Generic placeholder image

Current Chemical Biology

Editor-in-Chief

ISSN (Print): 2212-7968
ISSN (Online): 1872-3136

Review Article

Cyclin-Dependent Kinase as a Novel Therapeutic Target: An Endless Story

Author(s): Ahmed Mohamed Etman, Sherif Sabry Abdel Mageed, Mohamed Ahmed Ali and Mahmoud Abd El Monem El Hassab*

Volume 15, Issue 2, 2021

Published on: 23 November, 2020

Page: [139 - 162] Pages: 24

DOI: 10.2174/2212796814999201123194016

Price: $65

Abstract

Cyclin-Dependent Kinases (CDKs) are a family of enzymes that, along with their Cyclin partners, play a crucial role in cell cycle regulation at many biological functions such as proliferation, differentiation, DNA repair, and apoptosis. Thus, they are tightly regulated by a number of inhibitory and activating enzymes. Deregulation of these kinases’ activity either by amplification, overexpression or mutation of CDKs or Cyclins leads to uncontrolled proliferation of cancer cells. Hyperactivity of these kinases has been reported in a wide variety of human cancers. Hence, CDKs have been established as one of the most attractive pharmacological targets in the development of promising anticancer drugs. The elucidated structural features and the well-characterized molecular mechanisms of CDKs have been the guide in designing inhibitors to these kinases. Yet, they remain a challenging therapeutic class as they share conserved structure similarity in their active site. Several inhibitors have been discovered from natural sources or identified through high throughput screening and rational drug design approaches. Most of these inhibitors target the ATP binding pocket, therefore, they suffer from a number of limitations. Here, a growing number of ATP noncompetitive peptides and small molecules has been reported.

Keywords: CDK, cyclin, regulation, cell cycle, inhibitor, cancer.

Graphical Abstract
[1]
Malumbres M. Physiological relevance of cell cycle kinases. Physiol Rev 2011; 91(3): 973-1007.
[http://dx.doi.org/10.1152/physrev.00025.2010] [PMID: 21742793]
[2]
Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer 2009; 9(3): 153-66.
[http://dx.doi.org/10.1038/nrc2602] [PMID: 19238148]
[3]
Stone A, Sutherland RL, Musgrove EA. Inhibitors of cell cycle kinases: recent advances and future prospects as cancer therapeutics. Crit Rev Oncog 2012; 17(2): 175-98.
[http://dx.doi.org/10.1615/CritRevOncog.v17.i2.40] [PMID: 22471707]
[4]
Malumbres M. Cyclin-dependent kinases. Genome Biol 2014; 15(6): 122.
[http://dx.doi.org/10.1186/gb4184] [PMID: 25180339]
[5]
Ma Z, Wu Y, Jin J, et al. Phylogenetic analysis reveals the evolution and diversification of cyclins in eukaryotes. Mol Phylogenet Evol 2013; 66(3): 1002-10.
[http://dx.doi.org/10.1016/j.ympev.2012.12.007] [PMID: 23261709]
[6]
Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science 2002; 298(5600): 1912-34.
[http://dx.doi.org/10.1126/science.1075762] [PMID: 12471243]
[7]
Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov 2015; 14(2): 130-46.
[http://dx.doi.org/10.1038/nrd4504] [PMID: 25633797]
[8]
Harashima H, Dissmeyer N, Schnittger A. Cell cycle control across the eukaryotic kingdom. Trends Cell Biol 2013; 23(7): 345-56.
[http://dx.doi.org/10.1016/j.tcb.2013.03.002] [PMID: 23566594]
[9]
Otto T, Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nat Rev Cancer 2017; 17(2): 93-115.
[http://dx.doi.org/10.1038/nrc.2016.138] [PMID: 28127048]
[10]
Leal-Esteban LC, Fajas L. Cell cycle regulators in cancer cell metabolism. Biochim Biophys Acta Mol Basis Dis 2020; 1866(5): 165715.
[http://dx.doi.org/10.1016/j.bbadis.2020.165715] [PMID: 32035102]
[11]
Chan KS, Koh CG, Li HY. Mitosis-targeted anti-cancer therapies: where they stand. Cell Death Dis 2012; 3(10): e411.
[http://dx.doi.org/10.1038/cddis.2012.148] [PMID: 23076219]
[12]
Hydbring P, Malumbres M, Sicinski P. Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat Rev Mol Cell Biol 2016; 17(5): 280-92.
[http://dx.doi.org/10.1038/nrm.2016.27] [PMID: 27033256]
[13]
Peyressatre M, Prével C, Pellerano M, Morris MC. Targeting cyclin-dependent kinases in human cancers: from small molecules to Peptide inhibitors. Cancers (Basel) 2015; 7(1): 179-237.
[http://dx.doi.org/10.3390/cancers7010179] [PMID: 25625291]
[14]
Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 2013; 140(15): 3079-93.
[http://dx.doi.org/10.1242/dev.091744] [PMID: 23861057]
[15]
Nurse P. Universal control mechanism regulating onset of M-phase. Nature 1990; 344(6266): 503-8.
[http://dx.doi.org/10.1038/344503a0] [PMID: 2138713]
[16]
Morgan DO. Principles of CDK regulation. Nature 1995; 374: 131-4.
[http://dx.doi.org/10.1038/374131a0]
[17]
Malumbres M, Barbacid M. Mammalian cyclin-dependent kinases. Trends Biochem Sci 2005; 30(11): 630-41.
[http://dx.doi.org/10.1016/j.tibs.2005.09.005] [PMID: 16236519]
[18]
Santo L, Siu KT, Raje N. Targeting cyclin-dependent kinases and cell cycle progression in human cancers. Semin Oncol 2015; 42(6): 788-800.
[http://dx.doi.org/10.1053/j.seminoncol.2015.09.024] [PMID: 26615126]
[19]
Hunt T. Nobel Lecture. Protein synthesis, proteolysis, and cell cycle transitions. Biosci Rep 2002; 22(5-6): 465-86.
[http://dx.doi.org/10.1023/A:1022077317801] [PMID: 12635845]
[20]
Nurse PM. Cyclin dependent kinases and cell cycle control. Biosci Rep 2002; 22(5-6): 487-99.
[http://dx.doi.org/10.1023/A:1022017701871]
[21]
Hartwell LH. Yeast and cancer. Biosci Rep 2002; 22(3-6): 487-99.
[http://dx.doi.org/10.1023/A:1020918107706] [PMID: 12516780]
[22]
Malumbres M, Harlow E, Hunt T, et al. Cyclin-dependent kinases: a family portrait. Nat Cell Biol 2009; 11(11): 1275-6.
[http://dx.doi.org/10.1038/ncb1109-1275] [PMID: 19884882]
[23]
HUGO Gene Nomenclature Committee. HGNC database of human gene names. 2019. Available from: https://www.genenames.org/
[24]
Wood DJ, Endicott JA. Structural insights into the functional diversity of the CDK-cyclin family. Open Biol 2018; 8(9): 180112.
[http://dx.doi.org/10.1098/rsob.180112] [PMID: 30185601]
[25]
Nebreda AR. CDK activation by non-cyclin proteins. Curr Opin Cell Biol 2006; 18(2): 192-8.
[http://dx.doi.org/10.1016/j.ceb.2006.01.001] [PMID: 16488127]
[26]
Murray AW. Recycling the cell cycle: cyclins revisited. Cell 2004; 116(2): 221-34.
[http://dx.doi.org/10.1016/S0092-8674(03)01080-8] [PMID: 14744433]
[27]
Davidson G, Shen J, Huang YL, et al. Cell cycle control of wnt receptor activation. Dev Cell 2009; 17(6): 788-99.
[http://dx.doi.org/10.1016/j.devcel.2009.11.006] [PMID: 20059949]
[28]
Jiang M, Gao Y, Yang T, Zhu X, Chen J. Cyclin Y, a novel membrane-associated cyclin, interacts with PFTK1. FEBS Lett 2009; 583(13): 2171-8.
[http://dx.doi.org/10.1016/j.febslet.2009.06.010] [PMID: 19524571]
[29]
Fung TK, Poon RYC. A roller coaster ride with the mitotic cyclins. Semin Cell Dev Biol 2005; 16(3): 335-42.
[http://dx.doi.org/10.1016/j.semcdb.2005.02.014] [PMID: 15840442]
[30]
Coudreuse D, Nurse P. Driving the cell cycle with a minimal CDK control network. Nature 2010; 468(7327): 1074-9.
[http://dx.doi.org/10.1038/nature09543] [PMID: 21179163]
[31]
Ali F, Hindley C, McDowell G, et al. Cell cycle-regulated multi-site phosphorylation of Neurogenin 2 coordinates cell cycling with differentiation during neurogenesis. Development 2011; 138(19): 4267-77.
[http://dx.doi.org/10.1242/dev.067900] [PMID: 21852393]
[32]
Huertas P, Cortés-Ledesma F, Sartori AA, Aguilera A, Jackson SP. CDK targets Sae2 to control DNA-end resection and homologous recombination. Nature 2008; 455(7213): 689-92.
[http://dx.doi.org/10.1038/nature07215] [PMID: 18716619]
[33]
Chen S, Bohrer LR, Rai AN, et al. Cyclin-dependent kinases regulate epigenetic gene silencing through phosphorylation of EZH2. Nat Cell Biol 2010; 12(11): 1108-14.
[http://dx.doi.org/10.1038/ncb2116] [PMID: 20935635]
[34]
Kaneko S, Li G, Son J, Xu CF, Margueron R, Neubert TA. Phosphorylation of the PRC2 component Ezh2 is cell cycle-regulated and up-regulates its binding to ncRNA. genesdev.cshlp.org
[http://dx.doi.org/10.1101/gad.1983810]
[35]
Sherr CJ, Roberts JM. Living with or without cyclins and cyclin-dependent kinases. Genes & Dev 2004; 18: 2699-711.
[http://dx.doi.org/10.1101/gad.1256504]
[36]
Wang B, Song J. Structural basis for the ORC1-Cyclin A association. Protein Sci 2019; 28(9): 1727-33.
[http://dx.doi.org/10.1002/pro.3689] [PMID: 31309634]
[37]
Walter D, Hoffmann S, Komseli ES, Rappsilber J, Gorgoulis V, Sørensen CS. SCF(Cyclin F)-dependent degradation of CDC6 suppresses DNA re-replication. Nat Commun 2016; 7(1): 10530.
[http://dx.doi.org/10.1038/ncomms10530] [PMID: 26818844]
[38]
Ren S, Rollins BJ. Cyclin C/cdk3 promotes Rb-dependent G0 exit. Cell 2004; 117(2): 239-51.
[http://dx.doi.org/10.1016/S0092-8674(04)00300-9] [PMID: 15084261]
[39]
Zheng D, Cho YY, Lau ATY, et al. Cyclin-dependent kinase 3- mediated activating transcription factor 1 phosphorylation enhances cell transformation. Cancer Res 2008; 68(18): 7650-60.
[http://dx.doi.org/10.1158/0008-5472.CAN-08-1137] [PMID: 18794154]
[40]
Tomashevski A, Webster DR, Grammas P, Gorospe M, Kruman II. Cyclin-C-dependent cell-cycle entry is required for activation of non-homologous end joining DNA repair in postmitotic neurons. Cell Death Differ 2010; 17(7): 1189-98.
[http://dx.doi.org/10.1038/cdd.2009.221] [PMID: 20111042]
[41]
Aggarwal P, Vaites LP, Kim JK, et al. Nuclear cyclin D1/CDK4 kinase regulates CUL4 expression and triggers neoplastic growth via activation of the PRMT5 methyltransferase. Cancer Cell 2010; 18(4): 329-40.
[http://dx.doi.org/10.1016/j.ccr.2010.08.012] [PMID: 20951943]
[42]
Lavoie G, St-Pierre Y. Phosphorylation of human DNMT1: implication of cyclin-dependent kinases. Biochem Biophys Res Commun 2011; 409(2): 187-92.
[http://dx.doi.org/10.1016/j.bbrc.2011.04.115] [PMID: 21565170]
[43]
Roufayel R, Murshid N. CDK5: Key regulator of apoptosis and cell survival. Biomedicines 2019; 7(4): 88.
[http://dx.doi.org/10.3390/biomedicines7040088] [PMID: 31698798]
