]
[55]
Nikhil, K.; Raza, A.; Haymour, H.S.; Flueckiger, B.V.; Chu, J.; Shah, K. Aurora kinase A-YBX1 synergy fuels aggressive oncogenic phenotypes and chemoresistance in castration-resistant prostate cancer. Cancers, 2020, 12(3), 660.
[http://dx.doi.org/10.3390/cancers12030660] [PMID: 32178290]
[56]
Woodruff, J.B. Phase separation of BuGZ promotes Aurora A activation and spindle assembly. J. Cell. Biol., 2018, 217(1), 9.
[57]
Hirota, T.; Kunitoku, N.; Sasayama, T.; Marumoto, T.; Zhang, D.; Nitta, M.; Hatakeyama, K.; Saya, H. Aurora-A and an interacting activator, the LIM protein Ajuba, are required for mitotic commitment in human cells. Cell, 2003, 114(5), 585-598.
[http://dx.doi.org/10.1016/S0092-8674(03)00642-1] [PMID: 13678582]
[58]
Zhong, Y.; Yang, J.; Xu, W.W.; Wang, Y.; Zheng, C-C.; Li, B.; He, Q-Y. KCTD12 promotes tumorigenesis by facilitating CDC25B/CDK1/Aurora A-dependent G2/M transition. Oncogene, 2017, 36(44), 6177-6189.
[http://dx.doi.org/10.1038/onc.2017.287] [PMID: 28869606]
[59]
Wu, C.; Lyu, J.; Yang, E.J.; Liu, Y.; Zhang, B.; Shim, J.S. Targeting AURKA-CDC25C axis to induce synthetic lethality in ARID1A-deficient colorectal cancer cells. Nat. Commun., 2018, 9(1), 3212.
[http://dx.doi.org/10.1038/s41467-018-05694-4] [PMID: 30097580]
[60]
Yu, Z.; Sun, Y.; She, X.; Wang, Z.; Chen, S.; Deng, Z.; Zhang, Y.; Liu, Q.; Liu, Q.; Zhao, C.; Li, P.; Liu, C.; Feng, J.; Fu, H.; Li, G.; Wu, M. SIX3, a tumor suppressor, inhibits astrocytoma tumorigenesis by transcriptional repression of AURKA/B. J. Hematol. Oncol., 2017, 10(1), 115-115.
[http://dx.doi.org/10.1186/s13045-017-0483-2] [PMID: 28595628]
[61]
Nowak, I.; Boratyn, E.; Student, S.; Bernhart, S.F.; Fallmann, J.; Durbas, M.; Stadler, P.F.; Rokita, H. MCPIP1 ribonuclease can bind and cleave AURKA mRNA in MYCN amplified neuroblastoma cells. RNA Biol., 2021, 18(1), 144-156.
[http://dx.doi.org/10.1080/15476286.2020.1804698] [PMID: 32757706]
[62]
Taguchi, S.; Honda, K.; Sugiura, K.; Yamaguchi, A.; Furukawa, K.; Urano, T. Degradation of human Aurora-A protein kinase is mediated by hCdh1. FEBS Lett., 2002, 519(1-3), 59-65.
[http://dx.doi.org/10.1016/S0014-5793(02)02711-4] [PMID: 12023018]
[63]
Park, M.T.; Oh, E.T.; Song, M.J.; Lee, H.; Choi, E.K.; Park, H.J. NQO1 prevents radiation-induced aneuploidy by interacting with Aurora-A. Carcinogenesis, 2013, 34(11), 2470-2485.
[http://dx.doi.org/10.1093/carcin/bgt225] [PMID: 23803694]
[64]
SMAD4 suppresses ARK-A-induced metastatic phenotypes via degradation of ARK-Ain a TGFbeta-independent manner. Available from: https://search.yahoo.com/search?fr=mcafee&type=E210US1316G0&p=SMAD4+suppresses+ARK-A-induced+metastatic+phenotypes+via+degradation+of+ARK-Ain+a+TGFbeta-independent+manner(Accessed on: 2022 Aug 22).
[65]
Zhang, C.; Qu, L.; Lian, S.; Meng, L.; Min, L.; Liu, J.; Song, Q.; Shen, L.; Shou, C. PRL-3 promotes ubiquitination and degradation of AURKA and colorectal cancer progression via dephosphorylation of FZR1. Cancer Res., 2019, 79(5), 928-940.
[http://dx.doi.org/10.1158/0008-5472.CAN-18-0520] [PMID: 30498084]
[66]
Irelan, J.T.; Murphy, T.J.; DeJesus, P.D.; Teo, H.; Xu, D.; Gomez-Ferreria, M.A.; Zhou, Y.; Miraglia, L.J.; Rines, D.R.; Verma, I.M.; Sharp, D.J.; Tergaonkar, V.; Chanda, S.K. A role for IκB kinase 2 in bipolar spindle assembly. Proc. Natl. Acad. Sci. USA, 2007, 104(43), 16940-16945.
[http://dx.doi.org/10.1073/pnas.0706493104] [PMID: 17939994]
[67]
Lim, S.K.; Gopalan, G. Antizyme1 mediates AURKAIP1-dependent degradation of Aurora-A. Oncogene, 2007, 26(46), 6593-6603.
[http://dx.doi.org/10.1038/sj.onc.1210482] [PMID: 17452972]
[68]
Kiat, L.S.; Hui, K.M.; Gopalan, G. Aurora-A kinase interacting protein (AIP), a novel negative regulator of human Aurora-A kinase. J. Biol. Chem., 2002, 277(47), 45558-45565.
[http://dx.doi.org/10.1074/jbc.M206820200] [PMID: 12244051]
[69]
Hasanov, E.; Chen, G.; Chowdhury, P.; Weldon, J.; Ding, Z.; Jonasch, E.; Sen, S.; Walker, C.L.; Dere, R. Ubiquitination and regulation of AURKA identifies a hypoxia-independent E3 ligase activity of VHL. Oncogene, 2017, 36(24), 3450-3463.
[http://dx.doi.org/10.1038/onc.2016.495] [PMID: 28114281]
[70]
Meehan, M.; Parthasarathi, L.; Moran, N.; Jefferies, C.A.; Foley, N.; Lazzari, E. Protein tyrosine phosphatase receptor delta acts as a neuroblastoma tumor suppressor by destabilizing the aurora kinase A oncogene. Mol Cancer., 2012, 11, 6.
[http://dx.doi.org/10.1186/1476-4598-11-6]
[71]
Tong, Y.; Ben-Shlomo, A.; Zhou, C.; Wawrowsky, K.; Melmed, S. Pituitary tumor transforming gene 1 regulates Aurora kinase A activity. Oncogene, 2008, 27(49), 6385-6395.
[http://dx.doi.org/10.1038/onc.2008.234] [PMID: 18663361]
[72]
Kunitoku, N.; Sasayama, T.; Marumoto, T.; Zhang, D.; Honda, S.; Kobayashi, O.; Hatakeyama, K.; Ushio, Y.; Saya, H.; Hirota, T. CENP-A phosphorylation by Aurora-A in prophase is required for enrichment of Aurora-B at inner centromeres and for kinetochore function. Dev. Cell, 2003, 5(6), 853-864.
[http://dx.doi.org/10.1016/S1534-5807(03)00364-2] [PMID: 14667408]
[73]
Zheng, X.; Chi, J.; Zhi, J.; Zhang, H.; Yue, D.; Zhao, J.; Li, D.; Li, Y.; Gao, M.; Guo, J. Aurora-A-mediated phosphorylation of LKB1 compromises LKB1/AMPK signaling axis to facilitate NSCLC growth and migration. Oncogene, 2018, 37(4), 502-511.
[http://dx.doi.org/10.1038/onc.2017.354] [PMID: 28967900]
[74]
Sarkar, S.; Brautigan, D.L.; Larner, J.M. Aurora kinase A promotes AR degradation via the E3 ligase CHIP. Mol. Cancer Res., 2017, 15(8), 1063-1072.
[http://dx.doi.org/10.1158/1541-7786.MCR-17-0062] [PMID: 28536143]
[75]
Chang, S.S.; Yamaguchi, H.; Xia, W.; Lim, S.O.; Khotskaya, Y.; Wu, Y. Aurora A kinase activates YAP signaling in triple-negative breast cancer. Oncogene, 201736(9), 1265-75.
