Skip to main content

Molecular Regulation of Cell Cycle and Cell Cycle-Targeted Therapies in Head and Neck Squamous Cell Carcinoma (HNSCC)

  • Chapter
  • First Online:
Molecular Determinants of Head and Neck Cancer

Part of the book series: Current Cancer Research ((CUCR))

  • 623 Accesses

Abstract

Head and neck squamous cell carcinomas (HNSCCs) are clinically challenging. The molecular mechanisms and genetic changes that drive HNSCCs have been studied with the aim of developing better therapeutic strategies involving novel molecular targets. Genomic studies have identified mutations in genes that mediate cell cycle, and key differences in cell cycle regulation differentiate both human papillomavirus (HPV)-associated and HPV-negative HNSCC cases from normal tissue. Some of these differences may nominate specific therapeutic targets and impact treatment response in HNSCC. For example, one of the most frequent cell cycle alterations in HPV (−) HNSCC is the disruption of the p53 (TP53) pathway (over 80% of tumors, based on data in TCGA and other studies), which is involved in cell cycle control, DNA damage signaling, and overall maintenance of genome stability. Other frequent alterations disrupt the cell cycle regulator CDKN2A (28% alteration frequency), which encodes p16, an inhibitor of cell cycle kinases CDK4 and CDK6, and alters expression of CCND1, resulting in inactivation of the tumor suppressor Rb. Other mutations found less commonly in patients target elements of the cell cycle checkpoint and DNA damage response machinery. Such observations and a growing recognition of the importance of cell cycle regulatory defects in HNSCC response to typically DNA-damaging chemotherapies and radiation therapy have rationalized the development of novel cell cycle-targeted therapies for HNSCCs. We here provide a general overview of the process of cell cycle control, cell cycle checkpoints, and how these are dysregulated in HNSCC and other cancers and discuss current cell cycle-targeted therapies in development and in clinical trials for HNSCC. The ultimate goal of these efforts is to develop new, potent therapeutic agents and to identify patient subpopulations that will be more responsive to cell cycle-targeted therapies.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Dok R, Nuyts S. HPV positive head and neck cancers: molecular pathogenesis and evolving treatment strategies. Cancers (Basel). 2016;8:pii: E41. https://doi.org/10.3390/cancers8040041.

    Article  CAS  Google Scholar 

  2. Jenkins G, O’Byrne KJ, Panizza B, Richard DJ. Genome stability pathways in head and neck cancers. Int J Genomics. 2013;2013:464720. https://doi.org/10.1155/2013/464720.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Gillison ML, Lowy DR. A causal role for human papillomavirus in head and neck cancer. Lancet. 2004;363:1488–9. https://doi.org/10.1016/S0140-6736(04)16194-1.

    Article  PubMed  CAS  Google Scholar 

  4. Ang KK, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363:24–35. https://doi.org/10.1056/NEJMoa0912217.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Feldman R, et al. Molecular profiling of head and neck squamous cell carcinoma. Head Neck. 2016;38(Suppl 1):E1625–38. https://doi.org/10.1002/hed.24290.

    Article  PubMed  Google Scholar 

  6. Bingham HG, Copeland EM, Hackett R, Caffee HH. Breast cancer in a patient with silicone breast implants after 13 years. Ann Plast Surg. 1988;20:236–7.

    Article  CAS  PubMed  Google Scholar 

  7. Riaz N, Morris LG, Lee W, Chan TA. Unraveling the molecular genetics of head and neck cancer through genome-wide approaches. Genes Dis. 2014;1:75–86. https://doi.org/10.1016/j.gendis.2014.07.002.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Deshpande AM, Wong DT. Molecular mechanisms of head and neck cancer. Expert Rev Anticancer Ther. 2008;8:799–809. https://doi.org/10.1586/14737140.8.5.799.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Rothenberg SM, Ellisen LW. The molecular pathogenesis of head and neck squamous cell carcinoma. J Clin Invest. 2012;122:1951–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Worsham MJ, Ali H, Dragovic J, Schweitzer VP. Molecular characterization of head and neck cancer: how close to personalized targeted therapy? Mol Diagn Ther. 2012;16:209–22. https://doi.org/10.2165/11635330-000000000-00000.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Morgan DO. Cyclin-dependent kinases: engines, clocks, and microprocessors. Annu Rev Cell Dev Biol. 1997;13:261–91. https://doi.org/10.1146/annurev.cellbio.13.1.261.

    Article  PubMed  CAS  Google Scholar 

  12. Hartwell LH, Weinert TA. Checkpoints: controls that ensure the order of cell cycle events. Science. 1989;246:629–34.

    Article  PubMed  CAS  Google Scholar 

  13. Shapiro GI, Harper JW. Anticancer drug targets: cell cycle and checkpoint control. J Clin Invest. 1999;104:1645–53. https://doi.org/10.1172/JCI9054.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol. 2014;740:364–78. https://doi.org/10.1016/j.ejphar.2014.07.025.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Florea AM, Busselberg D. Cisplatin as an anti-tumor drug: cellular mechanisms of activity, drug resistance and induced side effects. Cancers (Basel). 2011;3:1351–71. https://doi.org/10.3390/cancers3011351.

    Article  CAS  Google Scholar 

  16. Rozencweig M, von Hoff DD, Slavik M, Muggia FM. Cis-diamminedichloroplatinum (II). A new anticancer drug. Ann Intern Med. 1977;86:803–12.

    Article  PubMed  CAS  Google Scholar 

  17. Weaver B, How A. Taxol/paclitaxel kills cancer cells. Mol Biol Cell. 2014;25:2677–81. https://doi.org/10.1091/mbc.E14-04-0916.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Herbst RS, Khuri FR. Mode of action of docetaxel – a basis for combination with novel anticancer agents. Cancer Treat Rev. 2003;29:407–15.

    Article  PubMed  CAS  Google Scholar 

  19. Bissery MC, Nohynek G, Sanderink GJ, Lavelle F. Docetaxel (Taxotere): a review of preclinical and clinical experience. Part I: preclinical experience. Anticancer Drugs. 1995;6:339–55., 363–338.

    Article  PubMed  CAS  Google Scholar 

  20. Altmann KH. Microtubule-stabilizing agents: a growing class of important anticancer drugs. Curr Opin Chem Biol. 2001;5:424–31.

    Article  CAS  PubMed  Google Scholar 

  21. Gabrielli B, Brooks K, Pavey S. Defective cell cycle checkpoints as targets for anti-cancer therapies. Front Pharmacol. 2012;3:9. https://doi.org/10.3389/fphar.2012.00009.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Kastan MB, Bartek J. Cell-cycle checkpoints and cancer. Nature. 2004;432:316–23. https://doi.org/10.1038/nature03097.

    Article  PubMed  CAS  Google Scholar 

  23. Visconti R, Della Monica R, Grieco D. Cell cycle checkpoint in cancer: a therapeutically targetable double-edged sword. J Exp Clin Cancer Res. 2016;35:153. https://doi.org/10.1186/s13046-016-0433-9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Loibl S, Turner NC, Ro J, Cristofanilli M, Iwata H, Im SA, Masuda N, Loi S, André F, Harbeck N, Verma S, Folkerd E, Puyana Theall K, Hoffman J, Zhang K, Bartlett CH, Dowsett M. Palbociclib Combined with Fulvestrant in Premenopausal Women with Advanced Breast Cancer and Prior Progression on Endocrine Therapy: PALOMA-3 Results. Oncologist. 2017 Sep;22(9):1028-1038. doi: 10.1634/theoncologist.2017-0072. Epub 2017 Jun 26.

    Google Scholar 

  25. Verma S, Bartlett CH, Schnell P, DeMichele AM, Loi S, Ro J, Colleoni M, Iwata H, Harbeck N, Cristofanilli M, Zhang K, Thiele A, Turner NC, Rugo HS. Palbociclib in Combination With Fulvestrant in Women With Hormone Receptor-Positive/HER2-Negative Advanced Metastatic Breast Cancer: Detailed Safety Analysis From a Multicenter, Randomized, Placebo-Controlled, Phase III Study (PALOMA-3). Oncologist. 2016 Oct;21(10):1165-1175. Epub 2016 Jul 1.

    Google Scholar 

  26. Hortobagyi G, Stemmer S, Burris H, et al. First-line ribociclib plus letrozole for postmenopausal women with HR+, HER2-, advanced breast cancer: first results from the phase III MONALEESA-2 study. Presented at the European Society for Medical Oncology (ESMO) Congress, Copenhagen, Denmark (October 8, 2016).

    Google Scholar 

  27. Shah A, Bloomquist E, Tang S, Fu W, Bi Y, Liu Q, Yu J, Zhao P, Palmby TR, Goldberg KB, CJG C, Patel P, Alebachew E, Tilley A, Pierce WF, Ibrahim A, Blumenthal GM, Sridhara R, Beaver JA, Pazdur R. FDA Approval: ribociclib for the treatment of postmenopausal women with hormone receptor-positive, HER2-Negative Advanced or Metastatic Breast Cancer. Clin Cancer Res. 2018. https://doi.org/10.1158/1078-0432.CCR-17-2369. clincanres.2369.2017. [Epub ahead of print]. PMID:29437768.

  28. Dempsey JA, et al. AACR Annual Meeting; April 6–10, 2013; Washington, DC. Abstract LB122.

    Google Scholar 

  29. Gelbert LM, et al. AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; November 12–16, 2011; San Francisco, CA. Abstract B233.

    Google Scholar 

  30. Bell T, et al. Impact of palbociclib plus letrozole on pain severity and pain interference with daily activities in patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer as first-line treatment. Curr Med Res Opin. 2016;32:959–65. https://doi.org/10.1185/03007995.2016.1157060.

    Article  CAS  PubMed  Google Scholar 

  31. Finn RS, 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. https://doi.org/10.1016/S1470-2045(14)71159-3.

    Article  CAS  PubMed  Google Scholar 

  32. Finn RS, et al. Palbociclib and letrozole in advanced breast cancer. N Engl J Med. 2016;375:1925–36. https://doi.org/10.1056/NEJMoa1607303.

    Article  CAS  PubMed  Google Scholar 

  33. Cristofanilli M, et al. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol. 2016;17:425–39. https://doi.org/10.1016/S1470-2045(15)00613-0.

    Article  CAS  PubMed  Google Scholar 

  34. Harbeck N, Iyer S, Turner N, Cristofanilli M, Ro J, André F, Loi S, Verma S, Iwata H, Bhattacharyya H, Puyana Theall K, Bartlett CH, Loibl S. Quality of life with palbociclib plus fulvestrant in previously treated hormone receptor-positive, HER2-negative metastatic breast cancer: patient-reported outcomes from the PALOMA-3 trial. Ann Oncol. 2016;27(6):1047–54. https://doi.org/10.1093/annonc/mdw139. Epub 2016 Mar 30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Turner NC, et al. Palbociclib in hormone-receptor-positive advanced breast cancer. N Engl J Med. 2015;373:209–19. https://doi.org/10.1056/NEJMoa1505270.

    Article  CAS  PubMed  Google Scholar 

  36. Verma S, Bartlett CH, Schnell P, DeMichele AM, Loi S, Ro J, Colleoni M, Iwata H, Harbeck N, Cristofanilli M, Zhang K, Thiele A, Turner NC, Rugo HS. Palbociclib in combination with fulvestrant in women with hormone receptor-positive/HER2-Negative Advanced Metastatic Breast Cancer: detailed safety analysis from a multicenter, randomized, placebo-controlled, phase III study (PALOMA-3). Oncologist. 2016;21(10):1165–75. Epub 2016 Jul 1

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Hortobagyi GN, et al. Ribociclib as first-line therapy for HR-positive, advanced breast cancer. N Engl J Med. 2016;375:1738–48. https://doi.org/10.1056/NEJMoa1609709.

    Article  CAS  PubMed  Google Scholar 

  38. de Carcer G, Manning G, Malumbres M. From Plk1 to Plk5: functional evolution of polo-like kinases. Cell Cycle. 2011;10:2255–62. https://doi.org/10.4161/cc.10.14.16494.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Glover DM, Hagan IM, Tavares AA. Polo-like kinases: a team that plays throughout mitosis. Genes Dev. 1998;12:3777–87.

    Article  CAS  PubMed  Google Scholar 

  40. Kops GJ, Weaver BA, Cleveland DW. On the road to cancer: aneuploidy and the mitotic checkpoint. Nat Rev Cancer. 2005;5:773–85. https://doi.org/10.1038/nrc1714.

    Article  CAS  PubMed  Google Scholar 

  41. Macurek L, et al. Polo-like kinase-1 is activated by aurora A to promote checkpoint recovery. Nature. 2008;455:119–23. https://doi.org/10.1038/nature07185.

    Article  CAS  PubMed  Google Scholar 

  42. Asteriti IA, De Mattia F, Guarguaglini G. Cross-talk between AURKA and Plk1 in mitotic entry and spindle assembly. Front Oncol. 2015;5:283. https://doi.org/10.3389/fonc.2015.00283.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Nikonova AS, Astsaturov I, Serebriiskii IG, Dunbrack RL Jr, Golemis EA. Aurora A kinase (AURKA) in normal and pathological cell division. Cell Mol Life Sci. 2013;70:661–87. https://doi.org/10.1007/s00018-012-1073-7.

    Article  CAS  PubMed  Google Scholar 

  44. Wirtz-Peitz F, Nishimura T, Knoblich JA. Linking cell cycle to asymmetric division: aurora-A phosphorylates the Par complex to regulate Numb localization. Cell. 2008;135:161–73. https://doi.org/10.1016/j.cell.2008.07.049.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Chan CS, Botstein D. Isolation and characterization of chromosome-gain and increase-in-ploidy mutants in yeast. Genetics. 1993;135:677–91.

    PubMed  PubMed Central  CAS  Google Scholar 

  46. Rhind N, Russell P. Signaling pathways that regulate cell division. Cold Spring Harb Perspect Biol. 2012;4:a005942. https://doi.org/10.1101/cshperspect.a005942.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Wells NJ, et al. The C-terminal domain of the Cdc2 inhibitory kinase Myt1 interacts with Cdc2 complexes and is required for inhibition of G(2)/M progression. J Cell Sci. 1999;112(Pt 19):3361–71.

    CAS  PubMed  Google Scholar 

  48. Booher RN, Holman PS, Fattaey A. Human Myt1 is a cell cycle-regulated kinase that inhibits Cdc2 but not Cdk2 activity. J Biol Chem. 1997;272:22300–6.

    Article  CAS  PubMed  Google Scholar 

  49. Liu F, Rothblum-Oviatt C, Ryan CE, Piwnica-Worms H. Overproduction of human Myt1 kinase induces a G2 cell cycle delay by interfering with the intracellular trafficking of Cdc2-cyclin B1 complexes. Mol Cell Biol. 1999;19:5113–23.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Den Haese GJ, Walworth N, Carr AM, Gould KL. The Wee1 protein kinase regulates T14 phosphorylation of fission yeast Cdc2. Mol Biol Cell. 1995;6:371–85.

    Article  Google Scholar 

  51. Coleman TR, Dunphy WG. Cdc2 regulatory factors. Curr Opin Cell Biol. 1994;6:877–82.

    Article  CAS  PubMed  Google Scholar 

  52. Rowley R, Hudson J, Young PG. The wee1 protein kinase is required for radiation-induced mitotic delay. Nature. 1992;356:353–5. https://doi.org/10.1038/356353a0.

    Article  CAS  PubMed  Google Scholar 

  53. Manic G, Obrist F, Sistigu A, Vitale I. Trial watch: targeting ATM-CHK2 and ATR-CHK1 pathways for anticancer therapy. Mol Cell Oncol. 2015;2:e1012976. https://doi.org/10.1080/23723556.2015.1012976.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Smith J, Tho LM, Xu N, Gillespie DA. The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Adv Cancer Res. 2010;108:73–112. https://doi.org/10.1016/B978-0-12-380888-2.00003-0.

    Article  CAS  PubMed  Google Scholar 

  55. Keith CT, Schreiber SL. PIK-related kinases: DNA repair, recombination, and cell cycle checkpoints. Science. 1995;270:50–1.

    Article  CAS  PubMed  Google Scholar 

  56. Lecona E, Fernandez-Capetillo O. Replication stress and cancer: it takes two to tango. Exp Cell Res. 2014;329:26–34. https://doi.org/10.1016/j.yexcr.2014.09.019.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Mazouzi A, Velimezi G, Loizou JI. DNA replication stress: causes, resolution and disease. Exp Cell Res. 2014;329:85–93. https://doi.org/10.1016/j.yexcr.2014.09.030.

    Article  CAS  PubMed  Google Scholar 

  58. Wallace MD, Southard TL, Schimenti KJ, Schimenti JC. Role of DNA damage response pathways in preventing carcinogenesis caused by intrinsic replication stress. Oncogene. 2014;33:3688–95. https://doi.org/10.1038/onc.2013.339.

    Article  CAS  PubMed  Google Scholar 

  59. Reinhardt HC, Yaffe MB. Kinases that control the cell cycle in response to DNA damage: Chk1, Chk2, and MK2. Curr Opin Cell Biol. 2009;21:245–55. https://doi.org/10.1016/j.ceb.2009.01.018.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. van den Heuvel S, Harlow E. Distinct roles for cyclin-dependent kinases in cell cycle control. Science. 1993;262:2050–4.

    Article  PubMed  Google Scholar 

  61. Malumbres M, Barbacid M. Mammalian cyclin-dependent kinases. Trends Biochem Sci. 2005;30:630–41. https://doi.org/10.1016/j.tibs.2005.09.005.

    Article  CAS  PubMed  Google Scholar 

  62. Gopinathan L, Ratnacaram CK, Kaldis P. Established and novel Cdk/cyclin complexes regulating the cell cycle and development. Results Probl Cell Differ. 2011;53:365–89. https://doi.org/10.1007/978-3-642-19065-0_16.

    Article  CAS  PubMed  Google Scholar 

  63. Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development. 2013;140:3079–93. https://doi.org/10.1242/dev.091744.

    Article  CAS  PubMed  Google Scholar 

  64. Cancer Genome Atlas Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517:576–82. https://doi.org/10.1038/nature14129.

    Article  CAS  Google Scholar 

  65. Seiwert TY, et al. Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas. Clin Cancer Res. 2015;21:632–41. https://doi.org/10.1158/1078-0432.CCR-13-3310.

    Article  CAS  PubMed  Google Scholar 

  66. Lin DC, et al. The genomic landscape of nasopharyngeal carcinoma. Nat Genet. 2014;46:866–71. https://doi.org/10.1038/ng.3006.

    Article  CAS  PubMed  Google Scholar 

  67. Pickering CR, et al. Squamous cell carcinoma of the oral tongue in young non-smokers is genomically similar to tumors in older smokers. Clin Cancer Res. 2014;20:3842–8. https://doi.org/10.1158/1078-0432.CCR-14-0565.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Pickering CR, et al. Integrative genomic characterization of oral squamous cell carcinoma identifies frequent somatic drivers. Cancer Discov. 2013;3:770–81. https://doi.org/10.1158/2159-8290.CD-12-0537.

    Article  PubMed  CAS  Google Scholar 

  69. Stransky N, et al. The mutational landscape of head and neck squamous cell carcinoma. Science. 2011;333:1157–60. https://doi.org/10.1126/science.1208130.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Agrawal N, et al. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. 2011;333:1154–7. https://doi.org/10.1126/science.1206923.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Russell P, Nurse P. cdc25+ functions as an inducer in the mitotic control of fission yeast. Cell. 1986;45:145–53.

    Article  CAS  PubMed  Google Scholar 

  72. Pines J. Four-dimensional control of the cell cycle. Nat Cell Biol. 1999;1:E73–9. https://doi.org/10.1038/11041.

    Article  CAS  PubMed  Google Scholar 

  73. Donzelli M, Draetta GF. Regulating mammalian checkpoints through Cdc25 inactivation. EMBO Rep. 2003;4:671–7. https://doi.org/10.1038/sj.embor.embor887.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Serrano M, Hannon GJ, Beach D. A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4. Nature. 1993;366:704–7. https://doi.org/10.1038/366704a0.

    Article  CAS  PubMed  Google Scholar 

  75. Sherr CJ, Roberts JM. Inhibitors of mammalian G1 cyclin-dependent kinases. Genes Dev. 1995;9:1149–63.

    Article  CAS  PubMed  Google Scholar 

  76. O’Connor MJ. Targeting the DNA damage response in cancer. Mol Cell. 2015;60:547–60. https://doi.org/10.1016/j.molcel.2015.10.040.

    Article  CAS  PubMed  Google Scholar 

  77. Geenen JJJ, Schellens JHM. Molecular pathways: targeting the protein kinase Wee1 in cancer. Clin Cancer Res. 2017;23:4540–4. https://doi.org/10.1158/1078-0432.CCR-17-0520.

    Article  CAS  PubMed  Google Scholar 

  78. Matheson CJ, Backos DS, Reigan P. Targeting WEE1 kinase in cancer. Trends Pharmacol Sci. 2016;37:872–81. https://doi.org/10.1016/j.tips.2016.06.006.

    Article  CAS  PubMed  Google Scholar 

  79. Otto T, Sicinski P. Cell cycle proteins as promising targets in cancer therapy. Nat Rev Cancer. 2017;17:93–115. https://doi.org/10.1038/nrc.2016.138.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Besson A, Dowdy SF, Roberts JM. CDK inhibitors: cell cycle regulators and beyond. Dev Cell. 2008;14:159–69. https://doi.org/10.1016/j.devcel.2008.01.013.

    Article  CAS  PubMed  Google Scholar 

  81. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 2009;9:153–66. https://doi.org/10.1038/nrc2602.

    Article  CAS  PubMed  Google Scholar 

  82. Sherr CJ, Beach D, Shapiro GI. Targeting CDK4 and CDK6: from discovery to therapy. Cancer Discov. 2016;6:353–67. https://doi.org/10.1158/2159-8290.CD-15-0894.

    Article  CAS  PubMed  Google Scholar 

  83. Sherr CJ, Bartek AJ. Cell cycle–targeted cancer therapies. Annu Rev Cancer Biol. 2017;1:41–57. https://doi.org/10.1146/annurev-cancerbio-040716-075628.

    Article  Google Scholar 

  84. Ewen ME, et al. Functional interactions of the retinoblastoma protein with mammalian D-type cyclins. Cell. 1993;73:487–97.

    Article  CAS  PubMed  Google Scholar 

  85. DeCaprio JA, et al. The product of the retinoblastoma susceptibility gene has properties of a cell cycle regulatory element. Cell. 1989;58:1085–95.

    Article  CAS  PubMed  Google Scholar 

  86. Weinberg RA. The retinoblastoma protein and cell cycle control. Cell. 1995;81:323–30.

    Article  CAS  PubMed  Google Scholar 

  87. Sherr CJ. The ins and outs of RB: coupling gene expression to the cell cycle clock. Trends Cell Biol. 1994;4:15–8.

    Article  CAS  PubMed  Google Scholar 

  88. Cobrinik D, Dowdy SF, Hinds PW, Mittnacht S, Weinberg RA. The retinoblastoma protein and the regulation of cell cycling. Trends Biochem Sci. 1992;17:312–5.

    Article  CAS  PubMed  Google Scholar 

  89. Chellappan SP, Hiebert S, Mudryj M, Horowitz JM, Nevins JR. The E2F transcription factor is a cellular target for the RB protein. Cell. 1991;65:1053–61.

    Article  CAS  PubMed  Google Scholar 

  90. Dyson N. The regulation of E2F by pRB-family proteins. Genes Dev. 1998;12:2245–62.

    Article  PubMed  CAS  Google Scholar 

  91. Lukas J, Bartkova J, Rohde M, Strauss M, Bartek J. Cyclin D1 is dispensable for G1 control in retinoblastoma gene-deficient cells independently of cdk4 activity. Mol Cell Biol. 1995;15:2600–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Rubin SM. Deciphering the retinoblastoma protein phosphorylation code. Trends Biochem Sci. 2013;38:12–9. https://doi.org/10.1016/j.tibs.2012.10.007.

    Article  CAS  PubMed  Google Scholar 

  93. Cappell SD, Chung M, Jaimovich A, Spencer SL, Meyer T. Irreversible APC(Cdh1) inactivation underlies the point of no return for cell-cycle entry. Cell. 2016;166:167–80. https://doi.org/10.1016/j.cell.2016.05.077.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Eguren M, Manchado E, Malumbres M. Non-mitotic functions of the anaphase-promoting complex. Semin Cell Dev Biol. 2011;22:572–8. https://doi.org/10.1016/j.semcdb.2011.03.010.

    Article  CAS  PubMed  Google Scholar 

  95. Rape M, Kirschner MW. Autonomous regulation of the anaphase-promoting complex couples mitosis to S-phase entry. Nature. 2004;432:588–95. https://doi.org/10.1038/nature03023.

    Article  CAS  PubMed  Google Scholar 

  96. Vodermaier HC. APC/C and SCF: controlling each other and the cell cycle. Curr Biol. 2004;14:R787–96. https://doi.org/10.1016/j.cub.2004.09.020.

    Article  CAS  PubMed  Google Scholar 

  97. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 1999;13:1501–12.

    Article  CAS  PubMed  Google Scholar 

  98. Kitagawa M, et al. Phosphorylation of E2F-1 by cyclin A-cdk2. Oncogene. 1995;10:229–36.

    CAS  PubMed  Google Scholar 

  99. Perry JA, Kornbluth S. Cdc25 and Wee1: analogous opposites? Cell Div. 2007;2:12. https://doi.org/10.1186/1747-1028-2-12.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Boutros R, Lobjois V, Ducommun B. CDC25 phosphatases in cancer cells: key players? Good targets? Nat Rev Cancer. 2007;7:495–507. https://doi.org/10.1038/nrc2169.

    Article  CAS  PubMed  Google Scholar 

  101. Heald R, McLoughlin M, McKeon F. Human wee1 maintains mitotic timing by protecting the nucleus from cytoplasmically activated Cdc2 kinase. Cell. 1993;74:463–74.

    Article  CAS  PubMed  Google Scholar 

  102. Kristjansdottir K, Rudolph J. Cdc25 phosphatases and cancer. Chem Biol. 2004;11:1043–51. https://doi.org/10.1016/j.chembiol.2004.07.007.

    Article  CAS  PubMed  Google Scholar 

  103. Lolli G, Johnson LN. CAK-Cyclin-dependent Activating Kinase: a key kinase in cell cycle control and a target for drugs? Cell Cycle. 2005;4:572–7.

    Article  CAS  PubMed  Google Scholar 

  104. Draetta GF. Cell cycle: will the real Cdk-activating kinase please stand up. Curr Biol. 1997;7:R50–2.

    Article  PubMed  CAS  Google Scholar 

  105. Shiekhattar R, et al. Cdk-activating kinase complex is a component of human transcription factor TFIIH. Nature. 1995;374:283–7. https://doi.org/10.1038/374283a0.

    Article  CAS  PubMed  Google Scholar 

  106. Kaldis P. The cdk-activating kinase (CAK): from yeast to mammals. Cell Mol Life Sci. 1999;55:284–96. https://doi.org/10.1007/s000180050290.

    Article  CAS  PubMed  Google Scholar 

  107. Palazzo RE, Vogel JM, Schnackenberg BJ, Hull DR, Wu X. Centrosome maturation. Curr Top Dev Biol. 2000;49:449–70.

    Article  CAS  PubMed  Google Scholar 

  108. Lera RF, et al. Decoding polo-like kinase 1 signaling along the kinetochore-centromere axis. Nat Chem Biol. 2016;12:411–8. https://doi.org/10.1038/nchembio.2060.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Dutertre S, et al. Phosphorylation of CDC25B by aurora-A at the centrosome contributes to the G2-M transition. J Cell Sci. 2004;117:2523–31. https://doi.org/10.1242/jcs.01108.

    Article  CAS  PubMed  Google Scholar 

  110. Lindqvist A, Rodriguez-Bravo V, Medema RH. The decision to enter mitosis: feedback and redundancy in the mitotic entry network. J Cell Biol. 2009;185:193–202. https://doi.org/10.1083/jcb.200812045.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  111. Shaltiel IA, Krenning L, Bruinsma W, Medema RH. The same, only different - DNA damage checkpoints and their reversal throughout the cell cycle. J Cell Sci. 2015;128:607–20. https://doi.org/10.1242/jcs.163766.

    Article  PubMed  CAS  Google Scholar 

  112. Nasmyth K. Disseminating the genome: joining, resolving, and separating sister chromatids during mitosis and meiosis. Annu Rev Genet. 2001;35:673–745. https://doi.org/10.1146/annurev.genet.35.102401.091334.

    Article  PubMed  CAS  Google Scholar 

  113. Peters JM. The anaphase promoting complex/cyclosome: a machine designed to destroy. Nat Rev Mol Cell Biol. 2006;7:644–56. https://doi.org/10.1038/nrm1988.

    Article  PubMed  CAS  Google Scholar 

  114. Engelbert D, Schnerch D, Baumgarten A, Wasch R. The ubiquitin ligase APC(Cdh1) is required to maintain genome integrity in primary human cells. Oncogene. 2008;27:907–17. https://doi.org/10.1038/sj.onc.1210703.

    Article  PubMed  CAS  Google Scholar 

  115. Garcia-Higuera I, et al. Genomic stability and tumour suppression by the APC/C cofactor Cdh1. Nat Cell Biol. 2008;10:802–11. https://doi.org/10.1038/ncb1742.

    Article  PubMed  CAS  Google Scholar 

  116. Li M, et al. The adaptor protein of the anaphase promoting complex Cdh1 is essential in maintaining replicative lifespan and in learning and memory. Nat Cell Biol. 2008;10:1083–9. https://doi.org/10.1038/ncb1768.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Ruas M, Peters G. The p16INK4a/CDKN2A tumor suppressor and its relatives. Biochim Biophys Acta. 1998;1378:F115–77.

    PubMed  CAS  Google Scholar 

  118. Sherr CJ. Tumor surveillance via the ARF-p53 pathway. Genes Dev. 1998;12:2984–91.

    Article  PubMed  CAS  Google Scholar 

  119. Sharpless NE, DePinho RA. The INK4A/ARF locus and its two gene products. Curr Opin Genet Dev. 1999;9:22–30.

    Article  PubMed  CAS  Google Scholar 

  120. Hirama T, Koeffler HP. Role of the cyclin-dependent kinase inhibitors in the development of cancer. Blood. 1995;86:841–54.

    PubMed  CAS  Google Scholar 

  121. Drexler HG. Review of alterations of the cyclin-dependent kinase inhibitor INK4 family genes p15, p16, p18 and p19 in human leukemia-lymphoma cells. Leukemia. 1998;12:845–59.

    Article  PubMed  CAS  Google Scholar 

  122. Xiong Y, et al. p21 is a universal inhibitor of cyclin kinases. Nature. 1993;366:701–4. https://doi.org/10.1038/366701a0.

    Article  PubMed  CAS  Google Scholar 

  123. el-Deiry WS, et al. WAF1, a potential mediator of p53 tumor suppression. Cell. 1993;75:817–25.

    Article  PubMed  CAS  Google Scholar 

  124. El-Deiry WS. p21(WAF1) mediates cell-cycle inhibition, relevant to cancer suppression and therapy. Cancer Res. 2016;76:5189–91. https://doi.org/10.1158/0008-5472.CAN-16-2055.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  125. Warfel NA, El-Deiry WS. p21WAF1 and tumourigenesis: 20 years after. Curr Opin Oncol. 2013;25:52–8. https://doi.org/10.1097/CCO.0b013e32835b639e.

    Article  PubMed  CAS  Google Scholar 

  126. Esashi F, et al. CDK-dependent phosphorylation of BRCA2 as a regulatory mechanism for recombinational repair. Nature. 2005;434:598–604. https://doi.org/10.1038/nature03404.

    Article  PubMed  CAS  Google Scholar 

  127. Wohlbold L, Fisher RP. Behind the wheel and under the hood: functions of cyclin-dependent kinases in response to DNA damage. DNA Repair (Amst). 2009;8:1018–24. https://doi.org/10.1016/j.dnarep.2009.04.009.

    Article  CAS  Google Scholar 

  128. Johnson N, et al. Compromised CDK1 activity sensitizes BRCA-proficient cancers to PARP inhibition. Nat Med. 2011;17:875–82. https://doi.org/10.1038/nm.2377.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  129. Foster SS, De S, Johnson LK, Petrini JH, Stracker TH. Cell cycle- and DNA repair pathway-specific effects of apoptosis on tumor suppression. Proc Natl Acad Sci U S A. 2012;109:9953–8. https://doi.org/10.1073/pnas.1120476109.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Kaufmann WK, Paules RS. DNA damage and cell cycle checkpoints. FASEB J. 1996;10:238–47.

    Article  PubMed  CAS  Google Scholar 

  131. Langerak P, Russell P. Regulatory networks integrating cell cycle control with DNA damage checkpoints and double-strand break repair. Philos Trans R Soc Lond B Biol Sci. 2011;366:3562–71. https://doi.org/10.1098/rstb.2011.0070.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  132. Buisson R, Boisvert JL, Benes CH, Zou L. Distinct but concerted roles of ATR, DNA-PK, and Chk1 in countering replication stress during S phase. Mol Cell. 2015;59:1011–24. https://doi.org/10.1016/j.molcel.2015.07.029.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Min W, et al. Poly(ADP-ribose) binding to Chk1 at stalled replication forks is required for S-phase checkpoint activation. Nat Commun. 2013;4:2993. https://doi.org/10.1038/ncomms3993.

    Article  PubMed  CAS  Google Scholar 

  134. Vitale I, Galluzzi L, Castedo M, Kroemer G. Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat Rev Mol Cell Biol. 2011;12:385–92. https://doi.org/10.1038/nrm3115.

    Article  PubMed  CAS  Google Scholar 

  135. Castedo M, et al. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004;23:2825–37. https://doi.org/10.1038/sj.onc.1207528.

    Article  PubMed  CAS  Google Scholar 

  136. Roninson IB, Broude EV, Chang BD. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist Updat. 2001;4:303–13. https://doi.org/10.1054/drup.2001.0213.

    Article  PubMed  CAS  Google Scholar 

  137. Vakifahmetoglu H, Olsson M, Zhivotovsky B. Death through a tragedy: mitotic catastrophe. Cell Death Differ. 2008;15:1153–62. https://doi.org/10.1038/cdd.2008.47.

    Article  PubMed  CAS  Google Scholar 

  138. Aarts M, Linardopoulos S, Turner NC. Tumour selective targeting of cell cycle kinases for cancer treatment. Curr Opin Pharmacol. 2013;13:529–35. https://doi.org/10.1016/j.coph.2013.03.012.

    Article  PubMed  CAS  Google Scholar 

  139. Dominguez-Brauer C, et al. Targeting mitosis in cancer: emerging strategies. Mol Cell. 2015;60:524–36. https://doi.org/10.1016/j.molcel.2015.11.006.

    Article  PubMed  CAS  Google Scholar 

  140. Keen N, Taylor S. Aurora-kinase inhibitors as anticancer agents. Nat Rev Cancer. 2004;4:927–36. https://doi.org/10.1038/nrc1502.

    Article  PubMed  CAS  Google Scholar 

  141. Strebhardt K, Ullrich A. Targeting polo-like kinase 1 for cancer therapy. Nat Rev Cancer. 2006;6:321–30. https://doi.org/10.1038/nrc1841.

    Article  PubMed  CAS  Google Scholar 

  142. Nigg EA. Mitotic kinases as regulators of cell division and its checkpoints. Nat Rev Mol Cell Biol. 2001;2:21–32. https://doi.org/10.1038/35048096.

    Article  PubMed  CAS  Google Scholar 

  143. Heijink AM, Krajewska M, van Vugt MA. The DNA damage response during mitosis. Mutat Res. 2013;750:45–55. https://doi.org/10.1016/j.mrfmmm.2013.07.003.

    Article  PubMed  CAS  Google Scholar 

  144. Orthwein A, et al. Mitosis inhibits DNA double-strand break repair to guard against telomere fusions. Science. 2014;344:189–93. https://doi.org/10.1126/science.1248024.

    Article  PubMed  CAS  Google Scholar 

  145. Mani RS, et al. Dual modes of interaction between XRCC4 and polynucleotide kinase/phosphatase: implications for nonhomologous end joining. J Biol Chem. 2010;285:37619–29. https://doi.org/10.1074/jbc.M109.058719.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Lees-Miller SP. DNA double strand break repair in mitosis is suppressed by phosphorylation of XRCC4. PLoS Genet. 2014;10:e1004598. https://doi.org/10.1371/journal.pgen.1004598.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  147. Hustedt N, Durocher D. The control of DNA repair by the cell cycle. Nat Cell Biol. 2016;19:1–9. https://doi.org/10.1038/ncb3452.

    Article  PubMed  CAS  Google Scholar 

  148. Rossio V, Galati E, Piatti S. Adapt or die: how eukaryotic cells respond to prolonged activation of the spindle assembly checkpoint. Biochem Soc Trans. 2010;38:1645–9. https://doi.org/10.1042/BST0381645.

    Article  PubMed  CAS  Google Scholar 

  149. Thompson RC, Dripps DJ, Eisenberg SP. Interleukin-1 receptor antagonist (IL-1ra) as a probe and as a treatment for IL-1 mediated disease. Int J Immunopharmacol. 1992;14:475–80.

    Article  PubMed  CAS  Google Scholar 

  150. Lawrence KS, Engebrecht J. The spindle assembly checkpoint: more than just keeping track of the spindle. Trends Cell Mol Biol. 2015;10:141–50.

    PubMed  PubMed Central  Google Scholar 

  151. Musacchio A. The molecular biology of spindle assembly checkpoint signaling dynamics. Curr Biol. 2015;25:R1002–18. https://doi.org/10.1016/j.cub.2015.08.051.

    Article  PubMed  CAS  Google Scholar 

  152. O'Sullivan B, et al. Outcomes of HPV-related oropharyngeal cancer patients treated by radiotherapy alone using altered fractionation. Radiother Oncol. 2012;103:49–56. https://doi.org/10.1016/j.radonc.2012.02.009.

    Article  PubMed  Google Scholar 

  153. Richards L. Human papillomavirus-a powerful predictor of survival in patients with oropharyngeal cancer. Nat Rev Clin Oncol. 2010;7:481. https://doi.org/10.1038/nrclinonc.2010.123.

    Article  PubMed  Google Scholar 

  154. Zhou G, Liu Z, Myers JN. TP53 mutations in head and neck squamous cell carcinoma and their impact on disease progression and treatment response. J Cell Biochem. 2016;117:2682–92. https://doi.org/10.1002/jcb.25592.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Suh Y, Amelio I, Guerrero Urbano T, Tavassoli M. Clinical update on cancer: molecular oncology of head and neck cancer. Cell Death Dis. 2014;5:e1018. https://doi.org/10.1038/cddis.2013.548.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Aung KL, Siu LL. Genomically personalized therapy in head and neck cancer. Cancers Head Neck. 2016;1:2. https://doi.org/10.1186/s41199-016-0004-y.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Chung CH, et al. Genomic alterations in head and neck squamous cell carcinoma determined by cancer gene-targeted sequencing. Ann Oncol. 2015;26:1216–23. https://doi.org/10.1093/annonc/mdv109.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  158. Guerrero-Preston R, et al. Key tumor suppressor genes inactivated by “greater promoter” methylation and somatic mutations in head and neck cancer. Epigenetics. 2014;9:1031–46. https://doi.org/10.4161/epi.29025.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Lim AM, et al. Differential mechanisms of CDKN2A (p16) alteration in oral tongue squamous cell carcinomas and correlation with patient outcome. Int J Cancer. 2014;135:887–95. https://doi.org/10.1002/ijc.28727.

    Article  PubMed  CAS  Google Scholar 

  160. Wiest T, Schwarz E, Enders C, Flechtenmacher C, Bosch FX. Involvement of intact HPV16 E6/E7 gene expression in head and neck cancers with unaltered p53 status and perturbed pRb cell cycle control. Oncogene. 2002;21:1510–7. https://doi.org/10.1038/sj.onc.1205214.

    Article  PubMed  CAS  Google Scholar 

  161. Bhatia A, Burtness B. Human papillomavirus-associated oropharyngeal cancer: defining risk groups and clinical trials. J Clin Oncol. 2015;33:3243–50. https://doi.org/10.1200/JCO.2015.61.2358.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Chung CH, et al. p16 protein expression and human papillomavirus status as prognostic biomarkers of nonoropharyngeal head and neck squamous cell carcinoma. J Clin Oncol. 2014;32:3930–8. https://doi.org/10.1200/JCO.2013.54.5228.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Lechner M, et al. Targeted next-generation sequencing of head and neck squamous cell carcinoma identifies novel genetic alterations in HPV+ and HPV- tumors. Genome Med. 2013;5:49. https://doi.org/10.1186/gm453.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Leemans CR, Braakhuis BJ, Brakenhoff RH. The molecular biology of head and neck cancer. Nat Rev Cancer. 2011;11:9–22. https://doi.org/10.1038/nrc2982.

    Article  CAS  PubMed  Google Scholar 

  165. Mesplede T, et al. p53 degradation activity, expression, and subcellular localization of E6 proteins from 29 human papillomavirus genotypes. J Virol. 2012;86:94–107. https://doi.org/10.1128/JVI.00751-11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. McLaughlin-Drubin ME, Munger K. The human papillomavirus E7 oncoprotein. Virology. 2009;384:335–44. https://doi.org/10.1016/j.virol.2008.10.006.

    Article  PubMed  CAS  Google Scholar 

  167. Durzynska J, Lesniewicz K, Poreba E. Human papillomaviruses in epigenetic regulations. Mutat Res. 2017;772:36–50. https://doi.org/10.1016/j.mrrev.2016.09.006.

    Article  CAS  Google Scholar 

  168. Duensing S, et al. The human papillomavirus type 16 E6 and E7 oncoproteins cooperate to induce mitotic defects and genomic instability by uncoupling centrosome duplication from the cell division cycle. Proc Natl Acad Sci U S A. 2000;97:10002–7. https://doi.org/10.1073/pnas.170093297.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  169. Sano D, Oridate N. The molecular mechanism of human papillomavirus-induced carcinogenesis in head and neck squamous cell carcinoma. Int J Clin Oncol. 2016;21:819–26. https://doi.org/10.1007/s10147-016-1005-x.

    Article  PubMed  CAS  Google Scholar 

  170. Romanczuk H, Howley PM. Disruption of either the E1 or the E2 regulatory gene of human papillomavirus type 16 increases viral immortalization capacity. Proc Natl Acad Sci U S A. 1992;89:3159–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  171. Chow YP, et al. Exome sequencing identifies potentially druggable mutations in nasopharyngeal carcinoma. Sci Rep. 2017;7:42980. https://doi.org/10.1038/srep42980.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Forbes SA, et al. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res. 2017;45:D777–83. https://doi.org/10.1093/nar/gkw1121.

    Article  PubMed  CAS  Google Scholar 

  173. Beck TN, Golemis EA. Genomic insights into head and neck cancer. Cancers Head Neck. 2016;1:1. https://doi.org/10.1038/nature14129.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Riese U, et al. Tumor suppressor gene p16 (CDKN2A) mutation status and promoter inactivation in head and neck cancer. Int J Mol Med. 1999;4:61–5.

    PubMed  CAS  Google Scholar 

  175. Greenblatt MS, Bennett WP, Hollstein M, Harris CC. Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res. 1994;54:4855–78.

    CAS  PubMed  Google Scholar 

  176. Poeta ML, et al. TP53 mutations and survival in squamous-cell carcinoma of the head and neck. N Engl J Med. 2007;357:2552–61. https://doi.org/10.1056/NEJMoa073770.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Wong SQ, et al. Targeted-capture massively-parallel sequencing enables robust detection of clinically informative mutations from formalin-fixed tumours. Sci Rep. 2013;3:3494. https://doi.org/10.1038/srep03494.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Beck TN, et al. Phospho-T356RB1 predicts survival in HPV-negative squamous cell carcinoma of the head and neck. Oncotarget. 2015;6:18863–74. https://doi.org/10.18632/oncotarget.4321.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Liu Z, et al. Knocking down CDK4 mediates the elevation of let-7c suppressing cell growth in nasopharyngeal carcinoma. BMC Cancer. 2014;14:274. https://doi.org/10.1186/1471-2407-14-274.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  180. Birkeland AC, Ludwig ML, Spector ME, Brenner JC. The potential for tumor suppressor gene therapy in head and neck cancer. Discov Med. 2016;21:41–7.

    PubMed  PubMed Central  Google Scholar 

  181. Bhowmik A, et al. BRCA1 and MDM2 as independent blood-based biomarkers of head and neck cancer. Tumour Biol. 2016. https://doi.org/10.1007/s13277-016-5359-5.

  182. Wang X, et al. Amplification and overexpression of the cyclin D1 gene in head and neck squamous cell carcinoma. Clin Mol Pathol. 1995;48:M256–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Sheu LF, Chen A, Lee HS, Hsu HY, Yu DS. Cooperative interactions among p53, bcl-2 and Epstein-Barr virus latent membrane protein 1 in nasopharyngeal carcinoma cells. Pathol Int. 2004;54:475–85. https://doi.org/10.1111/j.1440-1827.2004.01654.x.

    Article  CAS  PubMed  Google Scholar 

  184. Niedobitek G, et al. P53 overexpression and Epstein-Barr virus infection in undifferentiated and squamous cell nasopharyngeal carcinomas. J Pathol. 1993;170:457–61. https://doi.org/10.1002/path.1711700409.

    Article  CAS  PubMed  Google Scholar 

  185. Crook T, Nicholls JM, Brooks L, O'Nions J, Allday MJ. High level expression of deltaN-p63: a mechanism for the inactivation of p53 in undifferentiated nasopharyngeal carcinoma (NPC)? Oncogene. 2000;19:3439–44. https://doi.org/10.1038/sj.onc.1203656.

    Article  CAS  PubMed  Google Scholar 

  186. Kwong J, et al. Promoter hypermethylation of multiple genes in nasopharyngeal carcinoma. Clin Cancer Res. 2002;8:131–7.

    PubMed  CAS  Google Scholar 

  187. Aas T, et al. Specific P53 mutations are associated with de novo resistance to doxorubicin in breast cancer patients. Nat Med. 1996;2:811–4.

    Article  PubMed  CAS  Google Scholar 

  188. Pellegata NS, Antoniono RJ, Redpath JL, Stanbridge EJ. DNA damage and p53-mediated cell cycle arrest: a reevaluation. Proc Natl Acad Sci U S A. 1996;93:15209–14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  189. Fridman JS, Lowe SW. Control of apoptosis by p53. Oncogene. 2003;22:9030–40. https://doi.org/10.1038/sj.onc.1207116.

    Article  PubMed  CAS  Google Scholar 

  190. Chipuk JE, et al. Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science. 2004;303:1010–4. https://doi.org/10.1126/science.1092734.

    Article  PubMed  CAS  Google Scholar 

  191. Harbour JW, Dean DC. Rb function in cell-cycle regulation and apoptosis. Nat Cell Biol. 2000;2:E65–7. https://doi.org/10.1038/35008695.

    Article  PubMed  CAS  Google Scholar 

  192. Laptenko O, Prives C. Transcriptional regulation by p53: one protein, many possibilities. Cell Death Differ. 2006;13:951–61. https://doi.org/10.1038/sj.cdd.4401916.

    Article  PubMed  CAS  Google Scholar 

  193. Varley J, Germline M. TP53 mutations and Li-Fraumeni syndrome. Hum Mutat. 2003;21:313–20. https://doi.org/10.1002/humu.10185.

    Article  PubMed  CAS  Google Scholar 

  194. Zhang Y, Coillie SV, Fang JY, Xu J. Gain of function of mutant p53: R282W on the peak? Oncogene. 2016;5:e196. https://doi.org/10.1038/oncsis.2016.8.

    Article  CAS  Google Scholar 

  195. Willis A, Jung EJ, Wakefield T, Chen X. Mutant p53 exerts a dominant negative effect by preventing wild-type p53 from binding to the promoter of its target genes. Oncogene. 2004;23:2330–8. https://doi.org/10.1038/sj.onc.1207396.

    Article  PubMed  CAS  Google Scholar 

  196. Soengas MS, et al. Apaf-1 and caspase-9 in p53-dependent apoptosis and tumor inhibition. Science. 1999;284:156–9.

    Article  PubMed  CAS  Google Scholar 

  197. Mandic R, et al. Reduced cisplatin sensitivity of head and neck squamous cell carcinoma cell lines correlates with mutations affecting the COOH-terminal nuclear localization signal of p53. Clin Cancer Res. 2005;11:6845–52. https://doi.org/10.1158/1078-0432.CCR-05-0378.

    Article  CAS  PubMed  Google Scholar 

  198. Osman AA, et al. Evolutionary action score of TP53 coding variants is predictive of platinum response in head and neck cancer patients. Cancer Res. 2015;75:1205–15. https://doi.org/10.1158/0008-5472.CAN-14-2729.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  199. Gadhikar MA, et al. Chk1/2 inhibition overcomes the cisplatin resistance of head and neck cancer cells secondary to the loss of functional p53. Mol Cancer Ther. 2013;12:1860–73. https://doi.org/10.1158/1535-7163.MCT-13-0157.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  200. Bradford CR, et al. P53 mutation correlates with cisplatin sensitivity in head and neck squamous cell carcinoma lines. Head Neck. 2003;25:654–61. https://doi.org/10.1002/hed.10274.

    Article  PubMed  Google Scholar 

  201. Hoffmann TK, et al. Alterations in the p53 pathway and their association with radio- and chemosensitivity in head and neck squamous cell carcinoma. Oral Oncol. 2008;44:1100–9. https://doi.org/10.1016/j.oraloncology.2008.02.006.

    Article  CAS  PubMed  Google Scholar 

  202. Andrews GA, et al. Mutation of p53 in head and neck squamous cell carcinoma correlates with Bcl-2 expression and increased susceptibility to cisplatin-induced apoptosis. Head Neck. 2004;26:870–7. https://doi.org/10.1002/hed.20029.

    Article  PubMed  Google Scholar 

  203. Ekshyyan O, et al. Comparison of radiosensitizing effects of the mammalian target of rapamycin inhibitor CCI-779 to cisplatin in experimental models of head and neck squamous cell carcinoma. Mol Cancer Ther. 2009;8:2255–65. https://doi.org/10.1158/1535-7163.MCT-08-1184.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  204. Tokalov SV, Abolmaali N. Radiosensitization of p53-deficient lung cancer cells by pre-treatment with cytostatic compounds. Anticancer Res. 2012;32:1239–43.

    CAS  PubMed  Google Scholar 

  205. Adelstein DJ, et al. An intergroup phase III comparison of standard radiation therapy and two schedules of concurrent chemoradiotherapy in patients with unresectable squamous cell head and neck cancer. J Clin Oncol. 2003;21:92–8. https://doi.org/10.1200/JCO.2003.01.008.

    Article  PubMed  Google Scholar 

  206. Cooper JS, et al. Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med. 2004;350:1937–44. https://doi.org/10.1056/NEJMoa032646.

    Article  PubMed  Google Scholar 

  207. Forastiere AA, et al. Concurrent chemotherapy and radiotherapy for organ preservation in advanced laryngeal cancer. N Engl J Med. 2003;349:2091–8. https://doi.org/10.1056/NEJMoa031317.

    Article  PubMed  CAS  Google Scholar 

  208. Mullighan CG, Williams RT, Downing JR, Sherr CJ. Failure of CDKN2A/B (INK4A/B-ARF)-mediated tumor suppression and resistance to targeted therapy in acute lymphoblastic leukemia induced by BCR-ABL. Genes Dev. 2008;22:1411–5. https://doi.org/10.1101/gad.1673908.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  209. Williams RT, den Besten W, Sherr CJ. Cytokine-dependent imatinib resistance in mouse BCR-ABL+, Arf-null lymphoblastic leukemia. Genes Dev. 2007;21:2283–7. https://doi.org/10.1101/gad.1588607.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  210. Dhingra V, Verma J, Misra V, Srivastav S, Hasan F. Evaluation of cyclin D1 expression in head and neck squamous cell carcinoma. J Clin Diagn Res. 2017;11:EC01–4. https://doi.org/10.7860/JCDR/2017/21760.9329.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Kalish LH, et al. Deregulated cyclin D1 expression is associated with decreased efficacy of the selective epidermal growth factor receptor tyrosine kinase inhibitor gefitinib in head and neck squamous cell carcinoma cell lines. Clin Cancer Res. 2004;10:7764–74. https://doi.org/10.1158/1078-0432.CCR-04-0012.

    Article  CAS  PubMed  Google Scholar 

  212. Brockstein BE, Vokes EE, Yoo DS, Posner MR, Brizel DM, Ross ME. Methods to overcome radiation resistance in head and neck cancer. https://www.uptodate.com/contents/methods-to-overcome-radiation-resistance-in-head-and-neck-cancer. UpToDate, Wolters Kluwer Health – 2012-04-20.

  213. Pendleton KP, Grandis JR. Cisplatin-based chemotherapy options for recurrent and/or metastatic squamous cell cancer of the head and neck. Clin Med Insights Ther. 2013. https://doi.org/10.4137/CMT.S10409.

  214. Posner MR. Paradigm shift in the treatment of head and neck cancer: the role of neoadjuvant chemotherapy. Oncologist. 2005;10(Suppl 3):11–9. https://doi.org/10.1634/theoncologist.10-90003-11.

    Article  PubMed  CAS  Google Scholar 

  215. Zhu H, et al. Molecular mechanisms of cisplatin resistance in cervical cancer. Drug Des Devel Ther. 2016;10:1885–95. https://doi.org/10.2147/DDDT.S106412.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  216. Shapiro GI. Cyclin-dependent kinase pathways as targets for cancer treatment. J Clin Oncol. 2006;24:1770–83. https://doi.org/10.1200/JCO.2005.03.7689.

    Article  PubMed  CAS  Google Scholar 

  217. Sedlacek H, et al. Flavopiridol (L86 8275; NSC 649890), a new kinase inhibitor for tumor therapy. Int J Oncol. 1996;9:1143–68.

    PubMed  CAS  Google Scholar 

  218. 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;14:130–46. https://doi.org/10.1038/nrd4504.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  219. Le Tourneau C, et al. Phase I evaluation of seliciclib (R-roscovitine), a novel oral cyclin-dependent kinase inhibitor, in patients with advanced malignancies. Eur J Cancer. 2010;46:3243–50. https://doi.org/10.1016/j.ejca.2010.08.001.

    Article  PubMed  CAS  Google Scholar 

  220. Meijer L, Raymond E. Roscovitine and other purines as kinase inhibitors. From starfish oocytes to clinical trials. Acc Chem Res. 2003;36:417–25. https://doi.org/10.1021/ar0201198.

    Article  CAS  PubMed  Google Scholar 

  221. Whittaker SR, Walton MI, Garrett MD, Workman P. The Cyclin-dependent kinase inhibitor CYC202 (R-roscovitine) inhibits retinoblastoma protein phosphorylation, causes loss of Cyclin D1, and activates the mitogen-activated protein kinase pathway. Cancer Res. 2004;64:262–72.

    Article  CAS  PubMed  Google Scholar 

  222. Malumbres M, et al. Mammalian cells cycle without the D-type cyclin-dependent kinases Cdk4 and Cdk6. Cell. 2004;118:493–504. https://doi.org/10.1016/j.cell.2004.08.002.

    Article  CAS  PubMed  Google Scholar 

  223. O'Leary B, Finn RS, Turner NC. Treating cancer with selective CDK4/6 inhibitors. Nat Rev Clin Oncol. 2016;13:417–30. https://doi.org/10.1038/nrclinonc.2016.26.

    Article  CAS  PubMed  Google Scholar 

  224. VanArsdale T, Boshoff C, Arndt KT, Abraham RT. Molecular pathways: targeting the cyclin D-CDK4/6 axis for cancer treatment. Clin Cancer Res. 2015;21:2905–10. https://doi.org/10.1158/1078-0432.CCR-14-0816.

    Article  CAS  PubMed  Google Scholar 

  225. Finn RS, 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. https://doi.org/10.1186/bcr2419.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  226. Lukas J, Bartkova J, Bartek J. Convergence of mitogenic signalling cascades from diverse classes of receptors at the cyclin D-cyclin-dependent kinase-pRb-controlled G1 checkpoint. Mol Cell Biol. 1996;16:6917–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  227. Kim S, et al. Abstract PR02: LEE011: an orally bioavailable, selective small molecule inhibitor of CDK4/6– reactivating Rb in cancer. Mol Cancer Ther. 2013;12:PR02. https://doi.org/10.1158/1535-7163.targ-13-pr02.

    Article  Google Scholar 

  228. Schrijvers D, Vermorken JB. Role of taxoids in head and neck cancer. Oncologist. 2000;5:199–208.

    Article  CAS  PubMed  Google Scholar 

  229. Marzo I, Naval J. Antimitotic drugs in cancer chemotherapy: promises and pitfalls. Biochem Pharmacol. 2013;86:703–10. https://doi.org/10.1016/j.bcp.2013.07.010.

    Article  CAS  PubMed  Google Scholar 

  230. Fathi AT, et al. Phase I study of the aurora A kinase inhibitor alisertib with induction chemotherapy in patients with acute myeloid leukemia. Haematologica. 2017;102:719–27. https://doi.org/10.3324/haematol.2016.158394.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  231. Ding YH, et al. Alisertib, an Aurora kinase A inhibitor, induces apoptosis and autophagy but inhibits epithelial to mesenchymal transition in human epithelial ovarian cancer cells. Drug Des Devel Ther. 2015;9:425–64. https://doi.org/10.2147/DDDT.S74062.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  232. Matulonis UA, et al. Phase II study of MLN8237 (alisertib), an investigational Aurora A kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol Oncol. 2012;127:63–9. https://doi.org/10.1016/j.ygyno.2012.06.040.

    Article  PubMed  CAS  Google Scholar 

  233. Melichar B, et al. Safety and activity of alisertib, an investigational aurora kinase A inhibitor, in patients with breast cancer, small-cell lung cancer, non-small-cell lung cancer, head and neck squamous-cell carcinoma, and gastro-oesophageal adenocarcinoma: a five-arm phase 2 study. Lancet Oncol. 2015;16:395–405. https://doi.org/10.1016/S1470-2045(15)70051-3.

    Article  CAS  PubMed  Google Scholar 

  234. Thall PF, Cook JD. Dose-finding based on efficacy-toxicity trade-offs. Biometrics. 2004;60:684–93. https://doi.org/10.1111/j.0006-341X.2004.00218.x.

    Article  PubMed  Google Scholar 

  235. Hoellein A, et al. Aurora kinase inhibition overcomes cetuximab resistance in squamous cell cancer of the head and neck. Oncotarget. 2011;2:599–609. https://doi.org/10.18632/oncotarget.311.

    Article  PubMed  PubMed Central  Google Scholar 

  236. Collins S, Blair D, Zarycki J, Szynal C, Gangolli E, Vincent P, Chakravarty A, Ecsedy J. Abstract 3738: a rationale for combining the targeted investigational agents TAK-733, a MEK1/2 inhibitor, with alisertib (MLN8237), an Aurora A kinase inhibitor, for cancer therapy. Cancer Res. 2012;72(8 Suppl):3738. https://doi.org/10.1158/1538-7445.AM2012-3738.

    Article  Google Scholar 

  237. Yu MG, Zheng HY. Acute myeloid leukemia: advancements in diagnosis and treatment. Chin Med J (Engl). 2017;130:211–8. https://doi.org/10.4103/0366-6999.198004.

    Article  Google Scholar 

  238. Kantarjian HM, et al. Phase I study assessing the safety and tolerability of barasertib (AZD1152) with low-dose cytosine arabinoside in elderly patients with AML. Clin Lymphoma Myeloma Leuk. 2013;13:559–67. https://doi.org/10.1016/j.clml.2013.03.019.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  239. Kantarjian HM, et al. Stage I of a phase 2 study assessing the efficacy, safety, and tolerability of barasertib (AZD1152) versus low-dose cytosine arabinoside in elderly patients with acute myeloid leukemia. Cancer. 2013;119:2611–9. https://doi.org/10.1002/cncr.28113.

    Article  PubMed  CAS  Google Scholar 

  240. Foran J, et al. A phase I and pharmacodynamic study of AT9283, a small-molecule inhibitor of aurora kinases in patients with relapsed/refractory leukemia or myelofibrosis. Clin Lymphoma Myeloma Leuk. 2014;14:223–30. https://doi.org/10.1016/j.clml.2013.11.001.

    Article  PubMed  Google Scholar 

  241. Schoffski P, et al. Efficacy and safety of biweekly i.v. administrations of the Aurora kinase inhibitor danusertib hydrochloride in independent cohorts of patients with advanced or metastatic breast, ovarian, colorectal, pancreatic, small-cell and non-small-cell lung cancer: a multi-tumour, multi-institutional phase II study. Ann Oncol. 2015;26:598–607. https://doi.org/10.1093/annonc/mdu566.

    Article  PubMed  CAS  Google Scholar 

  242. Meulenbeld HJ, et al. Randomized phase II study of danusertib in patients with metastatic castration-resistant prostate cancer after docetaxel failure. BJU Int. 2013;111:44–52. https://doi.org/10.1111/j.1464-410X.2012.11404.x.

    Article  PubMed  CAS  Google Scholar 

  243. Liu Z, Sun Q, Wang X. PLK1, a potential target for cancer therapy. Transl Oncol. 2017;10:22–32. https://doi.org/10.1016/j.tranon.2016.10.003.

    Article  PubMed  Google Scholar 

  244. Schoffski P, et al. A phase I, dose-escalation study of the novel Polo-like kinase inhibitor volasertib (BI 6727) in patients with advanced solid tumours. Eur J Cancer. 2012;48:179–86. https://doi.org/10.1016/j.ejca.2011.11.001.

    Article  PubMed  CAS  Google Scholar 

  245. Gutteridge RE, Ndiaye MA, Liu X, Ahmad N. Plk1 inhibitors in cancer therapy: from laboratory to clinics. Mol Cancer Ther. 2016;15:1427–35. https://doi.org/10.1158/1535-7163.MCT-15-0897.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  246. Yu B, Yu Z, Qi PP, Yu DQ, Liu HM. Discovery of orally active anticancer candidate CFI-400945 derived from biologically promising spirooxindoles: success and challenges. Eur J Med Chem. 2015;95:35–40. https://doi.org/10.1016/j.ejmech.2015.03.020.

    Article  PubMed  CAS  Google Scholar 

  247. Hirai H, et al. Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents. Mol Cancer Ther. 2009;8:2992–3000. https://doi.org/10.1158/1535-7163.MCT-09-0463.

    Article  PubMed  CAS  Google Scholar 

  248. Sorensen CS, Syljuasen RG. Safeguarding genome integrity: the checkpoint kinases ATR, CHK1 and WEE1 restrain CDK activity during normal DNA replication. Nucleic Acids Res. 2012;40:477–86. https://doi.org/10.1093/nar/gkr697.

    Article  PubMed  CAS  Google Scholar 

  249. Van Linden AA, et al. Inhibition of Wee1 sensitizes cancer cells to antimetabolite chemotherapeutics in vitro and in vivo, independent of p53 functionality. Mol Cancer Ther. 2013;12:2675–84. https://doi.org/10.1158/1535-7163.MCT-13-0424.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  250. Zeng L, Beggs RR, Cooper TS, Weaver AN, Yang ES. Combining Chk1/2 inhibition with cetuximab and radiation enhances in vitro and in vivo cytotoxicity in head and neck squamous cell carcinoma. Mol Cancer Ther. 2017;16:591–600. https://doi.org/10.1158/1535-7163.MCT-16-0352.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  251. Kato S, et al. Cyclin-dependent kinase pathway aberrations in diverse malignancies: clinical and molecular characteristics. Cell Cycle. 2015;14:1252–9. https://doi.org/10.1080/15384101.2015.1014149.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  252. Osman AA, et al. Wee-1 kinase inhibition overcomes cisplatin resistance associated with high-risk TP53 mutations in head and neck cancer through mitotic arrest followed by senescence. Mol Cancer Ther. 2015;14:608–19. https://doi.org/10.1158/1535-7163.MCT-14-0735-T.

    Article  PubMed  CAS  Google Scholar 

  253. Do K, et al. Phase I study of single-agent AZD1775 (MK-1775), a Wee1 kinase inhibitor, in patients with refractory solid tumors. J Clin Oncol. 2015;33:3409–15. https://doi.org/10.1200/JCO.2014.60.4009.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  254. Ledford H. CRISPR, the disruptor. Nature. 2015;522:20–4. https://doi.org/10.1038/522020a.

    Article  PubMed  CAS  Google Scholar 

  255. Cyranoski D. CRISPR gene-editing tested in a person for the first time. Nature. 2016;539:479. https://doi.org/10.1038/nature.2016.20988.

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sanjeevani Arora .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG, part of Springer Nature

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Demidova, E.V., Iqbal, W., Arora, S. (2018). Molecular Regulation of Cell Cycle and Cell Cycle-Targeted Therapies in Head and Neck Squamous Cell Carcinoma (HNSCC). In: Burtness, B., Golemis, E. (eds) Molecular Determinants of Head and Neck Cancer. Current Cancer Research. Humana Press, Cham. https://doi.org/10.1007/978-3-319-78762-6_7

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-78762-6_7

  • Published:

  • Publisher Name: Humana Press, Cham

  • Print ISBN: 978-3-319-78761-9

  • Online ISBN: 978-3-319-78762-6

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics