GET THE APP

The chemotherapeutic effects of lapacho tree extract: β-la
Chemotherapy: Open Access

Chemotherapy: Open Access
Open Access

ISSN: 2167-7700

+44 1223 790975

Review Article - (2014) Volume 3, Issue 2

The chemotherapeutic effects of lapacho tree extract: β-lapachone

Hsiu-Ni Kung1*, Kuo-Shyan Lu1 and Yat-Pang Chau2
1Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
2Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
*Corresponding Author: Hsiu-Ni Kung, Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, Tel: 886-911750616 Email:

Abstract

Cancer, a serious problem throughout the world, needs more efficient drugs for clinical applications. One potential chemotherapeutic drug is β-lapachone. The study of β-lapachone begins from 1977. This commentary is going to discuss the current understanding for the therapeutic uses of β-lapachone

Keywords: β-lapachone; Chemotherapy; Apoptosis; Necroptosis; Anti-tumorL; NQO1

The Introduction of β-lapachone

β-lapachone, (3,4-dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran- 5,6-dione), is a natural extract from lapacho tree (Handroanthus impetiginosus, (Mart. ex DC.) Mattos; Synonym: Tabebuia avellanedae). It has been reported to have many physiological effects, including anti-fungal [1,2], anti-bacterial [3,4], anti-parasitic [5,6], anti-inflammation [7], and anti-cancer [8,9] effects. Among all these effects, the most attention-grabbing effect of β-lapachone is the anticancer effect. β-lapachone has been demonstrated to elicit significant anti-tumor activity against many cancer cells, including leukemia [10,11], prostate cancer [12], osteosarcoma [13], breast cancer [14], bladder cancer [15], lung cancer [16], hepatoma [9], and even the worst pancreatic cancer [17-19]. There are at least three phase I and three phase II clinical trials of β-lapachone, as the name of ARQ 501, on treating patients with various cancers (National Cancer Institute) [20-22].

The in Vitro Effect Of β-lapachone On Tumor Cells

Many experiments showed that β-lapachone undergoes a redox cycle by NAD(P)H:quinine oxidoreductase (NQO1) [23], which reduces β-lapachone to an unstable semiquinone. Semiquinone then rapidly undergoes a two-step oxidation back to the parent stable compound, perpetuating a futile redox cycle (Figure 1). While NQO1 is highly expressed in cancer cells, futile cycle leads to the unbalance of intracellular reactive oxygen species (ROS) [24] and subsequently induces the cell death in cancer cells, and thus be considered as a potential chemotherapeutic drug.

chemotherapy-Futile-cycle

Figure 1: Futile cycle of ß-lapachone. ß-lapachone was reduced to an unstable semiquinone by NQO1. Semiquinone then rapidly undergoes a two-step oxidation back to the parent stable compound, perpetuating a futile redox cycle.

Does β-lapachone really induce apoptosis in cancer cells? Most researchers reported that β-lapachone leads to cell death through apoptosis [25], however a few studies indicated the necrotic signaling [13,26-29] involving in the cell death mediated by β-lapachone. This kind of cell death, combined both apoptotic and necrotic signals, is now called necroptosis [27,29], a programmed form of necrotic cell death which initiates an extensive discussion recently [16,18]. Moreover, β-lapachone is also shown to induce autophagic cell death in glioma cells [30].

β-lapachone triggers cell death with various signaling pathways in different types of cancer cells. Two main hypothesis of how β-lapachone causes cell death are proposed: one is the typical apoptosis and the other is atypical apoptosis, which is the necroptosis mentioned above. According to recent researches, β-lapachone may trigger tumor cells undergoing apoptosis through many pathways. First, the oxidoredox cycle of β-lapachone within cells resulting in the generation of reactive oxygen species (ROS) and superoxide anion [11,31-33]. These reactive oxidative species can oxidize thiol groups of the mitochondrial potential transition pore complex, leading to an increase in permeability of the mitochondrial inner membrane, a reduction in mitochondrial membrane depolarization and the release of cytochrome c, causes the activation of poly ADP ribose polymerase (PARP) and caspase cascade and subsequently induces cell death [9,12,29,34-36] (Figure 2). Second, β-lapachone triggers intracellular calcium influx [37], and a large amount of calcium releases from endoplasmic reticulum (ER) causing ER stress [38] in breast and prostate cancers (Figure 2). Third, β-lapachone produces DNA strand breaks [39], impedes DNA repair [40] through inhibiting topoisomerase I [41], II [42] and PARP [39] (Figure 2). Fourth, β-lapachone leads to cell cycle arrest by activating p21 and p27 and suppressing PI3K and AKT [12], the main signaling molecules of cell proliferation. Recently, JNK activation has also been demonstrated to be involved in β-lapachone-induced apoptosis (Figure 2).

chemotherapy-scheme-lapachone

Figure 2: The scheme showing the effects of ß-lapachone.

In the necroptosis, a large amount of intracellular calcium influx induced by β-lapachone has been observed. The unbalance of intracellular calcium level leads to the activation of calpain, a caspase-independent and neutral Ca2+dependent cysteine protease. Calpain has been shown to be the key executioner in β-lapachone-induced atypical apoptotic cell death in lung, breast and prostate cancers [16,31,34,43-45] (Figure 2).

The Synergistic of β-lapachone on Tumors with Radiotherapy

The most effective application currently for tumors is radiotherapy. Radiotherapy leads to cell death in most cancer cells, but unfortunately not in all cancer cells. Some tumor cells have very low sensitivity to radiotherapy. Radiation strength for cancer patients can be increased, but the normal cells inside human bodies will be damaged as well, especially the immune cells. Researchers tend to dig out new methods to increase the radiosensitivity of cancer cells those can survive well under radiotherapy. From 1987, researchers found that β-lapachone increases sensitivity of cancer cells to radiotherapy in various cancers including laryngeal, colon, breast, lung, and prostate cancers [40,46-48]. While radiation breaks DNA in cancer cell, those cells with low radiosensitivity can repair the DNA breaks with some unclear mechanisms, which may be involved in p53 [49], telomerase [50], and topoisomerase, and escape cell death. Since p53 and telomerase are specific targets for β-lapachone [51-56], β-lapachone enhances the lethality of X-rays and radiomimetic agents in cancer cells by inhibiting potentially lethal DNA damage repair (PLDR) [40,57,58] and radiation-induced neoplastic transformation [59].

Additionally, NQO1 is also shown to be upregulated under radiation in cancer cells. In normal condition, NQO1 is a potent antioxidative enzyme that increases its enzyme activity and expression level to protect cells against the radiation-induced oxidative stress. However, the elevation of NQO1 plays a key role in the redox cycle of β-lapachone and increases the β-lapachone cytotoxic effect [31,46-48,58,60-63]. Thus, the more NQO1 in cells, the higher futile cycle of β-lapachone occurs [23], leading a serious oxidative stress inside cells that triggers cell death in cancer cells under radiation.

Taken together, we conclude that β-lapachone and radiation exert synergistic effects on cancer cells; β-lapachone sensitizes cells to radiation by inhibiting DNA repair, and radiation sensitizes cells to β-lapachone by increasing NQO1 level in tumor cells.

The Synergistic of Drugs with β-lapachone on Tumors

In addition to radiotherapy, chemotherapy is also a key treatment for cancer patients. Searching new drug to raise the survival and life quality of patients is an urgent mission in the clinical use. With the apoptotic or necroptotic effects of β-lapachone, it has the great potential to be a good therapeutic reagent in various cancers.

It is known that NQO1 is the principal determinant of β-lapachone cytotoxicity. Highly expressed NQO1 cells are more sensitive to β-lapachone, cells with low NQO1 level can escape from β-lapachone toxicity. Thus, the main task of increasing β-lapachone sensitivity is to raise the NQO1 level or activity in cancer cells. Recently some chemicals or many physical methods have been demonstrated to upregulate NQO1 level or increase NQO1 activity to enhance the cytotoxic effect of β-lapachone [17,64-67]. In the chemical methods, many drugs including cisplatin [68], resveratrol [69,70], dimethyl fumarate [71], taxifolin [72], sulforaphane [73] etc. are suggested to raise NQO1 level or activity, that may enhance the anticancer effect of β-lapachone. In recent, a Food and Drug Administration (FDA)-approved non-steroidal anti-inflammation drug (NSAID), sulindac [35], is demonstrated by Kung to raise both the NQO1 level and activity, leading to higher sensitivity of cancer cells to β-lapachone cytotoxicity. With long investigation and observation time in clinical use, sulindac and its metabolites are well known about the safe dosage range, disadvantages, and side effects. It can increase the cytotoxic effect and decrease the dosage use of β-lapachone, which makes sulindac become a safer choice for synergistic treatment with β-lapachone.

For the physical methods, hyperthermia [74], heat shock [75,76], and photodynamic therapy (PDT) [77] showed synergistic enhancement of antitumor effect of β-lapachone by induction of NQO1 production. Under proper control of β-lapachone dosage and localization of the tumor, these physical methods can be good for β-lapachone treatment.

Other Targets of β-lapachone

Chemotherapy is a painful process for cancer patients. Although most patients are beneficial from chemotherapy, some are still suffered with the low efficiency or high side effects of chemotherapeutic drugs. One way to rescue those patients is finding the synergistic reagents to not only increase the sensitivity of chemotherapeutic drugs, but also lower the dosage of drugs to decrease the side effects. Except NQO1, there are still other targets of β-lapachone. Paclitaxel [78] or genistein isoflavone [79] combined with β-lapachone respectively are good examples that the toxicities of β-lapachone are significantly increased by targeting molecules other than NQO1. Paclitaxel-lapachone combination kills human retinoblastoma cells by targeting phospho-AKT and increasing nuclear p53 level [78]. Genistein isoflavone and β-lapachone synergistically target cell cycle at various critical checkpoints (G2/M checkpoints for genistein isoflavone; G1/S checkpoints for β-lapachone), that block the cell cycles and cause cell death [79]. In addition to NQO1, the indoleamine 2, 3-dioxygenase 1 (IDO1) enzyme, which has been demonstrated compelling anti-tumor property by modulating tumor immunity [80], is another potential target of β-lapachone. Inhibition of IDO1 has been shown to constrain the immune system from being able to mount an effective anti-tumor response, lower down the resistance to β-lapachone. β-lapachone inhibited the IDO1 activity is totally distinct from its cytotoxic property, and may add a new role of β-lapachone in anti-immunity.

Conclusion

β-lapachone now becomes a popular study object for its promising anti-tumor activity. There will be more targets of β-lapachone found in cancer cells. The more effects of β-lapachone found, the more powerful of β-lapachone. Although normal cells have lower NQO1 and are more insensitive to β-lapachone than cancer cells, the toxicity of β-lapachone toward normal human tissue is still not clear. With more clinical trials, we may find the best dosage range and more clinical uses to increase the efficiency and reduce the side effect of β-lapachone.

References

  1. Gafner S, Wolfender JL, Nianga M, Stoeckli-Evans H, Hostettmann K (1996) Antifungal and antibacterial naphthoquinones from Newbouldia laevis roots. Phytochemistry 42: 1315-1320.
  2. Guiraud P, Steiman R, Campos-Takaki GM, Seigle-Murandi F, Simeon de Buochberg M (1994) Comparison of antibacterial and antifungal activities of lapachol and beta-lapachone. Planta Med 60: 373-374.
  3. Otten S, Rosazza JP (1979) Microbial transformations of natural antitumor agents: conversion of lapachol to dehydro-alpha-lapachone by Curvularia lunata. Appl Environ Microbiol 38: 311-313.
  4. Cruz FS, Docampo R, de Souza W (1978) Effect of beta-lapachone on hydrogen peroxide production in Trypanosoma cruzi. Acta Trop 35: 35-40.
  5. Docampo R, Lopes JN, Cruz FS, Souza W (1977) Trypanosoma cruzi: ultrastructural and metabolic alterations of epimastigotes by beta-lapachone. Exp Parasitol 42: 142-149.
  6. Boveris A, Docampo R, Turrens JF, Stoppani AO (1978) Effect of beta-lapachone on superoxide anion and hydrogen peroxide production in Trypanosoma cruzi. Biochem J 175: 431-439.
  7. Tzeng HP, Ho FM, Chao KF, Kuo ML, Lin-Shiau SY, et al. (2003) beta-Lapachone reduces endotoxin-induced macrophage activation and lung edema and mortality. Am J Respir Crit Care Med 168: 85-91.
  8. Dolan ME, Frydman B, Thompson CB, Diamond AM, Garbiras BJ, et al. (1998) Effects of 1,2-naphthoquinones on human tumor cell growth and lack of cross-resistance with other anticancer agents. Anticancer Drugs 9: 437-448.
  9. Lai CC, Liu TJ, Ho LK, Don MJ, Chau YP (1998) beta-Lapachone induced cell death in human hepatoma (HepA2) cells. Histol Histopathol 13: 89-97.
  10. Shiah SG, Chuang SE, Chau YP, Shen SC and Kuo ML (1999) Activation of c-Jun NH2-terminal kinase and subsequent CPP32/Yama during topoisomerase inhibitor beta-lapachone-induced apoptosis through an oxidation-dependent pathway. Cancer Res 59: 391-398.
  11. Chau YP, Shiah SG, Don MJ, Kuo ML (1998) Involvement of hydrogen peroxide in topoisomerase inhibitor beta-lapachone-induced apoptosis and differentiation in human leukemia cells. Free Radic Biol Med 24: 660-670.
  12. Don MJ, Chang YH, Chen KK, Ho LK, Chau YP (2001) Induction of CDK inhibitors (p21(WAF1) and p27(Kip1)) and Bak in the beta-lapachone-induced apoptosis of human prostate cancer cells. Mol Pharmacol 59: 784-794.
  13. Liu TJ, Lin SY, Chau YP (2002) Inhibition of poly(ADP-ribose) polymerase activation attenuates beta-lapachone-induced necrotic cell death in human osteosarcoma cells. Toxicol Appl Pharmacol 182: 116-125.
  14. Lin MT, Chang CC, Chen ST, Chang HL, Su JL, et al. (2004) Cyr61 expression confers resistance to apoptosis in breast cancer MCF-7 cells by a mechanism of NF-kappaB-dependent XIAP up-regulation. J Biol Chem 279: 24015-24023.
  15. Lee JI, Choi DY, Chung HS, Seo HG, Woo HJ, et al. (2006) beta-lapachone induces growth inhibition and apoptosis in bladder cancer cells by modulation of Bcl-2 family and activation of caspases. Exp Oncol 28: 30-35.
  16. Bey EA, Bentle MS, Reinicke KE, Dong Y, Yang CR, et al. (2007) An NQO1- and PARP-1-mediated cell death pathway induced in non-small-cell lung cancer cells by beta-lapachone. Proc Natl Acad Sci U S A 104: 11832-11837.
  17. Li LS, Bey EA, Dong Y, Meng J, Patra B, et al. (2011) Modulating endogenous NQO1 levels identifies key regulatory mechanisms of action of β-lapachone for pancreatic cancer therapy. Clin Cancer Res 17: 275-285.
  18. Middleton G, Ghaneh P, Costello E, Greenhalf W, Neoptolemos JP (2008) New treatment options for advanced pancreatic cancer. Expert Rev Gastroenterol Hepatol 2: 673-696.
  19. Ough M, Lewis A, Bey EA, Gao J, Ritchie JM, et al. (2005) Efficacy of beta-lapachone in pancreatic cancer treatment: exploiting the novel, therapeutic target NQO1. Cancer Biol Ther 4: 95-102.
  20. Miao XS, Zhong C, Wang Y, Savage RE, Yang RY, et al. (2009) In vitro metabolism of beta-lapachone (ARQ 501) in mammalian hepatocytes and cultured human cells. Rapid Commun Mass Spectrom 23: 12-22.
  21. Yang RY, Kizer D, Wu H, Volckova E, Miao XS, et al. (2008) Synthetic methods for the preparation of ARQ 501 (beta-Lapachone) human blood metabolites. Bioorg Med Chem 16: 5635-5643.
  22. Clinical Trials Search Results: ARQ 501, beta-lapachone (National Cancer Institute).
  23. Reinicke KE, Bey EA, Bentle MS, Pink JJ, Ingalls ST, et al. (2005) Development of beta-lapachone prodrugs for therapy against human cancer cells with elevated NAD(P)H:quinone oxidoreductase 1 levels. Clin Cancer Res 11: 3055-3064.
  24. Docampo R, Cruz FS, Boveris A, Muniz RP, Esquivel DM (1979) beta-Lapachone enhancement of lipid peroxidation and superoxide anion and hydrogen peroxide formation by sarcoma 180 ascites tumor cells. Biochem Pharmacol 28: 723-728.
  25. Li Y, Li CJ, Yu D, Pardee AB (2000) Potent induction of apoptosis by beta-lapachone in human multiple myeloma cell lines and patient cells. Mol Med 6: 1008-1015.
  26. Bey EA, Reinicke KE, Srougi MC, Varnes M, Anderson VE, et al. (2013) Catalase abrogates β-lapachone-induced PARP1 hyperactivation-directed programmed necrosis in NQO1-positive breast cancers. Mol Cancer Ther 12: 2110-2120.
  27. Blanco E, Bey EA, Khemtong C, Yang SG, Setti-Guthi J, et al. (2010) Beta-lapachone micellar nanotherapeutics for non-small cell lung cancer therapy. Cancer Res 70: 3896-3904.
  28. Huang X, Dong Y, Bey EA, Kilgore JA, Bair JS, et al. (2012) An NQO1 substrate with potent antitumor activity that selectively kills by PARP1-induced programmed necrosis. Cancer Res 72: 3038-3047.
  29. Li YZ, Li CJ, Pinto AV, Pardee AB (1999) Release of mitochondrial cytochrome C in both apoptosis and necrosis induced by beta-lapachone in human carcinoma cells. Mol Med 5: 232-239.
  30. Park EJ, Choi KS, Kwon TK (2011) β-Lapachone-induced reactive oxygen species (ROS) generation mediates autophagic cell death in glioma U87 MG cells. Chem Biol Interact 189: 37-44.
  31. Pink JJ, Planchon SM, Tagliarino C, Varnes ME, Siegel D, et al. (2000) NAD(P)H:Quinone oxidoreductase activity is the principal determinant of beta-lapachone cytotoxicity. J Biol Chem 275: 5416-5424.
  32. Planchon SM, Pink JJ, Tagliarino C, Bornmann WG, Varnes ME, et al. (2001) beta-Lapachone-induced apoptosis in human prostate cancer cells: involvement of NQO1/xip3. Exp Cell Res 267: 95-106.
  33. Kim SO, Kwon J, Jeong YK, Kim GY, Kim ND, et al. (2007) Induction of Egr-1 is associated with anti-metastatic and anti-invasive ability of beta-lapachone in human hepatocarcinoma cells. Biosci Biotechnol Biochem 71: 2169-2176.
  34. Lien YC, Kung HN, Lu KS, Jeng CJ, Chau YP (2008) Involvement of endoplasmic reticulum stress and activation of MAP kinases in beta-lapachone-induced human prostate cancer cell apoptosis. Histol Histopathol 23: 1299-1308.
  35. Kung HN, Weng TY2, Liu YL2, Lu KS1, Chau YP3 (2014) Sulindac compounds facilitate the cytotoxicity of β-lapachone by up-regulation of NAD(P)H quinone oxidoreductase in human lung cancer cells. PLoS One 9: e88122.
  36. de Witte NV, Stoppani AO, Dubin M (2004) 2-Phenyl-beta-lapachone can affect mitochondrial function by redox cycling mediated oxidation. Arch Biochem Biophys 432: 129-135.
  37. Tagliarino C, Pink JJ, Dubyak GR, Nieminen AL, Boothman DA (2001) Calcium is a key signaling molecule in beta-lapachone-mediated cell death. J Biol Chem 276: 19150-19159.
  38. Lee H, Park MT, Choi BH, Oh ET, Song MJ, et al. (2011) Endoplasmic reticulum stress-induced JNK activation is a critical event leading to mitochondria-mediated cell death caused by β-lapachone treatment. PLoS One 6: e21533.
  39. Vanni A, Fiore M, De Salvia R, Cundari E, Ricordy R, et al. (1998) DNA damage and cytotoxicity induced by beta-lapachone: relation to poly(ADP-ribose) polymerase inhibition. Mutat Res 401: 55-63.
  40. Boothman DA, Greer S and Pardee AB (1987) Potentiation of halogenated pyrimidine radiosensitizers in human carcinoma cells by beta-lapachone (3,4-dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran- 5,6-dione), a novel DNA repair inhibitor. Cancer Res 47: 5361-5366.
  41. Hueber A, Esser P, Heimann K, Kociok N, Winter S, et al. (1998) The topoisomerase I inhibitors, camptothecin and beta-lapachone, induce apoptosis of human retinal pigment epithelial cells. Exp Eye Res 67: 525-530.
  42. Neder K, Marton LJ, Liu LF, Frydman B (1998) Reaction of beta-lapachone and related naphthoquinones with 2-mercaptoethanol: a biomimetic model of topoisomerase II poisoning by quinones. Cell Mol Biol (Noisy-le-grand) 44: 465-474.
  43. Guicciardi ME, Gores GJ (2003) Calpains can do it alone: implications for cancer therapy. Cancer Biol Ther 2: 153-154.
  44. Pink JJ, Wuerzberger-Davis S, Tagliarino C, Planchon SM, Yang X, et al. (2000) Activation of a cysteine protease in MCF-7 and T47D breast cancer cells during beta-lapachone-mediated apoptosis. Exp Cell Res 255: 144-155.
  45. Tagliarino C, Pink JJ, Reinicke KE, Simmers SM, Wuerzberger-Davis SM, et al. (2003) Mu-calpain activation in beta-lapachone-mediated apoptosis. Cancer Biol Ther 2: 141-152.
  46. Kim EJ, Ji IM, Ahn KJ, Choi EK, Park HJ, et al. (2005) Synergistic effect of ionizing radiation and beta-Lapachone against RKO human colon adenocarcinoma cells. Cancer Res Treat 37: 183-190.
  47. Park MT, Song MJ, Lee H, Oh ET, Choi BH, et al (2011) beta-lapachone significantly increases the effect of ionizing radiation to cause mitochondrial apoptosis via JNK activation in cancer cells. PLoS One 6: e25976.
  48. Ahn KJ, Lee HS, Bai SK, Song CW (2013) Enhancement of radiation effect using beta-lapachone and underlying mechanism. Radiat Oncol J 31: 57-65.
  49. Smith ML, Ford JM, Hollander MC, Bortnick RA, Amundson SA, et al. (2000) p53-mediated DNA repair responses to UV radiation: studies of mouse cells lacking p53, p21, and/or gadd45 genes. Mol Cell Biol 20: 3705-3714.
  50. Wong KK, Chang S, Weiler SR, Ganesan S, Chaudhuri J, et al. (2000) Telomere dysfunction impairs DNA repair and enhances sensitivity to ionizing radiation. Nat Genet 26: 85-88.
  51. Li Y, Huang W, Huang S, Du J, Huang C (2012) Screening of anti-cancer agent using zebrafish: comparison with the MTT assay. Biochem Biophys Res Commun 422: 85-90.
  52. Weller M, Winter S, Schmidt C, Esser P, Fontana A, et al. (1997) 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 73: 707-714.
  53. Choi YH, Kim MJ, Lee SY, Lee YN, Chi GY, et al. (2002) Phosphorylation of p53, induction of Bax and activation of caspases during beta-lapachone-mediated apoptosis in human prostate epithelial cells. Int J Oncol 21: 1293-1299.
  54. Moon DO, Kang CH, Kim MO, Jeon YJ, Lee JD, et al. (2010) Beta-lapachone (LAPA) decreases cell viability and telomerase activity in leukemia cells: suppression of telomerase activity by LAPA. J Med Food 13: 481-488.
  55. Lee JH, Cheong J, Park YM, Choi YH (2005) Down-regulation of cyclooxygenase-2 and telomerase activity by beta-lapachone in human prostate carcinoma cells. Pharmacol Res 51: 553-560.
  56. Woo HJ, Choi YH (2005) Growth inhibition of A549 human lung carcinoma cells by beta-lapachone through induction of apoptosis and inhibition of telomerase activity. Int J Oncol 26: 1017-1023.
  57. Boothman DA, Trask DK, Pardee AB (1989) Inhibition of potentially lethal DNA damage repair in human tumor cells by beta-lapachone, an activator of topoisomerase I. Cancer Res 49: 605-612.
  58. Choi EK, Terai K, Ji IM, Kook YH, Park KH, et al. (2007) Upregulation of NAD(P)H:quinone oxidoreductase by radiation potentiates the effect of bioreductive beta-lapachone on cancer cells. Neoplasia 9: 634-642.
  59. Boothman DA, Pardee AB (1989) Inhibition of radiation-induced neoplastic transformation by beta-lapachone. Proc Natl Acad Sci U S A 86: 4963-4967.
  60. Szumiel I, Buraczewska I, Gradzka I, Gasinska A (1995) Effects of topoisomerase I-targeted drugs on radiation response of L5178Y sublines differentially radiation and drug sensitive. Int J Radiat Biol 67: 441-448.
  61. Miyamoto S, Huang TT, Wuerzberger-Davis S, Bornmann WG, Pink JJ, et al. (2000) Cellular and molecular responses to topoisomerase I poisons. Exploiting synergy for improved radiotherapy. Ann N Y Acad Sci 922: 274-292.
  62. Park HJ, Ahn KJ, Ahn SD, Choi E, Lee SW, et al. (2005) Susceptibility of cancer cells to beta-lapachone is enhanced by ionizing radiation. Int J Radiat Oncol Biol Phys 61: 212-219.
  63. Suzuki M, Amano M, Choi J, Park HJ, Williams BW, et al. (2006) Synergistic effects of radiation and beta-lapachone in DU-145 human prostate cancer cells in vitro. Radiat Res 165: 525-531.
  64. Tan XL, Marquardt G, Massimi AB, Shi M, Han W, et al. (2012) High-throughput library screening identifies two novel NQO1 inducers in human lung cells. Am J Respir Cell Mol Biol 46: 365-371.
  65. Satsu H, Chidachi E, Hiura Y, Ogiwara H, Gondo Y, et al. (2012) Induction of NAD(P)H:quinone oxidoreductase 1 expression by cysteine via Nrf2 activation in human intestinal epithelial LS180 cells. Amino Acids 43: 1547-1555.
  66. Tsai CW, Lin CY, Wang YJ (2011) Carnosic acid induces the NAD(P)H: quinone oxidoreductase 1 expression in rat clone 9 cells through the p38/nuclear factor erythroid-2 related factor 2 pathway. J Nutr 141: 2119-2125.
  67. Bottone FG, Martinez JM, Collins JB, Afshari CA, Eling TE (2003) Gene modulation by the cyclooxygenase inhibitor, sulindac sulfide, in human colorectal carcinoma cells: possible link to apoptosis. J Biol Chem 278: 25790-25801.
  68. Terai K, Dong GZ, Oh ET, Park MT, Gu Y, et al. (2009) Cisplatin enhances the anticancer effect of beta-lapachone by upregulating NQO1. Anticancer Drugs 20: 901-909.
  69. Hsieh TC, Lu X, Wang Z, Wu JM (2006) Induction of quinone reductase NQO1 by resveratrol in human K562 cells involves the antioxidant response element ARE and is accompanied by nuclear translocation of transcription factor Nrf2. Med Chem 2: 275-285.
  70. Li Y, Cao Z and Zhu H (2006) Upregulation of endogenous antioxidants and phase 2 enzymes by the red wine polyphenol, resveratrol in cultured aortic smooth muscle cells leads to cytoprotection against oxidative and electrophilic stress. Pharmacol Res 53: 6-15.
  71. Begleiter A, Leith MK, Thliveris JA, Digby T (2004) Dietary induction of NQO1 increases the antitumour activity of mitomycin C in human colon tumours in vivo. Br J Cancer 91: 1624-1631.
  72. Lee SB, Cha KH, Selenge D, Solongo A, Nho CW (2007) The chemopreventive effect of taxifolin is exerted through ARE-dependent gene regulation. Biol Pharm Bull 30: 1074-1079.
  73. Wagner AE, Ernst I, Iori R, Desel C, Rimbach G (2010) Sulforaphane but not ascorbigen, indole-3-carbinole and ascorbic acid activates the transcription factor Nrf2 and induces phase-2 and antioxidant enzymes in human keratinocytes in culture. Exp Dermatol 19: 137-144.
  74. Hori T, Kondo T, Lee H, Song CW, Park HJ (2011) Hyperthermia enhances the effect of β-lapachone to cause γH2AX formations and cell death in human osteosarcoma cells. Int J Hyperthermia 27: 53-62.
  75. Dong GZ, Youn H, Park MT, Oh ET, Park KH, et al. (2009) 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 25: 477-487.
  76. Park HJ, Choi EK, Choi J, Ahn KJ, Kim EJ, et al. (2005) Heat-induced up-regulation of NAD(P)H:quinone oxidoreductase potentiates anticancer effects of beta-lapachone. Clin Cancer Res 11: 8866-8871.
  77. Lamberti MJ, Vittar NB, da Silva Fde C, Ferreira VF, Rivarola VA (2013) Synergistic enhancement of antitumor effect of β-Lapachone by photodynamic induction of quinone oxidoreductase (NQO1). Phytomedicine 20: 1007-1012.
  78. D'Anneo A, Augello G, Santulli A, Giuliano M, di Fiore R, et al. (2010) Paclitaxel and beta-lapachone synergistically induce apoptosis in human retinoblastoma Y79 cells by downregulating the levels of phospho-Akt. J Cell Physiol 222: 433-443.
  79. Kumi-Diaka J (2002) Chemosensitivity of human prostate cancer cells PC3 and LNCaP to genistein isoflavone and beta-lapachone. Biol Cell 94: 37-44.
  80. Flick HE, Lalonde JM, Malachowski WP, Muller AJ (2013) The Tumor-Selective Cytotoxic Agent β-Lapachone is a Potent Inhibitor of IDO1. Int J Tryptophan Res 6: 35-45.
Citation: Kung HN, Lu KS, Chau YP (2014) The Chemotherapeutic Effects of Lapacho Tree Extract: β-Lapachone. Chemotherapy 3:131.

Copyright: © 2014 Kung HN, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top