Open Access Open Access  Restricted Access Subscription or Fee Access

Anticancer Effects of Beta-Lapachone

Uma Sharma

Abstract


Beta-lapachone is a naturally occurring ortho-napthoquinone originated from lapacho tree i.e. Tabebuia avelloandae. It is a bioreductive anticancer drug that synergistically interacts with ionizing radiation and sensitivity of cells to beta-lapachone is related to the activity of NAD (P) H: quinone oxidoreductase 1(NQO1). Beta-lapachone shows a variety of pharmacological effects. The combined effects of ionizing radiation and beta-lapachone that increases mitochondrial apoptotic cell death in an NQO1 dependent manner. And the positive feedback regulation between ERK and ROS induced by combined treatment play an important role in the induction of ER stress. ER stress is required for JNK activation, that leads to subsequent mitochondrial apoptotic cell death. In this paper we can read about the anticancer activity of beta-lapachon, and effects of beta-lapchone with other components on cancer cells. Various anticancer activity of beta-lapachone. Beta-lapchone is inhibited the topoisomerasses I and II. NAD (P) H: Quinone oxidoreductase (NQO1) activity is a principle determinant of beta-lapachone. Effect of NQO1 on cancer cells, highly expressed NQO1 cells are more sensitive to beta-lapachone and cells with lower NQO1 level can escape from beta-lapachone toxicity. Beta-lapachone has various chemotherapeutic effects such as, synergistic effects,cytotoxic effects,and neoplastic effects etc. Most researchers said that beta-lapachone leads to cell death through apoptosis but few studied indicate the necrotic signaling involving in cell death mediated by beta-lapachone. This type cell death called necroptosis combined both apoptotic and necrotic signal beta-lapachone also infers the replication of HIV I through the inhibition of beta-lapachone inhibiting DNA. Beta-lapachone cause apoptosis in various tumor cells.

Full Text:

PDF

References


M.-T. Park, M.-J. Song, H. Lee, E.-T. Oh, B.-H. Choi, et al. Beta-lapachone significantly increases the effect of ionizing radiation to cause mitochondrial apoptosis via JNK activation in cancer cells, PLoS One. 2011; 6(10): e25976p. doi: 10.1371/journal.pone.0025976.

J.B. Schulz, D. Bremen, J.C. Reed, J. Lommatzsch, S. Takayama, et al. Cooperative interception of neuronal apoptosis by BCL-2 and BAG1expression: prevention of caspase activation and reduced production of reactive oxygen species, J Neurochem. 1997; 69: 2075–86p.

M.T. Park, M.J. Kim, Y.H. Kang, S.Y. Choi, J.H. Lee, et al. Phytosphingosine in combination with ionizing radiation enhances apoptotic cell death in radiation-resistant cancer cells through ROS-dependent and -independent AIF release, Blood. 2005; 105: 1724–33p.

Y.H. Kang, M.J. Yi, M.J. Kim, M.T. Park, S. Bae, et al. Caspase-independent cell death by arsenic trioxide in human cervical cancer cells: reactive oxygen species-mediated poly(ADP-ribose) polymerase-1 activation signals apoptosis inducing factor release from mitochondria, Cancer Res. 2004; 64: 8960–7p.

V. Goossens, K. De Vos, D. Vercammen, M. Steemans, K. Vancompernolle, et al. Redox regulation of TNF signaling, Biofactors. 1999; 10: 145–56p.

N.S. Chandel, P.T. Schumacker, R.H. Arch. Reactive oxygen species are downstream products of TRAF-mediated signal transduction, J Biol Chem. 2001; 276: 42728–36p.

Y.C. Lien, H.N. Kung, K.S. Lu, C.J. Jeng, Y.P. Chau. Involvement of endoplasmic reticulum stress and activation of MAP kinases in beta-lapachone induced human prostate cancer cell apoptosis, Histol Histopathol. 2008; 23: 1299–1308p.

T.J. Liu, S.Y. Lin, Y.P. Chau. Inhibition of poly (ADP-ribose) polymerase activation attenuates beta-lapachone-induced necrotic celldeath in human osteosarcoma cells, Toxicol Appl Pharmacol. 2002; 182: 116–25p.

E.A. Bey, M.S. Bentle, K.E. Reinicke, Y. Dong, C.R. Yang, et al. 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. 2007; 104: 11832–7p.

G. Middleton, P. Ghaneh, E. Costello, W. Greenhalf, J.P. Neoptolemos. New treatment options for advanced pancreatic cancer, Expert Rev Gastroenterol Hepatol. 2008; 2: 673–96p.

E.A. Bey, K.E. Reinicke, M.C. Srougi, M. Varnes, V.E. Anderson, et al. Catalase abrogates β-lapachone-induced PARP1 hyperactivationdirected programmed necrosis in NQO1-positive breast cancers, Mol Cancer Ther. 2013; 12: 2110–20p.

E. Blanco, E.A. Bey, C. Khemtong, S.G. Yang, J. Setti-Guthi, et al. Beta-lapachone micellar nanotherapeutics for non-small cell lung cancer therapy, Cancer Res. 2010; 70: 3896–904p.

Y.Z. Li, C.J. Li, A.V. Pinto, A.B. Pardee. Release of mitochondrial cytochrome C in both apoptosis and necrosis induced by beta-lapachone in human carcinoma cells, Mol Med. 1999; 5: 232–9p.

E.J. Park, K.S. Choi, T.K. Kwon. β-Lapachone-induced reactive oxygen species (ROS) generation mediates autophagic cell death in glioma U87 MG cells, Chem Biol Interact. 2011; 189: 37–44p.

Y.P. Chau, S.G. Shiah, M.J. Don, M.L. Kuo. Involvement of hydrogen peroxide in topoisomerase inhibitor beta-lapachone-induced apoptosis and differentiation in human leukemia cells, Free Radic Biol Med. 1998; 24: 660–70p.

J.J. Pink, S.M. Planchon, C. Tagliarino, M.E. Varnes, D. Siegel, et al. NAD(P)H:Quinone oxidoreductase activity is the principal determinant of beta-lapachone cytotoxicity, J Biol Chem. 2000; 275: 5416–24p.

S.M. Planchon, J.J. Pink, C. Tagliarino, W.G. Bornmann, M.E. Varnes, et al. Beta-Lapachone-induced apoptosis in human prostate cancer cells: involvement of NQO1/xip3, Exp Cell Res. 2001; 267: 95–106p.

C.C. Lai, T.J. Liu, L.K. Ho, M.J. Don, Y.P. Chau. beta-Lapachone induced cell death in human hepatoma (HepA2) cells, Histol Histopathol. 1998; 13: 89–97p.

M.J. Don, Y.H. Chang, K.K. Chen, L.K. Ho, Y.P. Chau. Induction of CDK inhibitors (p21(WAF1) and p27(Kip1)) and Bak in the betalapachone- induced apoptosis of human prostate cancer cells, Mol Pharmacol. 2001; 59: 784–94p.

Y.C. Lien, H.N. Kung, K.S. Lu, C.J. Jeng, Y.P. Chau. Involvement of endoplasmic reticulum stress and activation of MAP kinases in betalapachone- induced human prostate cancer cell apoptosis, Histol Histopathol. 2008; 23: 1299–308p.

H.N. Kung, T.Y. Weng, Y.L. Liu, K.S. Lu, Y.P. Chau. Sulindac compounds facilitate the cytotoxicity of β-lapachone by up-regulation of NAD(P)H quinone oxidoreductase in human lung cancer cells, PLoS One. 2014; 9: e88122.

N.V. De Witte, A.O. Stoppani, M. Dubin. 2-Phenyl-beta-lapachone can affect mitochondrial function by redox cycling mediated oxidation, Arch Biochem Biophys. 2004; 432: 129–35p.

C. Tagliarino, J.J. Pink, G.R. Dubyak, A.L. Nieminen, D.A. Boothman. Calcium is a key signaling molecule in beta-lapachone-mediated cell death, J Biol Chem. 2001; 276: 19150–9p.

H. Lee, M.T. Park, B.H. Choi, E.T. Oh, M.J. Song, et al. Endoplasmic reticulum stress-induced JNK activation is a critical event leading to mitochondria-mediated cell death caused by β-lapachone treatment, PLoS One. 2011; 6: e21533p.

D.A. Boothman, S. Greer, A.B. Pardee. 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. 1987; 47: 5361–6p.

M.L. Smith, J.M. Ford, M.C. Hollander, R.A. Bortnick, S.A. Amundson, et al. p53-mediated DNA repair responses to UV radiation: studies of mouse cells lacking p53, p21, and/or gadd45 genes, Mol Cell Biol. 2000; 20: 3705–14p.

K.K. Wong, S. Chang, S.R. Weiler, S. Ganesan, J. Chaudhuri, et al. Telomere dysfunction impairs DNA repair and enhances sensitivity to ionizing radiation, Nat Genet. 2000; 26: 85–8p.

D.A. Boothman, D.K. Trask, A.K. Pardee. Inhibition of potentially lethal DNA damage repair in human tumor cells by beta-lapachone, an activator of topoisomerase I, Cancer Res. 1989; 49: 605–12p.

E.K. Choi, K. Terai, I.M. Ji, Y.H. Kook, K.H. Park, et al. Upregulation of NAD(P)H:quinone oxidoreductase by radiation potentiates the effect of bioreductive beta-lapachone on cancer cells, Neoplasia. 2007; 9: 634–42p.

L.S. Li, E.A. Bey, Y. Dong, J. Meng, B. Patra, et al. Modulating endogenous NQO1 levels identifies key regulatory mechanisms of action of β-lapachone for pancreatic cancer therapy, Clin Cancer Res. 2011; 17: 275–85p.

X.L. Tan, G. Marquardt, A.B. Massimi, M. Shi, W. Han, et al. Highthroughput library screening identifies two novel NQO1 inducers in human lung cells, Am J Respir Cell Mol Biol. 2012; 46: 365–71p.

H. Satsu, E. Chidachi, Y. Hiura, H. Ogiwara, Y. Gondo, et al. Induction of NAD(P)H:quinone oxidoreductase 1 expression by cysteine via Nrf2 activation in human intestinal epithelial LS180 cells, Amino Acids. 2012; 43: 1547–55p.

CW Tsai, C.Y. Lin, Y.J. Wang. 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. 2011; 141: 2119–25p.

F.G. Bottone, J.M. Martinez, J.B. Collins, C.A. Afshari, T.E. Eling. Gene modulation by the cyclooxygenase inhibitor, sulindac sulfide, in human colorectal carcinoma cells: possible link to apoptosis, J Biol Chem. 2003; 278: 25790–801p.

K. Terai, G.Z. Dong, E.T. Oh, M.T. Park, Y. Gu, et al. Cisplatin enhances the anticancer effect of beta-lapachone by upregulating NQO1, Anticancer Drugs. 2009; 20: 901–9p.

S.B. Lee, K.H. Cha, D. Selenge, A. Solongo, C.W. Nho. The chemopreventive effect of taxifolin is exerted through ARE-dependent gene regulation, Biol Pharm Bull. 2007; 30: 1074–9p.

A.E. Wagner, I. Ernst, R. Iori, C. Desel, G. Rimbach. 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. 2010; 19: 137–44p.

T. Hori, T. Kondo, H. Lee, C.W. Song, H.J. Park. Hyperthermia enhances the effect of β-lapachone to cause γH2AX formations and cell death in human osteosarcoma cells, Int J Hyperther. 2011; 27: 53–62p.

G.Z. Dong, Youn H, Park MT, Oh ET, Park KH, et al. (2009) Heat shock increases expression of NAD(P)H:quinone oxidoreductase (NQO1), 1

H.J. Park, E.K. Choi, J. Choi, K.J. Ahn, E.J. Kim, et al. Heat-induced upregulation of NAD(P)H:quinone oxidoreductase potentiates anticancer effects of beta-lapachone, Clin Cancer Res. 11: 8866–71p.

M.J. Lamberti, N.B. Vittar, C. da Silva Fde, V.F. Ferreira, V.A. Rivarola. Synergistic enhancement of antitumor effect of β-Lapachone by photodynamic induction of quinone oxidoreductase (NQO1). Phytomedicine. 2013; 20: 1007–12p.

P. Tugwell, D. Ludwin, M. Gent, R. Roberts, W. Bensen, et al. Interaction between cyclosporin A and nonsteroidal antiinflammatory drugs, J Rheumatol. 1997; 24: 1122–5p.

P. Tugwell, D. Ludwin, M. Gent, R. Roberts, W. Bensen, et al. Interaction between cyclosporin A and nonsteroidal antiinflammatory drugs, J Rheumatol. 1997; 24: 1122–5p.

H.L. Muncie, Jr. Medical aspects of the multidisciplinary assessment and management of osteoarthritis, Clin Ther. 1986; 9 Suppl B: 4–13p.

Fernandes E, Toste SA, Lima JL, Reis S (2003) The metabolism of sulindac enhances its scavenging activity against reactive oxygen and nitrogen species. Free Radic Biol Med 35: 1008–1017. doi: 10.1016/s0891-5849(03)00437-4

G.N. Karachalios, G. Donas. Sulindac in the treatment of acute gout arthritis, Int J Tissue React. 1982; 4: 297–9p.

D.K. Klassen, R.L. Stout, P.S. Spilman, A. Whelton. Sulindac kinetics and effects on renal function and prostaglandin excretion in renal insufficiency, J Clin Pharmacol. 1989; 29: 1037–42p. doi: 10.1002/j.1552-4604. 1989.tb03275.x.

M.J. Thun, S.J. Henley, C. Patrono. Nonsteroidal anti-inflammatory drugs as anticancer agents: mechanistic, pharmacologic, and clinical issues, J Natl Cancer Inst. 2002; 94: 252–66p. doi: 10.1093/jnci/94.4.252.

C. Ruegg, J. Zaric, R. Stupp. Nonsteroidal anti-inflammatory drugs and COX-2 inhibitors as anti-cancer therapeutics: hypes, hopes and reality, Ann Med. 2003; 35: 476–87p. doi: 10.1080/07853890310017053.

Hollander, P. M., and Ernster, L. (1975) Arch. Biochem. Biophys.169. 560–567

S. Hosoda, W. Nakamura, K. Hayashi. J Biol Chem. 1974; 249: 6416–23p.

J.J. Pink, S.M. Planchon, C. Tagliarino, M.E. Varnes, D. Siegeis, D.A. Boothman. NAD(P) H:Quinone oxidoreductase activity is the principle determinant of beta–lapachone cytotoxicity.

J.I. Lee, D.Y. Choi, H.S. Chung, H.G. Seo, H.J. Woo, B.T. Choi, Y.H. Choi. Beta-lapachone induces growth inhibition and apoptosis in bladder cancer cells by modulation of Bcl-2 family and activation of caspases, Exp Oncol. 2006a; 28: 30–5p.

H.Y. Yu, S.O. Kim, C.-Y. Jin, W.-J. Kim, Y.H. Yoo, Y.H. Choi. Beta–lapachone-induce apoptosis of Huma gastric carcinoma AGS cells is caspase-dependent and Regulate by the P13K/AKT Pathway.

Inhibition of radiation –induce neoplastic transformation by beta-lapachone. Proc.Natl.Acad Sci.U S A 1987 Jul;86(13) :4963-7.

A. D'Anneo, G. Augello, A. Santulli, M. Giuliano, R. di Fiore, et al. Paclitaxel and beta-lapachone synergistically induce apoptosis in human retinoblastoma Y79 cells by downregulating the levels of phospho-Akt, J Cell Physiol. 2010; 222: 433–43p.

J. Kumi-Diaka. Chemosensitivity of human prostate cancer cells PC3 and LNCaP to genistein isoflavone and beta-lapachone, Biol Cell. 2002; 94: 37–44p.

H.N. Kung, K.S. Lu, Y.P. Chau The chemotherapeutic effects of lapacho tree extract: beta-lapachone, Chemotherapy. 2014; 3: 131p.

P. Krishnan, K.F. Bastow. Noval mechanism of topoisomerase II inhibition by pyranonphthoquinone derivatives–eleutherin, alpha–lapachone, beta-lapachone.

A. Carnero, C. Blanco-Aparicio, O. Renner, W. Link, J.F. Leal. The PTEN/PI3K/AKT signalling pathway in cancer, therapeutic implications, Curr Cancer Drug Targets. 2008; 8: 187–98p. J.J. Champoux. In: DNA Topology and Its Biological Effects. Wang J.C., Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 1990.

J.C. Wang. Annu Reu Biochem. 1985; 54: 665–97p; Mediator of beta-lapachone cytotoxicity, by increasing NQO1 gene activity and via Hsp70-mediated stabilisation of NQO1 protein, Int J Hyperther. 25: 477–87p.

K.M. Tewev, G.L. Chem, E.M. Nelson, L.F. Liu, J Biol C m. 1984; 259: 9182–7p.

K.M. Tewev, T.C. Rowe, L. Yang, B.D. Halligan, L.F. Lin. Science. 1984; 226: 466–8p.

Y. Pommier, J.K. Minford, R.E. Schwartz, L.A. Zwelling, K.W. Khon. Biochemistry. 1985; 24: 6410–6p.

G.L. Chem, L. Yang, T.C. Rowe, B.D. Halligan, K.M. Tewey, L.F. Liu. J Biol Chem. 1985; 259: 13560–6p.




DOI: https://doi.org/10.37628/ijmb.v3i2.225

Refbacks

  • There are currently no refbacks.