[44]
Su SC, Tsai LH. Cyclin-dependent kinases in brain development and disease. Annu Rev Cell Dev Biol 2011; 27(1): 465-91.
[http://dx.doi.org/10.1146/annurev-cellbio-092910-154023] [PMID: 21740229]
[45]
Nikolic M, Dudek H, Kwon YT, Ramos YFM, Tsai LH. The cdk5/p35 kinase is essential for neurite outgrowth during neuronal differentiation. Genes Dev 1996; 10(7): 816-25.
[http://dx.doi.org/10.1101/gad.10.7.816] [PMID: 8846918]
[46]
Lilja L, Yang SN, Webb DL, Juntti-Berggren L, Berggren PO, Bark C. Cyclin-dependent kinase 5 promotes insulin exocytosis. J Biol Chem 2001; 276(36): 34199-205.
[http://dx.doi.org/10.1074/jbc.M103776200] [PMID: 11443123]
[47]
Cheung ZH, Ip NY. Cdk5: a multifaceted kinase in neurodegenerative diseases. Trends Cell Biol 2012; 22(3): 169-75.
[http://dx.doi.org/10.1016/j.tcb.2011.11.003] [PMID: 22189166]
[48]
Ou CY, Poon VY, Maeder CI, et al. Two cyclin-dependent kinase pathways are essential for polarized trafficking of presynaptic components. Cell 2010; 141(5): 846-58.
[http://dx.doi.org/10.1016/j.cell.2010.04.011] [PMID: 20510931]
[49]
Park M, Watanabe S, Poon VYN, Ou CY, Jorgensen EM, Shen K. CYY-1/cyclin Y and CDK-5 differentially regulate synapse elimination and formation for rewiring neural circuits. Neuron 2011; 70(4): 742-57.
[http://dx.doi.org/10.1016/j.neuron.2011.04.002] [PMID: 21609829]
[50]
Mikolcevic P, Sigl R, Rauch V, Hess MW, Pfaller K, Barisic M. Cyclin-dependent kinase 16/PCTAIRE kinase 1 is activated by cyclin Y and is essential for spermatogenesis. Am Soc Micro 2012; 32(4): 868-79.
[http://dx.doi.org/10.1128/MCB.06261-11]
[51]
Fisher RP. Secrets of a double agent: CDK7 in cell-cycle control and transcription. J Cell Sci 2005; 118(Pt 22): 5171-80.
[http://dx.doi.org/10.1242/jcs.02718] [PMID: 16280550]
[52]
Szilagyi Z, Gustafsson CM. Emerging roles of Cdk8 in cell cycle control. Biochim Biophys Acta 2013; 1829(9): 916-20.
[http://dx.doi.org/10.1016/j.bbagrm.2013.04.010] [PMID: 23643644]
[53]
Firestein R, Bass AJ, Kim SY, et al. CDK8 is a colorectal cancer oncogene that regulates β-catenin activity. Nature 2008; 455(7212): 547-51.
[http://dx.doi.org/10.1038/nature07179] [PMID: 18794900]
[54]
Menzl I, Witalisz-Siepracka A, Sexl V. CDK8-Novel therapeutic opportunities. Pharmaceuticals (Basel) 2019; 12(2): 92.
[http://dx.doi.org/10.3390/ph12020092] [PMID: 31248103]
[55]
Battista N, Di Tommaso M, Bari M, Maccarrone M. The endocannabinoid system: an overview. Front Behav Neurosci 2012; 6: 9.
[http://dx.doi.org/10.3389/fnbeh.2012.00009] [PMID: 22457644]
[56]
Zhao X, Feng D, Wang Q, et al. Regulation of lipogenesis by cyclin-dependent kinase 8-mediated control of SREBP-1. J Clin Invest 2012; 122(7): 2417-27.
[http://dx.doi.org/10.1172/JCI61462] [PMID: 22684109]
[57]
Wang S, Fischer PM. Cyclin-dependent kinase 9: a key transcriptional regulator and potential drug target in oncology, virology and cardiology. Trends Pharmacol Sci 2008; 29(6): 302-13.
[http://dx.doi.org/10.1016/j.tips.2008.03.003] [PMID: 18423896]
[58]
Yu DS, Zhao R, Hsu EL, et al. Cyclin-dependent kinase 9-cyclin K functions in the replication stress response. EMBO Rep 2010; 11(11): 876-82.
[http://dx.doi.org/10.1038/embor.2010.153] [PMID: 20930849]
[59]
Chen HH, Wang YC, Fann MJ. Identification and characterization of the CDK12/cyclin L1 complex involved in alternative splicing regulation. Mol Cell Biol 2006; 26(7): 2736-45.
[http://dx.doi.org/10.1128/MCB.26.7.2736-2745.2006] [PMID: 16537916]
[60]
Bartkowiak B, Liu P, Phatnani HP, et al. CDK12 is a transcription elongation-associated CTD kinase, the metazoan ortholog of yeast Ctk1. Genes Dev 2010; 24(20): 2303-16.
[http://dx.doi.org/10.1101/gad.1968210] [PMID: 20952539]
[61]
Blazek D, Kohoutek J, Bartholomeeusen K, et al. The Cyclin K/Cdk12 complex maintains genomic stability via regulation of expression of DNA damage response genes. Genes Dev 2011; 25(20): 2158-72.
[http://dx.doi.org/10.1101/gad.16962311] [PMID: 22012619]
[62]
Chen HH, Wong YH, Geneviere AM, Fann MJ. CDK13/CDC2L5 interacts with L-type cyclins and regulates alternative splicing. Biochem Biophys Res Commun 2007; 354(3): 735-40.
[http://dx.doi.org/10.1016/j.bbrc.2007.01.049] [PMID: 17261272]
[63]
Guen VJ, Gamble C, Flajolet M, et al. CDK10/cyclin M is a protein kinase that controls ETS2 degradation and is deficient in STAR syndrome. Proc Natl Acad Sci USA 2013; 110(48): 19525-30.
[http://dx.doi.org/10.1073/pnas.1306814110] [PMID: 24218572]
[64]
Iorns E, Turner NC, Elliott R, et al. Identification of CDK10 as an important determinant of resistance to endocrine therapy for breast cancer. Cancer Cell 2008; 13(2): 91-104.
[http://dx.doi.org/10.1016/j.ccr.2008.01.001] [PMID: 18242510]
[65]
Trembley JH, Loyer P, Hu D, et al. Cyclin dependent kinase 11 in RNA transcription and splicing. Prog Nucleic Acid Res Mol Biol 2004; 77: 263-88.
[http://dx.doi.org/10.1016/S0079-6603(04)77007-5] [PMID: 15196895]
[66]
Loyer P, Trembley JH, Katona R, Kidd VJ, Lahti JM. Role of CDK/cyclin complexes in transcription and RNA splicing. Cell Signal 2005; 17(9): 1033-51.
[http://dx.doi.org/10.1016/j.cellsig.2005.02.005] [PMID: 15935619]
[67]
Loyer P, Trembley JH. Roles of CDK/Cyclin complexes in transcription and pre-mRNA splicing: Cyclins L and CDK11 at the cross-roads of cell cycle and regulation of gene expression. Semin Cell Dev Biol 2020; 107: 36-45.
[http://dx.doi.org/10.1016/j.semcdb.2020.04.016] [PMID: 32446654]
[68]
Cell-cycle regulation. WormBook: The online review of C. elegans biology. Pasadena (CA): WormBook; 2005-2018.
[69]
Loyer P, Trembley JH, Grenet JA, et al. Characterization of cyclin L1 and L2 interactions with CDK11 and splicing factors: influence of cyclin L isoforms on splice site selection. J Biol Chem 2008; 283(12): 7721-32.
[http://dx.doi.org/10.1074/jbc.M708188200] [PMID: 18216018]
[70]
Zhou Y, Shen JK, Hornicek FJ, Kan Q, Duan Z. The emerging roles and therapeutic potential of cyclin-dependent kinase 11 (CDK11) in human cancer. Oncotarget 2016; 7(26): 40846-59.
[http://dx.doi.org/10.18632/oncotarget.8519] [PMID: 27049727]
[71]
Donzelli M, Draetta GF. Regulating mammalian checkpoints through Cdc25 inactivation. EMBO Rep 2003; 4(7): 671-7.
[http://dx.doi.org/10.1038/sj.embor.embor887] [PMID: 12835754]
[72]
Vella S, Tavanti E, Hattinger CM, et al. Targeting CDKs with roscovitine increases sensitivity to DNA damaging drugs of human osteosarcoma cells. PLoS One 2016; 11(11): e0166233.
[http://dx.doi.org/10.1371/journal.pone.0166233] [PMID: 27898692]
[73]
Saeed U, Jalal N, Ashraf M. Roles of cyclin dependent kinase and CDK-activating kinase in cell cycle regulation: contemplation of intracellular interactions and functional characterization. Global J Med Res 2012; 12: 1-7.
[74]
Mariaule G, Belmont P. Cyclin-dependent kinase inhibitors as marketed anticancer drugs: where are we now? A short survey. Molecules 2014; 19(9): 14366-82.
[http://dx.doi.org/10.3390/molecules190914366] [PMID: 25215591]
[75]
Lubischer JL. The cell cycle, principles of control. David O. Morgan. Integr Comp Biol 2007; 47(5): 794-5.
[http://dx.doi.org/10.1093/icb/icm066]
[76]
Di Giovanni C, Novellino E, Chilin A, Lavecchia A, Marzaro G. Investigational drugs targeting cyclin-dependent kinases for the treatment of cancer: an update on recent findings (2013-2016). Expert Opin Investig Drugs 2016; 25(10): 1215-30.
[http://dx.doi.org/10.1080/13543784.2016.1234603] [PMID: 27606939]
[77]
Vermeulen K, Van Bockstaele DR, Berneman ZN. The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer. Cell Prolif 2003; 36(3): 131-49.
[http://dx.doi.org/10.1046/j.1365-2184.2003.00266.x] [PMID: 12814430]
[78]
Pavletich NP. Mechanisms of cyclin-dependent kinase regulation: structures of Cdks, their cyclin activators, and Cip and INK4 inhibitors. J Mol Biol 1999; 287(5): 821-8.
[http://dx.doi.org/10.1006/jmbi.1999.2640] [PMID: 10222191]
[79]
Law ME, Corsino PE, Narayan S, Law BK. Cyclin-dependent kinase inhibitors as anticancer therapeutics. Mol Pharmacol 2015; 88(5): 846-52.
[http://dx.doi.org/10.1124/mol.115.099325] [PMID: 26018905]
