DOI QR코드

DOI QR Code

Enhancement of radiation effect using beta-lapachone and underlying mechanism

  • Ahn, Ki Jung (Department of Radiation Oncology, Inje University College of Medicine) ;
  • Lee, Hyung Sik (Department of Radiation Oncology, Dong-A University College of Medicine) ;
  • Bai, Se Kyung (Department of Biochemistry, Kangwon National University) ;
  • Song, Chang Won (Radiobiology Laboratory, Department of Therapeutic Radiology, University of Minnesota Medical School)
  • Received : 2013.02.01
  • Accepted : 2013.06.04
  • Published : 2013.06.30

Abstract

Beta-lapachone (${\beta}$-Lap; 3,4-dihydro-2, 2-dimethyl-2H-naphthol[1, 2-b]pyran-5,6-dione) is a novel anti-cancer drug under phase I/II clinical trials. ${\beta}$-Lap has been demonstrated to cause apoptotic and necrotic death in a variety of human cancer cells in vitro and in vivo. The mechanisms underlying the ${\beta}$-Lap toxicity against cancer cells has been controversial. The most recent view is that ${\beta}$-Lap, which is a quinone compound, undergoes two-electron reduction to hydroquinone form utilizing NAD(P)H or NADH as electron source. This two-electron reduction of ${\beta}$-Lap is mediated by NAD(P)H:quinone oxidoreductase (NQO1), which is known to mediate the reduction of many quinone compounds. The hydroquinone forms of ${\beta}$-Lap then spontaneously oxidizes back to the original oxidized ${\beta}$-Lap, creating futile cycling between the oxidized and reduced forms of ${\beta}$-Lap. It is proposed that the futile recycling between oxidized and reduced forms of ${\beta}$-Lap leads to two distinct cell death pathways. First one is that the two-electron reduced ${\beta}$-Lap is converted first to one-electron reduced ${\beta}$-Lap, i.e., semiquinone ${\beta}$-Lap $(SQ)^{{\cdot}-}$ causing production of reactive oxygen species (ROS), which then causes apoptotic cell death. The second mechanism is that severe depletion of NAD(P)H and NADH as a result of futile cycling between the quinone and hydroquinone forms of ${\beta}$-Lap causes severe disturbance in cellular metabolism leading to apoptosis and necrosis. The relative importance of the aforementioned two mechanisms, i.e., generation of ROS or depletion of NAD(P)H/NADH, may vary depending on cell type and environment. Importantly, the NQO1 level in cancer cells has been found to be higher than that in normal cells indicating that ${\beta}$-Lap may be preferentially toxic to cancer cells relative to non-cancer cells. The cellular level of NQO1 has been found to be significantly increased by divergent physical and chemical stresses including ionizing radiation. Recent reports clearly demonstrated that ${\beta}$-Lap and ionizing radiation kill cancer cells in a synergistic manner. Indications are that irradiation of cancer cells causes long-lasting elevation of NQO1, thereby sensitizing the cells to ${\beta}$-Lap. In addition, ${\beta}$-Lap has been shown to inhibit the repair of sublethal radiation damage. Treating experimental tumors growing in the legs of mice with irradiation and intraperitoneal injection of ${\beta}$-Lap suppressed the growth of the tumors in a manner more than additive. Collectively, ${\beta}$-Lap is a potentially useful anti-cancer drug, particularly in combination with radiotherapy.

Keywords

References

  1. Pardee AB, Li YZ, Li CJ. Cancer therapy with beta-lapachone. Curr Cancer Drug Targets 2002;2:227-42. https://doi.org/10.2174/1568009023333854
  2. Planchon SM, Wuerzberger S, Frydman B, et al. Beta-lapachone-mediated apoptosis in human promyelocytic leukemia (HL-60) and human prostate cancer cells: a p53-independent response. Cancer Res 1995;55:3706-11.
  3. Park HJ, Ahn KJ, Ahn SD, et al. Susceptibility of cancer cells to beta-lapachone is enhanced by ionizing radiation. Int J Radiat Oncol Biol Phys 2005;61:212-9. https://doi.org/10.1016/j.ijrobp.2004.09.018
  4. Kim EJ, Ji IM, Ahn KJ, et al. Synergistic effect of ionizing radiation and beta-Lapachone against RKO human colon adenocarcinoma cells. Cancer Res Treat 2005;37:183-90. https://doi.org/10.4143/crt.2005.37.3.183
  5. Suzuki M, Amano M, Choi J, et al. Synergistic effects of radiation and beta-lapachone in DU-145 human prostate cancer cells in vitro. Radiat Res 2006;165:525-31. https://doi.org/10.1667/RR3554.1
  6. Choi EK, Terai K, Ji IM, et al. Upregulation of NAD(P)H:quinine oxidoreductase by radiation potentiates the effect of bioreductive beta-lapachone on cancer cells. Neoplasia 2007;9:634-42. https://doi.org/10.1593/neo.07397
  7. Terai K, Dong GZ, Oh ET, et al. Cisplatin enhances the anticancer effect of beta-lapachone by upregulating NQO1. Anticancer Drugs 2009;20:901-9. https://doi.org/10.1097/CAD.0b013e328330098d
  8. Park HJ, Choi EK, Choi J, et al. Heat-induced up-regulation of NAD(P)H:quinone oxidoreductase potentiates anticancer effects of beta-lapachone. Clin Cancer Res 2005;11:8866-71. https://doi.org/10.1158/1078-0432.CCR-05-0818
  9. Song CW, Chae JJ, Choi EK, et al. Anti-cancer effect of bioreductive drug beta-lapachon is enhanced by activating NQO1 with heat shock. Int J Hyperthermia 2008;24:161-9. https://doi.org/10.1080/02656730701781895
  10. Dong GZ, Youn H, Park MT, et al. Heat shock increases expression of NAD(P)H:quinone oxidoreductase (NQO1), mediator of beta-lapachone cytotoxicity, by increasing NQO1 gene activity and via Hsp70-mediated stabilisation of NQO1 protein. Int J Hyperthermia 2009;25:477-87. https://doi.org/10.1080/02656730903049836
  11. Hori T, Kondo T, Lee H, Song CW, Park HJ. Hyperthermia enhances the effect of ${\beta}$ -Lapachone to cause ${\gamma}H2AX$ formations and cell death in human osteosarcoma cells. Int J Hyperthermia 2011;27:53-62. https://doi.org/10.3109/02656736.2010.513361
  12. Planchon SM, Pink JJ, Tagliarino C, Bornmann WG, Varnes ME, Boothman DA. beta-Lapachone-induced apoptosis in human prostate cancer cells: involvement of NQO1/xip3. Exp Cell Res 2001;267:95-106. https://doi.org/10.1006/excr.2001.5234
  13. Pink JJ, Planchon SM, Tagliarino C, Varnes ME, Siegel D, Boothman DA. NAD(P)H:quinone oxidoreductase activity is the principal determinant of beta-lapachone cytotoxicity. J Biol Chem 2000;275:5416-24. https://doi.org/10.1074/jbc.275.8.5416
  14. Pink JJ, Wuerzberger-Davis S, Tagliarino C, et al. Activation of a cysteine protease in MCF-7 and T47D breast cancer cells during beta-lapachone-mediated apoptosis. Exp Cell Res 2000;255:144-55. https://doi.org/10.1006/excr.1999.4790
  15. Tagliarino C, Pink JJ, Dubyak GR, Nieminen AL, Boothman DA. Calcium is a key signaling molecule in beta-lapachonemediated cell death. J Biol Chem 2001;276:19150-9. https://doi.org/10.1074/jbc.M100730200
  16. Wuerzberger SM, Pink JJ, Planchon SM, Byers KL, Bornmann WG, Boothman DA. Induction of apoptosis in MCF-7:WS8 breast cancer cells by beta-lapachone. Cancer Res 1998;58:1876-85.
  17. Huang L, Pardee AB. beta-lapachone induces cell cycle arrest and apoptosis in human colon cancer cells. Mol Med 1999;5:711-20.
  18. Li Y, Sun X, LaMont JT, Pardee AB, Li CJ. Selective killing of cancer cells by beta-lapachone: direct checkpoint activation as a strategy against cancer. Proc Natl Acad Sci U S A 2003;100:2674-8. https://doi.org/10.1073/pnas.0538044100
  19. Boothman DA, Trask DK, Pardee AB. Inhibition of potentially lethal DNA damage repair in human tumor cells by betalapachone, an activator of topoisomerase I. Cancer Res 1989;49:605-12.
  20. Hueber A, Esser P, Heimann K, Kociok N, Winter S, Weller M. The topoisomerase I inhibitors, camptothecin and betalapachone, induce apoptosis of human retinal pigment epithelial cells. Exp Eye Res 1998;67:525-30. https://doi.org/10.1006/exer.1998.0544
  21. Li CJ, Averboukh L, Pardee AB. beta-Lapachone, a novel DNA topoisomerase I inhibitor with a mode of action different from camptothecin. J Biol Chem 1993;268:22463-8.
  22. Weller M, Winter S, Schmidt C, et al. Topoisomerase-I inhibitors for human malignant glioma: differential modulation of p53, p21, bax and bcl-2 expression and of CD95-mediated apoptosis by camptothecin and beta-lapachone. Int J Cancer 1997;73:707-14. https://doi.org/10.1002/(SICI)1097-0215(19971127)73:5<707::AID-IJC16>3.0.CO;2-2
  23. Frydman B, Marton LJ, Sun JS, et al. Induction of DNA topoisomerase II-mediated DNA cleavage by beta-lapachone and related naphthoquinones. Cancer Res 1997;57:620-7.
  24. Krishnan P, Bastow KF. Novel mechanism of cellular DNA topoisomerase II inhibition by the pyranonaphthoquinone derivatives alpha-lapachone and beta-lapachone. Cancer Chemother Pharmacol 2001;47:187-98. https://doi.org/10.1007/s002800000221
  25. Li CJ, Li YZ, Pinto AV, Pardee AB. Potent inhibition of tumor survival in vivo by beta-lapachone plus taxol: combining drugs imposes different artificial checkpoints. Proc Natl Acad Sci U S A 1999;96:13369-74. https://doi.org/10.1073/pnas.96.23.13369
  26. Begleiter A, Fourie J. Induction of NQO1 in cancer cells. Methods Enzymol 2004;382:320-51. https://doi.org/10.1016/S0076-6879(04)82018-4
  27. Joseph P, Xie T, Xu Y, Jaiswal AK. NAD(P)H:quinone oxidoreductase1 (DT-diaphorase): expression, regulation, and role in cancer. Oncol Res 1994;6:525-32.
  28. Joseph P, Jaiswal AK. NAD(P)H:quinone oxidoreductase1 (DT diaphorase) specifically prevents the formation of benzo[a] pyrene quinone-DNA adducts generated by cytochrome P4501A1 and P450 reductase. Proc Natl Acad Sci U S A 1994;91:8413-7. https://doi.org/10.1073/pnas.91.18.8413
  29. Jaiswal AK. Characterization and partial purification of microsomal NAD(P)H:quinone oxidoreductases. Arch Biochem Biophys 2000;375:62-8. https://doi.org/10.1006/abbi.1999.1650
  30. Cullen JJ, Hinkhouse MM, Grady M, et al. Dicumarol inhibition of NADPH:quinone oxidoreductase induces growth inhibition of pancreatic cancer via a superoxide-mediated mechanism. Cancer Res 2003;63:5513-20.
  31. Monks TJ, Hanzlik RP, Cohen GM, Ross D, Graham DG. Quinone chemistry and toxicity. Toxicol Appl Pharmacol 1992;112:2-16. https://doi.org/10.1016/0041-008X(92)90273-U
  32. Riley RJ, Workman P. DT-diaphorase and cancer chemotherapy. Biochem Pharmacol 1992;43:1657-69. https://doi.org/10.1016/0006-2952(92)90694-E
  33. Rauth AM, Goldberg Z, Misra V. DT-diaphorase: possible roles in cancer chemotherapy and carcinogenesis. Oncol Res 1997;9:339-49.
  34. Ross D, Siegel D. NAD(P)H:quinone oxidoreductase 1 (NQO1, DT-diaphorase), functions and pharmacogenetics. Methods Enzymol 2004;382:115-44. https://doi.org/10.1016/S0076-6879(04)82008-1
  35. Talalay P. Mechanisms of induction of enzymes that protect against chemical carcinogenesis. Adv Enzyme Regul 1989;28:237-50. https://doi.org/10.1016/0065-2571(89)90074-5
  36. Jaiswal AK. Jun and Fos regulation of NAD(P)H: quinone oxidoreductase gene expression. Pharmacogenetics 1994;4:1-10. https://doi.org/10.1097/00008571-199402000-00001
  37. Keyes SR, Fracasso PM, Heimbrook DC, Rockwell S, Sligar SG, Sartorelli AC. Role of NADPH:cytochrome c reductase and DTdiaphorase in the biotransformation of mitomycin C1. Cancer Res 1984;44:5638-43.
  38. Pan SS, Andrews PA, Glover CJ, Bachur NR. Reductive activation of mitomycin C and mitomycin C metabolites catalyzed by NADPH-cytochrome P-450 reductase and xanthine oxidase. J Biol Chem 1984;259:959-66.
  39. Hodnick WF, Sartorelli AC. Reductive activation of mitomycin C by NADH:cytochrome b5 reductase. Cancer Res 1993;53:4907-12.
  40. Siegel D, Franklin WA, Ross D. Immunohistochemical detection of NAD(P)H:quinone oxidoreductase in human lung and lung tumors. Clin Cancer Res 1998;4:2065-70.
  41. Belinsky M, Jaiswal AK. NAD(P)H:quinone oxidoreductase1 (DT-diaphorase) expression in normal and tumor tissues. Cancer Metastasis Rev 1993;12:103-17. https://doi.org/10.1007/BF00689804
  42. Cresteil T, Jaiswal AK. High levels of expression of the NAD(P)H: quinine oxidoreductase (NQO1) gene in tumor cells compared to normal cells of the same origin. Biochem Pharmacol 1991;42:1021-7. https://doi.org/10.1016/0006-2952(91)90284-C
  43. Long DJ 2nd, Gaikwad A, Multani A, et al. Disruption of the NAD(P)H:quinone oxidoreductase 1 (NQO1) gene in mice causes myelogenous hyperplasia. Cancer Res 2002;62:3030-6.
  44. Dhakshinamoorthy S, Jaiswal AK. Functional characterization and role of INrf2 in antioxidant response element-mediated expression and antioxidant induction of NAD(P)H:quinone oxidoreductase1 gene. Oncogene 2001;20:3906-17. https://doi.org/10.1038/sj.onc.1204506
  45. Winski SL, Swann E, Hargreaves RH, et al. Relationship between NAD(P)H:quinone oxidoreductase 1 (NQO1) levels in a series of stably transfected cell lines and susceptibility to antitumor quinones. Biochem Pharmacol 2001;61:1509-16. https://doi.org/10.1016/S0006-2952(01)00631-1
  46. Docampo R, Cruz FS, Boveris A, Muniz RP, Esquivel DM. beta-Lapachone enhancement of lipid peroxidation and superoxide anion and hydrogen peroxide formation by sarcoma 180 ascites tumor cells. Biochem Pharmacol 1979;28:723-8. https://doi.org/10.1016/0006-2952(79)90348-4
  47. O'Brien PJ. Molecular mechanisms of quinone cytotoxicity. Chem Biol Interact 1991;80:1-41. https://doi.org/10.1016/0009-2797(91)90029-7
  48. Boveris A, Docampo R, Turrens JF, Stoppani AO. Effect of beta-lapachone on superoxide anion and hydrogen peroxide production in Trypanosoma cruzi. Biochem J 1978;175:431-9. https://doi.org/10.1042/bj1750431
  49. Chau YP, Shiah SG, Don MJ, Kuo ML. Involvement of hydrogen peroxide in topoisomerase inhibitor beta-lapachone-induced apoptosis and differentiation in human leukemia cells. Free Radic Biol Med 1998;24:660-70. https://doi.org/10.1016/S0891-5849(97)00337-7
  50. Molina Portela MP, Stoppani AO. Redox cycling of beta-lapachone and related o-naphthoquinones in the presence of dihydrolipoamide and oxygen. Biochem Pharmacol 1996;51:275-83. https://doi.org/10.1016/0006-2952(95)02168-X

Cited by

  1. UDP-Glucuronosyltransferase 1A Determinates Intracellular Accumulation and Anti-Cancer Effect of β-Lapachone in Human Colon Cancer Cells vol.10, pp.2, 2013, https://doi.org/10.1371/journal.pone.0117051
  2. Excessive Reactive Oxygen Species and Exotic DNA Lesions as an Exploitable Liability vol.55, pp.38, 2013, https://doi.org/10.1021/acs.biochem.6b00703
  3. Pharmacokinetics and derivation of an anticancer dosing regimen for the novel anti-cancer agent isobutyl-deoxynyboquinone (IB-DNQ), a NQO1 bioactivatable molecule, in the domestic felid species vol.35, pp.2, 2013, https://doi.org/10.1007/s10637-016-0414-z
  4. Phytotherapy and Nutritional Supplements on Breast Cancer vol.2017, pp.None, 2013, https://doi.org/10.1155/2017/7207983
  5. Trypanosoma cruzi: death phenotypes induced by ortho-naphthoquinone substrates of the aldo-keto reductase (TcAKR). Role of this enzyme in the mechanism of action of β-lapachone vol.145, pp.9, 2013, https://doi.org/10.1017/s0031182018000045
  6. Synthesis and biological evaluation of β-lapachone and nor-β-lapachone complexes with 2-hydroxypropyl-β-cyclodextrin as trypanocidal agents vol.52, pp.3, 2020, https://doi.org/10.1007/s10863-020-09826-8