Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007;128:683–692.
Lin HY, Chen CS, Lin SP, et al. Targeting histone deacetylase in cancer therapy. Med Res Rev 2006;26:397–413.
Conley BA, Wright JJ, Kummar S. Targeting epigenetic abnormalities with histone deacetylase inhibitors. Cancer 2006;107:832–840.
Marks P, Rifkind RA, Richon VM, et al. Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer 2001;1:194–202.
Cress WD, Seto E. Histone deacetylases, transcriptional control, and cancer. J Cell Physiol 2000;184:1–16.
Marks PA, Breslow R. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol 2007;25:84–90.
Marks PA. Discovery and development of SAHA as an anticancer agent. Oncogene 2007;26:1351–1356.
Lu Q, Yang YT, Chen CS, et al. Zn2+-chelating motif-tethered short-chain fatty acids as a novel class of histone deacetylase inhibitors. J Med Chem 2004;47:467–474.
Lu Q, Wang DS, Chen CS, et al. Structure-based optimization of phenylbutyrate-derived histone deacetylase inhibitors. J Med Chem 2005;48:5530–5535.
Kulp SK, Chen CS, Wang DS, et al. Antitumor effects of a novel phenylbutyrate-based histone deacetylase inhibitor, (S)-HDAC42, in prostate cancer. Clin Cancer Res 2006;12:5199–5206.
Lu YS, Kashida Y, Kulp SK, et al. Efficacy of OSU-HDAC42, a novel histone deacetylase inhibitor, in murine models of hepatocellular carcinoma. Hepatology 2007;46:1119–1130.
Cohen LA, Powers B, Amin S, et al. Treatment of canine haemangiosarcoma with suberoylanilide hydroamic acid, a histone deacetylase inhibitor. Vet Comp Oncol 2004;2:243–248.
Kisseberth WC, Nadella MV, Breen M, et al. A novel canine lymphoma cell line: a translational and comparative model for lymphoma research. Leuk Res. 2007;31:1709–1720.
LeRoy BE, Thudi NK, Nadella MV, et al. New bone formation and osteolysis by a metastatic, highly invasive canine prostate carcinoma xenograft. Prostate 2006;66:1213–1222.
Knapp DW, Chan TC, Kuczek T, et al. Evaluation of in vitro cytotoxicity of nonsteroidal anti-inflammatory drugs against canine tumor cells. Am J Vet Res 1995;56:801–805.
Forsberg LS, Lazarus SC, Seno N, et al. Dog mastocytoma proteoglycans: occurrence of heparin and oversulfated chondroitin sulfates, containing trisulfated disaccharides, in three cell lines. Biochim Biophys Acta 1988;967:416–428.
Shelly S, Chien MB, Yip B, et al. Exon 15 BRAF mutations are uncommon in canine oral malignant melanomas. Mamm Genome 2005;16:211–217.
Liao AT, McMahon M, London C. Characterization, expression, and function of c-Met in canine spontaneous cancers. Vet Comp Oncol 2005;3:61–72.
Papeleu P, Vanhaecke T, Elaut G, et al. Differential effects of histone deacetylase inhibitors in tumor and normal cells—what is the toxicological relevance? Crit Rev Toxicol 2005;35:363–378.
Garcia-Manero G, Issa JP. Histone deacetylase inhibitors: a review of their clinical status as antineoplastic agents. Cancer Invest 2005;23:635–642.
Kelly WK, Richon VM, O'Connor O, et al. Phase I clinical trial of histone deacetylase inhibitor: suberoylanilide hydroxamic acid administered intravenously. Clin Cancer Res 2003;9:3578–3588.
Kelly WK, O'Connor OA, Krug LM, et al. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol 2005;23:3923–3931.
Duvic M, Talpur R, Ni X, et al. Phase 2 trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) for refractory cutaneous T-cell lymphoma (CTCL). Blood 2007;109:31–39.
Piekarz RL, Robey R, Sandor V, et al. Inhibitor of histone deacetylation, depsipeptide (FR901228), in the treatment of peripheral and cutaneous T-cell lymphoma: a case report. Blood 2001;98:2865–2868.
Zhang C, Richon V, Ni X, et al. Selective induction of apoptosis by histone deacetylase inhibitor SAHA in cutaneous T-cell lymphoma cells: relevance to mechanism of therapeutic action. J Invest Dermatol 2005;125:1045–1052.
Piekarz RL, Robey RW, Zhan Z, et al. T-cell lymphoma as a model for the use of histone deacetylase inhibitors in cancer therapy: impact of depsipeptide on molecular markers, therapeutic targets, and mechanisms of resistance. Blood 2004;103:4636–4643.
Yee SB, Kim MS, Baek SJ, et al. Trichostatin A induces apoptosis of p815 mastocytoma cells in histone acetylation- and mitochondria-dependent fashion. Int J Oncol 2004;25:1431–1436.
Advertisement
Objective—To determine whether exposure of canine cancer cells to histone deacetylase (HDAC) inhibitors S(+)-N-hydroxy-4-(3-methyl-2-phenyl-butyrylamino)benzamide (OSU-HDAC42) or suberoylanilide hydroxamic acid (SAHA) results in increased histone acetylation and decreased cell viability and whether any changes in viability involve induction of apoptosis or alterations in progression of the cell cycle.
Sample Population—9 canine cancer cell lines.
Procedures—Cells from 9 canine cancer cell lines were treated with dimethyl sulfoxide vehicle, OSU-HDAC42, or SAHA, then assays of cell viability were performed. Histone acetylation was assessed by use of western blot analysis. Apoptosis was assessed via ELISA to detect fragmentation of cytoplasmic nucleosomal DNA and western blot analysis to detect cleavage of caspase 3. Cell cycle analysis was performed by use of propidium iodide staining and flow cytometry.
Results—Concentrations of OSU-HDAC42 and SAHA required to achieve 50% inhibition of cell viability (IC50) were reached in cells of 6 and 4 canine cancer cell lines, respectively, and ranged from approximately 0.4 to 1.3μM for OSU-HDAC42 and 0.6 to 4.8μM for SAHA. Cells from T-cell lymphoma, mast cell tumor, osteosarcoma, and histiocytic sarcoma lines were most sensitive to HDAC inhibition, with IC50s of < 1μM for OSU-HDAC42 and < 5μM for SAHA. Induction of apoptosis was indicated via cleavage of caspase 3 and increases in cytoplasmic nucleosomes and the subG1 cell population.
Conclusions and Clinical Relevance—Micromolar concentrations of HDAC inhibitors OSU-HDAC42 and SAHA induced histone acetylation, cytotoxicity, and apoptosis in canine cancer cells. In general, OSU-HDAC42 was more potent than SAHA.
The authors thank Marc Hardman and Tim Vojt for assistance with the illustration.