[80]
Mikolcevic P, Rainer J, Geley S. Orphan kinases turn eccentric: a new class of cyclin Y-activated, membrane-targeted CDKs. Cell Cycle 2012; 11(20): 3758-68.
[http://dx.doi.org/10.4161/cc.21592] [PMID: 22895054]
[81]
Arif A. Extraneuronal activities and regulatory mechanisms of the atypical cyclin-dependent kinase Cdk5. Biochem Pharmacol 2012; 84(8): 985-93.
[http://dx.doi.org/10.1016/j.bcp.2012.06.027] [PMID: 22795893]
[82]
Aga Y, Matsushita T, Ogi S, Onuma K, Sunamoto H, Ogawa A. Novel oral derivative UD-017, a highly selective CDK7 inhibitor, exhibits anticancer activity by inducing cell-cycle arrest and apoptosis in human colorectal cancer. Hiroshima J Med Sci 2020; 69(1): 23-31.
[http://dx.doi.org/10.24811/hjms.69.1_23]
[83]
Boutros R, Lobjois V, Ducommun B. CDC25 phosphatases in cancer cells: key players? Good targets? Nat Rev Cancer 2007; 7(7): 495-507.
[http://dx.doi.org/10.1038/nrc2169] [PMID: 17568790]
[84]
Echalier A, Endicott JA, Noble MEM. Recent developments in cyclin-dependent kinase biochemical and structural studies. Biochim Biophys Acta 2010; 1804(3): 511-9.
[http://dx.doi.org/10.1016/j.bbapap.2009.10.002] [PMID: 19822225]
[85]
Hoffmann I, Draetta G, Karsenti E. Activation of the phosphatase activity of human cdc25A by a cdk2-cyclin E dependent phosphorylation at the G1/S transition. EMBO J 1994; 13(18): 4302-10.
[http://dx.doi.org/10.1002/j.1460-2075.1994.tb06750.x] [PMID: 7523110]
[86]
Cui C, Zang T, Cao Y, Qin X, Zhang X. CDC25B is involved in the centrosomal microtubule nucleation in two-cell stage mouse embryos. Dev Growth Differ 2016; 58(9): 714-26.
[http://dx.doi.org/10.1111/dgd.12328] [PMID: 27885657]
[87]
Sur S, Agrawal DK. Phosphatases and kinases regulating CDC25 activity in the cell cycle: clinical implications of CDC25 overexpression and potential treatment strategies. Mol Cell Biochem 2016; 416(1-2): 33-46.
[http://dx.doi.org/10.1007/s11010-016-2693-2] [PMID: 27038604]
[88]
Shen Y, Sherman JW, Chen X, Wang R. Phosphorylation of CDC25C by AMP-activated protein kinase mediates a metabolic checkpoint during cell-cycle G2/M-phase transition. J Biol Chem 2018; 293(14): 5185-99.
[http://dx.doi.org/10.1074/jbc.RA117.001379] [PMID: 29467227]
[89]
Takizawa CG, Morgan DO. Control of mitosis by changes in the subcellular location of cyclin-B1-Cdk1 and Cdc25C. Curr Opin Cell Biol 2000; 12(6): 658-65.
[http://dx.doi.org/10.1016/S0955-0674(00)00149-6] [PMID: 11063929]
[90]
Malumbres M, Barbacid M. To cycle or not to cycle: a critical decision in cancer. Nat Rev Cancer 2001; 1(3): 222-31.
[http://dx.doi.org/10.1038/35106065] [PMID: 11902577]
[91]
Jeffrey PD, Tong L, Pavletich NP. Structural basis of inhibition of CDK-cyclin complexes by INK4 inhibitors. Genes Dev 2000; 14(24): 3115-25.
[http://dx.doi.org/10.1101/gad.851100] [PMID: 11124804]
[92]
Dorée M, Hunt T. From Cdc2 to Cdk1: when did the cell cycle kinase join its cyclin partner?. 2002; Vol. 115.
[93]
Harbour JW, Dean DC. The Rb/E2F pathway: expanding roles and emerging paradigms. Genes Dev 2000; 14(19): 2393-409.
[http://dx.doi.org/10.1101/gad.813200] [PMID: 11018009]
[94]
Adams PD. Regulation of the retinoblastoma tumor suppressor protein by cyclin/cdks. Biochim Biophys Acta 2001; 1471(3): M123-33.
[http://dx.doi.org/10.1016/S0304-419X(01)00019-1] [PMID: 11250068]
[95]
Ho A, Dowdy SF. Regulation of G(1) cell-cycle progression by oncogenes and tumor suppressor genes. Curr Opin Genet Dev 2002; 12(1): 47-52.
[http://dx.doi.org/10.1016/S0959-437X(01)00263-5] [PMID: 11790554]
[96]
Korah J, Canaff L, Lebrun JJ. The retinoblastoma tumor suppressor protein (pRb)/E2 promoter binding factor 1 (E2F1) pathway as a novel mediator of TGFβ-induced autophagy. J Biol Chem 2016; 291(5): 2043-54.
[http://dx.doi.org/10.1074/jbc.M115.678557] [PMID: 26598524]
[97]
Burkhart DL, Sage J. Cellular mechanisms of tumour suppression by the retinoblastoma gene. Nat Rev Cancer 2008; 8(9): 671-82.
[http://dx.doi.org/10.1038/nrc2399] [PMID: 18650841]
[98]
Stevaux O, Dyson NJ. A revised picture of the E2F transcriptional network and RB function. Curr Opin Cell Biol 2002; 14(6): 684-91.
[http://dx.doi.org/10.1016/S0955-0674(02)00388-5] [PMID: 12473340]
[99]
Uchida C. Roles of pRB in the regulation of nucleosome and chromatin structures. BioMed Res Int 2016; 2016: 5959721.
[http://dx.doi.org/10.1155/2016/5959721] [PMID: 28101510]
[100]
Thwaites MJ, Cecchini MJ, Passos DT, Welch I, Dick FA. Interchangeable roles for E2F transcriptional repression by the retinoblastoma protein and p27KIP1-cyclin-dependent kinase regulation in cell cycle control and tumor suppression. Mol Cell Biol 2017; 37(2): e00561-16.
[http://dx.doi.org/10.1128/MCB.00561-16] [PMID: 27821477]
[101]
Giacinti C, Giordano A. RB and cell cycle progression. Oncogene 2006; 25(38): 5220-7.
[http://dx.doi.org/10.1038/sj.onc.1209615] [PMID: 16936740]
[102]
Goel S, DeCristo MJ, McAllister SS, Zhao JJ. CDK4/6 Inhibition in Cancer: Beyond Cell Cycle Arrest. Trends Cell Biol 2018; 28(11): 911-25.
[http://dx.doi.org/10.1016/j.tcb.2018.07.002] [PMID: 30061045]
[103]
Zhang HS, Postigo AA, Dean DC. Active transcriptional repression by the Rb-E2F complex mediates G1 arrest triggered by p16INK4a, TGFbeta, and contact inhibition. Cell 1999; 97(1): 53-61.
[http://dx.doi.org/10.1016/S0092-8674(00)80714-X] [PMID: 10199402]
[104]
Karimian A, Ahmadi Y, Yousefi B. Multiple functions of p21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair (Amst) 2016; 42: 63-71.
[http://dx.doi.org/10.1016/j.dnarep.2016.04.008] [PMID: 27156098]
[105]
Muzi-Falconi M, Giannattasio M, Foiani M, Plevani P. The DNA polymerase α-primase complex: multiple functions and interactions. Scientific World J 2003; 3: 21-33.
[http://dx.doi.org/10.1100/tsw.2003.05] [PMID: 12806117]
[106]
Maziero RRD, Guaitolini CRF, Paschoal DM, et al. Effects of the addition of oocyte meiosis-inhibiting drugs on the expression of maturation-promoting factor components and organization of cytoplasmic organelles. Reprod Biol 2020; 20(1): 48-62.
[http://dx.doi.org/10.1016/j.repbio.2019.12.005] [PMID: 31889629]
[107]
Akaike Y, Chibazakura T. Aberrant activation of cyclin A-CDK induces G2/M-phase checkpoint in human cells. Cell Cycle 2020; 19(1): 84-96.
[http://dx.doi.org/10.1080/15384101.2019.1693119] [PMID: 31760882]
[108]
Kasten M, Giordano A. Cdk10, a Cdc2-related kinase, associates with the Ets2 transcription factor and modulates its transactivation activity. Oncogene 2001; 20(15): 1832-8.
[http://dx.doi.org/10.1038/sj.onc.1204295] [PMID: 11313931]
[109]
Nigg EA. Mitotic kinases as regulators of cell division and its checkpoints. Nat Rev Mol Cell Biol 2001; 2(1): 21-32.
[http://dx.doi.org/10.1038/35048096] [PMID: 11413462]
[110]
Peters JM. The anaphase-promoting complex: proteolysis in mitosis and beyond. Mol Cell 2002; 9(5): 931-43.
[http://dx.doi.org/10.1016/S1097-2765(02)00540-3] [PMID: 12049731]
[111]
Munro HN. Regulation of protein metabolism. Acta Anaesthesiol Scand Suppl 1974; 55: 66-73.
[http://dx.doi.org/10.1111/j.1399-6576.1974.tb00708.x] [PMID: 4531810]
[112]
Guiley KZ, Stevenson JW, Lou K, et al. p27 allosterically activates cyclin-dependent kinase 4 and antagonizes palbociclib inhibition. Science 2019; 366(6471): eaaw2106.
[http://dx.doi.org/10.1126/science.aaw2106] [PMID: 31831640]
[113]
Cheng M, Olivier P, Diehl JA, et al. The p21(Cip1) and p27(Kip1) CDK ‘inhibitors’ are essential activators of cyclin D-dependent kinases in murine fibroblasts. EMBO J 1999; 18(6): 1571-83.
[http://dx.doi.org/10.1093/emboj/18.6.1571] [PMID: 10075928]
[114]
Moser J, Miller I, Carter D, Spencer SL. Control of the restriction point by rb and p21. Proc Natl Acad Sci USA 2018; 115(35): E8219-27.
[http://dx.doi.org/10.1073/pnas.1722446115] [PMID: 30111539]
[115]
Dannenberg JH, van Rossum A, Schuijff L, te Riele H. Ablation of the retinoblastoma gene family deregulates G(1) control causing immortalization and increased cell turnover under growth-restricting conditions. Genes Dev 2000; 14(23): 3051-64.
[http://dx.doi.org/10.1101/gad.847700] [PMID: 11114893]
[116]
Bertoli C, Skotheim JM, de Bruin RAM. Control of cell cycle transcription during G1 and S phases. Nat Rev Mol Cell Biol 2013; 14(8): 518-28.
[http://dx.doi.org/10.1038/nrm3629] [PMID: 23877564]
[117]
Nurse P. A long twentieth century of the cell cycle and beyond. Cell 2000; 100(1): 71-8.
[http://dx.doi.org/10.1016/S0092-8674(00)81684-0] [PMID: 10647932]
[118]
Gordon EM, Ravicz JR, Liu S, Chawla SP, Hall FL. Cell cycle checkpoint control: The cyclin G1/Mdm2/p53 axis emerges as a strategic target for broad-spectrum cancer gene therapy - A review of molecular mechanisms for oncologists. Mol Clin Oncol 2018; 9(2): 115-34.
[http://dx.doi.org/10.3892/mco.2018.1657] [PMID: 30101008]
[119]
Bavle R. Enigmatic morpho insight: mitosis at a glance. J Oral Maxillofac Pathol 2014; 18(4): 2.
[http://dx.doi.org/10.4103/0973-029X.141175] [PMID: 24959028]
[120]
Meijer L, Jézéquel A, Ducommun B. Progress in cell cycle research Springer, US . 1996.
[http://dx.doi.org/10.1007/978-1-4615-5873-6]
[121]
Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000; 100(1): 57-70.
[http://dx.doi.org/10.1016/S0092-8674(00)81683-9] [PMID: 10647931]
[122]
Sánchez-Martínez C, Lallena MJ, Sanfeliciano SG, de Dios A. Cyclin dependent kinase (CDK) inhibitors as anticancer drugs: Recent advances (2015-2019). Bioorg Med Chem Lett 2019; 29(20): 126637.
[http://dx.doi.org/10.1016/j.bmcl.2019.126637] [PMID: 31477350]
[123]
Van Dross R, Browning PJ, Pelling JC. Do truncated cyclins contribute to aberrant cyclin expression in cancer? Cell Cycle 2006; 5(5): 472-7.
[http://dx.doi.org/10.4161/cc.5.5.2516] [PMID: 16552186]
[124]
Husdal A, Bukholm G, Bukholm IRK. The prognostic value and overexpression of cyclin A is correlated with gene amplification of both cyclin A and cyclin E in breast cancer patient. Cell Oncol 2006; 28(3): 107-16.
[http://dx.doi.org/10.1155/2006/721919] [PMID: 16823179]
[125]
Harwell RM, Mull BB, Porter DC, Keyomarsi K. Activation of cyclin-dependent kinase 2 by full length and low molecular weight forms of cyclin E in breast cancer cells. J Biol Chem 2004; 279(13): 12695-705.
[http://dx.doi.org/10.1074/jbc.M313407200] [PMID: 14701826]
[126]
Ekberg J, Holm C, Jalili S, et al. Expression of cyclin A1 and cell cycle proteins in hematopoietic cells and acute myeloid leukemia and links to patient outcome. Eur J Haematol 2005; 75(2): 106-15.
[http://dx.doi.org/10.1111/j.1600-0609.2005.00473.x] [PMID: 16004607]
[127]
Wang C, Yang Y, Zhang G, et al. Long noncoding RNA EMS connects c-Myc to cell cycle control and tumorigenesis. Proc Natl Acad Sci USA 2019; 116(29): 14620-9.
[http://dx.doi.org/10.1073/pnas.1903432116] [PMID: 31262817]
[128]
Dent P. Investigational CHK1 inhibitors in early phase clinical trials for the treatment of cancer. Expert Opin Investig Drugs 2019; 28(12): 1095-100.
[http://dx.doi.org/10.1080/13543784.2019.1694661] [PMID: 31783714]
[129]
Ding L, Cao J, Lin W, et al. The roles of cyclin-dependent kinases in cell-cycle progression and therapeutic strategies in human breast cancer. Int J Mol Sci 2020; 21(6): 1960.
[http://dx.doi.org/10.3390/ijms21061960] [PMID: 32183020]
[130]
Cheng W, Yang Z, Wang S, et al. Recent development of CDK inhibitors: An overview of CDK/inhibitor co-crystal structures. Eur J Med Chem 2019; 164: 615-39.
[http://dx.doi.org/10.1016/j.ejmech.2019.01.003] [PMID: 30639897]
[131]
Al Bitar S, Gali-Muhtasib H. The role of the cyclin dependent kinase inhibitor p21cip1/waf1 in targeting cancer: molecular mechanisms and novel therapeutics. Cancers (Basel) 2019; 11(10): 1475.
[http://dx.doi.org/10.3390/cancers11101475] [PMID: 31575057]
[132]
Venuto S, Merla G. E3 Ubiquitin ligase TRIM proteins, cell cycle and mitosis. Cells 2019; 8(5): 510.
[133]
Kalaszczynska I, Ciemerych MA. Mammalian development and cancer: a brief history of mice lacking D-type cyclins or CDK4/CDK6. In: Hinds P, Brown N, Eds. D-type cyclins and cancer Current cancer research.
[http://dx.doi.org/10.1007/978-3-319-64451-6_2]
[134]
Shi Y, Zou M, Farid NR, al-Sedairy ST. Evidence of gene deletion of p21 (WAF1/CIP1), a cyclin-dependent protein kinase inhibitor, in thyroid carcinomas. Br J Cancer 1996; 74(9): 1336-41.
[http://dx.doi.org/10.1038/bjc.1996.546] [PMID: 8912526]
[135]
Senderowicz AM, Sausville EA. Preclinical and clinical development of cyclin-dependent kinase modulators. J Natl Cancer Inst 2000; 92(5): 376-87.
[http://dx.doi.org/10.1093/jnci/92.5.376] [PMID: 10699068]
[136]
Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J Clin Oncol 2006; 24(11): 1770-83.
[http://dx.doi.org/10.1200/JCO.2005.03.7689] [PMID: 16603719]
[137]
Abate AA, Pentimalli F, Esposito L, Giordano A. ATP-noncompetitive CDK inhibitors for cancer therapy: an overview. Expert Opin Investig Drugs 2013; 22(7): 895-906.
[http://dx.doi.org/10.1517/13543784.2013.798641] [PMID: 23735075]
[138]
Austreid E, Lonning PE, Eikesdal HP. The emergence of targeted drugs in breast cancer to prevent resistance to endocrine treatment and chemotherapy. Expert Opin Pharmacother 2014; 15(5): 681-700.
[http://dx.doi.org/10.1517/14656566.2014.885952] [PMID: 24579888]
[139]
Cicenas J, Valius M. The CDK inhibitors in cancer research and therapy. J Cancer Res Clin Oncol 2011; 137(10): 1409-18.
[http://dx.doi.org/10.1007/s00432-011-1039-4] [PMID: 21877198]
[140]
Rizzolio F, Tuccinardi T, Caligiuri I, Lucchetti C, Giordano A. CDK inhibitors: from the bench to clinical trials. Curr Drug Targets 2010; 11(3): 279-90.
[http://dx.doi.org/10.2174/1389210200306074501] [PMID: 20210753]
[141]
Alexander B, Coppola G, Di Berardino D, et al. The effect of 6-dimethylaminopurine (6-DMAP) and cycloheximide (CHX) on the development and chromosomal complement of sheep parthenogenetic and nuclear transfer embryos. Mol Reprod Dev 2006; 73(1): 20-30.
[http://dx.doi.org/10.1002/mrd.20372] [PMID: 16211597]
[142]
Tararov VI, Tijsma A, Kolyachkina SV, et al. Chemical modification of the plant isoprenoid cytokinin N(6)-isopentenyladenosine yields a selective inhibitor of human enterovirus 71 replication. Eur J Med Chem 2015; 90: 406-13.
[http://dx.doi.org/10.1016/j.ejmech.2014.11.048] [PMID: 25461889]
[143]
Sharma S, Mehndiratta S, Kumar S, Singh J, Bedi PM, Nepali K. Purine Analogues as Kinase Inhibitors: A Review. Recent Patents Anticancer Drug Discov 2015; 10(3): 308-41.
[http://dx.doi.org/10.2174/1574892810666150617112230] [PMID: 26081925]
[144]
Sakurikar N, Thompson R, Montano R, Eastman A. A subset of cancer cell lines is acutely sensitive to the Chk1 inhibitor MK-8776 as monotherapy due to CDK2 activation in S phase. Oncotarget 2016; 7(2): 1380-94.
[http://dx.doi.org/10.18632/oncotarget.6364] [PMID: 26595527]
[145]
Shi XN, Li H, Yao H, et al. Adapalene inhibits the activity of cyclin-dependent kinase 2 in colorectal carcinoma. Mol Med Rep 2015; 12(5): 6501-8.
[http://dx.doi.org/10.3892/mmr.2015.4310] [PMID: 26398439]
[146]
Phoomvuthisarn P, Cross A, Glennon-Alty L, Wright HL, Edwards SW. The CDK inhibitor purvalanol A induces neutrophil apoptosis and increases the turnover rate of Mcl-1: potential role of p38-MAPK in regulation of Mcl-1 turnover. Clin Exp Immunol 2018; 192(2): 171-80.
[http://dx.doi.org/10.1111/cei.13107] [PMID: 29377076]
[147]
Marti GE, Stetler-Stevenson M, Grant ND, et al. Phase I trial of 7-hydroxystaurosporine and fludararbine phosphate: in vivo evidence of 7-hydroxystaurosporine induced apoptosis in chronic lymphocytic leukemia. Leuk Lymphoma 2011; 52(12): 2284-92.
[http://dx.doi.org/10.3109/10428194.2011.589547] [PMID: 21745173]
[148]
Li T, Christensen SD, Frankel PH, et al. A phase II study of cell cycle inhibitor UCN-01 in patients with metastatic melanoma: a California Cancer Consortium trial. Invest New Drugs 2012; 30(2): 741-8.
[http://dx.doi.org/10.1007/s10637-010-9562-8] [PMID: 20967484]
[149]
An X, Feng BM, Chen G, et al. Isolation and identification of phase I metabolites of butyrolactone I in rats. Xenobiotica 2017; 47(3): 236-44.
[http://dx.doi.org/10.3109/00498254.2016.1172280] [PMID: 27604497]
[150]
Ahn S, Jang DM, Park SC, et al. Cyclin-dependent kinase 5 inhibitor butyrolactone I elicits a partial agonist activity of peroxisome proliferator-activated receptor γ. Biomolecules 2020; 10(2): 275.
[http://dx.doi.org/10.3390/biom10020275] [PMID: 32054125]
[151]
Hofmeister CC, Poi M, Bowers MA, et al. A phase I trial of flavopiridol in relapsed multiple myeloma. Cancer Chemother Pharmacol 2014; 73(2): 249-57.
[http://dx.doi.org/10.1007/s00280-013-2347-y] [PMID: 24241210]
[152]
Medeiros A, Benítez D, Korn RS, et al. Mechanistic and biological characterisation of novel N5-substituted paullones targeting the biosynthesis of trypanothione in Leishmania. J Enzyme Inhib Med Chem 2020; 35(1): 1345-58.
[http://dx.doi.org/10.1080/14756366.2020.1780227] [PMID: 32588679]
[153]
Song XY, Kong LL, Chen NH. Indirubin. In: Du HS, Ed. Natural small molecule drugs from plants. 2018; pp. 529-32.
[154]
Pandey V, Ranjan N, Narne P, Babu PP. Roscovitine effectively enhances antitumor activity of temozolomide in vitro and in vivo mediated by increased autophagy and Caspase-3 dependent apoptosis. Sci Rep 2019; 9(1): 5012.
[http://dx.doi.org/10.1038/s41598-019-41380-1] [PMID: 30899038]
[155]
Wang Q, Chen D, Jin H, et al. Hymenialdisine: a marine natural product that acts on both osteoblasts and osteoclasts and prevents estrogen-dependent bone loss in mice. J Bone Miner Res 2020; 35(8): 1582-96.
[http://dx.doi.org/10.1002/jbmr.4025] [PMID: 32286705]
[156]
Pan H, Qiu H, Zhang K, et al. Fascaplysin derivatives are potent multitarget agents against alzheimer’s disease: in vitro and in vivo evidence. ACS Chem Neurosci 2019; 10(11): 4741-56.
[http://dx.doi.org/10.1021/acschemneuro.9b00503] [PMID: 31639294]
[157]
Singh U, Chashoo G, Khan SU, et al. Design of novel 3-Pyrimidinylazaindole CDK2/9 inhibitors with potent in vitro and in vivo antitumor efficacy in a triple-negative breast cancer model. J Med Chem 2017; 60(23): 9470-89.
[http://dx.doi.org/10.1021/acs.jmedchem.7b00663] [PMID: 29144137]
[158]
Cassandri M, Fioravanti R, Pomella S, et al. CDK9 as a valuable target in cancer: from natural compounds inhibitors to current treatment in pediatric soft tissue sarcomas. Front Pharmacol 2020; 11: 1230.
[http://dx.doi.org/10.3389/fphar.2020.01230] [PMID: 32903585]
[159]
Ognibene M, Pezzolo A. Roniciclib down-regulates stemness and inhibits cell growth by inducing nucleolar stress in neuroblastoma. Sci Rep 2020; 10(1): 12902.
[http://dx.doi.org/10.1038/s41598-020-69499-6] [PMID: 32737364]
[160]
Lam F, Abbas AY, Shao H, et al. Targeting RNA transcription and translation in ovarian cancer cells with pharmacological inhibitor CDKI-73. Oncotarget 2014; 5(17): 7691-704.
[http://dx.doi.org/10.18632/oncotarget.2296] [PMID: 25277198]
[161]
Walsby E, Pratt G, Shao H, et al. A novel Cdk9 inhibitor preferentially targets tumor cells and synergizes with fludarabine. Oncotarget 2014; 5(2): 375-85.
[http://dx.doi.org/10.18632/oncotarget.1568] [PMID: 24495868]
[162]
Rahaman MH, Yu Y, Zhong L, et al. CDKI-73: an orally bioavailable and highly efficacious CDK9 inhibitor against acute myeloid leukemia. Invest New Drugs 2019; 37(4): 625-35.
[http://dx.doi.org/10.1007/s10637-018-0661-2] [PMID: 30194564]
[163]
Rahaman MH, Lam F, Zhong L, et al. Targeting CDK9 for treatment of colorectal cancer. Mol Oncol 2019; 13(10): 2178-93.
[http://dx.doi.org/10.1002/1878-0261.12559] [PMID: 31398271]
[164]
DePinto W, Chu XJ, Yin X, et al. In vitro and in vivo activity of R547: a potent and selective cyclin-dependent kinase inhibitor currently in phase I clinical trials. Mol Cancer Ther 2006; 5(11): 2644-58.
[http://dx.doi.org/10.1158/1535-7163.MCT-06-0355] [PMID: 17121911]
[165]
Chu XJ, DePinto W, Bartkovitz D, et al. Discovery of [4-Amino-2-(1-methanesulfonylpiperidin-4-ylamino)pyrimidin-5-yl](2,3-difluoro-6- methoxyphenyl)methanone (R547), a potent and selective cyclin-dependent kinase inhibitor with significant in vivo antitumor activity. J Med Chem 2006; 49(22): 6549-60.
[http://dx.doi.org/10.1021/jm0606138] [PMID: 17064073]
[166]
Hacioğlu B, Kuş G, Kutlu HM, Kabadere S. The effect of R547, a cyclin-dependent kinase inhibitor, on hepatocellular carcinoma cell death. Turk J Biol 2020; 44(1): 24-33.
[PMID: 32123493]
[167]
Pennati M, Campbell AJ, Curto M, et al. Potentiation of paclitaxel-induced apoptosis by the novel cyclin-dependent kinase inhibitor NU6140: a possible role for survivin down-regulation. Mol Cancer Ther 2005; 4(9): 1328-37.
[http://dx.doi.org/10.1158/1535-7163.MCT-05-0022] [PMID: 16170024]
[168]
Arris CE, Boyle FT, Calvert AH, et al. Identification of novel purine and pyrimidine cyclin-dependent kinase inhibitors with distinct molecular interactions and tumor cell growth inhibition profiles. J Med Chem 2000; 43(15): 2797-804.
[http://dx.doi.org/10.1021/jm990628o] [PMID: 10956187]
[169]
Davies TG, Bentley J, Arris CE, et al. Structure-based design of a potent purine-based cyclin-dependent kinase inhibitor. Nat Struct Biol 2002; 9(10): 745-9.
[http://dx.doi.org/10.1038/nsb842] [PMID: 12244298]
[170]
Tripathy D, Im SA, Colleoni M, et al. Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): a randomised phase 3 trial. Lancet Oncol 2018; 19(7): 904-15.
[http://dx.doi.org/10.1016/S1470-2045(18)30292-4] [PMID: 29804902]
[171]
Hurvitz SA. Phase III MONALEESA-7 trial of premenopausal patients with HR+/HER2− advanced breast cancer (ABC) treated with endocrine therapy ± ribociclib: Overall survival (OS) results. J Clin Oncol 2019; 37(18_suppl)
[172]
Slamon DJ, Neven P, Chia S, et al. Phase III randomized study of ribociclib and fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: MONALEESA-3. J Clin Oncol 2018; 36(24): 2465-72.
[http://dx.doi.org/10.1200/JCO.2018.78.9909] [PMID: 29860922]
[173]
Sledge GW Jr, Toi M, Neven P, et al. MONARCH 2: abemaciclib in combination with fulvestrant in women with HR+/HER2− advanced breast cancer who had progressed while receiving endocrine therapy. J Clin Oncol 2017; 35(25): 2875-84.
[http://dx.doi.org/10.1200/JCO.2017.73.7585] [PMID: 28580882]
[174]
Dickler MN, Tolaney SM, Rugo HS, et al. MONARCH 1, a phase II study of abemaciclib, a CDK4 and CDK6 inhibitor, as a single agent, in patients with refractory HR+/HER2− metastatic breast cancer. Clin Cancer Res 2017; 23(17): 5218-24.
[http://dx.doi.org/10.1158/1078-0432.CCR-17-0754] [PMID: 28533223]
[175]
Goetz MP, Toi M, Campone M, et al. MONARCH 3: abemaciclib as initial therapy for advanced breast cancer. J Clin Oncol 2017; 35(32): 3638-46.
[http://dx.doi.org/10.1200/JCO.2017.75.6155] [PMID: 28968163]
[176]
Vassilev LT, Tovar C, Chen S, et al. Selective small-molecule inhibitor reveals critical mitotic functions of human CDK1. Proc Natl Acad Sci USA 2006; 103(28): 10660-5.
[http://dx.doi.org/10.1073/pnas.0600447103] [PMID: 16818887]
[177]
Kamath AV, Chong S, Chang M, Marathe PH. P-glycoprotein plays a role in the oral absorption of BMS-387032, a potent cyclin-dependent kinase 2 inhibitor, in rats. Cancer Chemother Pharmacol 2005; 55(2): 110-6.
[http://dx.doi.org/10.1007/s00280-004-0873-3] [PMID: 15338193]
[178]
Clark AS, Karasic TB, DeMichele A, et al. Palbociclib (PD0332991)—a selective and potent cyclin-dependent kinase inhibitor: a review of pharmacodynamics and clinical development. JAMA Oncol 2016; 2(2): 253-60.
[http://dx.doi.org/10.1001/jamaoncol.2015.4701] [PMID: 26633733]
[179]
DeMichele A, Clark AS, Tan KS, et al. CDK 4/6 inhibitor palbociclib (PD0332991) in Rb+ advanced breast cancer: phase II activity, safety, and predictive biomarker assessment. Clin Cancer Res 2015; 21(5): 995-1001.
[http://dx.doi.org/10.1158/1078-0432.CCR-14-2258] [PMID: 25501126]
[180]
de Bruijn P, Moghaddam-Helmantel IMG, de Jonge MJA, et al. Validated bioanalytical method for the quantification of RGB-286638, a novel multi-targeted protein kinase inhibitor, in human plasma and urine by liquid chromatography/tandem triple-quadrupole mass spectrometry. J Pharm Biomed Anal 2009; 50(5): 977-82.
[http://dx.doi.org/10.1016/j.jpba.2009.06.048] [PMID: 19628352]
[181]
Morales F, Giordano A. Overview of CDK9 as a target in cancer research. Cell Cycle 2016; 15(4): 519-27.
[http://dx.doi.org/10.1080/15384101.2016.1138186] [PMID: 26766294]
[182]
Joshi KS, Rathos MJ, Mahajan P, et al. P276-00, a novel cyclin-dependent inhibitor induces G1-G2 arrest, shows antitumor activity on cisplatin-resistant cells and significant in vivo efficacy in tumor models. Mol Cancer Ther 2007; 6(3): 926-34.
[http://dx.doi.org/10.1158/1535-7163.MCT-06-0614] [PMID: 17363487]
[183]
Poulsen A, William A, Blanchard S, et al. Structure-based design of nitrogen-linked macrocyclic kinase inhibitors leading to the clinical candidate SB1317/TG02, a potent inhibitor of cyclin dependant kinases (CDKs), Janus kinase 2 (JAK2), and Fms-like tyrosine kinase-3 (FLT3). J Mol Model 2013; 19(1): 119-30.
[http://dx.doi.org/10.1007/s00894-012-1528-7] [PMID: 22820730]
[184]
Benouaich-Amiel A, Mazza E, Massard C, Gaviani P, Fiorentini F, Scaburri A. Phase I study of the oral CDK-TRKA inhibitor PHA-848125 in recurrent malignant glioma (MG). J Clin Oncol 2010; 28: 2087.
[185]
Fry DW, Bedford DC, Harvey PH, et al. Cell cycle and biochemical effects of PD 0183812. A potent inhibitor of the cyclin D-dependent kinases CDK4 and CDK6. J Biol Chem 2001; 276(20): 16617-23.
[http://dx.doi.org/10.1074/jbc.M008867200] [PMID: 11278443]
[186]
O’Shaughnessy J, Wright GS, Thummala A, Danso M, Popovic LS, Pluard T. Trilaciclib improves overall survival when given with gemcitabine/carboplatin (GC) in patients with metastatic triple negative breast cancer (mTNBC) in a randomized phase II trial. Ann Oncol 2019; 30: v860-1.
[http://dx.doi.org/10.1093/annonc/mdz394.011]
[187]
Tan AR, Wright GS, Thummala AR, et al. Trilaciclib plus chemotherapy versus chemotherapy alone in patients with metastatic triple-negative breast cancer: a multicentre, randomised, open-label, phase 2 trial. Lancet Oncol 2019; 20(11): 1587-601.
[http://dx.doi.org/10.1016/S1470-2045(19)30616-3] [PMID: 31575503]
[188]
Sorrentino JA, Lai A, Weiss JM, Dragnev KH, Owonikoko TK, Adler S. 1671P Trilaciclib (trila) preserves and enhances immune system function in extensive-stage small cell lung cancer (ES-SCLC) patients receiving first-line chemotherapy. Annals Oncol 2018; 29(suppl_8): viii596-602.
[189]
Lima C, Roberts PJ, Priego VM, Divers SG. Trilaciclib (G1T28): a cyclin dependent kinase 4/6 inhibitor, in combination with etoposide and carboplatin (EP) for extensive stage small cell lung cancer (ES-SCLC)-phase 1b results. J Clin Oncol 2017; 35(Suppl.): 8568.
[http://dx.doi.org/10.1200/JCO.2017.35.15_suppl.8568]
[190]
Hart LI, Andric ZG, Hussein MA, Ferrarotto R, Beck JT, Subramanian J. Effect of trilaciclib, a CDK 4/6 inhibitor, on myelosuppression in patients with previously treated extensive-stage small cell lung cancer receiving topotecan. J Clin Oncol 2019; 37: 15.
[http://dx.doi.org/10.1200/JCO.2019.37.15_suppl.8505]
[191]
Berz D, Spira A, Gadgeel SM, Anderson IC, Goldman JW, Thompson J. Lerociclib (G1T38), an oral CDK4/6 inhibitor, dosed continuously in combination with osimertinib for EGFRmut non-small cell lung cancer: Initial phase Ib results. Ann Oncol 2019; 30: v631.
[http://dx.doi.org/10.1093/annonc/mdz260.059]
[192]
Freed DM, Hall CR, Strum JC, Roberts PJ. CDK4/6 inhibition with lerociclib (G1T38) delays acquired resistance to targeted therapies in preclinical models of non-small cell lung cancer. Proceedings of the American Association for Cancer Research Annual Meeting. Atlanta, GA. 2019.
[193]
Korb V, Tep K, Escriou V, et al. Current data on ATP-containing liposomes and potential prospects to enhance cellular energy status for hepatic applications. Crit Rev Ther Drug Carrier Syst 2008; 25(4): 305-45.
[http://dx.doi.org/10.1615/CritRevTherDrugCarrierSyst.v25.i4.10] [PMID: 18540841]
[194]
Adams PD, Sellers WR, Sharma SK, Wu AD, Nalin CM, Kaelin WGJ Jr. Identification of a cyclin-cdk2 recognition motif present in substrates and p21-like cyclin-dependent kinase inhibitors. Mol Cell Biol 1996; 16(12): 6623-33.
[http://dx.doi.org/10.1128/MCB.16.12.6623] [PMID: 8943316]
[195]
Sabt A, Eldehna WM, Al-Warhi T, et al. Discovery of 3,6-disubstituted pyridazines as a novel class of anticancer agents targeting cyclin-dependent kinase 2: synthesis, biological evaluation and in silico insights. J Enzyme Inhib Med Chem 2020; 35(1): 1616-30.
[http://dx.doi.org/10.1080/14756366.2020.1806259] [PMID: 32781872]
[196]
Al-Warhi T, Abo-Ashour MF, Almahli H, et al. Novel [(N-alkyl-3-indolylmethylene)hydrazono]oxindoles arrest cell cycle and induce cell apoptosis by inhibiting CDK2 and Bcl-2: synthesis, biological evaluation and in silico studies. J Enzyme Inhib Med Chem 2020; 35(1): 1300-9.
[http://dx.doi.org/10.1080/14756366.2020.1773814] [PMID: 32522063]
[197]
Al-Warhi T, Said MA, El Hassab MA, Aljaeed N, Ghabour HA, Almahli H. Unexpected synthesis, single-crystal X-ray structure, anticancer activity, and molecular docking studies of certain 2-((Imidazole/Benzimidazol-2-yl)thio)-1-arylethanones. Crystals (Basel) 2020; 10(6): 446.
[http://dx.doi.org/10.3390/cryst10060446]
[198]
Al-Warhi T, El Kerdawy AM, Aljaeed N, et al. Synthesis, biological evaluation and in silico studies of certain oxindole-indole conjugates as anticancer CDK inhibitors. Molecules 2020; 25(9): 2031.
[http://dx.doi.org/10.3390/molecules25092031] [PMID: 32349307]
[199]
Bagella L, Sun A, Tonini T, et al. A small molecule based on the pRb2/p130 spacer domain leads to inhibition of cdk2 activity, cell cycle arrest and tumor growth reduction in vivo. Oncogene 2007; 26(13): 1829-39.
[http://dx.doi.org/10.1038/sj.onc.1209987] [PMID: 17043661]
[200]
Ferguson M, Luciani MG, Finlan L, et al. The development of a CDK2-docking site peptide that inhibits p53 and sensitizes cells to death. Cell Cycle 2004; 3(1): 80-9.
[http://dx.doi.org/10.4161/cc.3.1.603] [PMID: 14657672]
[201]
Gondeau C, Gerbal-Chaloin S, Bello P, Aldrian-Herrada G, Morris MC, Divita G. Design of a novel class of peptide inhibitors of cyclin-dependent kinase/cyclin activation. J Biol Chem 2005; 280(14): 13793-800.
[http://dx.doi.org/10.1074/jbc.M413690200] [PMID: 15649889]
[202]
Canela N, Orzáez M, Fucho R, et al. Identification of an hexapeptide that binds to a surface pocket in cyclin A and inhibits the catalytic activity of the complex cyclin-dependent kinase 2-cyclin A. J Biol Chem 2006; 281(47): 35942-53.
[http://dx.doi.org/10.1074/jbc.M603511200] [PMID: 17001081]
[203]
Gius DR, Ezhevsky SA, Becker-Hapak M, Nagahara H, Wei MC, Dowdy SF. Transduced p16INK4a peptides inhibit hypophosphorylation of the retinoblastoma protein and cell cycle progression prior to activation of Cdk2 complexes in late G1. Cancer Res 1999; 59(11): 2577-80.
[PMID: 10363976]
[204]
Andrews MJI, McInnes C, Kontopidis G, et al. Design, synthesis, biological activity and structural analysis of cyclic peptide inhibitors targeting the substrate recruitment site of cyclin-dependent kinase complexes. Org Biomol Chem 2004; 2(19): 2735-41.
[http://dx.doi.org/10.1039/b409157d] [PMID: 15455144]
[205]
Takeda DY, Wohlschlegel JA, Dutta A. A bipartite substrate recognition motif for cyclin-dependent kinases. J Biol Chem 2001; 276(3): 1993-7.
[http://dx.doi.org/10.1074/jbc.M005719200] [PMID: 11067844]
[206]
Dai L, Liu Y, Liu J, et al. A novel cyclinE/cyclinA-CDK inhibitor targets p27(Kip1) degradation, cell cycle progression and cell survival: implications in cancer therapy. Cancer Lett 2013; 333(1): 103-12.
[http://dx.doi.org/10.1016/j.canlet.2013.01.025] [PMID: 23354589]
[207]
Fåhraeus R, Paramio JM, Ball KL, Laín S, Lane DP. Inhibition of pRb phosphorylation and cell-cycle progression by a 20-residue peptide derived from p16CDKN2/INK4A. Curr Biol 1996; 6(1): 84-91.
[http://dx.doi.org/10.1016/S0960-9822(02)00425-6] [PMID: 8805225]
[208]
Warenius HM, Kilburn JD, Essex JW, et al. Selective anticancer activity of a hexapeptide with sequence homology to a non-kinase domain of Cyclin Dependent Kinase 4. Mol Cancer 2011; 10(1): 72.
[http://dx.doi.org/10.1186/1476-4598-10-72] [PMID: 21668989]
[209]
Zheng YL, Li BS, Amin ND, Albers W, Pant HC. A peptide derived from cyclin-dependent kinase activator (p35) specifically inhibits Cdk5 activity and phosphorylation of tau protein in transfected cells. Eur J Biochem 2002; 269(18): 4427-34.
[http://dx.doi.org/10.1046/j.1432-1033.2002.03133.x] [PMID: 12230554]
[210]
Zheng YL, Amin ND, Hu YF, et al. A 24-residue peptide (p5), derived from p35, the Cdk5 neuronal activator, specifically inhibits Cdk5-p25 hyperactivity and tau hyperphosphorylation. J Biol Chem 2010; 285(44): 34202-12.
[http://dx.doi.org/10.1074/jbc.M110.134643] [PMID: 20720012]
[211]
Shukla V, Zheng YL, Mishra SK, et al. A truncated peptide from p35, a Cdk5 activator, prevents Alzheimer’s disease phenotypes in model mice. FASEB J 2013; 27(1): 174-86.
[http://dx.doi.org/10.1096/fj.12-217497] [PMID: 23038754]
[212]
Sundaram JR, Poore CP, Sulaimee NH, et al. Specific inhibition of p25/Cdk5 activity by the Cdk5 inhibitory peptide reduces neurodegeneration in vivo. J Neurosci 2013; 33(1): 334-43.
[http://dx.doi.org/10.1523/JNEUROSCI.3593-12.2013] [PMID: 23283346]
[213]
Lo MC, Ngo R, Dai K, et al. Development of a time-resolved fluorescence resonance energy transfer assay for cyclin-dependent kinase 4 and identification of its ATP-noncompetitive inhibitors. Anal Biochem 2012; 421(2): 368-77.
[http://dx.doi.org/10.1016/j.ab.2011.10.014] [PMID: 22056947]
[214]
Ohren JF, Chen H, Pavlovsky A, et al. Structures of human MAP kinase kinase 1 (MEK1) and MEK2 describe novel noncompetitive kinase inhibition. Nat Struct Mol Biol 2004; 11(12): 1192-7.
[http://dx.doi.org/10.1038/nsmb859] [PMID: 15543157]
[215]
Kubo A, Nakagawa K, Varma RK, et al. The p16 status of tumor cell lines identifies small molecule inhibitors specific for cyclin-dependent kinase 4. Clin Cancer Res 1999; 5(12): 4279-86.
[PMID: 10632371]
[216]
Diccianni MB, Yu J, Meppelink G, et al. 3-amino thioacridone inhibits DNA synthesis and induce DNA damage in T-cell acute lymphoblastic leukemia (T-ALL) in a p16-dependent manner. J Exp Ther Oncol 2004; 4(3): 223-37.
[PMID: 15724842]
[217]
Diccianni MB, Batova A, Yu J, et al. Shortened survival after relapse in T-cell acute lymphoblastic leukemia patients with p16/p15 deletions. Leuk Res 1997; 21(6): 549-58.
[http://dx.doi.org/10.1016/S0145-2126(97)00007-6] [PMID: 9279366]
[218]
Uchiyama H, Sowa Y, Wakada M, et al. Cyclin-dependent kinase inhibitor SU9516 enhances sensitivity to methotrexate in human T-cell leukemia Jurkat cells. Cancer Sci 2010; 101(3): 728-34.
[http://dx.doi.org/10.1111/j.1349-7006.2009.01449.x] [PMID: 20059476]
[219]
D’assoro AB, Busby R, Acu ID, Quatraro C, Reinholz MM, Farrugia DJ. Impaired p53 function leads to centrosome amplification, acquired ERα phenotypic heterogeneity and distant metastases in breast cancer MCF-7 xenografts. Oncogene 2008; 27(28): 3901-11.
[220]
Takagi K, Sowa Y, Cevik OM, Nakanishi R, Sakai T. CDK inhibitor enhances the sensitivity to 5-fluorouracil in colorectal cancer cells. Int J Oncol 2008; 32(5): 1105-10.
[http://dx.doi.org/10.3892/ijo.32.5.1105] [PMID: 18425338]
[221]
Gao N, Kramer L, Rahmani M, Dent P, Grant S. The three-substituted indolinone cyclin-dependent kinase 2 inhibitor 3-[1-(3H-imidazol-4-yl)-meth-(Z)-ylidene]-5-methoxy-1,3-dihydro-indol-2-one (SU9516) kills human leukemia cells via down-regulation of Mcl-1 through a transcriptional mechanism. Mol Pharmacol 2006; 70(2): 645-55.
[http://dx.doi.org/10.1124/mol.106.024505] [PMID: 16672643]
[222]
Lane ME, Yu B, Rice A, et al. A novel cdk2-selective inhibitor, SU9516, induces apoptosis in colon carcinoma cells. Cancer Res 2001; 61(16): 6170-7.
[PMID: 11507069]
[223]
Corsino P, Horenstein N, Ostrov D, et al. A novel class of cyclin-dependent kinase inhibitors identified by molecular docking act through a unique mechanism. J Biol Chem 2009; 284(43): 29945-55.
[http://dx.doi.org/10.1074/jbc.M109.055251] [PMID: 19710018]
[224]
Zhang B, Corbel C, Guéritte F, Couturier C, Bach S, Tan VBC. An in silico approach for the discovery of CDK5/p25 interaction inhibitors. Biotechnol J 2011; 6(7): 871-81.
[http://dx.doi.org/10.1002/biot.201100139] [PMID: 21681969]
[225]
Corbel C, Wang Q, Bousserouel H, et al. First BRET-based screening assay performed in budding yeast leads to the discovery of CDK5/p25 interaction inhibitors. Biotechnol J 2011; 6(7): 860-70.
[http://dx.doi.org/10.1002/biot.201100138] [PMID: 21681968]
[226]
Betzi S, Alam R, Martin M, et al. Discovery of a potential allosteric ligand binding site in CDK2. ACS Chem Biol 2011; 6(5): 492-501.
[http://dx.doi.org/10.1021/cb100410m] [PMID: 21291269]
[227]
Liu H, Liu K, Huang Z, et al. A chrysin derivative suppresses skin cancer growth by inhibiting cyclin-dependent kinases. J Biol Chem 2013; 288(36): 25924-37.
[http://dx.doi.org/10.1074/jbc.M113.464669] [PMID: 23888052]
[228]
Beaver JA, Amiri-Kordestani L, Charlab R, et al. FDA approval: Palbociclib for the treatment of postmenopausal patients with estrogen receptor-positive, HER2-negative metastatic breast cancer. Clin Cancer Res 2015; 21(21): 4760-6.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-1185] [PMID: 26324739]
[229]
McCain J. First-in-class CDK4/6 inhibitor palbociclib could usher in a new wave of combination therapies for HR+, HER2− breast cancer. P&T 2015; 40(8): 511-20.
[PMID: 26236140]
[230]
Finn RS, Dering J, Conklin D, et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res 2009; 11(5): R77.
[http://dx.doi.org/10.1186/bcr2419] [PMID: 19874578]
[231]
Konecny GE, Winterhoff B, Kolarova T, et al. Expression of p16 and retinoblastoma determines response to CDK4/6 inhibition in ovarian cancer. Clin Cancer Res 2011; 17(6): 1591-602.
[http://dx.doi.org/10.1158/1078-0432.CCR-10-2307] [PMID: 21278246]
[232]
Otterson GA, Kratzke RA, Coxon A, Kim YW, Kaye FJ. Absence of p16INK4 protein is restricted to the subset of lung cancer lines that retains wildtype RB. Oncogene 1994; 9(11): 3375-8.
[http://dx.doi.org/10.1016/0169-5002(95)98734-r] [PMID: 7936665]
[233]
Shapiro GI, Park JE, Edwards CD, et al. Multiple mechanisms of p16INK4A inactivation in non-small cell lung cancer cell lines. Cancer Res 1995; 55(24): 6200-9.
[PMID: 8521414]
[234]
Wang L, Wang J, Blaser BW, et al. Pharmacologic inhibition of CDK4/6: mechanistic evidence for selective activity or acquired resistance in acute myeloid leukemia. Blood 2007; 110(6): 2075-83.
[http://dx.doi.org/10.1182/blood-2007-02-071266] [PMID: 17537993]
[235]
Biankin AV, Hudson TJ. Somatic variation and cancer: therapies lost in the mix. Hum Genet 2011; 130(1): 79-91.
[http://dx.doi.org/10.1007/s00439-011-1010-0] [PMID: 21643984]
[236]
Dean JL, Thangavel C, McClendon AK, Reed CA, Knudsen ES. Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure. Oncogene 2010; 29(28): 4018-32.
[http://dx.doi.org/10.1038/onc.2010.154] [PMID: 20473330]
[237]
Barrière C, Santamaría D, Cerqueira A, et al. Mice thrive without Cdk4 and Cdk2. Mol Oncol 2007; 1(1): 72-83.
[http://dx.doi.org/10.1016/j.molonc.2007.03.001] [PMID: 19383288]
[238]
Cánepa ET, Scassa ME, Ceruti JM, et al. INK4 proteins, a family of mammalian CDK inhibitors with novel biological functions. IUBMB Life 2007; 59(7): 419-26.
[http://dx.doi.org/10.1080/15216540701488358] [PMID: 17654117]
[239]
Santamaría D, Barrière C, Cerqueira A, et al. Cdk1 is sufficient to drive the mammalian cell cycle. Nature 2007; 448(7155): 811-5.
[http://dx.doi.org/10.1038/nature06046] [PMID: 17700700]
[240]
Davis ST, Benson BG, Bramson HN, et al. Prevention of chemotherapy-induced alopecia in rats by CDK inhibitors. Science 2001; 291(5501): 134-7.
[http://dx.doi.org/10.1126/science.291.5501.134] [PMID: 11141566]
[241]
DiPippo AJ, Patel NK, Barnett CM. Cyclin-dependent kinase inhibitors for the treatment of breast cancer: past, present, and future. Pharmacotherapy 2016; 36(6): 652-67.
[http://dx.doi.org/10.1002/phar.1756] [PMID: 27087139]
[242]
Vora SR, Juric D, Kim N, et al. CDK 4/6 inhibitors sensitize PIK3CA mutant breast cancer to PI3K inhibitors. Cancer Cell 2014; 26(1): 136-49.
[http://dx.doi.org/10.1016/j.ccr.2014.05.020] [PMID: 25002028]

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