6,729
Views
90
CrossRef citations to date
0
Altmetric
Review

Targeting the cell cycle in breast cancer: towards the next phase

, , & ORCID Icon
Pages 1871-1885 | Received 02 May 2018, Accepted 07 Jul 2018, Published online: 11 Sep 2018

References

  • Arteaga CL, Sliwkowski MX, Osborne CK, et al. Treatment of HER2-positive breast cancer: current status and future perspectives. Nat Rev Clin Oncol. 2012;9:16–32.
  • Russnes HG, Lingjærde OC, Børresen-Dale A-L CC. Breast cancer molecular stratification: from intrinsic subtypes to integrative clusters. Am J Pathol. 2017;187:2152–2162.
  • Gingras I, Gebhart G, De Azambuja E, et al. HER2-positive breast cancer is lost in translation: time for patient-centered research. Nat Rev Clin Oncol. 2017;14:669–681.
  • Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363:1938–1948.
  • Ignatiadis M, Sotiriou C. Luminal breast cancer: from biology to treatment. Nat Rev Clin Oncol. 2013;10:494–506.
  • Bianchini G, Balko JM, Mayer IA, et al. Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 2016;13:674–690.
  • Osborne CK, Schiff R. Mechanisms of endocrine resistance in breast cancer. Annu Rev Med. 2011;62:233–247.
  • Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9:153–166.
  • Malumbres M. Cyclin-dependent kinases. Genome Biol. 2014;15:122.
  • Sivakumar S, Gorbsky GJ. Spatiotemporal regulation of the anaphase-promoting complex in mitosis. Nat Rev Mol Cell Biol. 2015;16:82–94.
  • Zhou Z, He M, Shah AA, et al. Insights into APC/C: from cellular function to diseases and therapeutics. Cell Div. 2016;11:9.
  • Senft D, Qi J, Ronai ZA. Ubiquitin ligases in oncogenic transformation and cancer therapy. Nat Rev Cancer. 2018;18:69–88.
  • Platet N, Cathiard AM, Gleizes M, et al. Estrogens and their receptors in breast cancer progression: a dual role in cancer proliferation and invasion. Crit Rev Oncol Hematol. 2004;51:55–67.
  • Klein EA, Assoian RK. Transcriptional regulation of the cyclin D1 gene at a glance. J Cell Sci. 2008;121:3853–3857.
  • Sabbah M, Courilleau D, Mester J, et al. Estrogen induction of the cyclin D1 promoter: involvement of a cAMP response-like element. Proc Natl Acad Sci USA. 1999;96:11217–11222.
  • Bertoli C, Skotheim JM, De Bruin RAM. Control of cell cycle transcription during G1 and S phases. Nat Rev Mol Cell Biol. 2013;14:518–528.
  • Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature. 2004;432:316–323.
  • Musacchio A. The molecular biology of spindle assembly checkpoint signaling dynamics. Curr Biol. 2015;25:R1002–18.
  • Lara-Gonzalez P, Westhorpe FG, Taylor SS. The spindle assembly checkpoint. Curr Biol. 2012;22:R966–80.
  • Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.
  • Xu H, Eirew P, Mullaly SC, et al. The omics of triple-negative breast cancers. Clin Chem. 2014;60:122–133.
  • Curtis C, Shah SP, Chin S-F, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352.
  • Daniel J, Coulter J, Woo J-H, et al. High levels of the Mps1 checkpoint protein are protective of aneuploidy in breast cancer cells. Proc Natl Acad Sci USA. 2011;108:5384–5389.
  • Yuan B, Xu Y, Woo J-H, et al. Increased expression of mitotic checkpoint genes in breast cancer cells with chromosomal instability. Clin Cancer Res. 2006;12:405–410.
  • Patel N, Weekes D, Drosopoulos K, et al. Integrated genomics and functional validation identifies malignant cell specific dependencies in triple negative breast cancer. Nat Commun. 2018;9:1044.
  • Brough R, Frankum JR, Sims D, et al. Functional viability profiles of breast cancer. Cancer Discov. 2011;1:260–273.
  • Dominguez-Brauer C, Thu KL, Mason JM, et al. Targeting mitosis in cancer: emerging strategies. Mol Cell. 2015;60:524–536.
  • Dumontet C, Jordan MA. Microtubule-binding agents: a dynamic field of cancer therapeutics. Nat Rev Drug Discov. 2010;9:790–803.
  • De Laurentiis M, Cancello G, D’Agostino D, et al. Taxane-based combinations as adjuvant chemotherapy of early breast cancer: a meta-analysis of randomized trials. J Clin Oncol. 2008;26:44–53.
  • Gradishar WJ. Taxanes for the treatment of metastatic breast cancer. Breast Cancer. 2012;6:159–171.
  • Finn RS, Aleshin A, Slamon DJ. Targeting the cyclin-dependent kinases (CDK) 4/6 in estrogen receptor-positive breast cancers. Breast Cancer Res. 2016;18:17.
  • Johnson SF, Cruz C, Greifenberg AK, et al. CDK12 inhibition reverses de novo and acquired PARP inhibitor resistance in BRCA wild-type and mutated models of triple-negative breast cancer. Cell Rep. 2016;17:2367–2381.
  • Reddy HKDL, Mettus RV, Rane SG, et al. Cyclin-dependent kinase 4 expression is essential for neu-induced breast tumorigenesis. Cancer Res. 2005;65:10174–10178.
  • Yu Q, Sicinska E, Geng Y, et al. Requirement for CDK4 kinase function in breast cancer. Cancer Cell. 2006;9:23–32.
  • Yu Q, Geng Y, Sicinski P. Specific protection against breast cancers by cyclin D1 ablation. Nature. 2001;411:1017–1021.
  • Landis MW, Pawlyk BS, Li T, et al. Cyclin D1-dependent kinase activity in murine development and mammary tumorigenesis. Cancer Cell. 2006;9:13–22.
  • Malumbres M, Sotillo R, Santamaría D, et al. Mammalian cells cycle without the D-type cyclin-dependent kinases Cdk4 and Cdk6. Cell. 2004;118:493–504.
  • Kozar K, Ciemerych MA, Rebel VI, et al. Mouse development and cell proliferation in the absence of D-cyclins. Cell. 2004;118:477–491.
  • Rocca A, Schirone A, Maltoni R, et al. Progress with palbociclib in breast cancer: latest evidence and clinical considerations. Ther Adv Med Oncol. 2016;9:83–105.
  • O’Leary B, Finn RS, Turner NC. Treating cancer with selective CDK4/6 inhibitors. Nat Rev Clin Oncol. 2016;13:417–430.
  • Barroso-Sousa R, Shapiro GI, Tolaney SM. Clinical development of the CDK4/6 inhibitors ribociclib and abemaciclib in breast cancer. Breast Care. 2016;11:167–173.
  • Klein ME, M K, Davis LE, et al. CDK4/6 inhibitors: the mechanism of action may not be as simple as once thought. Cancer Cell. 2018. DOI:10.1016/j.ccell.2018.03.023.
  • 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:R77.
  • Finn RS, Crown JP, Lang I, et al. The cyclin-dependent kinase 4/6 inhibitor palbociclib in combination with letrozole versus letrozole alone as first-line treatment of oestrogen receptor-positive, HER2-negative, advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2 study. Lancet Oncol. 2015;16:25–35.
  • Fang H, Huang D, Yang F, et al. Potential biomarkers of CDK4/6 inhibitors in hormone receptor-positive advanced breast cancer. Breast Cancer Res Treat. 2017. DOI:10.1007/s10549-017-4612-y
  • Knudsen ES, Witkiewicz AK. The strange case of CDK4/6 inhibitors: mechanisms, resistance, and combination strategies. Trends Cancer Res. 2017;3:39–55.
  • Turner NC, Liu Y, Zhu Z, et al. Abstract CT039 - Cyclin E1 (CCNE1) expression associates with benefit from palbociclib in metastatic breast cancer (MBC) in the PALOMA3 trial [Internet]. American Association for Cancer Research, Annual Conference; 2018 Apr 15, Chicago, IL [cited 2018 May 1]. Available from: http://cancerres.aacrjournals.org/content/78/13_Supplement/CT039
  • Mt H-A, Palafox M, Asghar U, et al. Early adaptation and acquired resistance to CDK4/6 inhibition in estrogen receptor-positive breast cancer. Cancer Res. 2016;76:2301–2313.
  • Jansen VM, Bhola NE, Bauer JA, et al. Kinome-wide RNA interference screen reveals a role for PDK1 in acquired resistance to CDK4/6 inhibition in ER-positive breast cancer. Cancer Res. 2017;77:2488–2499.
  • Lenihan C, Bouchekioua-Bouzaghou K, Shia A, et al. Abstract P3-06-02: characterization of resistance to the selective CDK4/6 inhibitor palbociclib in ER positive breast cancer. Cancer Res. 2016;76:P3–06–02–P3–06–02.
  • Asghar U, Barr A, Cutts R, et al. 44PUnravelling mechanisms of resistance to CDK4/6 inhibitors using triple negative breast cancer (TNBC). Ann Oncol. 2017 [ cited 2018 Mar 18];28. Available from: https://academic.oup.com/annonc/article-abstract/doi/10.1093/annonc/mdx145/3792864
  • Dean JL, Thangavel C, McClendon AK, et al. Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure. Oncogene. 2010;29:4018–4032.
  • Mao P, Kusiel J, Cohen O, et al. Abstract PD4-01: the role of FGF/FGFR axis in resistance to SERDs and CDK4/6 inhibitors in ER+ breast cancer. Cancer Res. 2018;78:PD4–01–PD4–01.
  • Formisano L, Lu Y, Jansen VM, et al. Abstract 1008: gain-of-function kinase library screen identifies FGFR1 amplification as a mechanism of resistance to antiestrogens and CDK4/6 inhibitors in ER+ breast cancer. Cancer Res. 2017;77:1008.
  • Pan Q, Sathe A, Tong Z, et al. 37Identification of molecular mechanisms that confer therapy response to CDK4/6 inhibition using a genome-wide CRIPR-dCsa9 gain-of-function screen. Ann Oncol. 2017 [ cited 2018 Mar 18];28. Available from: https://academic.oup.com/annonc/article/doi/10.1093/annonc/mdx511.003/4469352/37Identification-of-molecular-mechanisms-that
  • Condorelli R, Spring L, O’Shaughnessy J, Lacroix L, Bailleux C, Scott V, Dubois J, Nagy Rj, Lanman Rb, Lafrate Aj, et al. Polyclonal RB1 mutations and acquired resistance to CDK 4/6 inhibitors in patients with metastatic breast cancer. Ann Oncol. 2017. DOI:10.1093/annonc/mdx784
  • Michaloglou C, Crafter C, Siersbæk R, et al. Combined inhibition of mTOR and CDK4/6 is required for optimal blockade of E2F function and long term growth inhibition in estrogen receptor positive breast cancer. Mol Cancer Ther. 2018. DOI:10.1158/1535-7163.MCT-17-0537
  • Nikitorowicz-Buniak J, Pancholi S, Simigdala N, et al. Abstract P1-09-03: global knockdown of cellular kinases identifies MPS1 as a novel modulator of endocrine and palbociclib resistance highlighting a new role for MPS1 inhibitors. Cancer Res. 2018;78:P1–09–03–P1–09–03.
  • Nikitorowicz-Buniak J, Pancholi S, Simigdala N, et al. Abstract 4950 - MPS1 as a novel target in endocrine- and palbociclib-resistant estrogen receptor positive breast cancer [Internet]. American Association for Cancer Research, Annual Conference; 2018 Apr 15, Chicago, IL [cited 2018 May 1]. Available from: http://cancerres.aacrjournals.org/content/78/13_Supplement/CT039
  • Liu X, The WM. MPS1 family of protein kinases. Annu Rev Biochem. 2012;81:561–585.
  • Al-Ejeh F, Simpson PT, Sanus JM, et al. Meta-analysis of the global gene expression profile of triple-negative breast cancer identifies genes for the prognostication and treatment of aggressive breast cancer. Oncogenesis. 2014;3:e100.
  • Mason JM, Wei X, Fletcher GC, et al. Functional characterization of CFI-402257, a potent and selective Mps1/TTK kinase inhibitor, for the treatment of cancer. Proc Natl Acad Sci USA. 2017;114:3127–3132.
  • Wengner AM, Siemeister G, Koppitz M, et al. Novel Mps1 kinase inhibitors with potent antitumor activity. Mol Cancer Ther. 2016;15:583–592.
  • Tannous BA, Kerami M, Van Der Stoop PM, et al. Effects of the selective MPS1 inhibitor MPS1-IN-3 on glioblastoma sensitivity to antimitotic drugs. J Natl Cancer Inst. 2013;105:1322–1331.
  • Naud S, Westwood IM, Faisal A, et al. Structure-based design of orally bioavailable 1H-pyrrolo[3,2-c]pyridine inhibitors of mitotic kinase monopolar spindle 1 (MPS1). J Med Chem. 2013;56:10045–10065.
  • Faisal A, Mak GWY, Gurden MD, et al. Characterisation of CCT271850, a selective, oral and potent MPS1 inhibitor, used to directly measure in vivo MPS1 inhibition vs therapeutic efficacy. Br J Cancer. 2017;116:1166–1176.
  • Tardif KD, Rogers A, Cassiano J, et al. Characterization of the cellular and antitumor effects of MPI-0479605, a small-molecule inhibitor of the mitotic kinase Mps1. Mol Cancer Ther. 2011;10:2267–2275.
  • Colombo R, Caldarelli M, Mennecozzi M, et al. Targeting the mitotic checkpoint for cancer therapy with NMS-P715, an inhibitor of MPS1 kinase. Cancer Res. 2010;70:10255–10264.
  • Hewitt L, Tighe A, Santaguida S, et al. Sustained Mps1 activity is required in mitosis to recruit O-Mad2 to the Mad1-C-Mad2 core complex. J Cell Biol. 2010;190:25–34.
  • Jemaà M, Galluzzi L, Kepp O, et al. Characterization of novel MPS1 inhibitors with preclinical anticancer activity. Cell Death Differ. 2013;20:1532–1545.
  • Kusakabe K-I, Ide N, Daigo Y, et al. Discovery of Imidazo[1,2-b]pyridazine derivatives: selective and orally available Mps1 (TTK) kinase inhibitors exhibiting remarkable antiproliferative activity. J Med Chem. 2015;58:1760–1775.
  • Koch A, Maia A, Janssen A, et al. Molecular basis underlying resistance to Mps1/TTK inhibitors. Oncogene. 2016;35:2518–2528.
  • Thu KL, Silvester J, Elliott MJ, et al. Disruption of the anaphase-promoting complex confers resistance to TTK inhibitors in triple-negative breast cancer. Proc Natl Acad Sci USA. 2018;115:E1570–7.
  • Burkard ME, Weaver BA. Tuning chromosomal instability to optimize tumor fitness. Cancer Discov. 2017;7:134–136.
  • Sansregret L, Patterson JO, Dewhurst S, et al. APC/C dysfunction limits excessive cancer chromosomal instability. Cancer Discov. 2017;7:218–233.
  • Zaman GJR, Jadm DR, Libouban MAA, et al. Inhibitors as a targeted therapy for CTNNB1 (β-catenin) mutant cancers. Mol Cancer Ther. 2017;16:2609–2617.
  • Jemaà M, Vitale I, Kepp O, et al. Selective killing of p53-deficient cancer cells by SP600125. EMBO Mol Med. 2012;4:500–514.
  • Mendes-Pereira AM, Lord CJ, Ashworth A. NLK is a novel therapeutic target for PTEN deficient tumour cells. PLoS One. 2012;7:e47249.
  • Ea N, Aj H. Once and only once: mechanisms of centriole duplication and their deregulation in disease. Nat Rev Mol Cell Biol. 2018. DOI:10.1038/nrm.2017.127
  • Wang G, Jiang Q, Zhang C. The role of mitotic kinases in coupling the centrosome cycle with the assembly of the mitotic spindle. J Cell Sci. 2014;127:4111–4122.
  • Maniswami RR, Prashanth S, Karanth AV, et al. PLK4: a link between centriole biogenesis and cancer. Expert Opin Ther Targets. 2018;22:59–73.
  • Denu RA, Zasadil LM, Kanugh C, et al. Centrosome amplification induces high grade features and is prognostic of worse outcomes in breast cancer. BMC Cancer. 2016;16:47.
  • Pannu V, Mittal K, Cantuaria G, et al. Rampant centrosome amplification underlies more aggressive disease course of triple negative breast cancers. Oncotarget. 2015;6:10487–10497.
  • Chan JY. A clinical overview of centrosome amplification in human cancers. Int J Biol Sci. 2011;7:1122–1144.
  • Marteil G, Guerrero A, Vieira AF, et al. Over-elongation of centrioles in cancer promotes centriole amplification and chromosome missegregation. Nat Commun. 2018;9:1258.
  • Vitre B, Aj H, Kulukian A, et al. Chronic centrosome amplification without tumorigenesis. Proc Natl Acad Sci USA. 2015;112:E6321–30.
  • Levine MS, Bakker B, B B, et al. Centrosome amplification is sufficient to promote spontaneous tumorigenesis in mammals. Dev Cell. 2017;40:313–322.e5.
  • Mason JM, Lin D-C-C, Wei X, et al. Functional characterization of CFI-400945, a Polo-like kinase 4 inhibitor, as a potential anticancer agent. Cancer Cell. 2014;26:163–176.
  • Bedard PL, Cescon DW, Fletcher G, et al. Abstract CT066: first-in-human phase I trial of the oral PLK4 inhibitor CFI-400945 in patients with advanced solid tumors. Cancer Res. 2016;76:CT066–CT066.
  • Cunha-Ferreira I, Bento I, Pimenta-Marques A, et al. Regulation of autophosphorylation controls PLK4 self-destruction and centriole number. Curr Biol. 2013;23:2245–2254.
  • Aj H, Fachinetti D, Zhu Q, et al. The autoregulated instability of Polo-like kinase 4 limits centrosome duplication to once per cell cycle. Genes Dev. 2012;26:2684–2689.
  • Kawakami M, Mustachio LM, Zheng L, et al. Polo-like kinase 4 inhibition produces polyploidy and apoptotic death of lung cancers. Proc Natl Acad Sci USA. 2018;115:1913–1918.
  • Lohse I, Mason J, Cao PM, et al. Activity of the novel polo-like kinase 4 inhibitor CFI-400945 in pancreatic cancer patient-derived xenografts. Oncotarget. 2017;8:3064–3071.
  • Meitinger F, Anzola JV, Kaulich M, et al. 53BP1 and USP28 mediate p53 activation and G1 arrest after centrosome loss or extended mitotic duration. J Cell Biol. 2016;214:155–166.
  • Fong CS, Mazo G, Das T, et al. 53BP1 and USP28 mediate p53-dependent cell cycle arrest in response to centrosome loss and prolonged mitosis. Elife 2016;5. DOI:10.7554/eLife.16270
  • Bg L, Daggubati V, Uetake Y, et al. A USP28-53BP1-p53-p21 signaling axis arrests growth after centrosome loss or prolonged mitosis. J Cell Biol. 2016;214:143–153.
  • Doench JG. Am I ready for CRISPR? A user’s guide to genetic screens. Nat Rev Genet. 2018;19:67–80.
  • Cescon D, Siu LL. Cancer clinical trials: the rear-view mirror and the crystal ball. Cell. 2017;168:575–578.
  • El-Hachem N, Ba-Alawi W, Smith I, et al. Integrative cancer pharmacogenomics to establish drug mechanism of action: drug repurposing. Pharmacogenomics. 2017;18:1469–1472.
  • Madani Tonekaboni SA, Soltan Ghoraie L, Manem VSK, et al. Predictive approaches for drug combination discovery in cancer. Brief Bioinform. 2016. DOI:10.1093/bib/bbw104

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.