[76]
LeRoy, P.J.; Hunter, J.J.; Hoar, K.M.; Burke, K.E.; Shinde, V.; Ruan, J.; Bowman, D.; Galvin, K.; Ecsedy, J.A. Localization of human TACC3 to mitotic spindles is mediated by phosphorylation on Ser558 by Aurora A: a novel pharmacodynamic method for measuring Aurora A activity. Cancer Res., 2007, 67(11), 5362-5370.
[http://dx.doi.org/10.1158/0008-5472.CAN-07-0122] [PMID: 17545617]
[77]
Sehdev, V.; Katsha, A.; Arras, J.; Peng, D.; Soutto, M.; Ecsedy, J.; Zaika, A.; Belkhiri, A.; El-Rifai, W. HDM2 regulation by AURKA promotes cell survival in gastric cancer. Clin. Cancer Res., 2014, 20(1), 76-86.
[http://dx.doi.org/10.1158/1078-0432.CCR-13-1187] [PMID: 24240108]
[78]
Jin, S.; Wang, X.; Tong, T.; Zhang, D.; Shi, J.; Chen, J.; Zhan, Q. Aurora-A enhances malignant development of esophageal squamous cell carcinoma (ESCC) by phosphorylating β-catenin. Mol. Oncol., 2015, 9(1), 249-259.
[http://dx.doi.org/10.1016/j.molonc.2014.08.002] [PMID: 25217103]
[79]
Zheng, X.Q.; Guo, J.P.; Yang, H.; Kanai, M.; He, L.L.; Li, Y.Y.; Koomen, J.M.; Minton, S.; Gao, M.; Ren, X.B.; Coppola, D.; Cheng, J.Q. Aurora-A is a determinant of tamoxifen sensitivity through phosphorylation of ERα in breast cancer. Oncogene, 2014, 33(42), 4985-4996.
[http://dx.doi.org/10.1038/onc.2013.444] [PMID: 24166501]
[80]
Moustafa-Kamal, M.; Gamache, I.; Lu, Y.; Li, S.; Teodoro, J.G. BimEL is phosphorylated at mitosis by Aurora A and targeted for degradation by βTrCP1. Cell Death Differ., 2013, 20(10), 1393-1403.
[http://dx.doi.org/10.1038/cdd.2013.93] [PMID: 23912711]
[81]
Dar, A.A.; Belkhiri, A.; El-Rifai, W. The aurora kinase A regulates GSK-3β in gastric cancer cells. Oncogene, 2009, 28(6), 866-875.
[http://dx.doi.org/10.1038/onc.2008.434] [PMID: 19060929]
[82]
Macůrek, L.; Lindqvist, A.; Lim, D.; Lampson, M.A.; Klompmaker, R.; Freire, R.; Clouin, C.; Taylor, S.S.; Yaffe, M.B.; Medema, R.H. Polo-like kinase-1 is activated by aurora A to promote checkpoint recovery. Nature, 2008, 455(7209), 119-123.
[http://dx.doi.org/10.1038/nature07185] [PMID: 18615013]
[83]
Briassouli, P.; Chan, F.; Savage, K.; Reis-Filho, J.S.; Linardopoulos, S. Aurora-A regulation of nuclear factor-kappaB signaling by phosphorylation of IkappaBalpha. Cancer Res., 2007, 67(4), 1689-1695.
[http://dx.doi.org/10.1158/0008-5472.CAN-06-2272] [PMID: 17308110]
[84]
Martin, B.; Chesnel, F.; Delcros, J.G.; Jouan, F.; Couturier, A.; Dugay, F. Correction: Identification of pVHL as a novel substrate for aurora-A in clear cell renal cell carcinoma (ccRCC). PLoS One, 2014, 9(1)
[85]
Crosio, C.; Fimia, G.M.; Loury, R.; Kimura, M.; Okano, Y.; Zhou, H.; Sen, S.; Allis, C.D.; Sassone-Corsi, P. Mitotic phosphorylation of histone H3: spatio-temporal regulation by mammalian Aurora kinases. Mol. Cell. Biol., 2002, 22(3), 874-885.
[http://dx.doi.org/10.1128/MCB.22.3.874-885.2002] [PMID: 11784863]
[86]
Alexander, K.E.; Rizkallah, R. Aurora A phosphorylation of YY1 during mitosis inactivates its DNA binding activity. Sci. Reports, 2017, 7(1), 1-13.
[http://dx.doi.org/10.1038/s41598-017-10935-5]
[87]
Du, R.; Huang, C.; Chen, H.; Liu, K.; Xiang, P.; Yao, N.; Yang, L.; Zhou, L.; Wu, Q.; Zheng, Y.; Xin, M.; Dong, Z.; Li, X. SDCBP/MDA-9/syntenin phosphorylation by AURKA promotes esophageal squamous cell carcinoma progression through the EGFR-PI3K-Akt signaling pathway. Oncogene, 2020, 39(31), 5405-5419.
[http://dx.doi.org/10.1038/s41388-020-1369-2] [PMID: 32572158]
[88]
Qi, D.; Wang, Q.; Yu, M.; Lan, R.; Li, S.; Lu, F. Mitotic phosphorylation of SOX2 mediated by Aurora kinase A is critical for the stem-cell like cell maintenance in PA-1 cells. Cell Cycle, 2016, 15(15), 2009-2018.
[http://dx.doi.org/10.1080/15384101.2016.1192729] [PMID: 27249336]
[89]
Chou, E.J.; Hung, L.Y.; Tang, C.J.C.; Hsu, W. Phosphorylation of CPAP by Aurora-A maintains spindle pole integrity during mitosis. Cell. Rep., 2016, 14(12), 2975-87.
[90]
Wu, J.C.; Chen, T.Y.; Yu, C.T.R.; Tsai, S.J.; Hsu, J.M.; Tang, M.J.; Chou, C.K.; Lin, W.J.; Yuan, C.J.; Huang, C.Y.F. Identification of V23RalA-Ser194 as a critical mediator for Aurora-A-induced cellular motility and transformation by small pool expression screening. J. Biol. Chem., 2005, 280(10), 9013-9022.
[http://dx.doi.org/10.1074/jbc.M411068200] [PMID: 15637052]
[91]
Dutertre, S.; Cazales, M.; Quaranta, M.; Froment, C.; Trabut, V.; Dozier, C.; Mirey, G.; Bouché, J.P.; Theis-Febvre, N.; Schmitt, E.; Monsarrat, B.; Prigent, C.; Ducommun, B. Phosphorylation of CDC25B by Aurora-A at the centrosome contributes to the G2–M transition. J. Cell Sci., 2004, 117(12), 2523-2531.
[http://dx.doi.org/10.1242/jcs.01108] [PMID: 15128871]
[92]
Kollareddy, M.; Zheleva, D.; Dzubak, P.; Brahmkshatriya, P.S.; Lepsik, M.; Hajduch, M. Aurora kinase inhibitors: Progress towards the clinic. Invest. New Drugs, 2012, 30(6), 2411-2432.
[http://dx.doi.org/10.1007/s10637-012-9798-6] [PMID: 22350019]
[93]
Portier, N.; Audhya, A.; Maddox, P.S.; Green, R.A.; Dammermann, A.; Desai, A.; Oegema, K. A microtubule-independent role for centrosomes and aurora a in nuclear envelope breakdown. Dev. Cell, 2007, 12(4), 515-529.
[http://dx.doi.org/10.1016/j.devcel.2007.01.019] [PMID: 17419991]
[94]
Li, J.; Yan, Z.; Li, H.; Shi, Q.; Ahire, V.; Zhang, S.; Nimishetti, N.; Yang, D.; Allen, T.D.; Zhang, J. The phytochemical scoulerine inhibits aurora kinase activity to induce mitotic and cytokinetic defects. J. Nat. Prod. , 2021, 84(8), 2312-2320.
[http://dx.doi.org/10.1021/acs.jnatprod.1c00429]
[95]
Yan, Z.; Shi, Q.; Liu, X.; Li, J.; Ahire, V.; Zhang, S. The phytochemical, corynoline, diminishes Aurora kinase B activity to induce mitotic defect and polyploidy. Biomed. Pharmacother., 2022, 147, 112645.
[http://dx.doi.org/10.1016/j.biopha.2022.112645]
[96]
Hauf, S.; Cole, R.W.; LaTerra, S.; Zimmer, C.; Schnapp, G.; Walter, R.; Heckel, A.; van Meel, J.; Rieder, C.L.; Peters, J.M. The small molecule Hesperadin reveals a role for Aurora B in correcting kinetochore–microtubule attachment and in maintaining the spindle assembly checkpoint. J. Cell Biol., 2003, 161(2), 281-294.
[http://dx.doi.org/10.1083/jcb.200208092] [PMID: 12707311]
[97]
Fu, D.H.; Jiang, W.; Zheng, J.T.; Zhao, G.Y.; Li, Y.; Yi, H.; Li, Z.R.; Jiang, J.D.; Yang, K.Q.; Wang, Y.; Si, S.Y. Jadomycin B, an Aurora-B kinase inhibitor discovered through virtual screening. Mol. Cancer Ther., 2008, 7(8), 2386-2393.
[http://dx.doi.org/10.1158/1535-7163.MCT-08-0035] [PMID: 18723485]
[98]
Guo, J.; Anderson, M.G.; Tapang, P.; Palma, J.P.; Rodriguez, L.E.; Niquette, A.; Li, J.; Bouska, J.J.; Wang, G.; Semizarov, D.; Albert, D.H.; Donawho, C.K.; Glaser, K.B.; Shah, O.J. Identification of genes that confer tumor cell resistance to the Aurora B kinase inhibitor, AZD1152. Pharmacogenomics J., 2009, 9(2), 90-102.
[http://dx.doi.org/10.1038/tpj.2008.20] [PMID: 19188929]
[99]
Smith, M.L.; Murphy, K.; Doucette, C.D.; Greenshields, A.L.; Hoskin, D.W. The dietary flavonoid fisetin causes cell cycle arrest, caspase-dependent apoptosis, and enhanced cytotoxicity of chemotherapeutic drugs in triple-negative breast cancer cells. J. Cell. Biochem., 2016, 117(8), 1913-1925.
[http://dx.doi.org/10.1002/jcb.25490] [PMID: 26755433]
[100]
Lee, D.; Kim, C.; Lim, Y.; Shin, S. Aurora kinase A inhibitor TCS7010 demonstrates pro-apoptotic effect through the unfolded protein response pathway in HCT116 colon cancer cells. Oncol. Lett., 2017, 14(6), 6571-6577.
[http://dx.doi.org/10.3892/ol.2017.7023] [PMID: 29163689]
[101]
Nakashima, K.; Uematsu, T.; Takahashi, K.; Nishimura, S.; Tadokoro, Y.; Hayashi, T.; Sugino, T. Does breast cancer growth rate really depend on tumor subtype? Measurement of tumor doubling time using serial ultrasonography between diagnosis and surgery. Breast Cancer, 2019, 26(2), 206-214.
[http://dx.doi.org/10.1007/s12282-018-0914-0] [PMID: 30259332]
[102]
Ganjibakhsh, M.; Aminishakib, P.; Farzaneh, P.; Karimi, A.; Fazeli, S.A.S.; Rajabi, M.; Nasimian, A.; Naini, F.B.; Rahmati, H.; Gohari, N.S.; Mohebali, N.; Asadi, M.; Gorji, Z.E.; Izadpanah, M.; Moghanjoghi, S.M.; Ashouri, S. Establishment and characterization of primary cultures from iranian oral squamous cell carcinoma patients by enzymatic method and explant culture. J. Dent. (Tehran), 2017, 14(4), 191-202.
[PMID: 29285029]
[103]
Ruddarraju, R.R.; Murugulla, A.C.; Kotla, R.; Tirumalasetty, M.C.B.; Wudayagiri, R.; Donthabakthuni, S.; Maroju, R. Design, synthesis, anticancer activity and docking studies of theophylline containing 1,2,3-triazoles with variant amide derivatives. MedChemComm, 2017, 8(1), 176-183.
[http://dx.doi.org/10.1039/C6MD00479B] [PMID: 30108703]
[104]
Shan, B; Zhao, R; Zhou, J; Zhang, M; Qi, X; Wang, T AURKA increase the chemosensitivity of colon cancer cells to oxaliplatin by inhibiting the TP53-mediated DNA damage response genes. Biomed Res Int., 2020, 2020, 8916729.
[http://dx.doi.org/10.1155/2020/8916729]
[105]
Suman, S.; Mishra, A. Network analysis revealed aurora kinase dysregulation in five gynecological types of cancer. Oncol. Lett., 2018, 15(1), 1125-1132.
[PMID: 29391900]
[106]
Oliveira, R.C.; Abrantes, A.M.; Tralhão, J.G.; Botelho, M.F. The role of mouse models in colorectal cancer research-The need and the importance of the orthotopic models. Anim. Model. Exp. Med., 2020, 3(1), 1-8.
[107]
Muniyappan, G.; Kathavarayan, S.; Balachandran, C.; Kalliyappan, E.; Mahalingam, S.M.; Ajees Abdul Salam, A.; Aoki, S.; Arumugam, N.; Almansour, A.I.; Suresh Kumar, R. Synthesis, anticancer and molecular docking studies of new class of benzoisoxazolyl-piperidinyl-1, 2, 3-triazoles. J. King Saud Univ. Sci., 2020, 32(8), 3286-3292.
[http://dx.doi.org/10.1016/j.jksus.2020.09.012]
[108]
Pandya, P.N.; Mankad, A.U.; Raval, R.M. Role of aurora kinases in cancer: A comprehensive review. 2018, 4(80), 80-93.
[109]
Napier, K.J.; Scheerer, M.; Misra, S. Esophageal cancer: A Review of epidemiology, pathogenesis, staging workup and treatment modalities. World J. Gastrointest. Oncol., 2014, 6(5), 112.
[http://dx.doi.org/10.4251/wjgo.v6.i5.112]
[110]
Yang, Y-M.; Hong, P.; Xu, W.W.; He, Q-Y.; Li, B. Advances in targeted therapy for esophageal cancer. Signal Transduct Target Ther., 2020, 5(1), 1-11.
[http://dx.doi.org/10.1038/s41392-020-00323-3]
[111]
Nemoto, T.; Ohashi, K.; Akashi, T.; Johnson, J.D.; Hirokawa, K. Overexpression of protein tyrosine kinases in human esophageal cancer. Pathobiology, 1997, 65(4), 195-203.
[http://dx.doi.org/10.1159/000164123] [PMID: 9396043]
[112]
Júnior, A.P.; Costa, N.M.; Da; Esposito, F.; Fusco, A.; Pinto, L.F.R. High Mobility Group A proteins in esophageal carcinomas. Cell Cycle, 2016, 15(18), 2410.
[113]
He, F.; Ai, B.; Tian, L. Identification of genes and pathways in esophageal adenocarcinoma using bioinformatics analysis. Biomed. Rep., 2018, 9(4), 305-312.
[http://dx.doi.org/10.3892/br.2018.1134] [PMID: 30233782]
[114]
Su, P.; Wen, S.; Zhang, Y.; Li, Y.; Xu, Y.; Zhu, Y. Identification of the key genes and pathways in esophageal carcinoma. Gastroenterol. Res. Pract., 2016, 2016, 2968106.
[http://dx.doi.org/10.1155/2016/2968106]
[115]
Clemons, N.J.; Phillips, W.A.; Lord, R.V. Signaling pathways in the molecular pathogenesis of adenocarcinomas of the esophagus and gastroesophageal junction. Cancer Biol. Ther., 2013, 14(9), 782.
[http://dx.doi.org/10.4161/cbt.25362]
[116]
Raufi, A.G.; Klempner, S.J. Immunotherapy for advanced gastric and esophageal cancer: preclinical rationale and ongoing clinical investigations. J. Gastrointest. Oncol., 2015, 6(5), 561.
[117]
Pennathur, A.; Xi, L.; Litle, V.R.; Gooding, W.E.; Krasinskas, A.; Landreneau, R.J. Gene expression profiles in esophageal adenocarcinoma predict survival after resection. J. Thoracic Cardivasc. Surg., 2013, 145(2), 505-513.
[http://dx.doi.org/10.1016/j.jtcvs.2012.10.031]
[118]
Ando, N.; Ozawa, S.; Kitagawa, Y.; Shinozawa, Y.; Kitajima, M. Improvement in the results of surgical treatment of advanced squamous esophageal carcinoma during 15 consecutive years. Ann. Surg., 2000, 232(2), 225-232.
[http://dx.doi.org/10.1097/00000658-200008000-00013] [PMID: 10903602]
[119]
Sun, S.; Zhang, H.; Wang, Y.; Gao, J.; Zhou, S.; Li, Y. Proteomic analysis of human esophageal cancer using tandem mass tag quantifications. Biomed. Res. Int., 2020, 2020, 5849323.
[http://dx.doi.org/10.1155/2020/5849323]
[120]
Ilson, D.H.; Saltz, L.; Enzinger, P.; Huang, Y.; Kornblith, A.; Gollub, M.; O'Reilly, E.; Schwartz, G.; DeGroff, J.; Gonzalez, G.; Kelsen, D.P. Phase II trial of weekly irinotecan plus cisplatin in advanced esophageal cancer. J. Clin. Oncol., 1999, 17(10), 3270-5.
[http://dx.doi.org/10.1200/JCO.1999.17.10.3270]
[121]
Xia, J.L.; Fan, W.J.; Zheng, F.M.; Zhang, W.W.; Xie, J.J.; Yang, M.Y.; Kamran, M.; Wang, P.; Teng, H.M.; Wang, C.L.; Liu, Q. Inhibition of AURKA kinase activity suppresses collective invasion in a microfluidic cell culture platform. Sci. Rep., 2017, 7(1), 2973.
[http://dx.doi.org/10.1038/s41598-017-02623-1] [PMID: 28592839]
[122]
Sehdev, V.; Peng, D.; Soutto, M.; Washington, M.K.; Revetta, F.; Ecsedy, J. The Aurora kinase A inhibitor MLN8237 enhances cisplatin-induced cell death in esophageal adenocarcinoma cells. Mol. Cancer Ther., 2012, 11(3), 763.
[http://dx.doi.org/10.1158/1535-7163.MCT-11-0623]
[123]
Liao, Y.; Liao, Y.; Li, J.; Li, J.; Fan, Y.; Xu, B. Polymorphisms in AURKA and AURKB are associated with the survival of triple-negative breast cancer patients treated with taxane-based adjuvant chemotherapy. Cancer Manag. Res., 2018, 10, 3801-3808.
[http://dx.doi.org/10.2147/CMAR.S174735] [PMID: 30288111]
[124]
Kelly, C.M. A three-gene model to robustly identify breast cancer molecular subtypes. Breast Dis., 2013, 24(1), 36-38.
[http://dx.doi.org/10.1016/j.breastdis.2013.01.022]
[125]
Opyrchal, M.; Divya, K.; Sangameswaran, G.; Khoury, T. Aurora kinase inhibitors in breast cancer treatment. Cancer, 2015, 32, 34.
[126]
Zhao, C.H.; Qu, L.; Zhang, H.; Qu, R. Identification of breast cancer-related circRNAs by analysis of microarray and RNA-sequencing data. Medicine, 2019, 98(46), e18042.
[http://dx.doi.org/10.1097/MD.0000000000018042] [PMID: 31725681]
[127]
Engin, H.B.; Guney, E.; Keskin, O.; Oliva, B.; Gursoy, A. ntegrating structure to protein-protein interaction networks that drive metastasis to brain and lung in breast cancer. PLoS One, 2013, 8(11), e81035.
[http://dx.doi.org/10.1371/journal.pone.0081035]
[128]
Lassmann, S.; Shen, Y.; Wiehle, P.; Walch, A.; Gitsch, G. Predictive Value of Aurora-A/STK15 Expression for Late Stage Epithelial Ovarian Cancer Patients Treated by Adjuvant Chemotherapy. 2007. Available from: www.aacrjournals.org
[129]
Duckworth, C.; Zhang, L.; Carroll, S.L.; Ethier, S.P.; Cheung, H.W. Overexpression of GAB2 in ovarian cancer cells promotes tumor growth and angiogenesis by upregulating chemokine expression. Oncogene, 2016, 35(31), 4036-4047.
[http://dx.doi.org/10.1038/onc.2015.472] [PMID: 26657155]
[130]
Liu, X.; Gao, Y.; Zhao, B.; Li, X.; Lu, Y.; Zhang, J.; Li, D.; Li, L.; Yin, F. Discovery of microarray-identified genes associated with ovarian cancer progression. Int. J. Oncol., 2015, 46(6), 2467-2478.
[http://dx.doi.org/10.3892/ijo.2015.2971] [PMID: 25891226]
[131]
Li, W.; Liu, Z.; Liang, B.; Chen, S.; Zhang, X.; Tong, X.; Lou, W.; Le, L.; Tang, X.; Fu, F. Identification of core genes in ovarian cancer by an integrative meta-analysis. J. Ovarian Res., 2018, 11(1), 94.
[http://dx.doi.org/10.1186/s13048-018-0467-z] [PMID: 30453999]
[132]
Thorn, C.F.; Oshiro, C.; Marsh, S.; Hernandez-Boussard, T.; Mcleod, H.; Klein, T.E. Doxorubicin pathways: Pharmacodynamics and adverse effects. Pharmacogenet. Genomics., 2011, 21(7), 440.
[133]
Gan, X.; Zhu, H.; Jiang, X.; Obiegbusi, S.C.; Yong, M.; Long, X.; Hu, J. CircMUC16 promotes autophagy of epithelial ovarian cancer via interaction with ATG13 and miR-199a. Mol. Cancer, 2020, 19(1), 45.
[http://dx.doi.org/10.1186/s12943-020-01163-z] [PMID: 32111227]
[134]
Karnezis, A.N.; Cho, K.R. Preclinical models of ovarian cancer: pathogenesis, problems, and implications for prevention. In: Clinical Obstetrics and Gynecology; Lippincott Williams and Wilkins, 2017; 60, pp. 789-800. Available from: https://journals.lww.com/00003081- 201712000-00011
[135]
Alcaraz-Sanabria, A.; Nieto-Jim Enez, C. Cancer biology and translational studies synthetic lethality interaction between aurora kinases and chek1 inhibitors in ovarian cancer. 2017. Available from: https://www.oncomine.org/resource/ (Accessed on: 2021 Mar 13)
[136]
Carosati, E.; Tochowicz, A.; Marverti, G.; Guaitoli, G.; Benedetti, P.; Ferrari, S.; Stroud, R.M.; Finer-Moore, J.; Luciani, R.; Farina, D.; Cruciani, G.; Costi, M.P. Inhibitor of ovarian cancer cells growth by virtual screening: a new thiazole derivative targeting human thymidylate synthase. J. Med. Chem., 2012, 55(22), 10272-10276.
[http://dx.doi.org/10.1021/jm300850v] [PMID: 23075414]
[137]
An, Y.; Lee, E.; Yu, Y.; Yun, J.; Lee, M.Y.; Kang, J.S. Design and synthesis of novel benzoxazole analogs as Aurora B kinase inhibitors. Bioorganic. Med. Chem. Lett., 2016, 26(13), 3067-72.
[138]
Bavetsias, V.; Faisal, A.; Crumpler, S.; Brown, N.; Kosmopoulou, M.; Joshi, A.; Atrash, B.; Pérez-Fuertes, Y.; Schmitt, J.A.; Boxall, K.J.; Burke, R.; Sun, C.; Avery, S.; Bush, K.; Henley, A.; Raynaud, F.I.; Workman, P.; Bayliss, R.; Linardopoulos, S.; Blagg, J. Aurora isoform selectivity: design and synthesis of imidazo[4,5-b]pyridine derivatives as highly selective inhibitors of Aurora-A kinase in cells. J. Med. Chem., 2013, 56(22), 9122-9135.
[http://dx.doi.org/10.1021/jm401115g] [PMID: 24195668]
[139]
Protein-protein interaction spider of oral cancer. Available from: https://figshare.com/articles/figure/Protein-protein_interaction_spider_of_oral_cancer/6396827 (Accessed on: 2021 Mar 16)
[140]
Qi, G.; Ogawa, I.; Kudo, Y.; Miyauchi, M.; Siriwardena, B.S.M.S.; Shimamoto, F.; Tatsuka, M.; Takata, T. Aurora-B expression and its correlation with cell proliferation and metastasis in oral cancer. Virchows Archiv, 2021, 450(3), 297-302.
[http://dx.doi.org/10.1007/s00428-006-0360-9]
[141]
Wenzhao, L.; Jiangdong, N.; Deye, S.; Muliang, D.; Junjie, W.; Xianzhe, H.; Mingming, Y.; Jun, H. Dual regulatory roles of HMGB1 in inflammatory reaction of chondrocyte cells and mice. Cell Cycle, 2019, 18(18), 2268-2280.
[http://dx.doi.org/10.1080/15384101.2019.1642680] [PMID: 31313630]
[142]
Wang, Y.F.; Li, B.W.; Sun, S.; Li, X.; Su, W.; Wang, Z.H. Circular RNA expression in oral squamous cell carcinoma. Front Oncol., 2018, 8, 398.
[http://dx.doi.org/10.3389/fonc.2018.00398]
[143]
Christowitz, C.; Davis, T.; Isaacs, A.; van Niekerk, G.; Hattingh, S.; Engelbrecht, A.M. Mechanisms of doxorubicin-induced drug resistance and drug resistant tumour growth in a murine breast tumour model. BMC Cancer, 2019, 19(1), 757.
[http://dx.doi.org/10.1186/s12885-019-5939-z] [PMID: 31370818]
[144]
Saxena, V.L.; Gupta, S.; Srivastava, S. Study of ligand based virtual screening tools in computer aided drug designing for oral cancer. IOSR J. Pharm. Biol. Sci., 2010, 10, 65-74.
[145]
Bavetsias, V.; Linardopoulos, S. Aurora kinase inhibitors: Current status and outlook. Front. Oncol., 2015, 5, 278.
[146]
Twu, N.F.; Yuan, C.C.; Yen, M.S.; Lai, C.R.; Chao, K.C.; Wang, P.H.; Wu, H.H.; Chen, Y.J. Expression of Aurora kinase A and B in normal and malignant cervical tissue: High Aurora A kinase expression in squamous cervical cancer. Eur. J. Obstet. Gynecol. Reprod. Biol., 2009, 142(1), 57-63.
[http://dx.doi.org/10.1016/j.ejogrb.2008.09.012] [PMID: 19059698]
[147]
Dasari, S.; Bernard Tchounwou, P. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol., 2014, 740, 364-78.
[148]
Wei, J; Wang, Y; Shi, K; Wang, Y. Identification of core prognosis-related candidate genes in cervical cancer via integrated bioinformatical analysis. Biomed Res Int., 2020, 2020, 8959210.
[http://dx.doi.org/10.1155/2020/8959210]
[149]
Martin, D.; Fallaha, S.; Proctor, M.; Stevenson, A.; Perrin, L.; McMillan, N.; Gabrielli, B. Inhibition of Aurora A and Aurora B is required for the sensitivity of HPV-driven cervical cancers to Aurora kinase inhibitors. Mol. Cancer Ther., 2017, 16(9), 1934-1941.
[http://dx.doi.org/10.1158/1535-7163.MCT-17-0159] [PMID: 28522591]
[150]
Khan, A.A.; A Abuderman, A.; Ashraf, M.T.; Khan, Z. Protein–protein interactions of HPV-Chlamydia trachomatis human and their potential in cervical cancer. Future Microbiol., 2020, 15(7), 509-520.
[http://dx.doi.org/10.2217/fmb-2019-0242] [PMID: 32476479]
[151]
Ager, B.J.; Gallardo-Rincón, D.; de León, D.C.; Chávez-Blanco, A.; Chuang, L.; Dueñas-González, A. Advancing clinical research globally: Cervical cancer research network from Mexico. Gynecol. Oncol. Rep., 2018, 25, 90-3.
[152]
Sagae, S.; Monk, B.J.; Pujade-Lauraine, E.; Gaffney, D.K.; Narayan, K.; Ryu, S.Y. Advances and concepts in cervical cancer trials: A road map for the future. Int. J. Gynecol. Cancer, 2016, 199-207.
[http://dx.doi.org/10.1097/IGC.0000000000000587]
[153]
Kumar, A.; Rathi, E.; Kini, S.G. E-pharmacophore modelling, virtual screening, molecular dynamics simulations and in-silico ADME analysis for identification of potential E6 inhibitors against cervical cancer. J. Mol. Struct., 2019, 1189, 299-306.
[154]
Bengtsson, E.; Malm, P. Screening for cervical cancer using automated analysis of PAP-smears. Comput Math Methods Med., 2014, 2014, 2037.
[http://dx.doi.org/10.1155/2014/842037]
[155]
How Long Does it Take for Cervical Cancer to Develop?- Moffitt. Available from: https://moffitt.org/cancers/cervical-cancer/faqs/how-long-does-it-take-for-cervical-cancer- to-develop/ (Accessed on: 2021 Mar 19).
[156]
Borah, N.A.; Reddy, M.M. Aurora kinase B inhibition: A potential therapeutic strategy for cancer. Molecules, 2021, 26(7), 1981.
[http://dx.doi.org/10.3390/molecules26071981] [PMID: 33915740]
[157]
Cai, J.; Li, L.; Hong, K.H.; Wu, X.; Chen, J.; Wang, P. Discovery of 4-aminoquinazoline-urea derivatives as Aurora kinase inhibitors with antiproliferative activity. Bioorganic Med. Chem., 2014, 22(21), 5813-23.
[158]
Chate, A.V.; Tagad, P.A.; Bondle, G.M.; Sarkate, A.P.; Tiwari, S.V.; Azad, R. Design, synthesis and biological evaluation of tetrahydrodibenzo[b,g][1,8]napthyridinones as potential anticancer agents and novel aurora kinases inhibitors. ChemistrySelect, 2021, 6(14), 3444-3452.
[http://dx.doi.org/10.1002/slct.202004666]
[159]
Merriel, S.W.D.; Funston, G.; Hamilton, W. Prostate cancer in primary care. Adv. Ther., 2018, 35, 1285-94.
[http://dx.doi.org/10.1007/s12325-018-0766-1]
[160]
Prostate cancer: Symptoms, treatment, and causes Available from: https://www.medicalnewstoday.com/articles/150086#symptoms(Accessed on: 2021 Mar 22)
[161]
Wang, B.; Hasan, M.K.; Alvarado, E.; Yuan, H.; Wu, H.; Chen, W.Y. NAMPT overexpression in prostate cancer and its contribution to tumor cell survival and stress response. Oncogene, 2011, 30(8), 907-921.
[http://dx.doi.org/10.1038/onc.2010.468] [PMID: 20956937]
[162]
Why Are Prostate Cancer Preclinical Models Hard to Develop? Available from: https://blog.crownbio.com/prostate-cancer-preclinical-models(Accessed on: 2021 Mar 21)
[163]
Matos, B.; Howl, J.; Jerónimo, C.; Fardilha, M. The disruption of protein-protein interactions as a therapeutic strategy for prostate cancer. Pharmacological Research. Academic Press 2020, 161, 105145. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1043661820314535(Accessed on: 2021 Mar 21).
[http://dx.doi.org/10.1016/j.phrs.2020.105145]
[164]
High-Fat Diet Linked to Prostate Cancer Metastasis - National Cancer Institute. Available from: https://www.cancer.gov/news-events/cancer-currents-blog/2018/high- fat-diet-prostate-metastasis(Accessed on: 2021 Mar 21).
[165]
Srikantan, V.; Zou, Z.; Petrovics, G.; Xu, L.; Augustus, M.; Davis, L. PCGEM1, a prostate-specific gene, is overexpressed in prostate cancer. Proc. Natl. Acad. Sci., 2000, 97(22), 12216-21.
[http://dx.doi.org/10.1073/pnas.97.22.12216]
[166]
Lutz, S.Z.; Hennenlotter, J.; Scharpf, M.O.; Sailer, C.; Fritsche, L.; Schmid, V.; Kantartzis, K.; Wagner, R.; Lehmann, R.; Berti, L.; Peter, A.; Staiger, H.; Fritsche, A.; Fend, F.; Todenhöfer, T.; Stenzl, A.; Häring, H.U.; Heni, M. Androgen receptor overexpression in prostate cancer in type 2 diabetes. Mol. Metab., 2018, 8, 158-166.
[http://dx.doi.org/10.1016/j.molmet.2017.11.013] [PMID: 29249638]
[167]
Arjun, H.A.; Elancheran, R.; Manikandan, N.; Lakshmithendral, K.; Ramanathan, M.; Bhattacharjee, A. Design, synthesis, and biological evaluation of (E)-N’-((1-Chloro-3,4-Dihydronaphthalen-2-yl)Methylene)benzohydrazide derivatives as anti-prostate cancer agents. Front. Chem., 2019, 7
[168]
Zhang, Y.; Xu, Q.; Liu, G.; Huang, H.; Lin, W.; Huang, Y. Effect of histone deacetylase on prostate carcinoma. Int. J. Clin. Exp. Pathol., 2015, 8(11), 15030.
[169]
Park, J.H.; Jung, Y.; Kim, T.Y.; Kim, S.G.; Jong, H.S.; Lee, J.W.; Kim, D.K.; Lee, J.S.; Kim, N.K.; Kim, T.Y.; Bang, Y.J. Class I histone deacetylase-selective novel synthetic inhibitors potently inhibit human tumor proliferation. Clin. Cancer Res., 2004, 10(15), 5271-5281.
[http://dx.doi.org/10.1158/1078-0432.CCR-03-0709] [PMID: 15297431]
[170]
Fiskus, W.; Wang, Y.; Joshi, R.; Rao, R.; Yang, Y.; Chen, J.; Kolhe, R.; Balusu, R.; Eaton, K.; Lee, P.; Ustun, C.; Jillella, A.; Buser, C.A.; Peiper, S.; Bhalla, K. Cotreatment with vorinostat enhances activity of MK-0457 (VX-680) against acute and chronic myelogenous leukemia cells. Clin. Cancer Res., 2008, 14(19), 6106-6115.
[http://dx.doi.org/10.1158/1078-0432.CCR-08-0721] [PMID: 18829489]
[171]
Dai, Y.; Chen, S.; Pei, X.Y.; Almenara, J.A.; Kramer, L.B.; Venditti, C.A.; Dent, P.; Grant, S. Interruption of the Ras/MEK/ERK signaling cascade enhances Chk1 inhibitor–induced DNA damage in vitro and in vivo in human multiple myeloma cells. Blood, 2008, 112(6), 2439-2449.
[http://dx.doi.org/10.1182/blood-2008-05-159392] [PMID: 18614762]
[172]
Fiskus, W.; Hembruff, S.L.; Rao, R.; Sharma, P.; Balusu, R.; Venkannagari, S.; Smith, J.E.; Peth, K.; Peiper, S.C.; Bhalla, K.N. Co-treatment with vorinostat synergistically enhances activity of Aurora kinase inhibitor against human breast cancer cells. Breast Cancer Res. Treat., 2012, 135(2), 433-444.
[http://dx.doi.org/10.1007/s10549-012-2171-9] [PMID: 22825030]
[173]
Kretzner, L.; Scuto, A.; Dino, P.M.; Kowolik, C.M.; Wu, J.; Ventura, P.; Jove, R.; Forman, S.J.; Yen, Y.; Kirschbaum, M.H. Combining histone deacetylase inhibitor vorinostat with aurora kinase inhibitors enhances lymphoma cell killing with repression of c-Myc, hTERT, and microRNA levels. Cancer Res., 2011, 71(11), 3912-3920.
[http://dx.doi.org/10.1158/0008-5472.CAN-10-2259] [PMID: 21502403]
[174]
Zullo, K.M.; Guo, Y.; Cooke, L.; Jirau-Serrano, X.; Mangone, M.; Scotto, L.; Amengual, J.E.; Mao, Y.; Nandakumar, R.; Cremers, S.; Duong, J.; Mahadevan, D.; O’Connor, O.A. Aurora A kinase inhibition selectively synergizes with histone deacetylase inhibitor through cytokinesis failure in T-cell lymphoma. Clin. Cancer Res., 2015, 21(18), 4097-4109.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-0033] [PMID: 25878331]
[175]
Paller, C.J.; Wissing, M.D.; Mendonca, J.; Sharma, A.; Kim, E.; Kim, H.S.; Kortenhorst, M.S.Q.; Gerber, S.; Rosen, M.; Shaikh, F.; Zahurak, M.L.; Rudek, M.A.; Hammers, H.; Rudin, C.M.; Carducci, M.A.; Kachhap, S.K. Combining the pan-aurora kinase inhibitor AMG 900 with histone deacetylase inhibitors enhances antitumor activity in prostate cancer. Cancer Med., 2014, 3(5), 1322-1335.
[http://dx.doi.org/10.1002/cam4.289] [PMID: 24989836]
[176]
Shah, K.N.; Bhatt, R.; Rotow, J.; Rohrberg, J.; Olivas, V.; Wang, V.E.; Hemmati, G.; Martins, M.M.; Maynard, A.; Kuhn, J.; Galeas, J.; Donnella, H.J.; Kaushik, S.; Ku, A.; Dumont, S.; Krings, G.; Haringsma, H.J.; Robillard, L.; Simmons, A.D.; Harding, T.C.; McCormick, F.; Goga, A.; Blakely, C.M.; Bivona, T.G.; Bandyopadhyay, S. Aurora kinase A drives the evolution of resistance to third-generation EGFR inhibitors in lung cancer. Nat. Med., 2019, 25(1), 111-118.
[http://dx.doi.org/10.1038/s41591-018-0264-7] [PMID: 30478424]
[177]
Čančer, M.; Drews, L.F.; Bengtsson, J.; Bolin, S.; Rosén, G.; Westermark, B.; Nelander, S.; Forsberg-Nilsson, K.; Uhrbom, L.; Weishaupt, H.; Swartling, F.J. BET and Aurora Kinase A inhibitors synergize against MYCN-positive human glioblastoma cells. Cell Death Dis., 2019, 10(12), 881.
[http://dx.doi.org/10.1038/s41419-019-2120-1] [PMID: 31754113]
[178]
Felgenhauer, J.; Tomino, L.; Selich-Anderson, J.; Bopp, E.; Shah, N. Dual BRD4 and AURKA inhibition is synergistic against MYCN-amplified and nonamplified neuroblastoma. Neoplasia, 2018, 20(10), 965-974.
[http://dx.doi.org/10.1016/j.neo.2018.08.002] [PMID: 30153557]
[179]
Vilgelm, A.E.; Pawlikowski, J.S.; Liu, Y.; Hawkins, O.E.; Davis, T.A.; Smith, J.; Weller, K.P.; Horton, L.W.; McClain, C.M.; Ayers, G.D.; Turner, D.C.; Essaka, D.C.; Stewart, C.F.; Sosman, J.A.; Kelley, M.C.; Ecsedy, J.A.; Johnston, J.N.; Richmond, A. Mdm2 and aurora kinase A inhibitors synergize to block melanoma growth by driving apoptosis and immune clearance of tumor cells. Cancer Res., 2015, 75(1), 181-193.
[http://dx.doi.org/10.1158/0008-5472.CAN-14-2405] [PMID: 25398437]
[180]
Kojima, K.; Konopleva, M.; Tsao, T.; Nakakuma, H.; Andreeff, M. Concomitant inhibition of Mdm2-p53 interaction and Aurora kinases activates the p53-dependent postmitotic checkpoints and synergistically induces p53-mediated mitochondrial apoptosis along with reduced endoreduplication in acute myelogenous leukemia. Blood, 2008, 112(7), 2886-2895.
[http://dx.doi.org/10.1182/blood-2008-01-128611] [PMID: 18633130]
[181]
Ratushny, V.; Pathak, H.B.; Beeharry, N.; Tikhmyanova, N.; Xiao, F.; Li, T. Dual inhibition of SRC and Aurora kinases induces postmitotic attachment defects and cell death. Oncogene, 2012, 31(10), 1217-27.
[http://dx.doi.org/10.1038/onc.2011.314]
[182]
Alcaraz-Sanabria, A.; Nieto-Jimenez, C.; Corrales- Sanchez, V.; Serrano-Oviedo, L.; Andres-Pretel, F.; Montero, J.C. Synthetic lethality interaction between aurora kinases and CHEK1 inhibitors in ovarian cancer. Mol. Cancer Ther., 2017, 16(11), 2552-2562.
[183]
Lu, Y.; Liu, L.L.; Liu, S.S.; Fang, Z.G.; Zou, Y.; Deng, X. Celecoxib suppresses autophagy and enhances cytotoxicity of imatinib in imatinib-resistant chronic myeloid leukemia cells. J. Transl. Med., 2016, 14(1), 270.
[184]
Brewer Savannah, K.J.; Demicco, E.G.; Lusby, K.; Ghadimi, M.P.H.; Belousov, R.; Young, E.; Zhang, Y.; Huang, K.L.; Lazar, A.J.; Hunt, K.K.; Pollock, R.E.; Creighton, C.J.; Anderson, M.L.; Lev, D. Dual targeting of mTOR and aurora-A kinase for the treatment of uterine Leiomyosarcoma. Clin. Cancer Res., 2012, 18(17), 4633-4645.
[http://dx.doi.org/10.1158/1078-0432.CCR-12-0436] [PMID: 22821997]
[185]
Lee, J.W.; Parameswaran, J.; Sandoval-Schaefer, T.; Eoh, K.J.; Yang, D.; Zhu, F.; Mehra, R.; Sharma, R.; Gaffney, S.G.; Perry, E.B.; Townsend, J.P.; Serebriiskii, I.G.; Golemis, E.A.; Issaeva, N.; Yarbrough, W.G.; Koo, J.S.; Burtness, B. Combined Aurora Kinase A (AURKA) and WEE1 inhibition demonstrates synergistic antitumor effect in squamous cell carcinoma of the head and neck. Clin. Cancer Res., 2019, 25(11), 3430-3442.
[http://dx.doi.org/10.1158/1078-0432.CCR-18-0440] [PMID: 30755439]
[186]
Daniele, S.; Sestito, S.; Pietrobono, D.; Giacomelli, C.; Chiellini, G.; Di Maio, D. Dual inhibition of PDK1 and aurora kinase A: An effective strategy to induce differentiation and apoptosis of human glioblastoma multiforme stem cells. ACS Chem. Neurosci, 2017, 8(1), 100-114.
[http://dx.doi.org/10.1021/acschemneuro.6b00251]
[187]
Casari, I.; Domenichini, A.; Sestito, S.; Capone, E.; Sala, G.; Rapposelli, S.; Falasca, M. Dual PDK1/Aurora kinase A inhibitors reduce pancreatic cancer cell proliferation and colony formation. Cancers, 2019, 11(11), 1695.
[http://dx.doi.org/10.3390/cancers11111695] [PMID: 31683659]
[188]
Caputo, E.; Miceli, R.; Motti, M.L.; Taté, R.; Fratangelo, F.; Botti, G.; Mozzillo, N.; Carriero, M.V.; Cavalcanti, E.; Palmieri, G.; Ciliberto, G.; Pirozzi, G.; Ascierto, P.A. AurkA inhibitors enhance the effects of B-RAF and MEK inhibitors in melanoma treatment. J. Transl. Med., 2014, 12(1), 216.
[http://dx.doi.org/10.1186/s12967-014-0216-z] [PMID: 25074438]
[189]
Horwacik, I.; Durbas, M.; Boratyn, E.; Węgrzyn, P.; Rokita, H. Targeting GD2 ganglioside and aurora A kinase as a dual strategy leading to cell death in cultures of human neuroblastoma cells. Cancer Lett., 2013, 341(2), 248-264.
[http://dx.doi.org/10.1016/j.canlet.2013.08.018] [PMID: 23962557]
[190]
Durbas, M.; Pabisz, P.; Wawak, K.; Wiśniewska, A.; Boratyn, E.; Nowak, I.; Horwacik, I.; Woźnicka, O.; Rokita, H. GD2 ganglioside-binding antibody 14G2a and specific aurora A kinase inhibitor MK-5108 induce autophagy in IMR-32 neuroblastoma cells. Apoptosis, 2018, 23(9-10), 492-511.
[http://dx.doi.org/10.1007/s10495-018-1472-9] [PMID: 30027525]
[191]
Liu, Y.; Hawkins, O.E.; Vilgelm, A.E.; Pawlikowski, J.S.; Ecsedy, J.A.; Sosman, J.A.; Kelley, M.C.; Richmond, A. Combining an aurora kinase inhibitor and a death receptor ligand/agonist antibody triggers apoptosis in melanoma cells and prevents tumor growth in preclinical mouse models. Clin. Cancer Res., 2015, 21(23), 5338-5348.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-0293] [PMID: 26152738]
[192]
Aurora A Inhibition Eliminates Myeloid Cell-Mediated Immunosuppression and Enhances the Efficacy of Anti-PD-L1 Therapy in Breast Cancer. Available from: https://europepmc.org/article/MED/30902796 (Accessed on: 2022 Sep 28).
[193]
Du, R.; Huang, C.; Liu, K.; Li, X.; Dong, Z. Targeting AURKA in Cancer: Molecular mechanisms and opportunities for cancer therapy. Mol. Cancer, 2021, 20(1), 15.
[http://dx.doi.org/10.1186/s12943-020-01305-3] [PMID: 33451333]
[194]
Defaux, J.; Antoine, M.; Logé, C.; Le Borgne, M.; Schuster, T.; Seipelt, I. Discovery of (7-aryl-1,5-naphthyridin-2-yl)ureas as dual inhibitors of ERK2 and Aurora B kinases with antiproliferative activity against cancer cells. Bioorganic Med. Chem. Lett., 2014, 4(16), 3748-52.
[195]
Li, J.; Hu, H.; Lang, Q.; Zhang, H.; Huang, Q.; Wu, Y.; Yu, L. A thienopyrimidine derivative induces growth inhibition and apoptosis in human cancer cell lines via inhibiting Aurora B kinase activity. Eur. J. Med. Chem., 2013, 65, 151-157.
[http://dx.doi.org/10.1016/j.ejmech.2013.04.058] [PMID: 23707920]
[196]
Pradhan, T.; Gupta, O.; Singh, G.; Monga, V. Aurora kinase inhibitors as potential anticancer agents: Recent advances. Eur. J. Med. Chem., 2021, 221, 113495.
[http://dx.doi.org/10.1016/j.ejmech.2021.113495] [PMID: 34020340]
[197]
Boss, D.S.; Witteveen, P.O.; van der Sar, J.; Lolkema, M.P.; Voest, E.E.; Stockman, P.K.; Ataman, O.; Wilson, D.; Das, S.; Schellens, J.H. Clinical evaluation of AZD1152, an i.v. inhibitor of Aurora B kinase, in patients with solid malignant tumors. Ann. Oncol., 2011, 22(2), 431-437.
[http://dx.doi.org/10.1093/annonc/mdq344] [PMID: 20924078]
[198]
Traynor, A.M.; Hewitt, M.; Liu, G.; Flaherty, K.T.; Clark, J.; Freedman, S.J.; Scott, B.B.; Leighton, A.M.; Watson, P.A.; Zhao, B.; O’Dwyer, P.J.; Wilding, G. Phase I dose escalation study of MK-0457, a novel Aurora kinase inhibitor, in adult patients with advanced solid tumors. Cancer Chemother. Pharmacol., 2011, 67(2), 305-314.
[http://dx.doi.org/10.1007/s00280-010-1318-9] [PMID: 20386909]
[199]
Manfredi, M.G.; Ecsedy, J.A.; Meetze, K.A.; Balani, S.K.; Burenkova, O.; Chen, W.; Galvin, K.M.; Hoar, K.M.; Huck, J.J.; LeRoy, P.J.; Ray, E.T.; Sells, T.B.; Stringer, B.; Stroud, S.G.; Vos, T.J.; Weatherhead, G.S.; Wysong, D.R.; Zhang, M.; Bolen, J.B.; Claiborne, C.F. Antitumor activity of MLN8054, an orally active small-molecule inhibitor of Aurora A kinase. Proc. Natl. Acad. Sci. USA, 2007, 104(10), 4106-4111.
[http://dx.doi.org/10.1073/pnas.0608798104] [PMID: 17360485]
[200]
Görgün, G.; Calabrese, E.; Hideshima, T.; Ecsedy, J.; Perrone, G.; Mani, M.; Ikeda, H.; Bianchi, G.; Hu, Y.; Cirstea, D.; Santo, L.; Tai, Y.T.; Nahar, S.; Zheng, M.; Bandi, M.; Carrasco, R.D.; Raje, N.; Munshi, N.; Richardson, P.; Anderson, K.C. A novel Aurora-A kinase inhibitor MLN8237 induces cytotoxicity and cell-cycle arrest in multiple myeloma. Blood, 2010, 115(25), 5202-5213.
[http://dx.doi.org/10.1182/blood-2009-12-259523] [PMID: 20382844]
[201]
Soncini, C.; Carpinelli, P.; Gianellini, L.; Fancelli, D.; Vianello, P.; Rusconi, L.; Storici, P.; Zugnoni, P.; Pesenti, E.; Croci, V.; Ceruti, R.; Giorgini, M.L.; Cappella, P.; Ballinari, D.; Sola, F.; Varasi, M.; Bravo, R.; Moll, J. PHA-680632, a novel Aurora kinase inhibitor with potent antitumoral activity. Clin. Cancer Res., 2006, 12(13), 4080-4089.
[http://dx.doi.org/10.1158/1078-0432.CCR-05-1964] [PMID: 16818708]
[202]
Benten, D.; Keller, G.; Quaas, A.; Schrader, J.; Gontarewicz, A.; Balabanov, S. Aurora kinase inhibitor PHA-739358 suppresses growth of hepatocellular carcinoma in vitro and in a xenograft mouse model. Neoplasia, 200911(9), 934.
[203]
Joshi, S.; Dhingra, A.K.; Chopra, B.; Guarve, K.; Bhateja, D. Therapeutic potential and clinical evidence of hesperidin as neuroprotective agent. Cent. Nerv. Syst. Agents Med. Chem., 2022, 22(1), 5-14.
[http://dx.doi.org/10.2174/1871524922666220404164405] [PMID: 35379141]
[204]
Georgieva, I.; Koychev, D.; Wang, Y.; Holstein, J.; Hopfenmüller, W.; Zeitz, M.; Grabowski, P. ZM447439, a novel promising aurora kinase inhibitor, provokes antiproliferative and proapoptotic effects alone and in combination with bio- and chemotherapeutic agents in gastroenteropancreatic neuroendocrine tumor cell lines. Neuroendocrinology, 2010, 91(2), 121-130.
[http://dx.doi.org/10.1159/000258705] [PMID: 19923785]
[205]
Dar, A.A.; Goff, L.W.; Majid, S.; Berlin, J.; El-Rifai, W. Aurora kinase inhibitors-rising stars in cancer therapeutics? Mol. Cancer Ther., 2010, 9(2), 268-278.
[http://dx.doi.org/10.1158/1535-7163.MCT-09-0765] [PMID: 20124450]
[206]
Chung, M.S.; Han, S.J. Endometriosis-associated angiogenesis and anti-angiogenic therapy for endometriosis. Front. Global Women's Health., 2022, 3, 856316
[http://dx.doi.org/10.3389/fgwh.2022.856316]
[207]
Chan, F.; Sun, C.; Perumal, M.; Nguyen, Q.D.; Bavetsias, V.; McDonald, E.; Martins, V.; Wilsher, N.E.; Raynaud, F.I.; Valenti, M.; Eccles, S.; te Poele, R.; Workman, P.; Aboagye, E.O.; Linardopoulos, S. Mechanism of action of the Aurora kinase inhibitor CCT129202 and in vivo quantification of biological activity. Mol. Cancer Ther., 2007, 6(12), 3147-3157.
[http://dx.doi.org/10.1158/1535-7163.MCT-07-2156] [PMID: 18089709]
[208]
Moreno, L.; Marshall, L.V.; Pearson, A.D.J.; Morland, B.; Elliott, M.; Campbell-Hewson, Q. . A phase I trial of AT9283 (a selective inhibitor of aurora kinases) in children and adolescents with solid tumors: A Cancer Research UK study. Clin. Cancer Res., 2015, 21(2), 267-73.
[209]
Joshi-Hangal, R.; Tang, C.; Sadikin, S.; Inloes, R.; Shi, C.; Severson, P.; Lamb, J.; Bearss, D.; Redkar, S.; Kanekal, S. Pharmacokinetics of MP529, a selective Aurora A kinase inhibitor, in a novel subcutaneous delivery system . Exp. Mol. Med., 2008, 68(9_Supplement), 5729.
[210]
VanderPorten, E.C.; Taverna, P.; Hogan, J.N.; Ballinger, M.D.; Flanagan, W.M.; Fucini, R.V. The Aurora kinase inhibitor SNS-314 shows broad therapeutic potential with chemotherapeutics and synergy with microtubule-targeted agents in a colon carcinoma model. Mol. Cancer Ther., 2009, 8(4), 930-939.
[http://dx.doi.org/10.1158/1535-7163.MCT-08-0754] [PMID: 19372566]
[211]
McLaughlin, J.; Markovtsov, V.; Li, H.; Wong, S.; Gelman, M.; Zhu, Y.; Franci, C.; Lang, D.W.; Pali, E.; Lasaga, J.; Low, C.; Zhao, F.; Chang, B.; Gururaja, T.L.; Xu, W.; Baluom, M.; Sweeny, D.; Carroll, D.; Sran, A.; Thota, S.; Parmer, M.; Romane, A.; Clemens, G.; Grossbard, E.; Qu, K.; Jenkins, Y.; Kinoshita, T.; Taylor, V.; Holland, S.J.; Argade, A.; Singh, R.; Pine, P.; Payan, D.G.; Hitoshi, Y. Preclinical characterization of Aurora kinase inhibitor R763/AS703569 identified through an image-based phenotypic screen. J. Cancer Res. Clin. Oncol., 2010, 136(1), 99-113.
[http://dx.doi.org/10.1007/s00432-009-0641-1] [PMID: 19609559]
[212]
Matulonis, U.A.; Lee, J.; Lasonde, B.; Tew, W.P.; Yehwalashet, A.; Matei, D.; Behbakht, K.; Grothusen, J.; Fleming, G.; Lee, N.K.; Arnott, J.; Bray, M.R.; Fletcher, G.; Brokx, R.D.; Castonguay, V.; Mackay, H.; Sidor, C.F.; Oza, A.M. ENMD-2076, an oral inhibitor of angiogenic and proliferation kinases, has activity in recurrent, platinum resistant ovarian cancer. Eur. J. Cancer, 2013, 49(1), 121-131.
[http://dx.doi.org/10.1016/j.ejca.2012.07.020] [PMID: 22921155]
[213]
Study of XL228 in Subjects With Chronic Myeloid Leukemia or Philadelphia-Chromosome-Positive Acute Lymphocytic Leukemia - Full Text View - ClinicalTrials. ClinicalTrials.gov. Available from: https://clinicaltrials.gov/ct2/show/NCT00464113(Accessed on: 2022 Aug 25)
[214]
TTP607 Clinical Trials. Clincosm. 2022. Available from: https://www.clincosm.com/drug/TTP607(Acessed on: 2022 Aug 25)
[215]
Schöffski, P.; Jones, S.F.; Dumez, H.; Infante, J.R.; Van Mieghem, E.; Fowst, C.; Gerletti, P.; Xu, H.; Jakubczak, J.L.; English, P.A.; Pierce, K.J.; Burris, H.A. Phase I, open-label, multicentre, dose-escalation, pharmacokinetic and pharmacodynamic trial of the oral aurora kinase inhibitor PF-03814735 in advanced solid tumours. Eur. J. Cancer, 2011, 47(15), 2256-2264.
[http://dx.doi.org/10.1016/j.ejca.2011.07.008] [PMID: 21852114]
[216]
A phase I pharmacologic study of CYC116, an oral aurora kinase inhibitor, in patients with advanced solid tumors. Available from: https://clinicaltrials.gov/ct2/show/NCT00560716(Acessed on: 2022 Aug 25).
[217]
Vitetta, L; Hall, S; Coulson, S. Metabolic interactions in the Gastrointestinal Tract (GIT): Host, commensal, probiotics, and bacteriophage influences. Microorganisms, 2015, 20153(4), 913.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy