Skip to main content
Top
Published in: Cellular Oncology 2/2014

01-04-2014 | Original Paper

Combined arsenic trioxide-cisplatin treatment enhances apoptosis in oral squamous cell carcinoma cells

Authors: Toshiki Nakaoka, Akinobu Ota, Takayuki Ono, Sivasundaram Karnan, Hiroyuki Konishi, Akifumi Furuhashi, Yukinobu Ohmura, Yoichi Yamada, Yoshitaka Hosokawa, Yoshiaki Kazaoka

Published in: Cellular Oncology | Issue 2/2014

Login to get access

Abstract

Background

Oral squamous cell carcinoma (OSCC) accounts for the majority of oral cancers. Despite recent advances in OSCC diagnostics and therapeutics, the overall survival rate still remains low. Here, we assessed the efficacy of a combinatorial arsenic trioxide (ATO) and cisplatin (CDDP) treatment in human OSCC cells.

Methods

The combinatorial effect of ATO/CDDP on the growth and apoptosis of OSCC cell lines HSC-2, HSC-3, and HSC-4 was evaluated using MTT and annexin V assays, respectively. Chou–Talalay analyses were preformed to evaluate the combinatorial effects of ATO/CDDP on the dose-reduction index (DRI). To clarify the mechanism underlying the ATO/CDDP anticancer effect, we also examined the involvement of reactive oxygen species (ROS) in ATO/CDDP-induced apoptosis.

Results

Combination index (CI) analyses revealed that a synergistic interaction of ATO and CDDP elicits a wide range of effects in HSC-2 cells, with CI values ranging from 0.78 to 0.90, where CI < 1 defines synergism. The CI values in HSC-3 and HSC-4 cells ranged from 0.34 to 0.45 and from 0.60 to 0.92, respectively. In addition, ATO/CDDP yielded favorable DRI values ranging from 1.6-fold to 7.71-fold dose reduction. Compared to mono-therapy, ATO/CDDP combinatorial therapy significantly augmented the loss of mitochondrial potential, caspase-3/7 activity and subsequent apoptosis. These changes were all abrogated by the antioxidant N-acetylcysteine.

Conclusions

This study provides the first evidence for a synergistic ATO/CDDP anticancer (apoptotic) activity in OSCC cells with a favorable DRI, thereby highlighting its potential as a combinational therapeutic regime in OSCC.
Appendix
Available only for authorised users
Literature
1.
go back to reference D.M. Parkin, F. Bray, J. Ferlay, P. Pisani, Global cancer statistics. 2002. CA Cancer J. Clin. 55, 74–108 (2005)PubMedCrossRef D.M. Parkin, F. Bray, J. Ferlay, P. Pisani, Global cancer statistics. 2002. CA Cancer J. Clin. 55, 74–108 (2005)PubMedCrossRef
2.
go back to reference J. Xhu, Z. Chen, V. Lallemand-Breitenbach, H. de The, How acute promyelocytic leukaemia revived arsenic. Nat. Rev. Cancer 2, 705–713 (2002)CrossRef J. Xhu, Z. Chen, V. Lallemand-Breitenbach, H. de The, How acute promyelocytic leukaemia revived arsenic. Nat. Rev. Cancer 2, 705–713 (2002)CrossRef
3.
go back to reference Z.Y. Wang, Z. Chen, Acute promyelocytic leukemia: from highly fatal to highly curable. Blood 111, 2505–2515 (2008)PubMedCrossRef Z.Y. Wang, Z. Chen, Acute promyelocytic leukemia: from highly fatal to highly curable. Blood 111, 2505–2515 (2008)PubMedCrossRef
4.
go back to reference X.W. Zhang, X.J. Yan, Z.R. Zhou, F.F. Yang, Z.Y. Wu, H.B. Sun, W.X. Liang, A.X. Song, V. Lallemand-Breitenbach, M. Jeanne, Q.Y. Zhang, H.Y. Yang, Q.H. Huang, G.B. Zhou, J.H. Tong, Y. Zhang, J.H. Wu, H.Y. Hu, H. de The, S.J. Chen, Z. Chen, Arsenic trioxide controls the fate of the PML-RARalpha oncoprotein by directly binding PML. Science 328, 240–243 (2010)PubMedCrossRef X.W. Zhang, X.J. Yan, Z.R. Zhou, F.F. Yang, Z.Y. Wu, H.B. Sun, W.X. Liang, A.X. Song, V. Lallemand-Breitenbach, M. Jeanne, Q.Y. Zhang, H.Y. Yang, Q.H. Huang, G.B. Zhou, J.H. Tong, Y. Zhang, J.H. Wu, H.Y. Hu, H. de The, S.J. Chen, Z. Chen, Arsenic trioxide controls the fate of the PML-RARalpha oncoprotein by directly binding PML. Science 328, 240–243 (2010)PubMedCrossRef
5.
go back to reference V. Lallemand-Breitenbach, M. Jeanne, S. Benhenda, R. Nasr, M. Lei, L. Peres, J. Zhou, J. Zhu, B. Raught, H. de The, Arsenic degrades PML or PML-RARalpha through a SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat. Cell Biol. 10, 547–555 (2008)PubMedCrossRef V. Lallemand-Breitenbach, M. Jeanne, S. Benhenda, R. Nasr, M. Lei, L. Peres, J. Zhou, J. Zhu, B. Raught, H. de The, Arsenic degrades PML or PML-RARalpha through a SUMO-triggered RNF4/ubiquitin-mediated pathway. Nat. Cell Biol. 10, 547–555 (2008)PubMedCrossRef
6.
go back to reference M.H. Tatham, M.C. Geoffroy, L. Shen, A. Plechanovova, N. Hattersley, E.G. Jaffray, J.J. Palvimo, R.T. Hay, RNF4 is a poly-SUMO-specific E3 ubiquitin ligase required for arsenic-induced PML degradation. Nat. Cell Biol. 10, 538–546 (2008)PubMedCrossRef M.H. Tatham, M.C. Geoffroy, L. Shen, A. Plechanovova, N. Hattersley, E.G. Jaffray, J.J. Palvimo, R.T. Hay, RNF4 is a poly-SUMO-specific E3 ubiquitin ligase required for arsenic-induced PML degradation. Nat. Cell Biol. 10, 538–546 (2008)PubMedCrossRef
7.
go back to reference D. Doucer, M.S. Tallman, Arsenic trioxide: new clinical experience with an old medication in hematologic malignancies. J. Clin. Oncol. 23, 2396–2410 (2005)CrossRef D. Doucer, M.S. Tallman, Arsenic trioxide: new clinical experience with an old medication in hematologic malignancies. J. Clin. Oncol. 23, 2396–2410 (2005)CrossRef
8.
go back to reference M.R. Litzow, S. Lee, J.M. Bennett, G.W. Dewald, R.E. Gallagher, V. Jain, E.M. Paietta, J. Racevskis, S.R. Rousey, J.J. Mazza, M.S. Tallman, A phase II trial of arsenic trioxide for relapsed and refractory acute lymphoblastic leukemia. Haematologica 91, 1105–1108 (2006)PubMed M.R. Litzow, S. Lee, J.M. Bennett, G.W. Dewald, R.E. Gallagher, V. Jain, E.M. Paietta, J. Racevskis, S.R. Rousey, J.J. Mazza, M.S. Tallman, A phase II trial of arsenic trioxide for relapsed and refractory acute lymphoblastic leukemia. Haematologica 91, 1105–1108 (2006)PubMed
9.
go back to reference J.H. Park, E.J. Kim, H.Y. Jang, H. Shim, K.K. Lee, H.J. Jo, H.J. Kim, S.H. Yang, E.T. Jeong, H.R. Kim, Combination treatment with arsenic trioxide and sulindac enhances apoptotic cell death in lung cancer cells via activation of oxidative stress and mitogen-activated protein kinases. Oncol. Rep. 20, 379–384 (2008)PubMed J.H. Park, E.J. Kim, H.Y. Jang, H. Shim, K.K. Lee, H.J. Jo, H.J. Kim, S.H. Yang, E.T. Jeong, H.R. Kim, Combination treatment with arsenic trioxide and sulindac enhances apoptotic cell death in lung cancer cells via activation of oxidative stress and mitogen-activated protein kinases. Oncol. Rep. 20, 379–384 (2008)PubMed
10.
go back to reference X. Wang, G. Wang, D. Dong, S. Fu, B. Yang, Inhibition on LS-174T cell growth and activity of telomerase in vitro and in vivo by arsenic trioxide. Exp. Toxicol. Pathol. 60, 481–488 (2008)PubMedCrossRef X. Wang, G. Wang, D. Dong, S. Fu, B. Yang, Inhibition on LS-174T cell growth and activity of telomerase in vitro and in vivo by arsenic trioxide. Exp. Toxicol. Pathol. 60, 481–488 (2008)PubMedCrossRef
11.
go back to reference B. Liu, S. Pan, X. Dong, H. Qiao, H. Jiang, G.W. Krissansen, X. Sun, Opposing effects of arsenic trioxide on hepatocellular carcinoma in mice. Cancer Sci. 97, 675–681 (2006)PubMedCrossRef B. Liu, S. Pan, X. Dong, H. Qiao, H. Jiang, G.W. Krissansen, X. Sun, Opposing effects of arsenic trioxide on hepatocellular carcinoma in mice. Cancer Sci. 97, 675–681 (2006)PubMedCrossRef
12.
go back to reference S.Y. Woo, M.Y. Lee, Y.J. Jung, E.S. Yoo, J.Y. Seoh, H.Y. Shin, H.S. Ahn, K.H. Ryu, Arsenic trioxide inhibits cell growth in SH-SY5Y and SK-N-AS neuroblastoma cell lines by a different mechanism. Pediatr. Hematol. Oncol. 23, 231–243 (2006)PubMedCrossRef S.Y. Woo, M.Y. Lee, Y.J. Jung, E.S. Yoo, J.Y. Seoh, H.Y. Shin, H.S. Ahn, K.H. Ryu, Arsenic trioxide inhibits cell growth in SH-SY5Y and SK-N-AS neuroblastoma cell lines by a different mechanism. Pediatr. Hematol. Oncol. 23, 231–243 (2006)PubMedCrossRef
13.
go back to reference P. Kumar, Q. Gao, Y. Ning, Z. Wang, P.H. Krebsbach, P.J. Polverini, Arsenic trioxide enhances the therapeutic efficacy of radiation treatment of oral squamous carcinoma while protecting bone. Mol. Cancer Ther. 7, 2060–2069 (2008)PubMedCrossRef P. Kumar, Q. Gao, Y. Ning, Z. Wang, P.H. Krebsbach, P.J. Polverini, Arsenic trioxide enhances the therapeutic efficacy of radiation treatment of oral squamous carcinoma while protecting bone. Mol. Cancer Ther. 7, 2060–2069 (2008)PubMedCrossRef
14.
go back to reference N. Zhang, Z.M. Wu, E. McGowan, J. Shi, Z.B. Hong, C.W. Ding, P. Xia, W. Di, Arsenic trioxide and cisplatin synergism increase cytotoxicity in human ovarian cancer cells: therapeutic potential for ovarian cancer. Cancer Sci. 100, 2459–2464 (2009)PubMedCrossRef N. Zhang, Z.M. Wu, E. McGowan, J. Shi, Z.B. Hong, C.W. Ding, P. Xia, W. Di, Arsenic trioxide and cisplatin synergism increase cytotoxicity in human ovarian cancer cells: therapeutic potential for ovarian cancer. Cancer Sci. 100, 2459–2464 (2009)PubMedCrossRef
15.
go back to reference C. Nie, C. Tian, L. Zhao, P.X. Petit, M. Mehrpour, Q. Chen, Cysteine 62 of Bax is critical for its conformational activation and its proapoptotic activity in response to H2O2-induced apoptosis. J. Biol. Chem. 283, 15359–15369 (2008)PubMedCentralPubMedCrossRef C. Nie, C. Tian, L. Zhao, P.X. Petit, M. Mehrpour, Q. Chen, Cysteine 62 of Bax is critical for its conformational activation and its proapoptotic activity in response to H2O2-induced apoptosis. J. Biol. Chem. 283, 15359–15369 (2008)PubMedCentralPubMedCrossRef
16.
go back to reference A. Ota, M. Yamamoto, T. Hori, S. Miyai, Y. Naishiro, H. Sohma, M. Maeda, Y. Kokai, Upregulation of plasma CCL8 in mouse model of graft-vs-host disease. Exp. Hematol. 37, 525–531 (2009)PubMedCrossRef A. Ota, M. Yamamoto, T. Hori, S. Miyai, Y. Naishiro, H. Sohma, M. Maeda, Y. Kokai, Upregulation of plasma CCL8 in mouse model of graft-vs-host disease. Exp. Hematol. 37, 525–531 (2009)PubMedCrossRef
17.
go back to reference T.C. Chou, P. Talalay, Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv. Enzym. Regul. 22, 27–55 (1984)CrossRef T.C. Chou, P. Talalay, Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv. Enzym. Regul. 22, 27–55 (1984)CrossRef
18.
go back to reference T.C. Chou, Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol. Rev. 58, 621–681 (2006)PubMedCrossRef T.C. Chou, Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol. Rev. 58, 621–681 (2006)PubMedCrossRef
19.
go back to reference L. Zhang, K. Wang, F. Zhao, W. Hu, J. Chen, G.M. Lanza, B. Shen, B. Zhang, Near infrared imaging of EGFR of oral squamous cell carcinoma in mice administered arsenic trioxide. PLoS One 7, e46255 (2012)PubMedCentralPubMedCrossRef L. Zhang, K. Wang, F. Zhao, W. Hu, J. Chen, G.M. Lanza, B. Shen, B. Zhang, Near infrared imaging of EGFR of oral squamous cell carcinoma in mice administered arsenic trioxide. PLoS One 7, e46255 (2012)PubMedCentralPubMedCrossRef
20.
go back to reference C.W. Tsai, N.W. Chang, R.Y. Tsai, R.F. Wang, C.M. Hsu, S.S. Lin, C.N. Wu, S.S. Sun, M.H. Tsai, D.T. Bau, Synergistic cytotoxic effects of arsenic trioxide plus dithiothreitol on mice oral cancer cells. Anticancer Res. 30, 3655–3660 (2010)PubMed C.W. Tsai, N.W. Chang, R.Y. Tsai, R.F. Wang, C.M. Hsu, S.S. Lin, C.N. Wu, S.S. Sun, M.H. Tsai, D.T. Bau, Synergistic cytotoxic effects of arsenic trioxide plus dithiothreitol on mice oral cancer cells. Anticancer Res. 30, 3655–3660 (2010)PubMed
21.
go back to reference Y. Jing, J. Dai, R.M. Chalmers-Redman, W.G. Tatton, S. Waxman, Arsenic trioxide selectively induces acute promyelocytic leukemia cell apoptosis via a hydrogen peroxide-dependent pathway. Blood 94, 2102–2111 (1999)PubMed Y. Jing, J. Dai, R.M. Chalmers-Redman, W.G. Tatton, S. Waxman, Arsenic trioxide selectively induces acute promyelocytic leukemia cell apoptosis via a hydrogen peroxide-dependent pathway. Blood 94, 2102–2111 (1999)PubMed
22.
go back to reference S.H. Kang, J.H. Song, H.K. Kang, S.J. Kim, H.W. Kang, Y.K. Lee, D.B. Park, Arsenic trioxide-induced apoptosis is independent of stress-responsive signaling pathways but sensitive to inhibition of inducible nitric oxide synthase in HepG2 cells. Exp. Mol. Med. 35, 83–90 (2003)PubMedCrossRef S.H. Kang, J.H. Song, H.K. Kang, S.J. Kim, H.W. Kang, Y.K. Lee, D.B. Park, Arsenic trioxide-induced apoptosis is independent of stress-responsive signaling pathways but sensitive to inhibition of inducible nitric oxide synthase in HepG2 cells. Exp. Mol. Med. 35, 83–90 (2003)PubMedCrossRef
23.
go back to reference M. Okamura, K. Hashimoto, J. Shimada, H. Sakagami, Apoptosis-inducing activity of cisplatin (CDDP) against human hepatoma and oral squamous cell carcinoma cell lines. Anticancer Res. 24, 655–661 (2004)PubMed M. Okamura, K. Hashimoto, J. Shimada, H. Sakagami, Apoptosis-inducing activity of cisplatin (CDDP) against human hepatoma and oral squamous cell carcinoma cell lines. Anticancer Res. 24, 655–661 (2004)PubMed
24.
go back to reference Y. Jing, J. Dai, R.M. Chalmers-Redman, W.G. Tatton, S. Waxman, Arsenic trioxide selectively induces acute promyelocytic leukemia cell apoptosis via a hydrogen peroxide-dependent pathway. Blood 94, 2102–2111 (1999)PubMed Y. Jing, J. Dai, R.M. Chalmers-Redman, W.G. Tatton, S. Waxman, Arsenic trioxide selectively induces acute promyelocytic leukemia cell apoptosis via a hydrogen peroxide-dependent pathway. Blood 94, 2102–2111 (1999)PubMed
25.
go back to reference E. Brown, C.G. Yedjou, P.B. Tchounwou, Cytotoxicity and oxidative stress in human liver carcinoma cells exposed to arsenic trioxide (HepG(2)). Met. Ions Biol. Med. 10, 583–587 (2008)PubMedCentralPubMed E. Brown, C.G. Yedjou, P.B. Tchounwou, Cytotoxicity and oxidative stress in human liver carcinoma cells exposed to arsenic trioxide (HepG(2)). Met. Ions Biol. Med. 10, 583–587 (2008)PubMedCentralPubMed
26.
go back to reference S. Urba, G. Wolf, A. Eisbruch, F. Worden, J. Lee, C. Bradford, T. Teknos, D. Chepeha, M. Prince, N. Hogikyan, J. Taylor, Single-cycle induction chemotherapy selects patients with advanced laryngeal cancer for combined chemoradiation: a new treatment paradigm. J. Clin. Oncol. 24, 593–598 (2006)PubMedCrossRef S. Urba, G. Wolf, A. Eisbruch, F. Worden, J. Lee, C. Bradford, T. Teknos, D. Chepeha, M. Prince, N. Hogikyan, J. Taylor, Single-cycle induction chemotherapy selects patients with advanced laryngeal cancer for combined chemoradiation: a new treatment paradigm. J. Clin. Oncol. 24, 593–598 (2006)PubMedCrossRef
27.
go back to reference C. Andreadis, K. Vahtsevanos, T. Sidiras, I. Thomaidis, K. Antoniadis, D. Mouratidou, 5-Fluorouracil and cisplatin in the treatment of advanced oral cancer. Oral Oncol. 39, 380–385 (2003)PubMedCrossRef C. Andreadis, K. Vahtsevanos, T. Sidiras, I. Thomaidis, K. Antoniadis, D. Mouratidou, 5-Fluorouracil and cisplatin in the treatment of advanced oral cancer. Oral Oncol. 39, 380–385 (2003)PubMedCrossRef
28.
go back to reference E. Fox, B.I. Razzouk, B.C. Widemann, S. Xiao, M. O’Brien, W. Goodspeed, G.H. Reaman, S.M. Blaney, A.J. Murgo, F.M. Balis, P.C. Adamson, Phase 1 trial and pharmacokinetic study of arsenic trioxide in children and adolescents with refractory or relapsed acute leukemia, including acute promyelocytic leukemia or lymphoma. Blood 111, 566–573 (2008)PubMedCentralPubMedCrossRef E. Fox, B.I. Razzouk, B.C. Widemann, S. Xiao, M. O’Brien, W. Goodspeed, G.H. Reaman, S.M. Blaney, A.J. Murgo, F.M. Balis, P.C. Adamson, Phase 1 trial and pharmacokinetic study of arsenic trioxide in children and adolescents with refractory or relapsed acute leukemia, including acute promyelocytic leukemia or lymphoma. Blood 111, 566–573 (2008)PubMedCentralPubMedCrossRef
29.
go back to reference U. Kotowski, G. Heiduschka, M. Brunner, B.M. Erovic, H. Martinek, D. Thurnher, Arsenic trioxide enhances the cytotoxic effect of cisplatin in head and neck squamous cell carcinoma cell lines. Oncol. Lett. 3, 1326–1330 (2012)PubMedCentralPubMed U. Kotowski, G. Heiduschka, M. Brunner, B.M. Erovic, H. Martinek, D. Thurnher, Arsenic trioxide enhances the cytotoxic effect of cisplatin in head and neck squamous cell carcinoma cell lines. Oncol. Lett. 3, 1326–1330 (2012)PubMedCentralPubMed
30.
go back to reference K.Y. Kim, H.J. Cho, S.N. Yu, S.H. Kim, H.S. Yu, Y.M. Park, N. Mirkheshti, S.Y. Kim, C.S. Song, B. Chatterjee, S.C. Ahn, Interplay of reactive oxygen species, intracellular Ca(2+) surge and loss of mitochondrial homeostasis in the apoptotic ablation of prostate cancer cells by deoxypodophyllotoxin. J. Cell. Biochem. 114, 1124–1134 (2013)PubMedCrossRef K.Y. Kim, H.J. Cho, S.N. Yu, S.H. Kim, H.S. Yu, Y.M. Park, N. Mirkheshti, S.Y. Kim, C.S. Song, B. Chatterjee, S.C. Ahn, Interplay of reactive oxygen species, intracellular Ca(2+) surge and loss of mitochondrial homeostasis in the apoptotic ablation of prostate cancer cells by deoxypodophyllotoxin. J. Cell. Biochem. 114, 1124–1134 (2013)PubMedCrossRef
31.
go back to reference B.D. Bowling, N. Doudican, P. Manga, S.J. Orlow, Inhibition of mitochondrial protein translation sensitizes melanoma cells to arsenic trioxide cytotoxicity via a reactive oxygen species dependent mechanism. Cancer Chemother. Pharmacol. 63, 37–43 (2008)PubMedCentralPubMedCrossRef B.D. Bowling, N. Doudican, P. Manga, S.J. Orlow, Inhibition of mitochondrial protein translation sensitizes melanoma cells to arsenic trioxide cytotoxicity via a reactive oxygen species dependent mechanism. Cancer Chemother. Pharmacol. 63, 37–43 (2008)PubMedCentralPubMedCrossRef
32.
go back to reference I. Jutooru, G. Chadalapaka, S. Sreevalsan, P. Lei, R. Barhoumi, R. Burghardt, S. Safe, Arsenic trioxide downregulates specificity protein (Sp) transcription factors and inhibits bladder cancer cell and tumor growth. Exp. Cell Res. 316, 2174–2188 (2010)PubMedCentralPubMedCrossRef I. Jutooru, G. Chadalapaka, S. Sreevalsan, P. Lei, R. Barhoumi, R. Burghardt, S. Safe, Arsenic trioxide downregulates specificity protein (Sp) transcription factors and inhibits bladder cancer cell and tumor growth. Exp. Cell Res. 316, 2174–2188 (2010)PubMedCentralPubMedCrossRef
Metadata
Title
Combined arsenic trioxide-cisplatin treatment enhances apoptosis in oral squamous cell carcinoma cells
Authors
Toshiki Nakaoka
Akinobu Ota
Takayuki Ono
Sivasundaram Karnan
Hiroyuki Konishi
Akifumi Furuhashi
Yukinobu Ohmura
Yoichi Yamada
Yoshitaka Hosokawa
Yoshiaki Kazaoka
Publication date
01-04-2014
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 2/2014
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-014-0167-7

Other articles of this Issue 2/2014

Cellular Oncology 2/2014 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine