J Lipid Atheroscler.  2014 Dec;3(2):63-78. 10.12997/jla.2014.3.2.63.

Kinesin Spindle Protein Inhibition in Translational Research

Affiliations
  • 1National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Department of Health Sciences and Technology, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Korea. phyhanj@inje.ac.kr

Abstract

The kinesin superfamily is a class of motor proteins moving along microtubule filaments and playing essential roles in mitosis of eukaryotic cells. In the cancer biology, mitotic activity is an essential factor for development and metastasis of various cancers. Therefore, the inhibition of kinesin activity is suggested as an alternative cancer therapy. Accumulated clinical evidences have proved the potency of kinesin inhibitors in cancer treatments. In this review, we provided an overview of kinesins that play a critical role in the pathophysiology of various cancers and described the beneficial vs. side effects of their inhibitors that have been tested in both basic science and clinical studies.

Keyword

Kinesin; Kinesin spindle protein; KSP inhibitor; Cancer

MeSH Terms

Biology
Eukaryotic Cells
Kinesin*
Microtubules
Mitosis
Neoplasm Metastasis
Translational Medical Research*
Kinesin

Reference

1. Vale RD, Reese TS, Sheetz MP. Identification of a novel force-generating protein, kinesin, involved in microtubule-based motility. Cell. 1985; 42:39–50.
Article
2. Rowinsky EK, Chaudhry V, Cornblath DR, Donehower RC. Neurotoxicity of Taxol. J Natl Cancer Inst Monogr. 1993; 107–115.
3. Howard J. Mechanics of motor proteins and the cytoskeleton. Sunderland (MA): Sinauer Associates, Inc.;2001.
4. Goldstein LS, Philp AV. The road less traveled: emerging principles of kinesin motor utilization. Annu Rev Cell Dev Biol. 1999; 15:141–183.
Article
5. Wordeman L. How kinesin motor proteins drive mitotic spindle function: lessons from molecular assays. Semin Cell Dev Biol. 2010; 21:260–268.
Article
6. Miki H, Setou M, Kaneshiro K, Hirokawa N. All kinesin superfamily protein, KIF, genes in mouse and human. Proc Natl Acad Sci U S A. 2001; 98:7004–7011.
Article
7. Lawrence CJ, Dawe RK, Christie KR, Cleveland DW, Dawson SC, Endow SA, et al. A standardized kinesin nomenclature. J Cell Biol. 2004; 167:19–22.
Article
8. Yu Y, Feng YM. The role of kinesin family proteins in tumorigenesis and progression: potential biomarkers and molecular targets for cancer therapy. Cancer. 2010; 116:5150–5160.
Article
9. Li L, Li XQ, Pan XH, Feng YM. Prognostic prediction by detection of KIF1B mRNA level in breast cancer and its clinical significance. Chin J Breast Dis. 2009; 173–180.
10. Corson TW, Zhu CQ, Lau SK, Shepherd FA, Tsao MS, Gallie BL. KIF14 messenger RNA expression is independently prognostic for outcome in lung cancer. Clin Cancer Res. 2007; 13:3229–3234.
Article
11. Feng YM, Wan YF, Li XQ, Cao XC, Li X. Expression and clinical significance of KNSL4 in breast cancer. Ai Zheng. 2006; 25:744–748.
12. Taniwaki M, Takano A, Ishikawa N, Yasui W, Inai K, Nishimura H, et al. Activation of KIF4A as a prognostic biomarker and therapeutic target for lung cancer. Clin Cancer Res. 2007; 13:6624–6631.
Article
13. Schlisio S, Kenchappa RS, Vredeveld LC, George RE, Stewart R, Greulich H, et al. The kinesin KIF1Bbeta acts downstream from EglN3 to induce apoptosis and is a potential 1p36 tumor suppressor. Genes Dev. 2008; 22:884–893.
Article
14. Castillo A, Morse HC 3rd, Godfrey VL, Naeem R, Justice MJ. Overexpression of Eg5 causes genomic instability and tumor formation in mice. Cancer Res. 2007; 67:10138–10147.
Article
15. Cardoso CM, Groth-Pedersen L, Høyer-Hansen M, Kirkegaard T, Corcelle E, Andersen JS, et al. Depletion of kinesin 5B affects lysosomal distribution and stability and induces peri-nuclear accumulation of autophagosomes in cancer cells. PLoS One. 2009; 4:e4424.
Article
16. Narayan G, Bourdon V, Chaganti S, Arias-Pulido H, Nandula SV, Rao PH, et al. Gene dosage alterations revealed by cDNA microarray analysis in cervical cancer: identification of candidate amplified and overexpressed genes. Genes Chromosomes Cancer. 2007; 46:373–384.
Article
17. Rouam S, Moreau T, Broët P. Identifying common prognostic factors in genomic cancer studies: a novel index for censored outcomes. BMC Bioinformatics. 2010; 11:150.
Article
18. Liu X, Gong H, Huang K. Oncogenic role of kinesin proteins and targeting kinesin therapy. Cancer Sci. 2013; 104:651–656.
Article
19. Gao J, Sai N, Wang C, Sheng X, Shao Q, Zhou C, et al. Overexpression of chromokinesin KIF4 inhibits proliferation of human gastric carcinoma cells both in vitro and in vivo. Tumour Biol. 2011; 32:53–61.
Article
20. Corson TW, Gallie BL. KIF14 mRNA expression is a predictor of grade and outcome in breast cancer. Int J Cancer. 2006; 119:1088–1094.
Article
21. Madhavan J, Coral K, Mallikarjuna K, Corson TW, Amit N, Khetan V, et al. High expression of KIF14 in retinoblastoma: association with older age at diagnosis. Invest Ophthalmol Vis Sci. 2007; 48:4901–4906.
Article
22. Maney T, Hunter AW, Wagenbach M, Wordeman L. Mitotic centromere-associated kinesin is important for anaphase chromosome segregation. J Cell Biol. 1998; 142:787–801.
Article
23. Bie L, Zhao G, Wang YP, Zhang B. Kinesin family member 2C (KIF2C/MCAK) is a novel marker for prognosis in human gliomas. Clin Neurol Neurosurg. 2012; 114:356–360.
Article
24. Gnjatic S, Cao Y, Reichelt U, Yekebas EF, Nölker C, Marx AH, et al. NY-CO-58/KIF2C is overexpressed in a variety of solid tumors and induces frequent T cell responses in patients with colorectal cancer. Int J Cancer. 2010; 127:381–393.
Article
25. Nishidate T, Katagiri T, Lin ML, Mano Y, Miki Y, Kasumi F, et al. Genome-wide gene-expression profiles of breast-cancer cells purified with laser microbeam microdissection: identification of genes associated with progression and metastasis. Int J Oncol. 2004; 25:797–819.
26. Shimo A, Tanikawa C, Nishidate T, Lin ML, Matsuda K, Park JH, et al. Involvement of kinesin family member 2C/mitotic centromere-associated kinesin overexpression in mammary carcinogenesis. Cancer Sci. 2008; 99:62–70.
Article
27. Ishikawa K, Kamohara Y, Tanaka F, Haraguchi N, Mimori K, Inoue H, et al. Mitotic centromere-associated kinesin is a novel marker for prognosis and lymph node metastasis in colorectal cancer. Br J Cancer. 2008; 98:1824–1829.
Article
28. Kanehira M, Katagiri T, Shimo A, Takata R, Shuin T, Miki T, et al. Oncogenic role of MPHOSPH1, a cancer-testis antigen specific to human bladder cancer. Cancer Res. 2007; 67:3276–3285.
Article
29. Wang CQ, Qu X, Zhang XY, Zhou CJ, Liu GX, Dong ZQ, et al. Overexpression of Kif2a promotes the progression and metastasis of squamous cell carcinoma of the oral tongue. Oral Oncol. 2010; 46:65–69.
Article
30. Zhang C, Zhu C, Chen H, Li L, Guo L, Jiang W, et al. Kif18A is involved in human breast carcinogenesis. Carcinogenesis. 2010; 31:1676–1684.
Article
31. Nagahara M, Nishida N, Iwatsuki M, Ishimaru S, Mimori K, Tanaka F, et al. Kinesin 18A expression: clinical relevance to colorectal cancer progression. Int J Cancer. 2011; 129:2543–2552.
Article
32. Välk K, Vooder T, Kolde R, Reintam MA, Petzold C, Vilo J, et al. Gene expression profiles of non-small cell lung cancer: survival prediction and new biomarkers. Oncology. 2010; 79:283–292.
Article
33. Wang SM, Ooi LL, Hui KM. Upregulation of Rac GTPase-activating protein 1 is significantly associated with the early recurrence of human hepatocellular carcinoma. Clin Cancer Res. 2011; 17:6040–6051.
Article
34. Taniuchi K, Nakagawa H, Nakamura T, Eguchi H, Ohigashi H, Ishikawa O, 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. 2005; 65:105–112.
35. De S, Cipriano R, Jackson MW, Stark GR. Overexpression of kinesins mediates docetaxel resistance in breast cancer cells. Cancer Res. 2009; 69:8035–8042.
Article
36. Thériault BL, Pajovic S, Bernardini MQ, Shaw PA, Gallie BL. Kinesin family member 14: an independent prognostic marker and potential therapeutic target for ovarian cancer. Int J Cancer. 2012; 130:1844–1854.
Article
37. Yeh IT, Lenci RE, Qin Y, Buddavarapu K, Ligon AH, Leteurtre E, et al. A germline mutation of the KIF1B beta gene on 1p36 in a family with neural and nonneural tumors. Hum Genet. 2008; 124:279–285.
Article
38. Liu M, Wang X, Yang Y, Li D, Ren H, Zhu Q, et al. Ectopic expression of the microtubule-dependent motor protein Eg5 promotes pancreatic tumourigenesis. J Pathol. 2010; 221:221–228.
Article
39. Rath O, Kozielski F. Kinesins and cancer. Nat Rev Cancer. 2012; 12:527–539.
Article
40. Takahashi S, Fusaki N, Ohta S, Iwahori Y, Iizuka Y, Inagawa K, et al. Downregulation of KIF23 suppresses glioma proliferation. J Neurooncol. 2012; 106:519–529.
Article
41. Yan GR, Zou FY, Dang BL, Zhang Y, Yu G, Liu X, et al. Genistein-induced mitotic arrest of gastric cancer cells by downregulating KIF20A, a proteomics study. Proteomics. 2012; 12:2391–2399.
Article
42. Liu Z, Ling K, Wu X, Cao J, Liu B, Li S, et al. Reduced expression of cenp-e in human hepatocellular carcinoma. J Exp Clin Cancer Res. 2009; 28:156.
Article
43. Agarwal R, Gonzalez-Angulo AM, Myhre S, Carey M, Lee JS, Overgaard J, et al. Integrative analysis of cyclin protein levels identifies cyclin b1 as a classifier and predictor of outcomes in breast cancer. Clin Cancer Res. 2009; 15:3654–3662.
Article
44. Dyrskjøt L, Kruhøffer M, Thykjaer T, Marcussen N, Jensen JL, Møller K, et al. Gene expression in the urinary bladder: a common carcinoma in situ gene expression signature exists disregarding histopathological classification. Cancer Res. 2004; 64:4040–4048.
45. Richardson AL, Wang ZC, De Nicolo A, Lu X, Brown M, Miron A, et al. X chromosomal abnormalities in basallike human breast cancer. Cancer Cell. 2006; 9:121–132.
Article
46. Jimbo T, Kawasaki Y, Koyama R, Sato R, Takada S, Haraguchi K, et al. Identification of a link between the tumour suppressor APC and the kinesin superfamily. Nat Cell Biol. 2002; 4:323–327.
Article
47. Lukong KE, Richard S. Breast tumor kinase BRK requires kinesin-2 subunit KAP3A in modulation of cell migration. Cell Signal. 2008; 20:432–442.
Article
48. Corson T, Zhu C, Lau S, Shepherd F, Tsao M, Gallie B. KIF14 messenger RNA expression is independently prognostic for outcome in lung cancer. Clin Cancer Res [serial on the Internet]. 2007; 13:Available from: http://www.ncbi.nlm.nih.gov/pubmed/17545527.
Article
49. Tremblay MR, Lescarbeau A, Grogan MJ, Tan E, Lin G, Austad BC, et al. Discovery of a potent and orally active hedgehog pathway antagonist (IPI-926). J Med Chem. 2009; 52:4400–4418.
Article
50. Sarangi A, Valadez JG, Rush S, Abel TW, Thompson RC, Cooper MK. Targeted inhibition of the Hedgehog pathway in established malignant glioma xenografts enhances survival. Oncogene. 2009; 28:3468–3476.
Article
51. Nowicki MO, Pawlowski P, Fischer T, Hess G, Pawlowski T, Skorski T. Chronic myelogenous leukemia molecular signature. Oncogene. 2003; 22:3952–3963.
Article
52. Liu XR, Cai Y, Cao X, Wei RC, Li HL, Zhou XM, et al. A new oncolytic adenoviral vector carrying dual tumour suppressor genes shows potent anti-tumour effect. J Cell Mol Med. 2012; 16:1298–1309.
Article
53. Imai K, Hirata S, Irie A, Senju S, Ikuta Y, Yokomine K, et al. Identification of HLA-A2-restricted CTL epitopes of a novel tumour-associated antigen, KIF20A, overexpressed in pancreatic cancer. Br J Cancer. 2011; 104:300–307.
Article
54. Ganguly A, Yang H, Cabral F. Overexpression of mitotic centromere-associated Kinesin stimulates microtubule detachment and confers resistance to paclitaxel. Mol Cancer Ther. 2011; 10:929–937.
Article
55. Nakamura Y, Tanaka F, Haraguchi N, Mimori K, Matsumoto T, Inoue H, et al. Clinicopathological and biological significance of mitotic centromere-associated kinesin overexpression in human gastric cancer. Br J Cancer. 2007; 97:543–549.
Article
56. Schimizzi GV, Currie JD, Rogers SL. Expression levels of a kinesin-13 microtubule depolymerase modulates the effectiveness of anti-microtubule agents. PLoS One. 2010; 5:e11381.
Article
57. Grinberg-Rashi H, Ofek E, Perelman M, Skarda J, Yaron P, Hajdúch M, 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. 2009; 15:1755–1761.
Article
58. DeBonis S, Skoufias DA, Lebeau L, Lopez R, Robin G, Margolis RL, et al. In vitro screening for inhibitors of the human mitotic kinesin Eg5 with antimitotic and antitumor activities. Mol Cancer Ther. 2004; 3:1079–1090.
59. Sakowicz R, Finer JT, Beraud C, Crompton A, Lewis E, Fritsch A, et al. Antitumor activity of a kinesin inhibitor. Cancer Res. 2004; 64:3276–3280.
Article
60. Luo L, Parrish CA, Nevins N, McNulty DE, Chaudhari AM, Carson JD, et al. ATP-competitive inhibitors of the mitotic kinesin KSP that function via an allosteric mechanism. Nat Chem Biol. 2007; 3:722–726.
Article
61. Maliga Z, Kapoor TM, Mitchison TJ. Evidence that monastrol is an allosteric inhibitor of the mitotic kinesin Eg5. Chem Biol. 2002; 9:989–996.
Article
62. El-Nassan HB. Advances in the discovery of kinesin spindle protein (Eg5) inhibitors as antitumor agents. Eur J Med Chem. 2013; 62:614–631.
Article
63. Huszar D, Theoclitou ME, Skolnik J, Herbst R. Kinesin motor proteins as targets for cancer therapy. Cancer Metastasis Rev. 2009; 28:197–208.
Article
64. Sarli V, Giannis A. Targeting the kinesin spindle protein: basic principles and clinical implications. Clin Cancer Res. 2008; 14:7583–7587.
Article
65. Mayer TU, Kapoor TM, Haggarty SJ, King RW, Schreiber SL, Mitchison TJ. Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science. 1999; 286:971–974.
Article
66. Johnson RK, McCabe FL, Cauder E, Innlow L, Whitacre M, Winkler JD, et al. SB-715992, a potent and selective inhibitor of KSP mitotic kinesin, demonstrates broadspectrum activity in advanced murine tumors and human tumor xenografts. In : 93rd Annual Meeting of the American Association for Cancer Research; 2002 Apr 6-10; Moscone Convention Center, San Francisco (CA). Philadelphia (PA): American Association for Cancer Research;2002. Poster No. 1335.
67. Talapatra SK, Schüttelkopf AW, Kozielski F. The structure of the ternary Eg5-ADP-ispinesib complex. Acta Crystallogr D Biol Crystallogr. 2012; 68:1311–1319.
Article
68. Cox CD, Coleman PJ, Breslin MJ, Whitman DB, Garbaccio RM, Fraley ME, 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-(h ydroxymethyl)-N-methyl-2-phenyl-2,5-dihydro-1H-pyrrol e-1-carboxamide (MK-0731) for the treatment of taxane-refractory cancer. J Med Chem. 2008; 51:4239–4252.
Article
69. Jackson JR, Gilmartin A, Dhanak D, Knight S, Parrish C, Luo L, et al. A second generation KSP inhibitor, SB-743921, is a highly potent and active therapeutic in preclinical models of cancer. AACR Meet Abstr Online. 2006; 2006:B11.
70. Kim KH, Xie Y, Tytler EM, Woessner R, Mor G, Alvero AB. KSP inhibitor ARRY-520 as a substitute for Paclitaxel in Type I ovarian cancer cells. J Transl Med. 2009; 7:63.
Article
71. Woessner R, Tunquist B, Lemieux C, Chlipala E, Jackinsky S, Dewolf W Jr, et al. ARRY-520, a novel KSP inhibitor with potent activity in hematological and taxane-resistant tumor models. Anticancer Res. 2009; 29:4373–4380.
72. Carter BZ, Mak DH, Woessner R, Gross S, Schober WD, Estrov Z, et al. Inhibition of KSP by ARRY-520 induces cell cycle block and cell death via the mitochondrial pathway in AML cells. Leukemia. 2009; 23:1755–1762.
Article
73. Basso AD, Liu M, Dai C, Gray K, Nale L, Tevar S, et al. SCH 2047069, a novel oral kinesin spindle protein inhibitor, shows single-agent antitumor activity and enhances the efficacy of chemotherapeutics. Mol Cancer Ther. 2010; 9:2993–3002.
Article
74. Hotha S, Yarrow JC, Yang JG, Garrett S, Renduchintala KV, Mayer TU, et al. HR22C16: a potent small-molecule probe for the dynamics of cell division. Angew Chem Int Ed Engl. 2003; 42:2379–2382.
Article
75. Marcus AI, Peters U, Thomas SL, Garrett S, Zelnak A, Kapoor TM, et al. Mitotic kinesin inhibitors induce mitotic arrest and cell death in Taxol-resistant and -sensitive cancer cells. J Biol Chem. 2005; 280:11569–11577.
Article
76. Hwang MK, Min YK, Kim SH. Kinesin spindle protein inhibitor HR22C16 sensitizes TRAIL-induced apoptosis in human lung cancer H1299 cells. Bull Korean Chem Soc. 2011; 32:1737–1740.
Article
77. Yang L, Jiang C, Liu F, You QD, Wu WT. Cloning, enzyme characterization of recombinant human Eg5 and the development of a new inhibitor. Biol Pharm Bull. 2008; 31:1397–1402.
Article
78. Nakai R, Iida S, Takahashi T, Tsujita T, Okamoto S, Takada C, et al. K858, a novel inhibitor of mitotic kinesin Eg5 and antitumor agent, induces cell death in cancer cells. Cancer Res. 2009; 69:3901–3909.
Article
79. Jiang C, You Q. Kinesin spindle protein inhibitors in cancer: a patent review (2008 - present). Expert Opin Ther Pat. 2013; 23:1547–1560.
Article
80. Jiang C, Yang L, Wu WT, Guo QL, You QD. CPUYJ039, a newly synthesized benzimidazole-based compound, is proved to be a novel inducer of apoptosis in HCT116 cells with potent KSP inhibitory activity. J Pharm Pharmacol. 2011; 63:1462–1469.
Article
81. Chen LC, Rosen LS, Iyengar T, Goldman JW, Savage R, Kazakin J, et al. First in human study with ARQ 621, a novel inhibitor of Eg5: final results. J Clin Oncol (Meeting Abstracts). 2011; 29:3076.
82. Shimizu M, Ishii H, Ogo N, Unno Y, Matsuno K, Sawada J, et al. S-trityl-L-cysteine derivative induces caspase-independent cell death in K562 human chronic myeloid leukemia cell line. Cancer Lett. 2010; 298:99–106.
Article
83. Ogo N, Oishi S, Matsuno K, Sawada J, Fujii N, Asai A. Synthesis and biological evaluation of L-cysteine derivatives as mitotic kinesin Eg5 inhibitors. Bioorg Med Chem Lett. 2007; 17:3921–3924.
Article
84. Hayashi N, Koller E, Fazli L, Gleave ME. Effects of Eg5 knockdown on human prostate cancer xenograft growth and chemosensitivity. Prostate. 2008; 68:1283–1295.
Article
85. Liu M, Yu H, Huo L, Liu J, Li M, Zhou J. Validating the mitotic kinesin Eg5 as a therapeutic target in pancreatic cancer cells and tumor xenografts using a specific inhibitor. Biochem Pharmacol. 2008; 76:169–178.
Article
86. Leizerman I, Avunie-Masala R, Elkabets M, Fich A, Gheber L. Differential effects of monastrol in two human cell lines. Cell Mol Life Sci. 2004; 61:2060–2070.
Article
87. Vijapurkar U, Wang W, Herbst R. Potentiation of kinesin spindle protein inhibitor-induced cell death by modulation of mitochondrial and death receptor apoptotic pathways. Cancer Res. 2007; 67:237–245.
Article
88. Shi J, Orth JD, Mitchison T. Cell type variation in responses to antimitotic drugs that target microtubules and kinesin-5. Cancer Res. 2008; 68:3269–3276.
Article
89. Liu M, Aneja R, Liu C, Sun L, Gao J, Wang H, et al. Inhibition of the mitotic kinesin Eg5 up-regulates Hsp70 through the phosphatidylinositol 3-kinase/Akt pathway in multiple myeloma cells. J Biol Chem. 2006; 281:18090–18097.
Article
90. Tunquist BJ, Woessner RD, Walker DH. Mcl-1 stability determines mitotic cell fate of human multiple myeloma tumor cells treated with the kinesin spindle protein inhibitor ARRY-520. Mol Cancer Ther. 2010; 9:2046–2056.
Article
91. Tang Y, Orth JD, Xie T, Mitchison TJ. Rapid induction of apoptosis during Kinesin-5 inhibitor-induced mitotic arrest in HL60 cells. Cancer Lett. 2011; 310:15–24.
Article
92. Kashina AS, Baskin RJ, Cole DG, Wedaman KP, Saxton WM, Scholey JM. A bipolar kinesin. Nature. 1996; 379:270–272.
Article
93. Orth JD, Loewer A, Lahav G, Mitchison TJ. Prolonged mitotic arrest triggers partial activation of apoptosis, resulting in DNA damage and p53 induction. Mol Biol Cell. 2012; 23:567–576.
Article
94. Yin Y, Sun H, Xu J, Xiao F, Wang H, Yang Y, et al. Kinesin spindle protein inhibitor SB743921 induces mitotic arrest and apoptosis and overcomes imatinib resistance of chronic myeloid leukemia cells. Leuk Lymphoma. 2014; 1–8.
Article
95. Burris HA, Lorusso P, Jones S, Guthrie TM, Orr JB, Williams DD, et al. Phase I trial of novel kinesin spindle protein (KSP) inhibitor SB-715992 IV days 1, 8, 15 q 28 days. J Clin Oncol (Meeting Abstracts). 2004; 22:2004.
Article
96. Heath EI, Alousi A, Eder JP, Valdivieso M, Vasist LS, Appleman L, et al. A phase I dose escalation trial of ispinesib (SB-715992) administered days 1-3 of a 21-day cycle in patients with advanced solid tumors. J Clin Oncol (Meeting Abstracts). 2006; 24:2026.
Article
97. Chu QS, Holen KD, Rowinsky EK, Wilding G, Volkman JL, Orr JB, et al. Phase I trial of novel kinesin spindle protein (KSP) inhibitor SB-715992 IV Q 21 days. J Clin Oncol (Meeting Abstracts). 2004; 22:2078.
Article
98. Miller K, Ng C, Ang P, Brufsky AM, Lee SC, Dees EC, et al. Abstract 1089. Phase II, open label study of SB-715992 (Ispinesib) in subjects with advanced or metastatic breast cancer. Breast Cancer Res Treat. 2005; 94:S70.
99. Tang PA, Siu LL, Chen EX, Hotte SJ, Chia S, Schwarz JK, et al. Phase II study of ispinesib in recurrent or metastatic squamous cell carcinoma of the head and neck. Invest New Drugs. 2008; 26:257–264.
Article
100. Beer TM, Goldman B, Synold TW, Ryan CW, Vasist LS, Van Veldhuizen PJ Jr, et al. Southwest Oncology Group phase II study of ispinesib in androgen-independent prostate cancer previously treated with taxanes. Clin Genitourin Cancer. 2008; 6:103–109.
Article
101. Purcell JW, Davis J, Reddy M, Martin S, Samayoa K, Vo H, et al. Activity of the kinesin spindle protein inhibitor ispinesib (SB-715992) in models of breast cancer. Clin Cancer Res. 2010; 16:566–576.
Article
102. Carol H, Lock R, Houghton PJ, Morton CL, Kolb EA, Gorlick R, et al. Initial testing (stage 1) of the kinesin spindle protein inhibitor ispinesib by the pediatric preclinical testing program. Pediatr Blood Cancer. 2009; 53:1255–1263.
Article
103. Souid AK, Dubowy RL, Ingle AM, Conlan MG, Sun J, Blaney SM, et al. A pediatric phase I trial and pharmacokinetic study of ispinesib: a Children's Oncology Group phase I consortium study. Pediatr Blood Cancer. 2010; 55:1323–1328.
Article
104. Knox JJ, Gill S, Synold TW, Biagi JJ, Major P, Feld R, et al. A phase II and pharmacokinetic study of SB-715992, in patients with metastatic hepatocellular carcinoma: a study of the National Cancer Institute of Canada Clinical Trials Group (NCIC CTG IND.168). Invest New Drugs. 2008; 26:265–272.
Article
105. Lee CW, Bélanger K, Rao SC, Petrella TM, Tozer RG, Wood L, 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. 2008; 26:249–255.
Article
106. Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005; 5:275–284.
Article
107. Mimeault M, Hauke R, Batra SK. Recent advances on the molecular mechanisms involved in the drug resistance of cancer cells and novel targeting therapies. Clin Pharmacol Ther. 2008; 83:673–691.
Article
108. Khoury HJ, Garcia-Manero G, Borthakur G, Kadia T, Foudray MC, Arellano M, et al. A phase 1 dose-escalation study of ARRY-520, a kinesin spindle protein inhibitor, in patients with advanced myeloid leukemias. Cancer. 2012; 118:3556–3564.
Article
109. Kantarjian HM, Padmanabhan S, Stock W, Tallman MS, Curt GA, Li J, 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. 2012; 30:1107–1115.
Article
110. Infante JR, Kurzrock R, Spratlin J, Burris HA, Eckhardt SG, Li J, 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. 2012; 69:165–172.
Article
111. Holen K, DiPaola R, Liu G, Tan AR, Wilding G, Hsu K, et al. A phase I trial of MK-0731, a kinesin spindle protein (KSP) inhibitor, in patients with solid tumors. Invest New Drugs. 2012; 30:1088–1095.
Article
112. Lee RT, Beekman KE, Hussain M, Davis NB, Clark JI, Thomas SP, et al. A university of Chicago consortium phase II trial of SB-715992 in advanced renal cell cancer. Clin Genitourin Cancer. 2008; 6:21–24.
Article
113. Wakui H, Yamamoto N, Kitazono S, Mizugaki H, Nakamichi S, Fujiwara Y, et al. A phase 1 and dose-finding study of LY2523355 (litronesib), an Eg5 inhibitor, in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2014; 74:15–23.
Article
114. Shih KC, Infante JR, Papadopoulos KP, Bendell JC, Tolcher AW, Burris HA, et al. administered on Days 1, 2, and 3 with or without pegfilgrastim in patients with advanced malignancy (NCT01214629). J Clin Oncol (Meeting Abstracts). 2011; 29:2600.
115. Hentsch B. 4SC announces treatment of first patient in a phase I study of 4SC-205. Oral Eg5 kinesin spindle protein inhibitor to be evaluated in solid tumour and malignant lymphoma patients. Planegg-Martinsried: 4SC AG;2010.
116. Mross KB, Scharr D, Richly H, Frost A, Bauer S, Krauss B, et al. First in human study of 4SC-205 (AEGIS), a novel oral inhibitor of Eg5 kinesin spindle protein. J Clin Oncol (Meeting Abstracts). 2014; 32:2564.
Article
117. Burris HA 3rd, Jones SF, Williams DD, Kathman SJ, Hodge JP, Pandite L, 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. 2011; 29:467–472.
Article
118. Blagden SP, Molife LR, Seebaran A, Payne M, Reid AH, Protheroe AS, et al. A phase I trial of ispinesib, a kinesin spindle protein inhibitor, with docetaxel in patients with advanced solid tumours. Br J Cancer. 2008; 98:894–899.
Article
119. Holen KD, Belani CP, Wilding G, Ramalingam S, Volkman JL, Ramanathan RK, 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. 2011; 67:447–454.
Article
120. Rosen L, Chen LC, Iyengar T, Goldman J, Lahr S, Chen CR, et al. Abstract 2750: ARQ 621, a novel potent and selective inhibitor of Eg5: preclinical data and early results from a clinical phase 1 study. Cancer Res. 2010; 70:2750.
Article
121. Zelnak A. Overcoming taxane and anthracycline resistance. Breast J. 2010; 16:309–312.
Article
122. Tan MH, De S, Bebek G, Orloff MS, Wesolowski R, Downs-Kelly E, et al. Specific kinesin expression profiles associated with taxane resistance in basal-like breast cancer. Breast Cancer Res Treat. 2012; 131:849–858.
Article
123. S Kim . Protein structural biomarkers to guide targeted chemotherapies. United States patent US 20120122132 A1. 2012. 05. 17.
124. Boll IT, Fuchs G. A kinetic model of granulocytopoiesis. Exp Cell Res. 1970; 61:147–152.
Article
125. Gerecitano JF, Stephenson JJ, Lewis NL, Osmukhina A, Li J, Wu K, et al. A phase I trial of the kinesin spindle protein (Eg5) inhibitor AZD4877 in patients with solid and lymphoid malignancies. Invest New Drugs. 2013; 31:355–362.
Article
126. Lin S, Liu M, Son YJ, Timothy Himes B, Snow DM, Yu W, et al. Inhibition of Kinesin-5, a microtubule-based motor protein, as a strategy for enhancing regeneration of adult axons. Traffic. 2011; 12:269–286.
Article
127. Exertier P, Javerzat S, Wang B, Franco M, Herbert J, Platonova N, et al. Impaired angiogenesis and tumor development by inhibition of the mitotic kinesin Eg5. Oncotarget. 2013; 4:2302–2316.
Article
Full Text Links
  • JLA
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr