Skip to main content
Top
Published in: International Journal of Clinical Oncology 6/2011

01-12-2011 | Original Article

Histone deacetylase inhibitor augments anti-tumor effect of gemcitabine and pegylated interferon-α on pancreatic cancer cells

Authors: Shuichi Iwahashi, Mitsuo Shimada, Tohru Utsunomiya, Yuji Morine, Satoru Imura, Tetsuya Ikemoto, Hiroki Mori, Jun Hanaoka, Koji Sugimoto, Yu Saito

Published in: International Journal of Clinical Oncology | Issue 6/2011

Login to get access

Abstract

Background

Histone deacetylase (HDAC) is strongly associated with epigenetic regulation and carcinogenesis, and its inhibitor can induce the differentiation or apoptosis of cancer cells.

Methods

We investigated the anticancer effects of the HDAC inhibitor valproic acid (VPA) in combination with gemcitabine (GEM), an antimetabolic, and pegylated interferon-α2b (PEG-IFN-α2b) in a human pancreatic cancer cell line using a cell proliferation assay. The gene expressions of HDAC1, MTA1, p21Waf1, and HIF-1 were evaluated by reverse transcription-PCR.

Results

Valproic acid at 0.5 mM when used alone did not suppress cell proliferation. PEG–IFN-α2b at 10E/ml weakly suppressed cell proliferation in both the BxPC3 (by 28%) and SUIT-2 (by 17%) human pancreatic cancer cell lines. GEM at 5 nM when used alone suppressed cell proliferation by 36 and 61% in the BxPC3 and SUIT-2 cell lines, respectively. The combination treatment of GEM + PEG–IFN-α2b strongly suppressed cell proliferation in the SUIT-2 (82%) and BxPC3 (51%) cell lines, which was further reinforced by the addition of VPA up to 88 and 67%, respectively. The combination treatment of GEM + PEG–IFN-α2b enhanced the expression of p21Waf1, which was also reinforced by VPA.

Conclusion

VPA augmented the inhibitory effects of PEG–IFN-α2b alone or in combination with PEG–IFN-α2b and GEM on cell proliferation. Such inhibitory effects may be due to the up-regulation of p21Waf1 expression.
Literature
2.
go back to reference Burris HA III, Moore MJ, Andersen J et al (1997) Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 15:2403–2413PubMed Burris HA III, Moore MJ, Andersen J et al (1997) Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 15:2403–2413PubMed
3.
go back to reference Haberland M, Montgomery RL, Olson EN (2009) The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet 10:32–42PubMedCrossRef Haberland M, Montgomery RL, Olson EN (2009) The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet 10:32–42PubMedCrossRef
4.
go back to reference Minucci S, Pelicci PG (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6:38–51PubMedCrossRef Minucci S, Pelicci PG (2006) Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6:38–51PubMedCrossRef
6.
go back to reference Patra SK, Patra A, Dahiya R (2001) Histone deacetylase and DNA methyltransferase in human prostate cancer. Biochem Biophys Res Commun 287:705–713PubMedCrossRef Patra SK, Patra A, Dahiya R (2001) Histone deacetylase and DNA methyltransferase in human prostate cancer. Biochem Biophys Res Commun 287:705–713PubMedCrossRef
7.
go back to reference Zhu P, Martin E, Mengwasser J et al (2004) Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell 5:455–463PubMedCrossRef Zhu P, Martin E, Mengwasser J et al (2004) Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell 5:455–463PubMedCrossRef
8.
go back to reference Lin RJ, Nagy L, Inoue S et al (1998) Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature 391:811–814PubMedCrossRef Lin RJ, Nagy L, Inoue S et al (1998) Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature 391:811–814PubMedCrossRef
9.
go back to reference Miyake K, Yoshizumi T, Imura S et al (2008) Expression of hypoxia-inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreatic carcinoma: correlation with poor prognosis with possible regulation. Pancreas 36:e1–e9PubMedCrossRef Miyake K, Yoshizumi T, Imura S et al (2008) Expression of hypoxia-inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreatic carcinoma: correlation with poor prognosis with possible regulation. Pancreas 36:e1–e9PubMedCrossRef
10.
go back to reference Bolden JE, Peart MJ, Johnstone RW (2005) Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 5:769–784CrossRef Bolden JE, Peart MJ, Johnstone RW (2005) Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 5:769–784CrossRef
11.
go back to reference Warrell RP Jr, He LZ, Richon V et al (1998) Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J Natl Cancer Inst 90:1621–1625PubMedCrossRef Warrell RP Jr, He LZ, Richon V et al (1998) Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. J Natl Cancer Inst 90:1621–1625PubMedCrossRef
12.
go back to reference Göttlicher M, Minucci S, Zhu P et al (2001) Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J 20:6969–6978PubMedCrossRef Göttlicher M, Minucci S, Zhu P et al (2001) Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J 20:6969–6978PubMedCrossRef
13.
go back to reference Xia Q, Sung J, Chowdhury W et al (2006) Chronic administration of valproic acid inhibits prostate cancer cell growth in vitro and in vivo. Cancer Res 66:7237–7244PubMedCrossRef Xia Q, Sung J, Chowdhury W et al (2006) Chronic administration of valproic acid inhibits prostate cancer cell growth in vitro and in vivo. Cancer Res 66:7237–7244PubMedCrossRef
14.
go back to reference Catalano MG, Fortunati N, Pugliese M et al (2005) Valproic acid induces apoptosis and cell cycle arrest in poorly differentiated thyroid cancer cells. J Clin Endocrinol Metab 90:1383–1389PubMedCrossRef Catalano MG, Fortunati N, Pugliese M et al (2005) Valproic acid induces apoptosis and cell cycle arrest in poorly differentiated thyroid cancer cells. J Clin Endocrinol Metab 90:1383–1389PubMedCrossRef
15.
go back to reference Baron S, Dianzani F (1994) The interferons: a biological system with therapeutic potential in viral infections. Antiviral Res 24:97–110PubMedCrossRef Baron S, Dianzani F (1994) The interferons: a biological system with therapeutic potential in viral infections. Antiviral Res 24:97–110PubMedCrossRef
16.
go back to reference Hertzog PJ, Hwang SY, Kola I (1994) Role of interferons in the regulation of cell proliferation, differentiation and development. Mol Reprod Dev 39:226–232PubMedCrossRef Hertzog PJ, Hwang SY, Kola I (1994) Role of interferons in the regulation of cell proliferation, differentiation and development. Mol Reprod Dev 39:226–232PubMedCrossRef
17.
go back to reference Gutterman JU (1994) Cytokine therapeutics: lessons from interferon-α. Proc Natl Acad Sci USA 91:1198–1205PubMedCrossRef Gutterman JU (1994) Cytokine therapeutics: lessons from interferon-α. Proc Natl Acad Sci USA 91:1198–1205PubMedCrossRef
18.
go back to reference Krown SE (1988) Interferons in malignancy: biological products or biological response modifiers? J Natl Cancer Inst (Bethesda) 80:306–309CrossRef Krown SE (1988) Interferons in malignancy: biological products or biological response modifiers? J Natl Cancer Inst (Bethesda) 80:306–309CrossRef
19.
go back to reference Miyake K, Tsuchida K, Sugino H et al (2007) Combination therapy of human pancreatic cancer implanted in nude mice by oral fluoropyrimidine anticancer agent (S-1) with interferon-alpha. Cancer Chemother Pharmacol 59:113–126PubMedCrossRef Miyake K, Tsuchida K, Sugino H et al (2007) Combination therapy of human pancreatic cancer implanted in nude mice by oral fluoropyrimidine anticancer agent (S-1) with interferon-alpha. Cancer Chemother Pharmacol 59:113–126PubMedCrossRef
20.
go back to reference Baker DE (2003) Pegylated interferon plus ribavirin for the treatment of chronic hepatitis C. Rev Gastroenterol Disord 3:93–109PubMed Baker DE (2003) Pegylated interferon plus ribavirin for the treatment of chronic hepatitis C. Rev Gastroenterol Disord 3:93–109PubMed
21.
go back to reference Yamada T, Furukawa K, Yokoi K et al (2010) Effects of irinotecan and 5-FU combination therapy in gastric cancer—is combination therapy synergic (in Japanese)? Gan To Kagaku Ryoho 37:2125–2129PubMed Yamada T, Furukawa K, Yokoi K et al (2010) Effects of irinotecan and 5-FU combination therapy in gastric cancer—is combination therapy synergic (in Japanese)? Gan To Kagaku Ryoho 37:2125–2129PubMed
22.
go back to reference Yoo YG, Kong G, Lee MO (2006) Metastasis-associated protein 1 enhances stability of hypoxia-inducible factor-1alpha protein by recruiting histone deacetylase 1. EMBO J 25:1231–1241PubMedCrossRef Yoo YG, Kong G, Lee MO (2006) Metastasis-associated protein 1 enhances stability of hypoxia-inducible factor-1alpha protein by recruiting histone deacetylase 1. EMBO J 25:1231–1241PubMedCrossRef
23.
go back to reference Kelly WK, O’Connor OA, Krug LM et al (2005) Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol 23:3923–3931PubMedCrossRef Kelly WK, O’Connor OA, Krug LM et al (2005) Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol 23:3923–3931PubMedCrossRef
24.
go back to reference Fuino L, Bali P, Wittmann S et al (2003) Histone deacetylase inhibitor LAQ824 down-regulates Her-2 and sensitizes human breast cancer cells to trastuzumab, taxotere, gemcitabine, and epothilone B. Mol Cancer Ther 2:971–984PubMed Fuino L, Bali P, Wittmann S et al (2003) Histone deacetylase inhibitor LAQ824 down-regulates Her-2 and sensitizes human breast cancer cells to trastuzumab, taxotere, gemcitabine, and epothilone B. Mol Cancer Ther 2:971–984PubMed
25.
go back to reference Rundall BK, Denlinger CE, Jones DR (2005) Suberoylanilide hydroxamic acid combined with gemcitabine enhances apoptosis in non-small cell lung cancer. Surgery 138:360–367PubMedCrossRef Rundall BK, Denlinger CE, Jones DR (2005) Suberoylanilide hydroxamic acid combined with gemcitabine enhances apoptosis in non-small cell lung cancer. Surgery 138:360–367PubMedCrossRef
26.
go back to reference El-Deiry WS, Tokino T, Velculescu VE et al (1993) WAF1, a potential mediator of p53 tumor suppression. Cell 75:817–825PubMedCrossRef El-Deiry WS, Tokino T, Velculescu VE et al (1993) WAF1, a potential mediator of p53 tumor suppression. Cell 75:817–825PubMedCrossRef
27.
go back to reference El-Deiry WS, Harper JW, O’Connor PM et al (1994) WAF1/CIP1 is induced in p53-mediated G1 arrest and apoptosis. Cancer Res 54:1169–1174PubMed El-Deiry WS, Harper JW, O’Connor PM et al (1994) WAF1/CIP1 is induced in p53-mediated G1 arrest and apoptosis. Cancer Res 54:1169–1174PubMed
28.
go back to reference Deng C, Zhang P, Harper JW et al (1995) Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell 82:675–684PubMedCrossRef Deng C, Zhang P, Harper JW et al (1995) Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell 82:675–684PubMedCrossRef
29.
go back to reference Schwaller J, Koeffler HP, Niklaus G et al (1995) Posttranscriptional stabilization underlies p53-independent induction of p21WAF1/CIP1/SDI1 in differentiating human leukemic cells. J Clin Invest 95:973–979PubMedCrossRef Schwaller J, Koeffler HP, Niklaus G et al (1995) Posttranscriptional stabilization underlies p53-independent induction of p21WAF1/CIP1/SDI1 in differentiating human leukemic cells. J Clin Invest 95:973–979PubMedCrossRef
30.
go back to reference DiGiuseppe JA, Redston MS, Yeo CJ et al (1995) p53-independent expression of the cyclin-dependent kinase inhibitor p21 in pancreatic carcinoma. Am J Pathol 147:884–888PubMed DiGiuseppe JA, Redston MS, Yeo CJ et al (1995) p53-independent expression of the cyclin-dependent kinase inhibitor p21 in pancreatic carcinoma. Am J Pathol 147:884–888PubMed
31.
go back to reference Huang L, Sowa Y, Sakai T et al (2000) Activation of the p21WAF1/CIP1 promoter independent of p53 by the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) through the Sp1 sites. Oncogene 19:5712–5719PubMedCrossRef Huang L, Sowa Y, Sakai T et al (2000) Activation of the p21WAF1/CIP1 promoter independent of p53 by the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) through the Sp1 sites. Oncogene 19:5712–5719PubMedCrossRef
32.
go back to reference Das CM, Aguilera D, Vasquez H et al (2007) Valproic acid induces p21 and topoisomerase-II (alpha/beta) expression and synergistically enhances etoposide cytotoxicity in human glioblastoma cell lines. J Neurooncol 85:159–170PubMedCrossRef Das CM, Aguilera D, Vasquez H et al (2007) Valproic acid induces p21 and topoisomerase-II (alpha/beta) expression and synergistically enhances etoposide cytotoxicity in human glioblastoma cell lines. J Neurooncol 85:159–170PubMedCrossRef
33.
go back to reference Donadelli M, Costanzo C, Faggioli L et al (2003) Trichostatin A, an inhibitor of histone deacetylases, strongly suppresses growth of pancreatic adenocarcinoma cells. Mol Carcinog 38:59–69PubMedCrossRef Donadelli M, Costanzo C, Faggioli L et al (2003) Trichostatin A, an inhibitor of histone deacetylases, strongly suppresses growth of pancreatic adenocarcinoma cells. Mol Carcinog 38:59–69PubMedCrossRef
34.
go back to reference Richon VM, Sandhoff TW, Rifkind RA et al (2000) Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci USA 97:10014–10019PubMedCrossRef Richon VM, Sandhoff TW, Rifkind RA et al (2000) Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci USA 97:10014–10019PubMedCrossRef
35.
go back to reference Nichole B, Nohea A, Jason G et al (2007) The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces growth inhibition and enhances gemcitabine-induced cell death in pancreatic cancer. Clin Cancer Res 13:18–26CrossRef Nichole B, Nohea A, Jason G et al (2007) The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces growth inhibition and enhances gemcitabine-induced cell death in pancreatic cancer. Clin Cancer Res 13:18–26CrossRef
36.
go back to reference Tagliaferri P, Caraglia M, Budillon A et al (2005) New pharmacokinetic and pharmacodynamic tools for interferon-alpha (IFN-alpha) treatment of human cancer. Cancer Immunol Immunother 54:1–10PubMedCrossRef Tagliaferri P, Caraglia M, Budillon A et al (2005) New pharmacokinetic and pharmacodynamic tools for interferon-alpha (IFN-alpha) treatment of human cancer. Cancer Immunol Immunother 54:1–10PubMedCrossRef
37.
go back to reference Toh Y, Pencil SD, Nicolson GL (1994) A novel candidate metastasis-associated gene, mta1, differentially expressed in highly metastatic mammary adenocarcinoma cell lines. cDNA cloning, expression, and protein analyses. J Biol Chem 269:22958–22963PubMed Toh Y, Pencil SD, Nicolson GL (1994) A novel candidate metastasis-associated gene, mta1, differentially expressed in highly metastatic mammary adenocarcinoma cell lines. cDNA cloning, expression, and protein analyses. J Biol Chem 269:22958–22963PubMed
38.
39.
go back to reference Bowen NJ, Fujita N, Kajita M et al (2004) Mi-2/NuRD: multiple complexes for many purposes. Biochim Biophys Acta 1677:52–57PubMed Bowen NJ, Fujita N, Kajita M et al (2004) Mi-2/NuRD: multiple complexes for many purposes. Biochim Biophys Acta 1677:52–57PubMed
40.
go back to reference Picozzi VJ, Kozarek RA, Traverso LW (2003) Interferon based adjuvant chemoradiation therapy after pancreaticoduodenectomy for pancreatic adenocarcinoma. Am J Surg 185:4767–4780CrossRef Picozzi VJ, Kozarek RA, Traverso LW (2003) Interferon based adjuvant chemoradiation therapy after pancreaticoduodenectomy for pancreatic adenocarcinoma. Am J Surg 185:4767–4780CrossRef
41.
go back to reference Solorzano CC, Hwang R, Baker CH et al (2003) Administration of optimal biological dose and schedule of interferon alpha combined with gemcitabine induces apoptosis in tumor-associated endothelial cells and reduces growth of human pancreatic carcinoma implanted orthotopically in nude mice. Clin Cancer Res 9:1858–1867PubMed Solorzano CC, Hwang R, Baker CH et al (2003) Administration of optimal biological dose and schedule of interferon alpha combined with gemcitabine induces apoptosis in tumor-associated endothelial cells and reduces growth of human pancreatic carcinoma implanted orthotopically in nude mice. Clin Cancer Res 9:1858–1867PubMed
42.
go back to reference Perez-Zincer F, Olencki T, Budd GT et al (2002) A phase I trial of weekly gemcitabine and subcutaneous interferon alpha in patients with refractory renal cell carcinoma. Invest New Drugs 20:305–310PubMedCrossRef Perez-Zincer F, Olencki T, Budd GT et al (2002) A phase I trial of weekly gemcitabine and subcutaneous interferon alpha in patients with refractory renal cell carcinoma. Invest New Drugs 20:305–310PubMedCrossRef
43.
go back to reference Fuxius S, Mross K, Mansouri K et al (2002) Gemcitabine and interferon-alpha 2b in solid tumors: a phase I study in patients with advanced or metastatic non-small cell lung, ovarian, pancreatic or renal cancer. Anticancer Drugs 13:899–905PubMedCrossRef Fuxius S, Mross K, Mansouri K et al (2002) Gemcitabine and interferon-alpha 2b in solid tumors: a phase I study in patients with advanced or metastatic non-small cell lung, ovarian, pancreatic or renal cancer. Anticancer Drugs 13:899–905PubMedCrossRef
44.
go back to reference Amato RJ, Khan M (2008) A phase I clinical trial of low-dose interferon-alpha-2A, thalidomide plus gemcitabine and capecitabine for patients with progressive metastatic renal cell carcinoma. Cancer Chemother Pharmacol 61:1069–1073PubMedCrossRef Amato RJ, Khan M (2008) A phase I clinical trial of low-dose interferon-alpha-2A, thalidomide plus gemcitabine and capecitabine for patients with progressive metastatic renal cell carcinoma. Cancer Chemother Pharmacol 61:1069–1073PubMedCrossRef
45.
go back to reference Krepler C, Certa U, Wacheck V et al (2004) Pegylated and conventional interferon-alpha induce comparable transcriptional responses and inhibition of tumor growth in a human melanoma SCID mouse xenotransplantation model. J Invest Dermatol 123:664–669PubMedCrossRef Krepler C, Certa U, Wacheck V et al (2004) Pegylated and conventional interferon-alpha induce comparable transcriptional responses and inhibition of tumor growth in a human melanoma SCID mouse xenotransplantation model. J Invest Dermatol 123:664–669PubMedCrossRef
46.
go back to reference Blaheta RA, Michaelis M, Driever PH et al (2005) Evolving anticancer drug valproic acid: insights into the mechanism and clinical studies. Med Res Rev 25:383–397PubMedCrossRef Blaheta RA, Michaelis M, Driever PH et al (2005) Evolving anticancer drug valproic acid: insights into the mechanism and clinical studies. Med Res Rev 25:383–397PubMedCrossRef
47.
go back to reference Faiella A, Wernig M, Consalez GG et al (2000) A mouse model for valproate teratogenicity: parental effects, homeotic transformations, and altered HOX expression. Hum Mol Genet 9:227–236PubMedCrossRef Faiella A, Wernig M, Consalez GG et al (2000) A mouse model for valproate teratogenicity: parental effects, homeotic transformations, and altered HOX expression. Hum Mol Genet 9:227–236PubMedCrossRef
Metadata
Title
Histone deacetylase inhibitor augments anti-tumor effect of gemcitabine and pegylated interferon-α on pancreatic cancer cells
Authors
Shuichi Iwahashi
Mitsuo Shimada
Tohru Utsunomiya
Yuji Morine
Satoru Imura
Tetsuya Ikemoto
Hiroki Mori
Jun Hanaoka
Koji Sugimoto
Yu Saito
Publication date
01-12-2011
Publisher
Springer Japan
Published in
International Journal of Clinical Oncology / Issue 6/2011
Print ISSN: 1341-9625
Electronic ISSN: 1437-7772
DOI
https://doi.org/10.1007/s10147-011-0246-y

Other articles of this Issue 6/2011

International Journal of Clinical Oncology 6/2011 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