Lucanthone and hycanthone are thioxanthenone DNA intercalators used in the 1980s as antitumor agents. Lucanthone is in Phase I clinical trial, whereas hycanthone was pulled out of Phase II trials. Their potential mechanism of action includes DNA intercalation, inhibition of nucleic acid biosyntheses, and inhibition of enzymes like topoisomerases and the dual function base excision repair enzyme apurinic endonuclease 1 (APE1). Lucanthone inhibits the endonuclease activity of APE1, without affecting its redox activity. Our goal was to decipher the precise mechanism of APE1 inhibition as a prerequisite towards development of improved therapeutics that can counteract higher APE1 activity often seen in tumors. The IC50 values for inhibition of APE1 incision of depurinated plasmid DNA by lucanthone and hycanthone were 5 μM and 80 nM, respectively. The KD values (affinity constants) for APE1, as determined by BIACORE binding studies, were 89 nM for lucanthone/10 nM for hycanthone. APE1 structures reveal a hydrophobic pocket where hydrophobic small molecules like thioxanthenones can bind, and our modeling studies confirmed such docking. Circular dichroism spectra uncovered change in the helical structure of APE1 in the presence of lucanthone/hycanthone, and notably, this effect was decreased (Phe266Ala or Phe266Cys or Trp280Leu) or abolished (Phe266Ala/Trp280Ala) when hydrophobic site mutants were employed. Reduced inhibition by lucanthone of the diminished endonuclease activity of hydrophobic mutant proteins (as compared to wild type APE1) supports that binding of lucanthone to the hydrophobic pocket dictates APE1 inhibition. The DNA binding capacity of APE1 was marginally inhibited by lucanthone, and not at all by hycanthone, supporting our hypothesis that thioxanthenones inhibit APE1, predominantly, by direct interaction. Finally, lucanthone-induced degradation was drastically reduced in the presence of short and long lived free radical scavengers, e.g., TRIS and DMSO, suggesting that the mechanism of APE1 breakdown may involve free radical-induced peptide bond cleavage.
References
[1]
Izumi T, Brown B, Naidu CV, Bhakat KK, MacInnes M, et al. (2005) Two essential but distinct functions of the mammalian abasic endonuclease. Proc Natl Acad Sci U S A 102(16): 5739–5743.
[2]
Wilson DM III, Barsky D (2001) The major human abasic endonuclease: formation, consequences and repair of abasic lesions in DNA. Mutation Research/DNA Repair 485(4): 283–307.
[3]
Xanthoudakis S, Miao GG, Curran T (1994) The redox and DNA-repair activities of ref-1 are encoded by nonoverlapping domains. Proc Natl Acad Sci U S A 91: 23–27.
[4]
Chen DS, Herman T, Demple B (1991) Two distinct human DNA diesterases that hydrolyze 3′-blocking deoxyribose fragments from oxidized DNA. Nucleic Acids Res 19(21): 5907–5914.
[5]
Fishel M, Vasko MR, Kelley MR (2007) DNA repair in neurons: So if they don't divide what's to repair? Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis 614(1–2): 24–36.
[6]
Bobola MS, Finn LS, Ellenbogen RG, Geyer JR, Berger MS, et al. (2005) Apurinic/apyrimidinic endonuclease activity is associated with response to radiation and chemotherapy in medulloblastoma and primitive neuroectodermal tumors. Clin Cancer Res 11(20): 7405–14.
[7]
Silber JR, Bobola MS, Blank A, Schoeler KD, Haroldson PD, et al. (2002) The apurinic/apyrimidinic endonuclease activity of Ape1/Ref-1 contributes to human glioma cell resistance to alkylating agents and is elevated by oxidative stress. Clin Cancer Res 8(9): 3008–18.
[8]
Ono Y, Matsumoto K, Furata , Ohmoto T, Akiyama K, et al. (1995) Relationship between expression of a major apurinic/apyrimidinic endonuclease (APEX nuclease) and susceptibility to genotoxic agents in human glioma cell lines. J Neurooncol 25(3): 183–192.
[9]
Robertson KA, Bullock KA, Xu Y, Tritt R, Zimmerman E, et al. (2001) Altered expression of Ape1/ref-1 in germ cell tumors and overexpression in NT2 cells confers resistance to bleomycin and radiation. Cancer Res 61(5): 2220–5.
[10]
Naidu MD, Mason JM, Pica RV, Fung H, Pena LP (2010) Radiation resistance in glioma cells determined by DNA damage repair activity of Ape1/Ref-1. J Radiat Res (Tokyo) 51(4): 393–404.
[11]
Xiang DB, Chen ZT, Wang D, Li MX, Xie JY, et al. (2008) Chimeric adenoviral vector Ad5/F35-mediated APE1 siRNA enhances sensitivity of human colorectal cancer cells to radiotherapy in vitro and in vivo. Cancer Gene Ther 15(10): 625–635.
[12]
Luo M, Kelley MR (2004) Inhibition of the human apurinic/apyrimidinic endonuclease (APE1) repair activity and sensitization of breast cancer cells to DNA alkylating agents with lucanthone. Anticancer Res 24(4): 2127–34.
[13]
Madhusudan S, Smart F, Shrimpton S, Parsons GL, Gardiner L, et al. (2005) Isolation of a small molecule inhibitor of DNA base excision repair. Nucleic Acids Res 33(15): 4711–4724.
[14]
Horton JK, Prasad R, Hou E, Wilson SH (2000) Protection against methylation-induced cytotoxicity by DNA polymerase beta-dependent long patch base excision repair. J Biol Chem 275(3): 2211–2218.
[15]
Raffoul JJ, Banerjee S, Singh-Gupta V, Knoll ZE, Fite A, et al. (2007) Down-regulation of apurinic/apyrimidinic endonuclease 1/redox factor-1 expression by soy isoflavones enhances prostate cancer radiotherapy in vitro and in vivo. Cancer Res 67(5): 2141–2149.
[16]
Shimizu N, Sugimoto K, Tang J, Nishi T, Sato I, et al. (2000) High-performance affinity beads for identifying drug receptors. Nat Biotechnol 18(8): 877–881.
[17]
Kelley MR, Luo M, Reed A, Su D, Delaplane S, et al. (2010) Functional analysis of novel analogs of E3330 that block the redox signaling activity of the multifunctional AP endonuclease/redox signaling enzyme APE1/Ref-1. Sep 27 [Epub ahead of print].
[18]
Nyland RL, Luo M, Kelley MR, Broch RF (2010) Design and synthesis of novel quinone inhibitors targeted to the redox function of apurinic/apyrimidinic endonuclease 1/redox enhancing factor-1 (Ape1/ref-1). J Med Chem 53(3): 1200–1210.
[19]
Nguyen C, Teo JL, Matsuda A, Eguchi M, Chi EY, et al. (2003) Chemogenomic identification of Ref-1/AP-1 as a therapeutic target for asthma. Proc Natl Acad Sci U S A 100(3): 1169–1173.
[20]
Jiang Y, Guo C, Fishel M, Wang , Vasko MR, et al. (2009) Role of APE1 in differentiated neuroblastoma SH-SY5Y cells in response to oxidative stress: use of APE1 small molecule inhibitors to delineate APE1 functions. DNA Repair 8(11): 1273–1182.
[21]
Yang S, Irani K, Hefforn S, Jurnak F, Meyskens FL (2005) Alteration in the expression of Apurinic endonuclease-1/redox factor-1(APE/Ref-1) in human melanoma and identification of the therapeutic potential of resveratrol as an Ape/Ref-1 inhibitor. Mol Can Therap 12(4): 1930–1935.
[22]
Bases R, Mendez BA (1997) Topisomerase inhibition by Lucanthone, an adjuvant in rdaiation therapy. Int J Radiat Oncol Biol Phys 37(5): 1133–1137.
[23]
Rosi D, Perruzotti G, Dennis EW, Berberian DA, Freele H, et al. (1965) A New, Active metabolite of ‘Miracil D’. Nature 208(5014): 1005–1006.
[24]
Berberian DA, Freele H, Rosi D, Dennis EW, Archer S (1967) A Comparison of Oral and Parenteral Activity of Hycanthone and Lucanthone in Experimental Infections with Schistosoma Mansoni. Am J Trop Med Hyg 16(4): 487–491.
Russell WL (1975) Results of tests for possible transmitted genetic effects of hycanthone in mammals. J Toxicol Environ Health 1: 301–304.
[27]
Bulay O, Urman H, Path K, Clayton DB, Shubik P (1979) Carcinogenic potential of hycanthone in mice and hamsters. Int J Cancer 23: 97–104.
[28]
Turner S, Bases R, Pearlman A, Nobler M, Kabakow B (1975) The adjuvant effect of Lucanthone(Miracil D) in Clinical Radiation Therapy. Radiology 114: 729–731.
[29]
Del Rowe JD, Bello J, Mitnick R, Sood B, Filippi C, et al. (1999) Accelerated regression of brain metastases in patients receiving whole brain radiation and the topoisomerase II inhibitor, lucanthone. Int J Radiat Oncol Biol Phys 43(1): 89–93.
[30]
Mendez F, Goldman J, Dand Bases RBE (2002) Abasic sites in DNA of HeLa cells induced by lucanthone. Cancer Invest 20(7–8): 983–91.
[31]
Bailly C, Waring MJ (1993) Preferential Intercalation at AT sequences in DNA by Lucanthone, Hycanthone and Indazole Analogs. A Footprinting Study. Biochemistry 32: 5985–5993.
[32]
Zilversmit R (1971) Thioxanthones. II. Studies on the hydrogen-bonding capacity of lucanthone. Mol Pharmacol 7(6): 674–682.
[33]
Agarwal R, Eshwaramoorthy S, Kumaran D, Dunn JJ, Swaminathan S (2004) Cloning, high level expression, purification, and crystallization of the full length Clostridium botulinum neurotoxin type E light chain. Protein Expr Purif 34(1): 95–102.
[34]
Erzberger JP, Barsky D, Sch?rer OD, Colvin ME, Wilson DM III (1998) Elements in abasic site recognition by the major human and Escherichia coli apurinic/apyrimidinic endonucleases. Nucleic Acids Res 26(11): 2771–2778.
[35]
Bobola MS, Blank A, Beger MS, Stevens BA, Silber JR (2001) Apurinic/apyrimidinic endonuclease activity is elevated in human adult gliomas. Clin Cancer Res 7(11): 3510–8.
[36]
Futcher AB, Morgan AR (1979) A novel assay for endonucleases acting at apurinic sites and its use in measuring AP endonuclease activity in repair-deficient mutants of Saccharomyces cerevisiae. Can J Biochem 57(6): 932–7.
[37]
Sutherland JC, Sutherland BM, Emrick A, Monteleone DC, Riebero EA, et al. (1991) Quantitative electronic imaging of gel fluorescence with CCD cameras: applications in molecular biology. Biotechniques 10(4): 492–7.
[38]
Whitmore L, Wallace BM (2004) DICHROWEB: an online server for protein secondary structure analyses from circular dichroism spectroscopic data. Nucleic Acids Research 32(web serve issue): W 668–W 673.
[39]
Morris GM, Goodsell DS, Halliday RS, Huey R, Hart WE, et al. (1998) Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. Computational Chemistry 19(14): 1639–1642.
[40]
Goodsell DS, Morris GM, Olson AJ (1996) Automated Docking of Flexible Ligands: Applications of AutoDock. Journal of Molecular Recognition 9(1): 1–5.
[41]
Huey R, Morris GM, Olson AJ, Goodsell DS (2007) A semi-empirical free energy force field with charge-based desolvation. J Comp Chem 28: 1145–1152.
[42]
Mezei M, Zhou MM (2010) Dockres: a computer program that analyzes the output of virtual screening of small molecules. Source Code for Biology and Medicine 5: 2.
[43]
Hardgrove GL Jnr, Einstein JR, Wei CH (1983) Structure of p-(p-nitroanilino)phenyl isothiocyanate, C13H9N3O2S. Acta Cryst C39: 616–620.
[44]
Umphrey W, Dalke A, Schulten K (1996) VMD - Visual Molecular Dynamics. J Mol Graph 14: 33–38.
[45]
Jorgensen WL, Chandrashekhar J, Madhura JD, Impey RW, Klein ML (1983) Comparison of simple potential functions for simulating liquid water. J Chem Phys 79: 926–935.
[46]
Phillips JC, Braun R, Wang W, Gumbart J, Tajkhorshid E, et al. (2005) Scalable molecular dynamics with NAMD. J Comp Chem 26: 1781–1802.
[47]
Hornak V, Abel R, Okur A, Stockbine B, Roitberg A, et al. (2006) Comparison of multiple Amber force fields and development of improved protein backbone parameters. Proteins 65(3): 712–725.
[48]
Wang J, Wang W, Kollman PA, Case DA (2006) Automatic atom type and bond type perception in molecular mechanical calculations. J Mol Graph Mod 25: 247–260.
[49]
Wang J, Wolf RM, Caldwell JW, Kollman PA, Case DA (2004) Development and testing of a general AMBER force field. J Comp Chem 25: 1157–1174.
[50]
Liu Y, Prasad R, Beard WA, Kedar PS, Hou ES, et al. (2007) Coordination of Steps in Single-nucleotide Base Excision Repair Mediated by Apurinic/Apyrimidinic Endonuclease 1 and DNA Polymerase β. J Biol Chem, 2007 282(18): 13532–13541.
[51]
Liu Y, Beard WA, Shock PS, Prasad R, Hou ES, et al. (2005) DNA polymerase beta and flap endonuclease 1 enzymatic specificities sustain DNA synthesis for long patch base excision repair. J Biol Chem 280(5): 3665–3774.
[52]
Kingma PS, Corbett AH, Burcham PC, Marnett LJ, Osheroff N (1995) Abasic sites stimulate double-stranded DNA cleavage mediated by topoisomerase II. DNA lesions as endogenous topoisomerase II poisons. J Biol Chem 270(7): 21441–21444.
[53]
Bases R, Mendez F (1969) Reversible inhibition of Ribosomal RNA Synthesis in HeLa cells by Lucanthone(Miracil D) with continued synthesis of DNA -like RNA. J Cell Physiol 74: 283–294.
[54]
Epifanova OI, Makarova GF, Abuladze MK (1975) A comparative study of the effects of lucanthone 9Miracil D) and Actinomycin D on the Chinese Hamster cells grown in cultures. J Cell Physiol 86(2): 261–268.
[55]
Simeonov A, Kulkarni A, Dorjsuren D, Jadhav A, Shen M, et al. (2009) Identification and Characterization of Inhibitors of Human Apurinic/apyrimidinic Endonuclease APE1. PLoS ONE 4(6): 1–13.
[56]
Hadi M, Ginalski K, Nguyen LH, Wilson DM III (2002) Determinants in Nuclease specificity of Ape1 and Ape2, Human Homologues of Escherichia Coli Exonuclease III. J Biol Chem 316(3): 853–866.
[57]
Zawahir Z, Dayam R, Deng J, Pereira C, Neamati N (2009) Pharmacophore Guided Discovery of Small-Molecule Human Apurinic/Apyrimidinic Endonuclease 1 Inhibitors. J Med Chem 52(1): 22–32.
[58]
Fishel ML, Kelley MR (2007) The DNA base excision repair protein Ape1/Ref-1 as a therapeutic and chemopreventive target. Molecular Aspects of Medicine 28(3–4): 375–395.
[59]
Guikema JEJ, Liehan AK, Tsuchimoto D, Nakabeppu Y, Strauss PR, et al. (2007) APE1- and APE2-dependent DNA breaks in immunoglobulin class switch recombination. J Exp Med Vol. 204(12): 3017–3026.
[60]
Koll TT, Feis SS, Wright MH, Teniola MM, Richardson MM, et al. (2008) HSP90 inhibitor, DMAG, synergizes with radiation of lung cancer cells by interfering with base excision and ATM-mediated DNA repair. Mol Can Therap 7(7): 1985–1992.
[61]
Dassoneville L, Bailly C (1999) Stimulation of topoisomerase II-mediated DNA cleavage by an indazole analogue of lucanthone. Biochem Pharmacol 58(8): 1307–1312.
[62]
Archer S, Miller KJ, Rej R, Periana C, Fricker L (1982) Ring-Hydroxylated Analogues of Lucanthone as Antitumor Agents. J Med Chem 25(3): 220–227.
[63]
Kelley MR, Fishel ML (2008) DNA Repair Proteins as Molecular Targets for Cancer Therapeutics. Anticancer Agents Med Chem 8(4): 417–425.