Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Kinesins and cancer

Key Points

  • More than 650 members of the kinesin superfamily have been discovered in eukaryotic organisms. They are categorized into 14 subfamilies on the basis of sequence homology and classified as mitotic kinesins, which are involved in cell division, and non-mitotic kinesins, which are principally involved in intracellular transport.

  • The mitotic spindle is a validated target in cancer chemotherapy, and several agents that target tubulin and that interfere with microtubule dynamics are in clinical use. This success has triggered the search for additional mitotic spindle targets, including mitotic kinases (such as cyclin-dependent kinases, Aurora kinase A, Aurora kinase B and Polo-like kinase 1) and specific mitotic kinesins.

  • EG5 and centromere-associated protein E (CENPE) are two mitotic kinesins that have received much of this attention. Specific inhibitors have been developed and are currently in multiple Phase I or Phase II clinical trials. Other kinesins are also considered potential therapeutic targets and candidate inhibitors have been described.

  • Several non-mitotic kinesins are also involved in tumorigenesis and in the development of resistance to anticancer agents.

  • Several kinesins have multiple functions in mitosis or in intracellular transport. Therefore, careful validation is needed to identify any important side effects and to clarify whether they are viable cancer drug targets.

Abstract

Kinesins are a family of molecular motors that travel unidirectionally along microtubule tracks to fulfil their many roles in intracellular transport or cell division. Over the past few years kinesins that are involved in mitosis have emerged as potential targets for cancer drug development. Several compounds that inhibit two mitotic kinesins (EG5 (also known as KIF11) and centromere-associated protein E (CENPE)) have entered Phase I and II clinical trials either as monotherapies or in combination with other drugs. Additional mitotic kinesins are currently being validated as drug targets, raising the possibility that the range of kinesin-based drug targets may expand in the future.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Functions of human kinesins at different stages of mitosis and cytokinesis.
Figure 2: Simplified model of the roles of different mitotic kinesins during mitosis in a kinesin-centric view.
Figure 3: Kinesin inhibitors in clinical development.

Similar content being viewed by others

References

  1. Jordan, M. A. & Wilson, L. Microtubules as a target for anticancer drugs. Nature Rev. Cancer 4, 253–265 (2004).

    Article  CAS  Google Scholar 

  2. Orr, G. A., Verdier-Pinard, P., McDaid, H. & Horwitz, S. B. Mechanisms of Taxol resistance related to microtubules. Oncogene 22, 7280–7295 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kavallaris, M. Microtubules and resistance to tubulin-binding agents. Nature Rev. Cancer 10, 194–204 (2010).

    Article  CAS  Google Scholar 

  4. Wood, K. W., Cornwell, W. D. & Jackson, J. R. Past and future of the mitotic spindle as an oncology target. Curr. Opin. Pharmacol. 1, 370–377 (2001).

    Article  CAS  PubMed  Google Scholar 

  5. Miki, H., Okada, Y. & Hirokawa, N. Analysis of the kinesin superfamily: insights into structure and function. Trends Cell Biol. 15, 467–476 (2005).

    Article  CAS  PubMed  Google Scholar 

  6. Hirokawa, N., Niwa, S. & Tanaka, Y. Molecular motors in neurons: transport mechanisms and roles in brain function, development, and disease. Neuron 68, 610–638 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Lawrence, C. J. et al. A standardized kinesin nomenclature. J. Cell Biol. 167, 19–22 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wordeman, L. How kinesin motor proteins drive mitotic spindle function: lessons from molecular assays. Semin. Cell Dev. Biol. 21, 260–268 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hirokawa, N., Noda, Y., Tanaka, Y. & Niwa, S. Kinesin superfamily motor proteins and intracellular transport. Nature Rev. Mol. Cell Biol. 10, 682–696 (2009).

    Article  CAS  Google Scholar 

  10. Blangy, A. et al. Phosphorylation by p34cdc2 regulates spindle association of human Eg5, a kinesin-related motor essential for bipolar spindle formation in vivo. Cell 83, 1159–1169 (1995).

    Article  CAS  PubMed  Google Scholar 

  11. Mayer, T. U. et al. Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science 286, 971–974 (1999).

    Article  CAS  PubMed  Google Scholar 

  12. Kantarjian, H. M. et al. Phase I/II multicenter study to assess the safety, tolerability, pharmacokinetics and pharmacodynamics of AZD4877 in patients with refractory acute myeloid leukemia. Invest. New Drugs 30, 1107–111115 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Infante, J. R. et al. A Phase I study to assess the safety, tolerability, and pharmacokinetics of AZD4877, an intravenous Eg5 inhibitor in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 69, 165–172 (2011).

    Article  PubMed  CAS  Google Scholar 

  14. Purcell, J. W. et al. Activity of the kinesin spindle protein inhibitor ispinesib (SB-715992) in models of breast cancer. Clin. Cancer Res. 16, 566–576 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Burris, H. A. et al. A phase I study of ispinesib, a kinesin spindle protein inhibitor, administered weekly for three consecutive weeks of a 28-day cycle in patients with solid tumors. Invest. New Drugs 29, 467–472 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Knox, J. J. et al. A phase II and pharmacokinetic study of SB-715992, in patients with metastatic hepato-cellular carcinoma: a study of the National Cancer Institute of Canada Clinical Trials Group (NCIC CTG IND.168). Invest. New Drugs 26, 265–272 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Lee, R. T. et al. A University of Chicago consortium phase II trial of SB-715992 in advanced renal cell cancer. Clin. Genitourin Cancer 6, 21–24 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Holen, K. et al. A phase I trial of MK-0731, a Kinesin Spindle Protein (KSP) inhibitor, in patients with solid tumors. Invest. New Drugs 30, 1088–1095 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Holen, K. D. et al. A first in human study of SB-743921, a kinesin spindle protein inhibitor, to determine pharmacokinetics, biologic effects and establish a recommended phase II dose. Cancer Chemother. Pharmacol. 67, 447–454 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Lee, C. W. et al. A phase II study of ispinesib (SB-715992) in patients with metastatic or recurrent malignant melanoma: a National Cancer Institute of Canada Clinical Trials Group trial. Invest. New Drugs 26, 249–255 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Beer, T. M. et al. Southwest Oncology Group phase II study of ispinesib in androgen-independent prostate cancer previously treated with taxanes. Clin. Genitourin. Cancer 6, 103–109 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Tang, P. A. et al. Phase II study of ispinesib in recurrent or metastatic squamous cell carcinoma of the head and neck. Invest. New Drugs 26, 257–264 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Souid, A. K. et al. A pediatric phase I trial and pharmacokinetic study of ispinesib: a Children's Oncology Group phase I consortium study. Pediatr. Blood Cancer 55, 1323–1328 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Blagden, S. P. et al. A phase I trial of ispinesib, a kinesin spindle protein inhibitor, with docetaxel in patients with advanced solid tumours. Br. J. Cancer 98, 894–899 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gerecitano, J. F. et al. A Phase I trial of the kinesin spindle protein (Eg5) inhibitor AZD4877 in patients with solid and lymphoid malignancies. Invest. New Drugs 22 May 2012 (doi:10.1007/s10637-012-9821-y).

    Article  PubMed  CAS  Google Scholar 

  26. Miller, K. et al. Phase II, Open label study of ispinesib in patients with locally advanced or metastatic breast cancer. 28th Ann. San Antonio Breast Cancer Symp. Abstr. 1089 (San Antonio, 2005).

    Google Scholar 

  27. Lin, S. et al. Inhibition of Kinesin-5, a microtubule-based motor protein, as a strategy for enhancing regeneration of adult axons. Traffic 12, 269–286 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Komlodi-Pasztor, E., Sackett, D. L. & Fojo, A. T. Inhibitors targeting mitosis: tales of how great drugs against a promising target were brought down by a flawed rationale. Clin. Cancer Res. 18, 51–63 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Sueishi, M., Takagi, M. & Yoneda, Y. The forkhead-associated domain of Ki-67 antigen interacts with the novel kinesin-like protein Hklp2. J. Biol. Chem. 275, 28888–28892 (2000).

    Article  CAS  PubMed  Google Scholar 

  30. Tanenbaum, M. E. et al. Kif15 cooperates with eg5 to promote bipolar spindle assembly. Curr. Biol. 19, 1703–1711 (2009). This paper identified the functional redundancy of EG5 and KIF15, which provides a possible explanation for the limited success of EG5 inhibitors.

    Article  CAS  PubMed  Google Scholar 

  31. Vanneste, D., Takagi, M., Imamoto, N. & Vernos, I. The role of Hklp2 in the stabilization and maintenance of spindle bipolarity. Curr. Biol. 19, 1712–1717 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Scanlan, M. J. et al. Humoral immunity to human breast cancer: antigen definition and quantitative analysis of mRNA expression. Cancer Immun. 1, 4 (2001).

    CAS  PubMed  Google Scholar 

  33. Perera, C. N., Spalding, H. S., Mohammed, S. I. & Camarillo, I. G. Identification of proteins secreted from leptin stimulated MCF-7 breast cancer cells: a dual proteomic approach. Exp. Biol. Med. 233, 708–720 (2008).

    Article  CAS  Google Scholar 

  34. Nadar, V. C., Ketschek, A., Myers, K. A., Gallo, G. & Baas, P. W. Kinesin-5 is essential for growth-cone turning. Curr. Biol. 18, 1972–1977 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu, M. et al. Kinesin-12, a mitotic microtubule-associated motor protein, impacts axonal growth, navigation, and branching. J. Neurosci. 30, 14896–14906 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Jones, S. F. et al. Phase I study of ispinesib in combination with carboplatin in patients with advanced solid tumors. Proc. Am. Soc. Clin. Oncol. Abstr. 24, 2027 (2006).

    Google Scholar 

  37. Rhodon, J. et al. Phase I study of ispinesib (SB-715992), a kinesin spindle protein inhibitor, in combination with capecitabine in patients with advanced solid tumors. Eur. J. Canc. Suppl. 4, 193 (2006).

  38. Yen, T. J. et al. CENP-E, a novel human centromere-associated protein required for progression from metaphase to anaphase. EMBO J. 10, 1245–1254 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Mao, Y., Desai, A. & Cleveland, D. W. Microtubule capture by CENP-E silences BubR1-dependent mitotic checkpoint signaling. J. Cell Biol. 170, 873–880 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kim, Y., Holland, A. J., Lan, W. & Cleveland, D. W. Aurora kinases and protein phosphatase 1 mediate chromosome congression through regulation of CENP-E. Cell 142, 444–455 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chan, G. K., Jablonski, S. A., Sudakin, V., Hittle, J. C. & Yen, T. J. Human BUBR1 is a mitotic checkpoint kinase that monitors CENP-E functions at kinetochores and binds the cyclosome/APC. J. Cell Biol. 146, 941–954 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yang, C. P., Liu, L., Ikui, A. E. & Horwitz, S. B. The interaction between mitotic checkpoint proteins, CENP-E and BubR1, is diminished in epothilone B-resistant A549 cells. Cell Cycle 9, 1207–1213 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Chanel-Vos, C. & Giannakakou, P. CENP-E checks in microtubule-drug resistance. Cell Cycle 9, 1456–1465 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Putkey, F. R. et al. Unstable kinetochore-microtubule capture and chromosomal instability following deletion of CENP-E. Dev. Cell 3, 351–365 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Weaver, B. A. & Cleveland, D. W. Aneuploidy: instigator and inhibitor of tumorigenesis. Cancer Res. 67, 10103–10105 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Weaver, B. A., Silk, A. D., Montagna, C., Verdier-Pinard, P. & Cleveland, D. W. Aneuploidy acts both oncogenically and as a tumor suppressor. Cancer Cell 11, 25–36 (2007). This paper describes the involvement of CENPE in aneuploidy.

    Article  CAS  PubMed  Google Scholar 

  47. Agarwal, R. et al. Integrative analysis of cyclin protein levels identifies cyclin b1 as a classifier and predictor of outcomes in breast cancer. Clin. Cancer Res. 15, 3654–3662 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Liu, Z. et al. Reduced expression of cenp-e in human hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 28, 156 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Wood, K. W. et al. Antitumor activity of an allosteric inhibitor of centromere-associated protein-E. Proc. Natl Acad. Sci. USA 107, 5839–5844 (2010). This work describes the mechanism of action of the first CENPE inhibitor that displayed in vivo efficacy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wood, K. W., Chua, P., Sutton, D. & Jackson, J. R. Centromere-associated protein E: a motor that puts the brakes on the mitotic checkpoint. Clin. Cancer Res. 14, 7588–7592 (2008).

    Article  CAS  PubMed  Google Scholar 

  51. Chung, V. et al. First-time-in-human study of GSK923295, a novel antimitotic inhibitor of centromere-associated protein E (CENP-E), in patients with refractory cancer. Cancer Chemother. Pharmacol. 69, 733–741 (2012).

    Article  CAS  PubMed  Google Scholar 

  52. Ding, X. et al. Probing CENP-E function in chromosome dynamics using small molecule inhibitor syntelin. Cell Res. 20, 1386–1389 (2010).

    Article  PubMed  Google Scholar 

  53. Huang, Y. et al. Defects in chromosome congression and mitotic progression in KIF18A-deficient cells are partly mediated through impaired functions of CENP-E. Cell Cycle 8, 2643–2649 (2009).

    Article  CAS  PubMed  Google Scholar 

  54. Sharp, D. J., Rogers, G. C. & Scholey, J. M. Microtubule motors in mitosis. Nature 407, 41–47 (2000).

    Article  CAS  PubMed  Google Scholar 

  55. Ando, A. et al. Cloning of a new kinesin-related gene located at the centromeric end of the human MHC region. Immunogenetics 39, 194–200 (1994).

    Article  CAS  PubMed  Google Scholar 

  56. DeLuca, J. G., Newton, C. N., Himes, R. H., Jordan, M. A. & Wilson, L. Purification and characterization of native conventional kinesin, HSET, and CENP-E from mitotic hela cells. J. Biol. Chem. 276, 28014–28021 (2001).

    Article  CAS  PubMed  Google Scholar 

  57. Mountain, V. et al. The kinesin-related protein, HSET, opposes the activity of Eg5 and cross-links microtubules in the mammalian mitotic spindle. J. Cell Biol. 147, 351–366 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhu, C. et al. Functional analysis of human microtubule-based motor proteins, the kinesins and dyneins, in mitosis/cytokinesis using RNA interference. Mol. Biol. Cell 16, 3187–3199 (2005). This work describes a detailed study identifying 12 members of the kinesin superfamily involved in mitosis and cytokinesis using RNAi and high-resolution imaging.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Cai, S., Weaver, L. N., Ems-McClung, S. C. & Walczak, C. E. Kinesin-14 family proteins HSET/XCTK2 control spindle length by cross-linking and sliding microtubules. Mol. Biol. Cell 20, 1348–1359 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Cai, S., Weaver, L. N., Ems-McClung, S. C. & Walczak, C. E. Proper organization of microtubule minus ends is needed for midzone stability and cytokinesis. Curr. Biol. 20, 880–885 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Grinberg-Rashi, H. et al. The expression of three genes in primary non-small cell lung cancer is associated with metastatic spread to the brain. Clin. Cancer Res. 15, 1755–1761 (2009).

    Article  CAS  PubMed  Google Scholar 

  62. De, S., Cipriano, R., Jackson, M. W. & Stark, G. R. Overexpression of kinesins mediates docetaxel resistance in breast cancer cells. Cancer Res. 69, 8035–8042 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Kwon, M. et al. Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 22, 2189–2203 (2008). This work identified the role of KIFC1 in the chromosome clustering phenomenon that allows cancer cells to divide and evade cell death despite the presence of extra centrosomes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Chandhok, N. S. & Pellman, D. A little CIN may cost a lot: revisiting aneuploidy and cancer. Curr. Opin. Genet. Dev. 19, 74–81 (2009).

    Article  CAS  PubMed  Google Scholar 

  65. Ems-McClung, S. C. & Walczak, C. E. Kinesin-13s in mitosis: key players in the spatial and temporal organization of spindle microtubules. Semin. Cell Dev. Biol. 21, 276–282 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Jang, C. Y. et al. Plk1 and Aurora A regulate the depolymerase activity and the cellular localization of Kif2a. J. Cell Sci. 122, 1334–1341 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Knowlton, A. L., Vorozhko, V. V., Lan, W., Gorbsky, G. J. & Stukenberg, P. T. ICIS and Aurora B coregulate the microtubule depolymerase Kif2a. Curr. Biol. 19, 758–763 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Hood, E. A., Kettenbach, A. N., Gerber, S. A. & Compton, D. A. Plk1 regulates the kinesin-13 protein Kif2b to promote faithful chromosome segregation. Mol. Biol. Cell 25 Apr 2012 (doi:10.1091/mbc.E11-12-1013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Tanenbaum, M. E., Medema, R. H. & Akhmanova, A. Regulation of localization and activity of the microtubule depolymerase MCAK. Bioarchitecture 1, 80–87 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Ganem, N. J. & Compton, D. A. The KinI kinesin Kif2a is required for bipolar spindle assembly through a functional relationship with MCAK. J. Cell Biol. 166, 473–478 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Homma, N. et al. Kinesin superfamily protein 2A (KIF2A) functions in suppression of collateral branch extension. Cell 114, 229–239 (2003).

    Article  CAS  PubMed  Google Scholar 

  72. Wang, C. Q. et al. Overexpression of Kif2a promotes the progression and metastasis of squamous cell carcinoma of the oral tongue. Oral Oncol. 46, 65–69 (2010).

    Article  PubMed  CAS  Google Scholar 

  73. Schimizzi, G. V., Currie, J. D. & Rogers, S. L. Expression levels of a kinesin-13 microtubule depolymerase modulates the effectiveness of anti-microtubule agents. PLoS ONE 5, e11381 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Bakhoum, S. F., Thompson, S. L., Manning, A. L. & Compton, D. A. Genome stability is ensured by temporal control of kinetochore-microtubule dynamics. Nature Cell Biol. 11, 27–35 (2009).

    Article  CAS  PubMed  Google Scholar 

  75. Cimini, D., Moree, B., Canman, J. C. & Salmon, E. D. Merotelic kinetochore orientation occurs frequently during early mitosis in mammalian tissue cells and error correction is achieved by two different mechanisms. J. Cell Sci. 116, 4213–4225 (2003).

    Article  CAS  PubMed  Google Scholar 

  76. Manning, A. L. et al. The kinesin-13 proteins Kif2a, Kif2b, and Kif2c/MCAK have distinct roles during mitosis in human cells. Mol. Biol. Cell 18, 2970–2979 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Moore, A. T. et al. MCAK associates with the tips of polymerizing microtubules. J. Cell Biol. 169, 391–397 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Bakhoum, S. F., Genovese, G. & Compton, D. A. Deviant kinetochore microtubule dynamics underlie chromosomal instability. Curr. Biol. 19, 1937–1942 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Goldie, J. H. & Coldman, A. J. A mathematic model for relating the drug sensitivity of tumors to their spontaneous mutation rate. Cancer Treat. Rep. 63, 1727–1733 (1979).

    CAS  PubMed  Google Scholar 

  80. Ishikawa, K. et al. Mitotic centromere-associated kinesin is a novel marker for prognosis and lymph node metastasis in colorectal cancer. Br. J. Cancer 98, 1824–1829 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Nakamura, Y. et al. Clinicopathological and biological significance of mitotic centromere-associated kinesin overexpression in human gastric cancer. Br. J. Cancer 97, 543–549 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Shimo, A. et al. Involvement of kinesin family member 2C/mitotic centromere-associated kinesin overexpression in mammary carcinogenesis. Cancer Sci. 99, 62–70 (2008).

    CAS  PubMed  Google Scholar 

  83. Shimo, A. et al. Elevated expression of protein regulator of cytokinesis 1, involved in the growth of breast cancer cells. Cancer Sci. 98, 174–181 (2007).

    Article  CAS  PubMed  Google Scholar 

  84. Ganguly, A., Yang, H. & Cabral, F. Overexpression of mitotic centromere-associated Kinesin stimulates microtubule detachment and confers resistance to paclitaxel. Mol. Cancer Ther. 10, 929–937 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Ganguly, A., Yang, H., Pedroza, M., Bhattacharya, R. & Cabral, F. Mitotic centromere-associated kinesin (MCAK) mediates paclitaxel resistance. J. Biol. Chem. 286, 36378–36384 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Gonzalez-Garay, M. L., Chang, L., Blade, K., Menick, D. R. & Cabral, F. A β-tubulin leucine cluster involved in microtubule assembly and paclitaxel resistance. J. Biol. Chem. 274, 23875–23882 (1999).

    Article  CAS  PubMed  Google Scholar 

  87. Hedrick, D. G., Stout, J. R. & Walczak, C. E. Effects of anti-microtubule agents on microtubule organization in cells lacking the kinesin-13 MCAK. Cell Cycle 7, 2146–2156 (2008).

    Article  CAS  PubMed  Google Scholar 

  88. Aoki, S., Ohta, K., Yamazaki, T., Sugawara, F. & Sakaguchi, K. Mammalian mitotic centromere-associated kinesin (MCAK): a new molecular target of sulfoquinovosylacylglycerols novel antitumor and immunosuppressive agents. FEBS J. 272, 2132–2140 (2005).

    Article  CAS  PubMed  Google Scholar 

  89. Rickert, K. W. et al. Discovery and biochemical characterization of selective ATP competitive inhibitors of the human mitotic kinesin KSP. Arch. Biochem. Biophys. 469, 220–231 (2008).

    Article  CAS  PubMed  Google Scholar 

  90. Mazumdar, M. & Misteli, T. Chromokinesins: multitalented players in mitosis. Trends Cell Biol. 15, 349–355 (2005).

    Article  CAS  PubMed  Google Scholar 

  91. Zhu, C. & Jiang, W. Cell cycle-dependent translocation of PRC1 on the spindle by Kif4 is essential for midzone formation and cytokinesis. Proc. Natl Acad. Sci. USA 102, 343–348 (2005).

    Article  CAS  PubMed  Google Scholar 

  92. Kurasawa, Y., Earnshaw, W. C., Mochizuki, Y., Dohmae, N. & Todokoro, K. Essential roles of KIF4 and its binding partner PRC1 in organized central spindle midzone formation. EMBO J. 23, 3237–3248 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Mazumdar, M., Sundareshan, S. & Misteli, T. Human chromokinesin KIF4A functions in chromosome condensation and segregation. J. Cell Biol. 166, 613–620 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Mazumdar, M. et al. Tumor formation via loss of a molecular motor protein. Curr. Biol. 16, 1559–1564 (2006). The authors show that loss of KIF4 leads to aneuploidy and subsequent induction of tumorigenesis in vitro and in vivo.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Narayan, G. et al. Gene dosage alterations revealed by cDNA microarray analysis in cervical cancer: identification of candidate amplified and overexpressed genes. Genes Chromosom. Cancer 46, 373–384 (2007).

    Article  CAS  PubMed  Google Scholar 

  96. Taniwaki, M. et al. Activation of KIF4A as a prognostic biomarker and therapeutic target for lung cancer. Clin. Cancer Res. 13, 6624–6631 (2007).

    Article  CAS  PubMed  Google Scholar 

  97. Rouam, S., Moreau, T. & Broet, P. Identifying common prognostic factors in genomic cancer studies: a novel index for censored outcomes. BMC Bioinformatics 11, 150 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Gao, J. et al. Overexpression of chromokinesin KIF4 inhibits proliferation of human gastric carcinoma cells both in vitro and in vivo. Tumour Biol. 32, 53–61 (2011).

    Article  PubMed  CAS  Google Scholar 

  99. Lee, Y. M. & Kim, W. Association of human kinesin superfamily protein member 4 with BRCA2-associated factor 35. Biochem. J. 374, 497–503 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Wu, G. et al. A novel role of the chromokinesin Kif4A in DNA damage response. Cell Cycle 7, 2013–2020 (2008).

    Article  CAS  PubMed  Google Scholar 

  101. Midorikawa, R., Takei, Y. & Hirokawa, N. KIF4 motor regulates activity-dependent neuronal survival by suppressing PARP-1 enzymatic activity. Cell 125, 371–383 (2006).

    Article  CAS  PubMed  Google Scholar 

  102. Mayr, M. I. et al. The human kinesin Kif18A is a motile microtubule depolymerase essential for chromosome congression. Curr. Biol. 17, 488–498 (2007).

    Article  CAS  PubMed  Google Scholar 

  103. Tokai, N. et al. Kid, a novel kinesin-like DNA binding protein, is localized to chromosomes and the mitotic spindle. EMBO J. 15, 457–467 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Stumpff, J., von Dassow, G., Wagenbach, M., Asbury, C. & Wordeman, L. The kinesin-8 motor Kif18A suppresses kinetochore movements to control mitotic chromosome alignment. Dev. Cell 14, 252–262 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Stumpff, J. et al. A tethering mechanism controls the processivity and kinetochore-microtubule plus-end enrichment of the kinesin-8 Kif18A. Mol. Cell 43, 764–775 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Zhang, C. et al. Kif18A is involved in human breast carcinogenesis. Carcinogenesis 31, 1676–1684 (2010).

    Article  CAS  PubMed  Google Scholar 

  107. Nagahara, M. et al. Kinesin 18A expression: clinical relevance to colorectal cancer progression. Int. J. Cancer 129, 2543–2552 (2011).

    Article  CAS  PubMed  Google Scholar 

  108. Catarinella, M., Gruner, T., Strittmatter, T., Marx, A. & Mayer, T. U. BTB-1: a small molecule inhibitor of the mitotic motor protein Kif18A. Angew. Chem. Int. Ed Engl. 48, 9072–9076 (2009).

    Article  CAS  PubMed  Google Scholar 

  109. Lee, Y. M. et al. Cell cycle-regulated expression and subcellular localization of a kinesin-8 member human KIF18B. Gene 466, 16–25 (2010).

    Article  CAS  PubMed  Google Scholar 

  110. Stout, J. R. et al. Kif18B interacts with EB1 and controls astral microtubule length during mitosis. Mol. Biol. Cell 22, 3070–3080 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Tanenbaum, M. E. et al. A complex of Kif18b and MCAK promotes microtubule depolymerization and is negatively regulated by Aurora kinases. Curr. Biol. 21, 1356–1365 (2011).

    Article  CAS  PubMed  Google Scholar 

  112. Shiroguchi, K., Ohsugi, M., Edamatsu, M., Yamamoto, T. & Toyoshima, Y. Y. The second microtubule-binding site of monomeric Kid enhances the microtubule affinity. J. Biol. Chem. 278, 22460–22465 (2003).

    Article  CAS  PubMed  Google Scholar 

  113. Levesque, A. A. & Compton, D. A. The chromokinesin Kid is necessary for chromosome arm orientation and oscillation, but not congression, on mitotic spindles. J. Cell Biol. 154, 1135–1146 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Ohsugi, M. et al. Cdc2-mediated phosphorylation of Kid controls its distribution to spindle and chromosomes. EMBO J. 22, 2091–2103 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Brouhard, G. J. & Hunt, A. J. Microtubule movements on the arms of mitotic chromosomes: polar ejection forces quantified in vitro. Proc. Natl Acad. Sci. USA 102, 13903–13908 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Santamaria, A., Nagel, S., Sillje, H. H. & Nigg, E. A. The spindle protein CHICA mediates localization of the chromokinesin Kid to the mitotic spindle. Curr. Biol. 18, 723–729 (2008).

    Article  CAS  PubMed  Google Scholar 

  117. Ohsugi, M. et al. Kid-mediated chromosome compaction ensures proper nuclear envelope formation. Cell 132, 771–782 (2008).

    Article  CAS  PubMed  Google Scholar 

  118. Tokai-Nishizumi, N., Ohsugi, M., Suzuki, E. & Yamamoto, T. The chromokinesin Kid is required for maintenance of proper metaphase spindle size. Mol. Biol. Cell 16, 5455–5463 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Eggert, U. S. et al. Parallel chemical genetic and genome-wide RNAi screens identify cytokinesis inhibitors and targets. PLoS Biol. 2, e379 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Eggert, U. S., Mitchison, T. J. & Field, C. M. Animal cytokinesis: from parts list to mechanisms. Annu. Rev. Biochem. 75, 543–566 (2006).

    Article  CAS  PubMed  Google Scholar 

  121. Carleton, M. et al. RNA interference-mediated silencing of mitotic kinesin KIF14 disrupts cell cycle progression and induces cytokinesis failure. Mol. Cell. Biol. 26, 3853–3863 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Gruneberg, U. et al. KIF14 and citron kinase act together to promote efficient cytokinesis. J. Cell Biol. 172, 363–372 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Corson, T. W. & Gallie, B. L. KIF14 mRNA expression is a predictor of grade and outcome in breast cancer. Int. J. Cancer 119, 1088–1094 (2006).

    Article  CAS  PubMed  Google Scholar 

  124. Corson, T. W. et al. KIF14 messenger RNA expression is independently prognostic for outcome in lung cancer. Clin. Cancer Res. 13, 3229–3234 (2007).

    Article  CAS  PubMed  Google Scholar 

  125. Theriault, B. L., Pajovic, S., Bernardini, M. Q., Shaw, P. A. & Gallie, B. L. Kinesin family member 14: an independent prognostic marker and potential therapeutic target for ovarian cancer. Int. J. Cancer 130, 1844–1854 (2011).

    Article  PubMed  CAS  Google Scholar 

  126. Corson, T. W., Huang, A., Tsao, M. S. & Gallie, B. L. KIF14 is a candidate oncogene in the 1q minimal region of genomic gain in multiple cancers. Oncogene 24, 4741–4753 (2005).

    Article  CAS  PubMed  Google Scholar 

  127. Nislow, C., Lombillo, V. A., Kuriyama, R. & McIntosh, J. R. A plus-end-directed motor enzyme that moves antiparallel microtubules in vitro localizes to the interzone of mitotic spindles. Nature 359, 543–547 (1992).

    Article  CAS  PubMed  Google Scholar 

  128. Mishima, M., Kaitna, S. & Glotzer, M. Central spindle assembly and cytokinesis require a kinesin-like protein/RhoGAP complex with microtubule bundling activity. Dev. Cell 2, 41–54 (2002).

    Article  CAS  PubMed  Google Scholar 

  129. Zhu, C., Bossy-Wetzel, E. & Jiang, W. Recruitment of MKLP1 to the spindle midzone/midbody by INCENP is essential for midbody formation and completion of cytokinesis in human cells. Biochem. J. 389, 373–381 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Lee, K. S., Yuan, Y. L., Kuriyama, R. & Erikson, R. L. Plk is an M-phase-specific protein kinase and interacts with a kinesin-like protein, CHO1/MKLP-1. Mol. Cell. Biol. 15, 7143–7151 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Liu, X., Zhou, T., Kuriyama, R. & Erikson, R. L. Molecular interactions of Polo-like-kinase 1 with the mitotic kinesin-like protein CHO1/MKLP-1. J. Cell Sci. 117, 3233–3246 (2004).

    Article  CAS  PubMed  Google Scholar 

  132. Neef, R., Klein, U. R., Kopajtich, R. & Barr, F. A. Cooperation between mitotic kinesins controls the late stages of cytokinesis. Curr. Biol. 16, 301–307 (2006).

    Article  CAS  PubMed  Google Scholar 

  133. Takahashi, S. et al. Downregulation of KIF23 suppresses glioma proliferation. J. Neurooncol. 106, 519–529 (2012).

    Article  CAS  PubMed  Google Scholar 

  134. Valk, K. et al. Gene expression profiles of non-small cell lung cancer: survival prediction and new biomarkers. Oncology 79, 283–292 (2010).

    Article  PubMed  CAS  Google Scholar 

  135. Wang, S. M., Ooi, L. L. & Hui, K. M. Upregulation of Rac GTPase-activating protein 1 is significantly associated with the early recurrence of human hepatocellular carcinoma. Clin. Cancer Res. 17, 6040–6051 (2011).

    Article  CAS  PubMed  Google Scholar 

  136. Echard, A. et al. Interaction of a Golgi-associated kinesin-like protein with Rab6. Science 279, 580–585 (1998).

    Article  CAS  PubMed  Google Scholar 

  137. Fontijn, R. D. et al. The human kinesin-like protein RB6K is under tight cell cycle control and is essential for cytokinesis. Mol. Cell. Biol. 21, 2944–2955 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Hill, E., Clarke, M. & Barr, F. A. The Rab6-binding kinesin, Rab6-KIFL, is required for cytokinesis. EMBO J. 19, 5711–5719 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Simizu, S. & Osada, H. Mutations in the Plk gene lead to instability of Plk protein in human tumour cell lines. Nature Cell Biol. 2, 852–854 (2000).

    Article  CAS  PubMed  Google Scholar 

  140. Neef, R. et al. Phosphorylation of mitotic kinesin-like protein 2 by polo-like kinase 1 is required for cytokinesis. J. Cell Biol. 162, 863–875 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Gruneberg, U., Neef, R., Honda, R., Nigg, E. A. & Barr, F. A. Relocation of Aurora B from centromeres to the central spindle at the metaphase to anaphase transition requires MKlp2. J. Cell Biol. 166, 167–172 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Huemmer, S. & Mayer, T. Cdk1 negatively regulates midzone localization of the mitotic kinesin Mklp2 and the chromosomal passenger complex. Curr. Biol. 19, 607–612 (2009).

    Article  CAS  Google Scholar 

  143. Taniuchi, K. et al. Down-regulation of RAB6KIFL/KIF20A, a kinesin involved with membrane trafficking of discs large homologue 5, can attenuate growth of pancreatic cancer cell. Cancer Res. 65, 105–112 (2005).

    Article  CAS  PubMed  Google Scholar 

  144. Kamimoto, T., Zama, T., Aoki, R., Muro, Y. & Hagiwara, M. Identification of a novel kinesin-related protein, KRMP1, as a target for mitotic peptidyl-prolyl isomerase Pin1. J. Biol. Chem. 276, 37520–37528 (2001).

    Article  CAS  PubMed  Google Scholar 

  145. Kanehira, M. et al. Oncogenic role of MPHOSPH1, a cancer-testis antigen specific to human bladder cancer. Cancer Res. 67, 3276–3285 (2007).

    Article  CAS  PubMed  Google Scholar 

  146. Abaza, A. et al. M phase phosphoprotein 1 is a human plus-end-directed kinesin-related protein required for cytokinesis. J. Biol. Chem. 278, 27844–27852 (2003).

    Article  CAS  PubMed  Google Scholar 

  147. Krzywicka-Racka, A. & Sluder, G. Repeated cleavage failure does not establish centrosome amplification in untransformed human cells. J. Cell Biol. 194, 199–207 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Ganem, N. J., Godinho, S. A. & Pellman, D. A mechanism linking extra centrosomes to chromosomal instability. Nature 460, 278–282 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Fujiwara, T. et al. Cytokinesis failure generating tetraploids promotes tumorigenesis in p53-null cells. Nature 437, 1043–1047 (2005).

    Article  CAS  PubMed  Google Scholar 

  150. Ng, J. M. & Curran, T. The Hedgehog's tale: developing strategies for targeting cancer. Nature Rev. Cancer 11, 493–501 (2011).

    Article  CAS  Google Scholar 

  151. Wilson, C. W. & Chuang, P. T. Mechanism and evolution of cytosolic Hedgehog signal transduction. Development 137, 2079–2094 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Endoh-Yamagami, S. et al. The mammalian Cos2 homolog Kif7 plays an essential role in modulating Hh signal transduction during development. Curr. Biol. 19, 1320–1326 (2009).

    Article  CAS  PubMed  Google Scholar 

  153. Wong, S. Y. et al. Primary cilia can both mediate and suppress Hedgehog pathway-dependent tumorigenesis. Nature Med. 15, 1055–1061 (2009).

    Article  CAS  PubMed  Google Scholar 

  154. Cheung, H. O. et al. The kinesin protein Kif7 is a critical regulator of Gli transcription factors in mammalian hedgehog signaling. Sci. Signal. 2, ra29 (2009).

    Article  PubMed  CAS  Google Scholar 

  155. Hui, C. C. & Angers, S. Gli proteins in development and disease. Annu. Rev. Cell Dev. Biol. 27, 513–537 (2011).

    Article  CAS  PubMed  Google Scholar 

  156. Varjosalo, M. & Taipale, J. Hedgehog: functions and mechanisms. Genes Dev. 22, 2454–2472 (2008).

    Article  CAS  PubMed  Google Scholar 

  157. Hirokawa, N. Stirring up development with the heterotrimeric kinesin KIF3. Traffic 1, 29–34 (2000).

    Article  CAS  PubMed  Google Scholar 

  158. Huangfu, D. & Anderson, K. V. Signaling from Smo to Ci/Gli: conservation and divergence of Hedgehog pathways from Drosophila to vertebrates. Development 133, 3–14 (2006).

    Article  CAS  PubMed  Google Scholar 

  159. Kovacs, J. J. et al. β-arrestin-mediated localization of smoothened to the primary cilium. Science 320, 1777–1781 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Kolpakova-Hart, E., Jinnin, M., Hou, B., Fukai, N. & Olsen, B. R. Kinesin-2 controls development and patterning of the vertebrate skeleton by Hedgehog- and Gli3-dependent mechanisms. Dev. Biol. 309, 273–284 (2007).

    Article  CAS  PubMed  Google Scholar 

  161. Score, J. et al. Identification of a novel imatinib responsive KIF5B-PDGFRA fusion gene following screening for PDGFRA overexpression in patients with hypereosinophilia. Leukemia 20, 827–832 (2006).

    Article  CAS  PubMed  Google Scholar 

  162. Takeuchi, K. et al. KIF5B-ALK, a novel fusion oncokinase identified by an immunohistochemistry-based diagnostic system for ALK-positive lung cancer. Clin. Cancer Res. 15, 3143–3149 (2009).

    Article  CAS  PubMed  Google Scholar 

  163. Wong, D. W. et al. A novel KIF5B-ALK variant in nonsmall cell lung cancer. Cancer 117, 2709–2718 (2011).

    Article  CAS  PubMed  Google Scholar 

  164. Kwak, E. L. et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N. Engl. J. Med. 363, 1693–1703 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Tan, M. H. et al. Specific kinesin expression profiles associated with taxane resistance in basal-like breast cancer. Breast Cancer Res. Treat. 131, 849–858 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. Okada, Y., Yamazaki, H., Sekine-Aizawa, Y. & Hirokawa, N. The neuron-specific kinesin superfamily protein KIF1A is a unique monomeric motor for anterograde axonal transport of synaptic vesicle precursors. Cell 81, 769–780 (1995).

    Article  CAS  PubMed  Google Scholar 

  167. Loyo, M. et al. A survey of methylated candidate tumor suppressor genes in nasopharyngeal carcinoma. Int. J. Cancer 128, 1393–1403 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Ostrow, K. L. et al. Molecular analysis of plasma DNA for the early detection of lung cancer by quantitative methylation-specific PCR. Clin. Cancer Res. 16, 3463–3472 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Cheung, I. Y., Feng, Y., Gerald, W. & Cheung, N. K. Exploiting gene expression profiling to identify novel minimal residual disease markers of neuroblastoma. Clin. Cancer Res. 14, 7020–7027 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Yeh, I. T. et al. A germline mutation of the KIF1B β gene on 1p36 in a family with neural and nonneural tumors. Hum. Genet. 124, 279–285 (2008).

    Article  CAS  PubMed  Google Scholar 

  171. Zhang, H. et al. Genome-wide association study identifies 1p36.22 as a new susceptibility locus for hepatocellular carcinoma in chronic hepatitis B virus carriers. Nature Genet. 42, 755–758 (2010).

    Article  CAS  PubMed  Google Scholar 

  172. Matsuno, K., Sawada, J. S. & Asai, A. Therapeutic potential of mitotic kinesin inhibitors in cancer. Expert Opin. Ther. Patents 18, 253–274 (2008).

    Article  CAS  Google Scholar 

  173. McDonald, A., Bergnes, G. & Morgans, D. J. US Patents 514/266.2; 514/266.23; 514/309; 544/284 and 546/142 (2004).

  174. Good, J. A., Skoufias, D. A. & Kozielski, F. Elucidating the functionality of kinesins: an overview of small molecule inhibitors. Semin. Cell Dev. Biol. 22, 935–945 (2011).

    Article  CAS  PubMed  Google Scholar 

  175. Tcherniuk, S. et al. Relocation of Aurora B and survivin from centromeres to the central spindle impaired by a kinesin-specific MKLP-2 inhibitor. Angew. Chem. Int. Ed Engl. 49, 8228–8231 (2010).

    Article  CAS  PubMed  Google Scholar 

  176. Roche, H. et al. Phase II clinical trial of ixabepilone (BMS-247550), an epothilone B analog, as first-line therapy in patients with metastatic breast cancer previously treated with anthracycline chemotherapy. J. Clin. Oncol. 25, 3415–3420 (2007).

    Article  CAS  PubMed  Google Scholar 

  177. Perez, E. A. et al. Efficacy and safety of ixabepilone (BMS-247550) in a phase II study of patients with advanced breast cancer resistant to an anthracycline, a taxane, and capecitabine. J. Clin. Oncol. 25, 3407–3414 (2007).

    Article  CAS  PubMed  Google Scholar 

  178. Cavaletti, G. & Marmiroli, P. Chemotherapy-induced peripheral neurotoxicity. Nature Rev. Neurol. 6, 657–666 (2010).

    Article  Google Scholar 

  179. Ibrahim, N. K. et al. Multicenter phase II trial of ABI-007, an albumin-bound paclitaxel, in women with metastatic breast cancer. J. Clin. Oncol. 23, 6019–6026 (2005).

    Article  CAS  PubMed  Google Scholar 

  180. Gradishar, W. J. et al. Phase III trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based paclitaxel in women with breast cancer. J. Clin. Oncol. 23, 7794–7803 (2005).

    Article  CAS  PubMed  Google Scholar 

  181. Jackson, J. R., Patrick, D. R., Dar, M. M. & Huang, P. S. Targeted anti-mitotic therapies: can we improve on tubulin agents? Nature Rev. Cancer 7, 107–117 (2007).

    Article  CAS  Google Scholar 

  182. Strebhardt, K. & Ullrich, A. Targeting polo-like kinase 1 for cancer therapy. Nature Rev. Cancer 6, 321–330 (2006).

  183. Lens, S. M., Voest, E. E. & Medema, R. H. Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nature Rev. Cancer 10, 825–841 (2010).

    Article  CAS  Google Scholar 

  184. Dar, A. A., Goff, L. W., Majid, S., Berlin, J. & El-Rifai, W. Aurora kinase inhibitors--rising stars in cancer therapeutics? Mol. Cancer Ther. 9, 268–278 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Malumbres, M. & Barbacid, M. Cell cycle, CDKs and cancer: a changing paradigm. Nature Rev. Cancer 9, 153–166 (2009).

    Article  CAS  Google Scholar 

  186. Barsanti, P. A. et al. The discovery of tetrahydro-β-carbolines as inhibitors of the kinesin Eg5. Bioorg. Med. Chem. Lett. 20, 157–160 (2010).

    Article  CAS  PubMed  Google Scholar 

  187. Cox, C. D. et al. Kinesin spindle protein (KSP) inhibitors. 9. Discovery of (2S)-4-(2,5-difluorophenyl)-n-[(3R,4S)-3-fluoro-1-methylpiperidin-4-yl]-2- (hydroxymethyl)-N-methyl-2-phenyl-2,5-dihydro-1H-pyrrole-1-carboxamide (MK-0731) for the treatment of taxane-refractory cancer. J. Med. Chem. 51, 4239–4252 (2008).

    Article  CAS  PubMed  Google Scholar 

  188. Gartner, M. et al. Development and biological evaluation of potent and specific inhibitors of mitotic Kinesin Eg5. Chembiochem 6, 1173–1177 (2005).

    Article  CAS  PubMed  Google Scholar 

  189. Kim, K. S. et al. Synthesis and SAR of pyrrolotriazine-4-one based Eg5 inhibitors. Bioorg. Med. Chem. Lett. 16, 3937–3942 (2006).

    Article  CAS  PubMed  Google Scholar 

  190. Nakai, R. et al. K858, a novel inhibitor of mitotic kinesin Eg5 and antitumor agent, induces cell death in cancer cells. Cancer Res. 69, 3901–3909 (2009).

    Article  CAS  PubMed  Google Scholar 

  191. Pinkerton, A. B. et al. Synthesis and SAR of thiophene containing kinesin spindle protein (KSP) inhibitors. Bioorg. Med. Chem. Lett. 17, 3562–3569 (2007).

    Article  CAS  PubMed  Google Scholar 

  192. Schiemann, K. et al. The discovery and optimization of hexahydro-2H-pyrano[3,2-c]quinolines (HHPQs) as potent and selective inhibitors of the mitotic kinesin-5. Bioorg. Med. Chem. Lett. 20, 1491–1495 (2010).

    Article  CAS  PubMed  Google Scholar 

  193. Wang, F. et al. Triphenylbutanamines: kinesin spindle protein inhibitors with in vivo antitumor activity. J. Med. Chem. 55, 1511–1525 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Sakowicz, R. et al. Antitumor activity of a kinesin inhibitor. Cancer Res. 64, 3276–3280 (2004). This article was the first to describe in vivo activity of an EG5 inhibitor, thus manifesting kinesins as drug targets for cancer therapy.

    Article  CAS  PubMed  Google Scholar 

  195. Yan, Y. et al. Inhibition of a mitotic motor protein: where, how, and conformational consequences. J. Mol. Biol. 335, 547–554 (2004). This work presents the first crystal structure of an EG5 inhibitor complex and revealed details into the mechanism of action of this class of compounds.

    Article  CAS  PubMed  Google Scholar 

  196. Luo, L. et al. ATP-competitive inhibitors of the mitotic kinesin KSP that function via an allosteric mechanism. Nature Chem. Biol. 3, 722–726 (2007).

    Article  CAS  Google Scholar 

  197. Parrish, C. A. et al. Novel ATP-competitive kinesin spindle protein inhibitors. J. Med. Chem. 50, 4939–4952 (2007).

    Article  CAS  PubMed  Google Scholar 

  198. Learman, S. S. et al. NSC 622124 inhibits human Eg5 and other kinesins via interaction with the conserved microtubule-binding site. Biochemistry 48, 1754–1762 (2009).

    Article  CAS  PubMed  Google Scholar 

  199. Kaan, H. Y., Ulaganathan, V., Hackney, D. D. & Kozielski, F. An allosteric transition trapped in an intermediate state of a new kinesin-inhibitor complex. Biochem. J. 425, 55–60 (2009).

    Article  PubMed  CAS  Google Scholar 

  200. Maliga, Z. et al. A pathway of structural changes produced by monastrol binding to Eg5. J. Biol. Chem. 281, 7977–7982 (2006).

    Article  CAS  PubMed  Google Scholar 

  201. Gascoigne, K. E. & Taylor, S. S. Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs. Cancer Cell 14, 111–122 (2008). The authors present an intriguing systematic study that demonstrated the variability of cell fate after treatment with antimitotic drugs.

    Article  CAS  PubMed  Google Scholar 

  202. Shi, J., Orth, J. D. & Mitchison, T. Cell type variation in responses to antimitotic drugs that target microtubules and kinesin-5. Cancer Res. 68, 3269–3276 (2008).

    Article  CAS  PubMed  Google Scholar 

  203. Garcia-Saez, I., Yen, T., Wade, R. H. & Kozielski, F. Crystal structure of the motor domain of the human kinetochore protein CENP-E. J. Mol. Biol. 340, 1107–1116 (2004).

    Article  CAS  PubMed  Google Scholar 

  204. Turner, J. et al. Crystal structure of the mitotic spindle kinesin Eg5 reveals a novel conformation of the neck-linker. J. Biol. Chem. 276, 25496–25502 (2001).

    Article  CAS  PubMed  Google Scholar 

  205. Parke, C. L., Wojcik, E. J., Kim, S. & Worthylake, D. K. ATP hydrolysis in Eg5 kinesin involves a catalytic two-water mechanism. J. Biol. Chem. 285, 5859–5867 (2010).

    Article  CAS  PubMed  Google Scholar 

  206. Peters, C. et al. Insight into the molecular mechanism of the multitasking kinesin-8 motor. EMBO J. 29, 3437–3447 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Matuliene, J. & Kuriyama, R. Kinesin-like protein CHO1 is required for the formation of midbody matrix and the completion of cytokinesis in mammalian cells. Mol. Biol. Cell 13, 1832–1845 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Nigg, E. A., Mayer, T. U., Huemmer, S., Barr, F. & Bormann, J. EP1683523 (2006).

  209. Hotha, S. et al. HR22C16: a potent small-molecule probe for the dynamics of cell division. Angew. Chem. Int. Ed Engl. 42, 2379–2382 (2003).

    Article  CAS  PubMed  Google Scholar 

  210. Nowicki, M. O. et al. Chronic myelogenous leukemia molecular signature. Oncogene 22, 3952–3963 (2003).

    Article  CAS  PubMed  Google Scholar 

  211. Liu, M. et al. Ectopic expression of the microtubule-dependent motor protein Eg5 promotes pancreatic tumourigenesis. J. Pathol. 221, 221–228 (2010).

    Article  CAS  PubMed  Google Scholar 

  212. Imai, K. et al. Identification of HLA-A2-restricted CTL epitopes of a novel tumour-associated antigen, KIF20A, overexpressed in pancreatic cancer. Br. J. Cancer 104, 300–307 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Cancer Research UK for financial support. They regret that they were unable to include the work of many groups who have contributed to the understanding of the functions of various kinesins from non-human species.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Frank Kozielski.

Ethics declarations

Competing interests

Consultancy (F.K.); Honoraria for speaking (F.K.); Grants or support for research (F.K.); and Patents (F.K.).

Related links

Supplementary information

Supplementary information S1

Human kinesin nomenclature and function (PDF 451 kb)

Supplementary information S2

Published work describing kinesin-targeting inhibitors in clinical development (PDF 353 kb)

Supplementary information S3

Human mitotic kinesins and their motor classification. (PDF 393 kb)

Glossary

Monoastral spindle phenotype

An array of microtubules surrounded by a ring of chromosomes.

Merotelic kinetochore attachment

A single kinetochore becomes attached to microtubules from both spindle poles.

Optical tweezers

An instrument that uses a highly focused laser beam to manipulate microscopic objects (for example, biopolymers such as microtubules or molecular motors) and to measure forces.

Endoreduplication

Replication of the genome in the absence of cell division resulting in polyploidy.

Hypereosinophilia

A haematopoietic disease characterized by an unusually high eosinophil count.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rath, O., Kozielski, F. Kinesins and cancer. Nat Rev Cancer 12, 527–539 (2012). https://doi.org/10.1038/nrc3310

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc3310

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing