期刊论文详细信息
BMC Clinical Pharmacology
Efficacy of RNA polymerase II inhibitors in targeting dormant leukaemia cells
Nigel Russell2  Claire Seedhouse2  Nicholas Boddy2  Abigail Whittall2  Francis Burrows3  Monica Pallis1 
[1] Academic Haematology, Clinical Sciences Building, Nottingham University Hospitals City Campus, Nottingham NG5 1PB, UK;University of Nottingham, Nottingham, UK;Tragara Pharmaceuticals, San Diego, USA
关键词: RNA polymerase II inhibitors;    Dormancy;    Leukemia;   
Others  :  860581
DOI  :  10.1186/2050-6511-14-32
 received in 2013-02-21, accepted in 2013-06-05,  发布年份 2013
PDF
【 摘 要 】

Background

Dormant cells are characterised by low RNA synthesis. In contrast, cancer cells can be addicted to high RNA synthesis, including synthesis of survival molecules. We hypothesised that dormant cancer cells, already low in RNA, might be sensitive to apoptosis induced by RNA Polymerase II (RP2) inhibitors that further reduce RNA synthesis.

Methods

We cultured leukaemia cells continuously in vitro in the presence of an mTOR inhibitor to model dormancy. Apoptosis, damage, RNA content and reducing capacity were evaluated. We treated dormancy-enriched cells for 48 hours with the nucleoside analogues ara-C, 5-azacytidine and clofarabine, the topoisomerase targeting agents daunorubicin, etoposide and irinotecan and three multikinase inhibitors with activity against RP2 - flavopiridol, roscovitine and TG02, and we measured growth inhibition and apoptosis. We describe use of the parameter 2 × IC50 to measure residual cell targeting. RNA synthesis was measured with 5-ethynyl uridine. Drug-induced apoptosis was measured flow cytometrically in primary cells from patients with acute myeloid leukaemia using a CD34/CD71/annexinV gating strategy to identify dormant apoptotic cells.

Results

Culture of the KG1a cell line continuously in the presence of an mTOR inhibitor induced features of dormancy including low RNA content, low metabolism and low basal ROS formation in the absence of a DNA damage response or apoptosis. All agents were more effective against the unmanipulated than the dormancy-enriched cells, emphasising the chemoresistant nature of dormant cells. However, the percentage of cell reduction by RP2 inhibitors at 2 × IC50 was significantly greater than that of other agents. RP2 inhibitors strongly inhibited RNA synthesis compared with other drugs. We also showed that RP2 inhibitors induce apoptosis in proliferating and dormancy-enriched KG1a cells and in the CD71neg CD34pos subset of primary acute myeloid leukaemia cells.

Conclusion

We suggest that RP2 inhibitors may be a useful class of agent for targeting dormant leukaemia cells.

【 授权许可】

   
2013 Pallis et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20140724190357439.pdf 902KB PDF download
104KB Image download
131KB Image download
106KB Image download
89KB Image download
46KB Image download
【 图 表 】

【 参考文献 】
  • [1]Aguirre-Ghiso JA: Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer 2007, 7(11):834-846.
  • [2]Goss PE, Chambers AF: Does tumour dormancy offer a therapeutic target? Nat Rev Cancer 2010, 10(12):871-877.
  • [3]Blagosklonny MV: Cell senescence: hypertrophic arrest beyond the restriction point. J Cell Physiol 2006, 209(3):592-597.
  • [4]Koumenis C, Giaccia A: Transformed cells require continuous activity of RNA polymerase II to resist oncogene-induced apoptosis. Mol Cell Biol 1997, 17(12):7306-7316.
  • [5]Certo M, Del Gaizo MV, Nishino M, Wei G, Korsmeyer S, Armstrong SA, Letai A: Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell 2006, 9(5):351-365.
  • [6]Llambi F, Green DR: Apoptosis and oncogenesis: give and take in the BCL-2 family. Curr Opin Genet Dev 2011, 21(1):12-20.
  • [7]Komarnitsky P, Cho EJ, Buratowski S: Different phosphorylated forms of RNA polymerase II and associated mRNA processing factors during transcription. Genes Dev 2000, 14(19):2452-2460.
  • [8]Lam LT, Pickeral OK, Peng AC, Rosenwald A, Hurt EM, Giltnane JM, Averett LM, Zhao H, Davis RE, Sathyamoorthy M, et al.: Genomic-scale measurement of mRNA turnover and the mechanisms of action of the anti-cancer drug flavopiridol. Genome Biol 2001, 2(10):RESEARCH0041.
  • [9]Chen R, Keating MJ, Gandhi V, Plunkett W: Transcription inhibition by flavopiridol: mechanism of chronic lymphocytic leukemia cell death. Blood 2005, 106(7):2513-2519.
  • [10]Kitada S, Zapata JM, Andreeff M, Reed JC: Protein kinase inhibitors flavopiridol and 7-hydroxy-staurosporine down-regulate antiapoptosis proteins in B-cell chronic lymphocytic leukemia. Blood 2000, 96(2):393-397.
  • [11]Rosato RR, Almenara JA, Kolla SS, Maggio SC, Coe S, Gimenez MS, Dent P, Grant S: Mechanism and functional role of XIAP and Mcl-1 down-regulation in flavopiridol/vorinostat antileukemic interactions. Mol Cancer Ther 2007, 6(2):692-702.
  • [12]MacCallum DE, Melville J, Frame S, Watt K, Anderson S, Gianella-Borradori A, Lane DP, Green SR: Seliciclib (CYC202, R-Roscovitine) induces cell death in multiple myeloma cells by inhibition of RNA polymerase II-dependent transcription and down-regulation of Mcl-1. Cancer Res 2005, 65(12):5399-5407.
  • [13]Alvi AJ, Austen B, Weston VJ, Fegan C, MacCallum D, Gianella-Borradori A, Lane DP, Hubank M, Powell JE, Wei W, et al.: A novel CDK inhibitor, CYC202 (R-roscovitine), overcomes the defect in p53-dependent apoptosis in B-CLL by down-regulation of genes involved in transcription regulation and survival. Blood 2005, 105(11):4484-4491.
  • [14]Goh KC, Novotny-Diermayr V, Hart S, Ong LC, Loh YK, Cheong A, Tan YC, Hu C, Jayaraman R, William AD, et al.: TG02, a novel oral multi-kinase inhibitor of CDKs, JAK2 and FLT3 with potent anti-leukemic properties. Leukemia 2012, 26(2):236-243.
  • [15]Pallis M, Abdul-Aziz A, Burrows F, Seedhouse C, Grundy M, Russell N: The multi-kinase inhibitor TG02 overcomes signalling activation by survival factors to deplete MCL1 and XIAP and induce cell death in primary acute myeloid leukaemia cells. Br J Haematol 2012, 159(2):191-203.
  • [16]Fingar DC, Blenis J: Target of rapamycin (TOR): an integrator of nutrient and growth factor signals and coordinator of cell growth and cell cycle progression. Oncogene 2004, 23(18):3151-3171.
  • [17]Yang X, Yang C, Farberman A, Rideout TC, de Lange CF, France J, Fan MZ: The mammalian target of rapamycin-signaling pathway in regulating metabolism and growth. J Anim Sci 2008, 86(14 Suppl):E36-50.
  • [18]Bailly JD, Muller C, Jaffrezou JP, Demur C, Gassar G, Bordier C, Laurent G: Lack of correlation between expression and function of P-glycoprotein in acute myeloid leukemia cell lines. Leukemia 1995, 9(5):799-807.
  • [19]Schmid I, Cole SW, Korin YD, Zack JA, Giorgi JV: Detection of cell cycle subcompartments by flow cytometric estimation of DNA-RNA content in combination with dual-color immunofluorescence. Cytometry 2000, 39(2):108-116.
  • [20]Jao CY, Salic A: Exploring RNA transcription and turnover in vivo by using click chemistry. Proc Natl Acad Sci U S A 2008, 105(41):15779-15784.
  • [21]Scudiero DA, Shoemaker RH, Paull KD, Monks A, Tierney S, Nofziger TH, Currens MJ, Seniff D, Boyd MR: Evaluation of a soluble tetrazolium/formazan assay for cell growth and drug sensitivity in culture using human and other tumor cell lines. Cancer Res 1988, 48(17):4827-4833.
  • [22]Seedhouse C, Grundy M, Shang S, Ronan J, Pimblett H, Russell N, Pallis M: Impaired S-phase arrest in acute myeloid leukemia cells with a FLT3 internal tandem duplication treated with clofarabine. Clin Cancer Res 2009, 15(23):7291-7298.
  • [23]Jawad M, Seedhouse C, Mony U, Grundy M, Russell NH, Pallis M: Analysis of factors that affect in vitro chemosensitivity of leukaemic stem and progenitor cells to gemtuzumab ozogamicin (Mylotarg) in acute myeloid leukaemia. Leukemia 2010, 24:74-80.
  • [24]Mony U, Jawad M, Seedhouse C, Russell N, Pallis M: Resistance to FLT3 inhibition in an in vitro model of primary AML cells with a stem cell phenotype in a defined microenvironment. Leukemia 2008, 22(7):1395-1401.
  • [25]Yilmaz OH, Valdez R, Theisen BK, Guo W, Ferguson DO, Wu H, Morrison SJ: Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature 2006, 441(7092):475-482.
  • [26]Chen C, Liu Y, Liu R, Ikenoue T, Guan KL, Zheng P: TSC-mTOR maintains quiescence and function of hematopoietic stem cells by repressing mitochondrial biogenesis and reactive oxygen species. J Exp Med 2008, 205(10):2397-2408.
  • [27]Campbell TB, Basu S, Hangoc G, Tao W, Broxmeyer HE: Overexpression of Rheb2 enhances mouse hematopoietic progenitor cell growth while impairing stem cell repopulation. Blood 2009, 114(16):3392-3401.
  • [28]Ito K, Bernardi R, Morotti A, Matsuoka S, Saglio G, Ikeda Y, Rosenblatt J, Avigan DE, Teruya-Feldstein J, Pandolfi PP: PML targeting eradicates quiescent leukaemia-initiating cells. Nature 2008, 453(7198):1072-1078.
  • [29]Shang S, Seedhouse C, Russell N, Pallis M: Low dose rapamycin does not modulate p-glycoprotein function in acute myeloid leukaemia. Leuk Res 2008, 32(5):836-837.
  • [30]Gothot A, Pyatt R, McMahel J, Rice S, Srour EF: Functional heterogeneity of human CD34(+) cells isolated in subcompartments of the G0/G1 phase of the cell cycle. Blood 1997, 90(11):4384-4393.
  • [31]Guan Y, Gerhard B, Hogge DE: Detection, Isolation and Stimulation of Quiescent Primitive Leukemic Progenitor Cells from Patients with Acute Myeloid Leukemia (AML). Blood 2003, 101:3142-3149.
  • [32]Zandomeni R, Mittleman B, Bunick D, Ackerman S, Weinmann R: Mechanism of action of dichloro-beta-D-ribofuranosylbenzimidazole: effect on in vitro transcription. Proc Natl Acad Sci U S A 1982, 79(10):3167-3170.
  • [33]Cho EJ, Kobor MS, Kim M, Greenblatt J, Buratowski S: Opposing effects of Ctk1 kinase and Fcp1 phosphatase at Ser 2 of the RNA polymerase II C-terminal domain. Genes Dev 2001, 15(24):3319-3329.
  • [34]Carlson BA, Dubay MM, Sausville EA, Brizuela L, Worland PJ: Flavopiridol induces G1 arrest with inhibition of cyclin-dependent kinase (CDK) 2 and CDK4 in human breast carcinoma cells. Cancer Res 1996, 56(13):2973-2978.
  • [35]Meijer L, Borgne A, Mulner O, Chong JP, Blow JJ, Inagaki N, Inagaki M, Delcros JG, Moulinoux JP: Biochemical and cellular effects of roscovitine, a potent and selective inhibitor of the cyclin-dependent kinases cdc2, cdk2 and cdk5. Eur J Biochem 1997, 243(1–2):527-536.
  • [36]Neckers LM, Cossman J: Transferrin receptor induction in mitogen-stimulated human T lymphocytes is required for DNA synthesis and cell division and is regulated by interleukin 2. Proc Natl Acad Sci U S A 1983, 80(11):3494-3498.
  • [37]Ohkuma M, Haraguchi N, Ishii H, Mimori K, Tanaka F, Kim HM, Shimomura M, Hirose H, Yanaga K, Mori M: Absence of CD71 transferrin receptor characterizes human gastric adenosquamous carcinoma stem cells. Ann Surg Oncol 2012, 19(4):1357-1364.
  • [38]Cannistra SA, Groshek P, Griffin JD: Granulocyte-macrophage colony-stimulating factor enhances the cytotoxic effects of cytosine arabinoside in acute myeloblastic leukemia and in the myeloid blast crisis phase of chronic myeloid leukemia. Leukemia 1989, 3(5):328-334.
  • [39]Tafuri A, Andreeff M: Kinetic rationale for cytokine-induced recruitment of myeloblastic leukemia followed by cycle-specific chemotherapy in vitro. Leukemia 1990, 4(12):826-834.
  • [40]Saito Y, Uchida N, Tanaka S, Suzuki N, Tomizawa-Murasawa M, Sone A, Najima Y, Takagi S, Aoki Y, Wake A, et al.: Induction of cell cycle entry eliminates human leukemia stem cells in a mouse model of AML. Nat Biotechnol 2010, 28(3):275-280.
  • [41]Pabst T, Vellenga E, van Putten W, Schouten HC, Graux C, Vekemans MC, Biemond B, Sonneveld P, Passweg J, Verdonck L, et al.: Favorable effect of priming with granulocyte colony-stimulating factor in remission induction of acute myeloid leukemia restricted to dose-escalation of cytarabine. Blood 2012, 119:5367-5373.
  • [42]Hoshii T, Tadokoro Y, Naka K, Ooshio T, Muraguchi T, Sugiyama N, Soga T, Araki K, Yamamura K, Hirao A: mTORC1 is essential for leukemia propagation but not stem cell self-renewal. J Clin Invest 2012, 122(6):2114-2129.
  • [43]Bible KC, Kaufmann SH: Flavopiridol: a cytotoxic flavone that induces cell death in noncycling A549 human lung carcinoma cells. Cancer Res 1996, 56(21):4856-4861.
  • [44]Chao SH, Price DH: Flavopiridol inactivates P-TEFb and blocks most RNA polymerase II transcription in vivo. J Biol Chem 2001, 276(34):31793-31799.
  • [45]Rizzolio F, Tuccinardi T, Caligiuri I, Lucchetti C, Giordano A: CDK inhibitors: from the bench to clinical trials. Curr Drug Targets 2010, 11(3):279-290.
  • [46]Karp JE, Garrett-Mayer E, Estey EH, Rudek MA, Smith BD, Greer JM, Drye DM, Mackey K, Dorcy KS, Gore SD, et al.: Randomized phase II study of two schedules of flavopiridol given as timed sequential therapy with cytosine arabinoside and mitoxantrone for adults with newly diagnosed, poor-risk acute myelogenous leukemia. Haematologica 2012, 97(11):1736-1742.
  • [47]Lansdorp PM, Dragowska W: Long-term erythropoiesis from constant numbers of CD34+ cells in serum-free cultures initiated with highly purified progenitor cells from human bone marrow. J Exp Med 1992, 175(6):1501-1509.
  文献评价指标  
  下载次数:0次 浏览次数:11次