Skip to main content
Top
Published in: Annals of Surgical Oncology 5/2007

01-05-2007

Effect of Combined Therapy With Low-Dose 5-Aza-2′-Deoxycytidine and Irinotecan on Colon Cancer Cell Line HCT-15

Authors: Megumi Ishiguro, MD, Satoru Iida, MD, PhD, Hiroyuki Uetake, MD, PhD, Shinji Morita, MD, Hiroshi Makino, MD, Keiji Kato, MD, Yoko Takagi, Masayuki Enomoto, MD, PhD, Kenichi Sugihara, MD, PhD

Published in: Annals of Surgical Oncology | Issue 5/2007

Login to get access

Abstract

Background

Aberrant promoter hypermethylation is an epigenetic change that silences the expression of crucial genes, resulting in inactivation of the apoptotic pathway in various cancers. This hypermethylation can be restored by the demethylating agent 5-aza-2′-deoxycytidine (DAC). DAC might increase the tumor sensitivity to chemotherapy through demethylation and restoration of gene expression. We investigated the effect of combined therapy with DAC and irinotecan (CPT-11) on the human colon cancer cell line HCT-15.

Methods

Human colon cancer cell line HCT-15 was treated with DAC and/or CPT-11 both in vitro and in vivo. The changes in mRNA expression of several apoptosis-related genes were investigated by reverse transcriptase–polymerase chain reaction (PCR). Promoter methylation was detected by methylation-specific PCR and combined bisulfite restriction analysis. Suppression of tumor growth was observed during the treatment with DAC and/or CPT-11 and apoptosis in the tumors was investigated by TUNEL (terminal deoxynucleotidyl transferase dUTP nick-end labeling) assay.

Results

Promoter methylation of p14 ARF , p16 INK4a , BNIP3, and XAF1 was confirmed, and DAC restored mRNA expression of these genes. Demethylation and restoration of gene expression was observed with low-dose DAC, and demethylation status was sustained for several weeks. Combined therapy with DAC and CPT-11 produced marked suppression in tumor growth compared with DAC or CPT-11 alone, both in vitro and in vivo.

Conclusions

Pretreatment with low-dose DAC may have the potential to be used as a “biosensitizer” of DNA-damaging agents such as CPT-11 when the apoptotic pathway is inactivated as a result of aberrant promoter methylation in the cancer.
Literature
1.
go back to reference Weitz J, Koch M, Debus J, Höhler T, Galle PR, Büchler MW. Colorectal cancer. Lancet 2005; 365:153–65PubMedCrossRef Weitz J, Koch M, Debus J, Höhler T, Galle PR, Büchler MW. Colorectal cancer. Lancet 2005; 365:153–65PubMedCrossRef
2.
go back to reference Kawato Y, Aonuma M, Hirota Y, Kuga H, Sato K. Intracellular roles of SN-38, a metabolite of the camptothecin derivative CPT-11, in the antitumor effect of CPT-11. Cancer Res 1991; 51:4187–91PubMed Kawato Y, Aonuma M, Hirota Y, Kuga H, Sato K. Intracellular roles of SN-38, a metabolite of the camptothecin derivative CPT-11, in the antitumor effect of CPT-11. Cancer Res 1991; 51:4187–91PubMed
3.
go back to reference Hsiang YH, Lihou MG, Liu LF. arrest of replication forks by drug-stabilized topoisomerase I–DNA cleavable complexes as a mechanism of cell killing by camptothecin. Cancer Res 1989; 49:5077–82PubMed Hsiang YH, Lihou MG, Liu LF. arrest of replication forks by drug-stabilized topoisomerase I–DNA cleavable complexes as a mechanism of cell killing by camptothecin. Cancer Res 1989; 49:5077–82PubMed
4.
go back to reference Douillard JY, Cunningham D, Roth AD, et al. Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicentre randomized trial. Lancet 2000; 355:1041–7PubMedCrossRef Douillard JY, Cunningham D, Roth AD, et al. Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicentre randomized trial. Lancet 2000; 355:1041–7PubMedCrossRef
5.
go back to reference Rougier P, Bougat R, Douillard JY, et al. Phase II study of irinotecan in the treatment of advanced colorectal cancer in chemotherapy-naïve patients and patients pretreated with fluorouracil-based chemotherapy. J Clin Oncol 1997; 15:251–60PubMed Rougier P, Bougat R, Douillard JY, et al. Phase II study of irinotecan in the treatment of advanced colorectal cancer in chemotherapy-naïve patients and patients pretreated with fluorouracil-based chemotherapy. J Clin Oncol 1997; 15:251–60PubMed
6.
go back to reference Nishimura G, Satou T, Yoshimitsu Y, et al. Effect of chemotherapy using irinotecan (CPT-11) against recurrent colorectal cancer. Gan To Kagaku Ryoho 1995; 22:93–7PubMed Nishimura G, Satou T, Yoshimitsu Y, et al. Effect of chemotherapy using irinotecan (CPT-11) against recurrent colorectal cancer. Gan To Kagaku Ryoho 1995; 22:93–7PubMed
7.
go back to reference Tournigand C, André T, Achille E, et al. FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: a randomized GERCOR study. J Clin Oncol 2004; 22:229–37PubMedCrossRef Tournigand C, André T, Achille E, et al. FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: a randomized GERCOR study. J Clin Oncol 2004; 22:229–37PubMedCrossRef
8.
go back to reference Saltz LB, Cox JV, Blanke C, et al. Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. N Engl J Med 2000; 343:905–14PubMedCrossRef Saltz LB, Cox JV, Blanke C, et al. Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. N Engl J Med 2000; 343:905–14PubMedCrossRef
9.
go back to reference Ichikawa W, Uetake H, Shirota Y, et al. Combination of dihydropyrimidine dehydrogenase and thymidilate synthase gene expressions in primary tumors as predictive parameters for the efficacy of fluoropyrimidine-based chemotherapy for metastatic colorectal cancer. Clin Cancer Res 2003; 9:786–91PubMed Ichikawa W, Uetake H, Shirota Y, et al. Combination of dihydropyrimidine dehydrogenase and thymidilate synthase gene expressions in primary tumors as predictive parameters for the efficacy of fluoropyrimidine-based chemotherapy for metastatic colorectal cancer. Clin Cancer Res 2003; 9:786–91PubMed
10.
go back to reference Teodoridis JM, Strathdee G, Brown R. Epigenetic silencing mediated by CpG island methylation: potential as a therapeutic target and as a biomarker. Drug Resist Updat 2004; 7:267–78PubMedCrossRef Teodoridis JM, Strathdee G, Brown R. Epigenetic silencing mediated by CpG island methylation: potential as a therapeutic target and as a biomarker. Drug Resist Updat 2004; 7:267–78PubMedCrossRef
11.
go back to reference Paz MF, Fraga MF, Avila S, et al. A systematic profile of DNA methylation in human cancer cell lines. Cancer Res 2003; 63:1114–21PubMed Paz MF, Fraga MF, Avila S, et al. A systematic profile of DNA methylation in human cancer cell lines. Cancer Res 2003; 63:1114–21PubMed
12.
go back to reference Widschwendter M, Jones PA. The potential prognostic, predictive, and therapeutic values of DNA methylation in cancer. Clin Cancer Res 2002; 8:17–21PubMed Widschwendter M, Jones PA. The potential prognostic, predictive, and therapeutic values of DNA methylation in cancer. Clin Cancer Res 2002; 8:17–21PubMed
13.
go back to reference Goffin J, Eisenhauer E. DNA methyltransferase inhibitors: state of the art. Ann Oncol 2002; 13:1699–716PubMedCrossRef Goffin J, Eisenhauer E. DNA methyltransferase inhibitors: state of the art. Ann Oncol 2002; 13:1699–716PubMedCrossRef
14.
go back to reference Momparler RL. Molecular, cellular and animal pharmacology of 5-aza-2′-deoxycytidine. Pharmacol Ther 1985; 30:287–99PubMedCrossRef Momparler RL. Molecular, cellular and animal pharmacology of 5-aza-2′-deoxycytidine. Pharmacol Ther 1985; 30:287–99PubMedCrossRef
15.
go back to reference Jones PA, Taylor SM. Cellular differentiation, cytidine analogs, and DNA methylation. Cell 1980; 20:85–93PubMedCrossRef Jones PA, Taylor SM. Cellular differentiation, cytidine analogs, and DNA methylation. Cell 1980; 20:85–93PubMedCrossRef
16.
go back to reference Kuo ML, Duncavage EJ, Mathew R, et al. Arf induces p53-dependent and -independent antiproliferative genes. Cancer Res 2003; 63:1046–53PubMed Kuo ML, Duncavage EJ, Mathew R, et al. Arf induces p53-dependent and -independent antiproliferative genes. Cancer Res 2003; 63:1046–53PubMed
17.
go back to reference Normand G, Hemmati PG, Verdoodt B, et al. p14ARF induced g2 cell cycle arrest in p53- and p21-deficient cells by down-regulating p34cdc2 kinase activity. J Biol Chem 2005; 280:7118–30PubMedCrossRef Normand G, Hemmati PG, Verdoodt B, et al. p14ARF induced g2 cell cycle arrest in p53- and p21-deficient cells by down-regulating p34cdc2 kinase activity. J Biol Chem 2005; 280:7118–30PubMedCrossRef
18.
go back to reference Arnold CN, Goel A, Boland CR. Role of hMLH1 promoter hypermethylation in drug resistance to 5-fluorouracil in colorectal cancer cell lines. Int J Cancer 2003; 106:66–73PubMedCrossRef Arnold CN, Goel A, Boland CR. Role of hMLH1 promoter hypermethylation in drug resistance to 5-fluorouracil in colorectal cancer cell lines. Int J Cancer 2003; 106:66–73PubMedCrossRef
19.
go back to reference Plumb JA, Strahdee G, Sludden J, Kaye SB, Brown R. Reversal of drug resistance in human tumor xenografts by 2′-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter. Cancer Res 2000; 60:6039–44PubMed Plumb JA, Strahdee G, Sludden J, Kaye SB, Brown R. Reversal of drug resistance in human tumor xenografts by 2′-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter. Cancer Res 2000; 60:6039–44PubMed
20.
go back to reference Lavelle D, DeSimone J, Hankewych M, Kousnetzova T, Chen YH. Decitabine induces cell cycle arrest at G1 phase via p21WAF1 and G2/M phase via p38 MAP kinase pathway. Leuk Res 2003; 27:999–1007PubMedCrossRef Lavelle D, DeSimone J, Hankewych M, Kousnetzova T, Chen YH. Decitabine induces cell cycle arrest at G1 phase via p21WAF1 and G2/M phase via p38 MAP kinase pathway. Leuk Res 2003; 27:999–1007PubMedCrossRef
21.
go back to reference Yoshikawa H, Nagashima M, Khan MA, McMenamin MG, Hagiwara K, Harris CC. Mutation analysis of p73 and p53 in human cancer cell lines. Oncogene 1999; 18:3415–21PubMedCrossRef Yoshikawa H, Nagashima M, Khan MA, McMenamin MG, Hagiwara K, Harris CC. Mutation analysis of p73 and p53 in human cancer cell lines. Oncogene 1999; 18:3415–21PubMedCrossRef
22.
go back to reference Hagiwara K, McMenamin MG, Miura K, Harris CC. Mutation analysis of the p63/p73L/p51/p40/CUSP/KET gene in human cancer cell lines using intronic primers. Cancer Res 1999; 59:4165–9PubMed Hagiwara K, McMenamin MG, Miura K, Harris CC. Mutation analysis of the p63/p73L/p51/p40/CUSP/KET gene in human cancer cell lines using intronic primers. Cancer Res 1999; 59:4165–9PubMed
23.
go back to reference Aparicio A, Eads CA, Leong LA, et al. Phase I trial of continuous infusion 5-aza-2′-deoxycitidine. Cancer Chemother Pharmacol 2003; 51:231–9PubMed Aparicio A, Eads CA, Leong LA, et al. Phase I trial of continuous infusion 5-aza-2′-deoxycitidine. Cancer Chemother Pharmacol 2003; 51:231–9PubMed
24.
go back to reference Aoyagi Y, Kobunai T, Utsugi T, Wierzba K, Yamada Y. Establishment and characterization of 6-[[2-(dimethylamino)ethyl]amino]-3-hydroxy-7H-indeno[2,1-c]quinolin-7-one dihydrochloride (TAS-103)-resistant cell lines. Jpn J Cancer Res 2000; 91:543–50PubMed Aoyagi Y, Kobunai T, Utsugi T, Wierzba K, Yamada Y. Establishment and characterization of 6-[[2-(dimethylamino)ethyl]amino]-3-hydroxy-7H-indeno[2,1-c]quinolin-7-one dihydrochloride (TAS-103)-resistant cell lines. Jpn J Cancer Res 2000; 91:543–50PubMed
25.
go back to reference Ueno M, Nonaka S, Yamazaki R, Deguchi N, Murai M. SN-38 induces cell cycle arrest and apoptosis in human testicular cancer. Eur Urol 2002; 42:390–7PubMedCrossRef Ueno M, Nonaka S, Yamazaki R, Deguchi N, Murai M. SN-38 induces cell cycle arrest and apoptosis in human testicular cancer. Eur Urol 2002; 42:390–7PubMedCrossRef
26.
go back to reference Uetake H, Ichikawa W, Takechi T, Fukushima M, Nihei Z, Sugihara K. Relationship between intratumoral dihydropyrimidine dehydrogenase activity and gene expression in human colorectal cancer. Clin Cancer Res 1999; 5:2836–9PubMed Uetake H, Ichikawa W, Takechi T, Fukushima M, Nihei Z, Sugihara K. Relationship between intratumoral dihydropyrimidine dehydrogenase activity and gene expression in human colorectal cancer. Clin Cancer Res 1999; 5:2836–9PubMed
27.
go back to reference Bassett MH, Suzuki T, Sasano H, et al. The orphan nuclear receptor NGFIB regulates transcription of 3β-hydroxysteroid dehydrogenase. J Biol Chem 2004; 279:37622–30PubMedCrossRef Bassett MH, Suzuki T, Sasano H, et al. The orphan nuclear receptor NGFIB regulates transcription of 3β-hydroxysteroid dehydrogenase. J Biol Chem 2004; 279:37622–30PubMedCrossRef
28.
go back to reference Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 1996; 93:9821–6PubMedCrossRef Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 1996; 93:9821–6PubMedCrossRef
29.
go back to reference Iida S, Akiyama Y, Nakajima T, et al. Alterations and hypermethylation of the p14(ARF) gene in gastric cancer. Int J Cancer 2000; 87:654–8PubMedCrossRef Iida S, Akiyama Y, Nakajima T, et al. Alterations and hypermethylation of the p14(ARF) gene in gastric cancer. Int J Cancer 2000; 87:654–8PubMedCrossRef
30.
go back to reference Frommer M, McDonald LE, Millar DS, et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci U S A 1992; 89:1827–31PubMedCrossRef Frommer M, McDonald LE, Millar DS, et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc Natl Acad Sci U S A 1992; 89:1827–31PubMedCrossRef
31.
go back to reference Xiong Z, Laird PW. COBRA: a sensitive and quantitative DNA methylation assay. Nucleic Acids Res 1997;25:2532–4PubMedCrossRef Xiong Z, Laird PW. COBRA: a sensitive and quantitative DNA methylation assay. Nucleic Acids Res 1997;25:2532–4PubMedCrossRef
32.
go back to reference Petak I, Danam RP, Tillman DM, et al. Hypermethylation of the gene promoter and enhancer region can regulate Fas expression and sensitivity in colon carcinoma. Cell Death Differ 2003; 10:211–7PubMedCrossRef Petak I, Danam RP, Tillman DM, et al. Hypermethylation of the gene promoter and enhancer region can regulate Fas expression and sensitivity in colon carcinoma. Cell Death Differ 2003; 10:211–7PubMedCrossRef
33.
go back to reference Hemmati PG, Gillissen B, von Haefen C, et al. Adenovirus-mediated overexpression of p14ARF induces p53 and Bax-independent apoptosis. Oncogene 2002; 21:3149–61PubMedCrossRef Hemmati PG, Gillissen B, von Haefen C, et al. Adenovirus-mediated overexpression of p14ARF induces p53 and Bax-independent apoptosis. Oncogene 2002; 21:3149–61PubMedCrossRef
34.
go back to reference Chen G, Ray R, Dubik D, et al. The E1B 19K/bcl-2-binding protein Nip3 is a dimeric mitochondrial protein that activates apoptosis. J Exp Med 1997; 186:1975–83PubMedCrossRef Chen G, Ray R, Dubik D, et al. The E1B 19K/bcl-2-binding protein Nip3 is a dimeric mitochondrial protein that activates apoptosis. J Exp Med 1997; 186:1975–83PubMedCrossRef
35.
go back to reference Okami J, Simeone DM, Logsdon CD. Silencing of the hypoxia-inducible cell death protein BNIP3 in pancreatic cancer. Cancer Res 2004; 64:5338–46PubMedCrossRef Okami J, Simeone DM, Logsdon CD. Silencing of the hypoxia-inducible cell death protein BNIP3 in pancreatic cancer. Cancer Res 2004; 64:5338–46PubMedCrossRef
36.
go back to reference Vande Velde C, Cizeau J, Dubik D, et al. BNIP3 and genetic control of necrosis-like cell death through the mitochondrial permeability transition pore. Mol Cell Biol 2000; 20:5454–68PubMedCrossRef Vande Velde C, Cizeau J, Dubik D, et al. BNIP3 and genetic control of necrosis-like cell death through the mitochondrial permeability transition pore. Mol Cell Biol 2000; 20:5454–68PubMedCrossRef
37.
go back to reference Fong WG, Liston P, Rajcan-Separovic E, Jean MS, Craig C, Korneluk RG. Expression and genetic analysis of XIAP-associated factor 1 (XAF1) in cancer cell lines. Genomics 2000; 70:113–22PubMedCrossRef Fong WG, Liston P, Rajcan-Separovic E, Jean MS, Craig C, Korneluk RG. Expression and genetic analysis of XIAP-associated factor 1 (XAF1) in cancer cell lines. Genomics 2000; 70:113–22PubMedCrossRef
38.
go back to reference Ma TL, Ni PH, Zhohg J, Tan JH, Qiao MM, Jiang SH. Low expression of XIAP-associated factor 1 in human colorectal cancers. Chinese Journal of Digestive Diseases 2005; 6:10–4PubMedCrossRef Ma TL, Ni PH, Zhohg J, Tan JH, Qiao MM, Jiang SH. Low expression of XIAP-associated factor 1 in human colorectal cancers. Chinese Journal of Digestive Diseases 2005; 6:10–4PubMedCrossRef
39.
go back to reference Byun DS, Cho K, Ryu BK, et al. Hypermethylation of XIAP-associated factor 1, a putative tumor suppressor gene from the 17p13.2 locus, in human gastric adenocarcinomas. Cancer Res 2003; 63:7068–75PubMed Byun DS, Cho K, Ryu BK, et al. Hypermethylation of XIAP-associated factor 1, a putative tumor suppressor gene from the 17p13.2 locus, in human gastric adenocarcinomas. Cancer Res 2003; 63:7068–75PubMed
40.
go back to reference Violette S, Poulain L, Dussaulx E, et al. Resistance of colon cancer cells to long-term 5-fluorouracil exposure is correlated to the relative level of bcl-2 and bcl-XL in addition to bax and p53 status. Int J Cancer 2002; 98:498–504PubMedCrossRef Violette S, Poulain L, Dussaulx E, et al. Resistance of colon cancer cells to long-term 5-fluorouracil exposure is correlated to the relative level of bcl-2 and bcl-XL in addition to bax and p53 status. Int J Cancer 2002; 98:498–504PubMedCrossRef
41.
go back to reference Tan KB, Mattern MR, Eng WK, McCabe FL, Johnson RK. Nonproductive rearrangement of DNA Topoisomerase I and II Genes: correlation with resistance to topoisomerase inhibitors. J Natl Cancer Inst 1989; 81:1732–5PubMedCrossRef Tan KB, Mattern MR, Eng WK, McCabe FL, Johnson RK. Nonproductive rearrangement of DNA Topoisomerase I and II Genes: correlation with resistance to topoisomerase inhibitors. J Natl Cancer Inst 1989; 81:1732–5PubMedCrossRef
42.
go back to reference Rasheed ZA, Rubin EH. Mechanisms of resistance to topoisomerase I–targeting drugs. Oncogene 2003; 22:7296–304PubMedCrossRef Rasheed ZA, Rubin EH. Mechanisms of resistance to topoisomerase I–targeting drugs. Oncogene 2003; 22:7296–304PubMedCrossRef
43.
go back to reference Bras-Goncalves RA, Rosty C, Laurent-Puig P, Soulie P, Dutrillaux B, Poupon MF. Sensitivity to CPT-11 of xenografted human colorectal cancers as a function of microsatellite instability and p53 status. Br J Cancer 2000; 82:913–23PubMedCrossRef Bras-Goncalves RA, Rosty C, Laurent-Puig P, Soulie P, Dutrillaux B, Poupon MF. Sensitivity to CPT-11 of xenografted human colorectal cancers as a function of microsatellite instability and p53 status. Br J Cancer 2000; 82:913–23PubMedCrossRef
44.
go back to reference Issa JJ, Garcia-Manero G, Giles FJ, et al. Phase I study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies. Blood 2004; 103:1635–40PubMedCrossRef Issa JJ, Garcia-Manero G, Giles FJ, et al. Phase I study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies. Blood 2004; 103:1635–40PubMedCrossRef
45.
go back to reference Wijermans P, Lubbert M, Verhoef G, et al. Low-dose 5-aza-2′-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase II study in elderly patients. J Clin Oncol 2000; 18:956–62PubMed Wijermans P, Lubbert M, Verhoef G, et al. Low-dose 5-aza-2′-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase II study in elderly patients. J Clin Oncol 2000; 18:956–62PubMed
46.
go back to reference Samlowski WE, Leachman SA, Wade M, et al. Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. J Clin Oncol 2005; 23:3897–905PubMedCrossRef Samlowski WE, Leachman SA, Wade M, et al. Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. J Clin Oncol 2005; 23:3897–905PubMedCrossRef
47.
go back to reference Aparicio A, Weber JS. Review of the clinical experience with 5-azacytidine and 5-aza-2′-deoxycytidine in solid tumors. Curr Opin Invest Drugs 2002; 3:627–33 Aparicio A, Weber JS. Review of the clinical experience with 5-azacytidine and 5-aza-2′-deoxycytidine in solid tumors. Curr Opin Invest Drugs 2002; 3:627–33
48.
go back to reference Anzai H, Frost P, Abbruzzese JL. Synergistic cytotoxicity with 2′-deoxy-5-azacytidine and topotecan in vitro and in vivo. Cancer Res 1992; 52:2180–5PubMed Anzai H, Frost P, Abbruzzese JL. Synergistic cytotoxicity with 2′-deoxy-5-azacytidine and topotecan in vitro and in vivo. Cancer Res 1992; 52:2180–5PubMed
49.
go back to reference Pohlmann P, DiLeone LP, Cancella AI, et al. Phase II trial of cisplatin plus decitabine, a new DNA hypomethylating agent, in patients with advanced squamous cell carcinoma of the cervix. Am J Clin Oncol 2002; 25:496–501PubMedCrossRef Pohlmann P, DiLeone LP, Cancella AI, et al. Phase II trial of cisplatin plus decitabine, a new DNA hypomethylating agent, in patients with advanced squamous cell carcinoma of the cervix. Am J Clin Oncol 2002; 25:496–501PubMedCrossRef
Metadata
Title
Effect of Combined Therapy With Low-Dose 5-Aza-2′-Deoxycytidine and Irinotecan on Colon Cancer Cell Line HCT-15
Authors
Megumi Ishiguro, MD
Satoru Iida, MD, PhD
Hiroyuki Uetake, MD, PhD
Shinji Morita, MD
Hiroshi Makino, MD
Keiji Kato, MD
Yoko Takagi
Masayuki Enomoto, MD, PhD
Kenichi Sugihara, MD, PhD
Publication date
01-05-2007
Publisher
Springer-Verlag
Published in
Annals of Surgical Oncology / Issue 5/2007
Print ISSN: 1068-9265
Electronic ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-006-9285-4

Other articles of this Issue 5/2007

Annals of Surgical Oncology 5/2007 Go to the issue