71
Views
4
CrossRef citations to date
0
Altmetric
Review

Finding the place of histone deacetylase inhibitors in prostate cancer therapy

, , , &
Pages 619-630 | Published online: 10 Jan 2014

References

  • Jemal A, Siegel R, Ward E et al. Cancer statistics, 2008. CA Cancer J. Clin.58(2), 71–96 (2008).
  • Han M, Partin A, Pound C, Epstein J, Walsh P. Long-term biochemical disease-free and cancer-specific survival following anatomic radical retropubic prostatectomy. The 15-year Johns Hopkins experience. Urol. Clin. North Am.28(3), 555–565 (2001).
  • Scher HI, Halabi S, Tannock I et al. Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J. Clin. Oncol.26(7), 1148–1159 (2008).
  • Petrylak DP, Tangen CM, Hussain MH et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N. Engl. J. Med.351(15), 1513–1520 (2004).
  • Tannock IF, de Wit R, Berry WR et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N. Engl. J. Med.351(15), 1502–1512 (2004).
  • Attard G, Reid AHM, A’Hern R et al. Selective inhibition of CYP17 with abiraterone acetate is highly active in the treatment of castration-resistant prostate cancer. J. Clin. Oncol.27(23), 3742–3748 (2009).
  • Tran C, Ouk S, Clegg NJ et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science324(5928), 787–790 (2009).
  • Marks PA, Breslow R. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat. Biotechnol.25(1), 84–90 (2007).
  • Gleave ME, Sato N, Sadar M et al. Butyrate analogue, isobutyramide, inhibits tumor growth and time to androgen-independent progression in the human prostate LNCaP tumor model. J. Cell. Biochem.69(3), 271–281 (1998).
  • Butler LM, Agus DB, Scher HI et al. Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res.60(18), 5165–5170. (2000).
  • Butler LM, Webb Y, Agus DB et al. Inhibition of transformed cell growth and induction of cellular differentiation by pyroxamide, an inhibitor of histone deacetylase. Clin. Cancer Res.7(4), 962–970 (2001).
  • Camphausen K, Scott T, Sproull M, Tofilon PJ. Enhancement of xenograft tumor radiosensitivity by the histone deacetylase inhibitor MS-275 and correlation with histone hyperacetylation. Clin. Cancer Res.10(18 Pt 1), 6066–6071 (2004).
  • Kuefer R, Hofer MD, Altug V et al. Sodium butyrate and tributyrin induce in vivo growth inhibition and apoptosis in human prostate cancer. Br. J. Cancer90(2), 535–541 (2004).
  • Thelen P, Schweyer S, Hemmerlein B et al. Expressional changes after histone deacetylase inhibition by valproic acid in LNCaP human prostate cancer cells. Int. J. Oncol.24(1), 25–31 (2004).
  • Fronsdal K, Saatcioglu F. Histone deacetylase inhibitors differentially mediate apoptosis in prostate cancer cells. Prostate62(3), 299–306 (2005).
  • Roy S, Packman K, Jeffrey R, Tenniswood M. Histone deacetylase inhibitors differentially stabilize acetylated p53 and induce cell cycle arrest or apoptosis in prostate cancer cells. Cell Death Differ.12(5), 482–491 (2005).
  • Glaser KB. HDAC inhibitors: clinical update and mechanism-based potential. Biochem. Pharmacol.74(5), 659–671 (2007).
  • Agalioti T, Chen G, Thanos D. Deciphering the transcriptional histone acetylation code for a human gene. Cell111(3), 381–392 (2002).
  • Polevoda B, Sherman F. The diversity of acetylated proteins. Genome Biol.3(5), reviews0006.1–reviews0006.6 (2002).
  • Richards EJ, Elgin SC. Epigenetic codes for heterochromatin formation and silencing: rounding up the usual suspects. Cell108(4), 489–500 (2002).
  • Haberland M, Montgomery RL, Olson EN. The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat. Rev. Genet.10(1), 32–42 (2009).
  • Villagra A, Cheng F, Wang HW et al. The histone deacetylase HDAC11 regulates the expression of interleukin 10 and immune tolerance. Nat. Immunol.10(1), 92–100 (2009).
  • Glozak MA, Seto E. Acetylation/deacetylation modulates the stability of DNA replication licensing factor CDt1. J. Biol. Chem.284(17), 11446–11453 (2009).
  • Finnin M, Donigian J, Cohen A et al. Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors. Nature401(6749), 188–193 (1999).
  • Somoza JR, Skene RJ, Katz BA et al. Structural snapshots of human HDAC8 provide insights into the class I histone deacetylases. Structure12(7), 1325–1334 (2004).
  • Vannini A, Volpari C, Filocamo G et al. Crystal structure of a eukaryotic zinc-dependent histone deacetylase, human HDAC8, complexed with a hydroxamic acid inhibitor. Proc. Natl Acad. Sci. USA101(42), 15064–15069 (2004).
  • Haggarty SJ, Koeller KM, Wong JC, Grozinger CM, Schreiber SL. Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation. Proc. Natl Acad. Sci. USA100(8), 4389–4394 (2003).
  • Hu E, Dul E, Sung C-M et al. Identification of novel isoform-selective inhibitors within class I histone deacetylases. J. Pharmacol. Exp. Ther.307(2), 720–728 (2003).
  • Halkidou K, Gaughan L, Cook S et al. Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer. Prostate59(2), 177–189 (2004).
  • Waltregny D, North B, Van Mellaert F et al. Screening of histone deacetylases (HDAC) expression in human prostate cancer reveals distinct class I HDAC profiles between epithelial and stromal cells. Eur. J. Histochem.48(3), 273–290 (2004).
  • Nakagawa M, Oda Y, Eguchi T et al. Expression profile of class I histone deacetylases in human cancer tissues. Oncol. Rep.18(4), 769–774 (2007).
  • Weichert W, Roske A, Gekeler V et al. Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br. J. Cancer98(3), 604–610 (2008).
  • Halkidou K, Cook S, Leung HY, Neal DE, Robson CN. Nuclear accumulation of histone deacetylase 4 (HDAC4) coincides with the loss of androgen sensitivity in hormone refractory cancer of the prostate. Eur. Urol.45(3), 382–389 (2004).
  • Patra SK, Patra A, Dahiya R. Histone deacetylase and DNA methyltransferase in human prostate cancer. Biochem. Biophys. Res. Commun.287(3), 705–713 (2001).
  • Huffman DM, Grizzle WE, Bamman MM et al. SIRT1 Is Significantly Elevated in Mouse and Human Prostate Cancer. Cancer Res.67(14), 6612–6618 (2007).
  • Jung-Hynes B, Nihal M, Zhong W, Ahmad N. Role of Sirtuin Histone Deacetylase SIRT1 in prostate cancer: a target for prostate cancer management via its inhibition? J. Biol. Chem.284(6), 3823–3832 (2009).
  • Strahl BD, Allis CD. The language of covalent histone modifications. Nature403(6765), 41–45 (2000).
  • Kouzarides T. Chromatin modifications and their function. Cell128(4), 693–705 (2007).
  • Shahbazian MD, Grunstein M. Functions of site-specific histone acetylation and deacetylation. Annu. Rev. Biochem.76(75–100) (2007).
  • Seligson DB, Horvath S, Shi T et al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature435(7046), 1262–1266 (2005).
  • Mohamed MA, Greif PA, Diamond J et al. Epigenetic events, remodelling enzymes and their relationship to chromatin organization in prostatic intraepithelial neoplasia and prostatic adenocarcinoma. BJU Int.99(4), 908–915 (2007).
  • Gregoretti IV, Lee YM, Goodson HV. Molecular evolution of the histone deacetylase family: functional implications of phylogenetic analysis. J. Mol. Biol.338(1), 17–31 (2004).
  • Glozak MA, Sengupta N, Zhang X, Seto E. Acetylation and deacetylation of non-histone proteins. Gene363, 15–23 (2005).
  • Fuino L, Bali P, Wittmann S et al. 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(10), 971–984 (2003).
  • Nimmanapalli R, Fuino L, Bali P et al. Histone deacetylase inhibitor LAQ824 both lowers expression and promotes proteasomal degradation of Bcr–Abl and induces apoptosis of imatinib mesylate-sensitive or -refractory chronic myelogenous leukemia-blast crisis cells. Cancer Res.63(16), 5126–5135 (2003).
  • Bali P, Pranpat M, Bradner J et al. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J. Biol. Chem.280(29), 26729–26734 (2005).
  • Chen L, Meng S, Wang H et al. Chemical ablation of androgen receptor in prostate cancer cells by the histone deacetylase inhibitor LAQ824. Mol. Cancer Ther.4(9), 1311–1319 (2005).
  • Bali P, Pranpat M, Swaby R et al. Activity of suberoylanilide hydroxamic acid against human breast cancer cells with amplification of her-2. Clin. Cancer Res.11(17), 6382–6389 (2005).
  • Yu X, Guo ZS, Marcu MG et al. Modulation of p53, ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228. J. Natl Cancer Inst.94(7), 504–513 (2002).
  • Kovacs JJ, Murphy PJM, Gaillard S et al. HDAC6 regulates Hsp90 acetylation and chaperone-dependent activation of glucocorticoid receptor. Mol. Cell18(5), 601–607 (2005).
  • Murphy PJ, Morishima Y, Kovacs JJ, Yao TP, Pratt WB. Regulation of the dynamics of hsp90 action on the glucocorticoid receptor by acetylation/deacetylation of the chaperone. J. Biol. Chem.280(40), 33792–33799 (2005).
  • George P, Bali P, Annavarapu S et al. Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3. Blood.105(4), 1768–1776 (2005).
  • Rahmani M, Yu C, Dai Y et al. Coadministration of the heat shock protein 90 antagonist 17-allylamino- 17-demethoxygeldanamycin with suberoylanilide hydroxamic acid or sodium butyrate synergistically induces apoptosis in human leukemia cells. Cancer Res.63(23), 8420–8427 (2003).
  • Popov VM, Wang C, Shirley LA et al. The functional significance of nuclear receptor acetylation. Steroids72(2), 221–230 (2007).
  • Fu M, Wang C, Reutens AT et al. p300 and p300/cAMP-response element-binding protein-associated factor acetylate the androgen receptor at sites governing hormone-dependent transactivation. J. Biol. Chem.275(27), 20853–20860 (2000).
  • Fu M, Wang C, Wang J et al. Androgen receptor acetylation governs trans activation and MEKK1-induced apoptosis without affecting in vitro sumoylation and trans-repression function. Mol. Cell. Biol.22(10), 3373–3388 (2002).
  • Gaughan L, Logan IR, Cook S, Neal DE, Robson CN. Tip60 and histone deacetylase 1 regulate androgen receptor activity through changes to the acetylation status of the receptor. J. Biol. Chem.277(29), 25904–25913 (2002).
  • Gaughan L, Logan IR, Neal DE, Robson CN. Regulation of androgen receptor and histone deacetylase 1 by Mdm2-mediated ubiquitylation. Nucleic Acids Res.33(1), 13–26 (2005).
  • Thomas M, Dadgar N, Aphale A et al. Androgen receptor acetylation site mutations cause trafficking defects, misfolding, and aggregation similar to expanded glutamine tracts. J. Biol. Chem.279(9), 8389–8395 (2004).
  • Fu M, Rao M, Wang C et al. Acetylation of androgen receptor enhances coactivator binding and promotes prostate cancer cell growth. Mol. Cell. Biol.23(23), 8563–8575 (2003).
  • Melchior SW, Brown LG, Figg WD et al. Effects of phenylbutyrate on proliferation and apoptosis in human prostate cancer cells in vitro and in vivo. Int. J. Oncol.14(3), 501–508 (1999).
  • Qian DZ, Wang X, Kachhap SK et al. The histone deacetylase inhibitor NVP-LAQ824 inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584. Cancer Res.64(18), 6626–6634 (2004).
  • Chinnaiyan P, Vallabhaneni G, Armstrong E, Huang SM, Harari PM. Modulation of radiation response by histone deacetylase inhibition. Int. J. Radiat. Oncol. Biol. Phys.62(1), 223–229 (2005).
  • Gediya LK, Chopra P, Purushottamachar P, Maheshwari N, Njar VC. A new simple and high-yield synthesis of suberoylanilide hydroxamic acid and its inhibitory effect alone or in combination with retinoids on proliferation of human prostate cancer cells. J. Med. Chem.48(15), 5047–5051 (2005).
  • Kulp SK, Chen CS, Wang DS, Chen CY. Antitumor effects of a novel phenylbutyrate-based histone deacetylase inhibitor, (S)-HDAC-42, in prostate cancer. Clin. Cancer Res.12(17), 5199–5206 (2006).
  • Rokhlin OW, Glover RB, Guseva NV et al. Mechanisms of cell death induced by histone deacetylase inhibitors in androgen receptor-positive prostate cancer cells. Mol. Cancer Res.4(2), 113–123 (2006).
  • Xia Q, Sung J, Chowdhury W et al. Chronic administration of valproic acid inhibits prostate cancer cell growth in vitro and in vivo. Cancer Res.66(14), 7237–7244 (2006).
  • Xu W, Ngo L, Perez G, Dokmanovic M, Marks PA. Intrinsic apoptotic and thioredoxin pathways in human prostate cancer cell response to histone deacetylase inhibitor. Proc. Natl Acad. Sci. USA103(42), 15540–15545 (2006).
  • Lai MT, Yang CC, Lin TY, Tsai FJ, Chen WC. Depsipeptide (FK228) inhibits growth of human prostate cancer cells. Urol. Oncol.26(2), 182–189 (2008).
  • Qian X, Ara G, Mills E et al. Activity of the histone deacetylase inhibitor belinostat (PXD101) in preclinical models of prostate cancer. Int. J. Cancer122(6), 1400–1410 (2008).
  • Sargeant AM, Rengel RC, Kulp SK et al. OSU-HDAC42, a histone deacetylase inhibitor, blocks prostate tumor progression in the transgenic adenocarcinoma of the mouse prostate model. Cancer Res.68(10), 3999–4009 (2008).
  • Xu WS, Parmigiani RB, Marks PA. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene26(37), 5541–5552 (2007).
  • Sonnemann J, Hartwig M, Plath A et al. Histone deacetylase inhibitors require caspase activity to induce apoptosis in lung and prostate carcinoma cells. Cancer Lett.232(2), 148–160 (2006).
  • Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov.5(9), 769–784 (2006).
  • Valentini A, Biancolella M, Amati F et al. Valproic acid induces neuroendocrine differentiation and UGT2B7 up-regulation in human prostate carcinoma cell line. Drug Metab. Dispos.35(6), 968–972 (2007).
  • Frigo DE, McDonnell DP. Differential effects of prostate cancer therapeutics on neuroendocrine transdifferentiation. Mol. Cancer Ther.7(3), 659–669 (2008).
  • Marrocco DL, Tilley WD, Bianco-Miotto T et al. Suberoylanilide hydroxamic acid (vorinostat) represses androgen receptor expression and acts synergistically with an androgen receptor antagonist to inhibit prostate cancer cell proliferation. Mol. Cancer Ther.6(1), 51–60 (2007).
  • Roy S, Jeffrey R, Tenniswood M. Array-based analysis of the effects of trichostatin A and CG-1521 on cell cycle and cell death in LNCaP prostate cancer cells. Mol. Cancer Ther.7(7), 1931–1939 (2008).
  • Welsbie DS, Xu J, Chen Y et al. Histone deacetylases are required for androgen receptor function in hormone-sensitive and castrate-resistant prostate cancer. Cancer Res.69(3), 958–966 (2009).
  • Wang LG, Ossowski L, Ferrari AC. Androgen receptor level controlled by a suppressor complex lost in an androgen-independent prostate cancer cell line. Oncogene23(30), 5175–5184 (2004).
  • Goh M, Chen F, Paulsen MT et al. Phenylbutyrate attenuates the expression of Bcl-X(L), DNA-PK, caveolin-1, and VEGF in prostate cancer cells. Neoplasia3(4), 331–338 (2001).
  • Sasakawa Y, Naoe Y, Noto T et al. Antitumor efficacy of FK228, a novel histone deacetylase inhibitor, depends on the effect on expression of angiogenesis factors. Biochem. Pharmacol.66(6), 897–906 (2003).
  • Butler LM, Zhou X, Xu WS et al. The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc. Natl Acad. Sci. USA99(18), 11700–11705. (2002).
  • Roy S, Tenniswood M. Site specific acetylation of p53 directs selective transcription complex assembly. J. Biol. Chem.282(7), 4765–4771 (2007).
  • Butler LM, Agus DB, Scher HI et al. Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res.60(18), 5165–5170 (2000).
  • Bjorkman M, Iljin K, Halonen P et al. Defining the molecular action of HDAC inhibitors and synergism with androgen deprivation in ERG-positive prostate cancer. Int. J. Cancer123(12), 2774–2781 (2008).
  • Karagiannis TC, El-Osta A. Modulation of cellular radiation responses by histone deacetylase inhibitors. Oncogene25(28), 3885–3893 (2006).
  • Basch EM, Somerfield MR, Beer TM et al. American Society of Clinical Oncology endorsement of the Cancer Care Ontario Practice Guideline on nonhormonal therapy for men with metastatic hormone-refractory (castration-resistant) prostate cancer. J. Clin. Oncol.25(33), 5313–5318 (2007).
  • Kanzaki M, Kakinuma H, Kumazawa T et al. Low concentrations of the histone deacetylase inhibitor, depsipeptide, enhance the effects of gemcitabine and docetaxel in hormone refractory prostate cancer cells. Oncol. Rep.17(4), 761–767 (2007).
  • Zhang Z, Stanfield J, Frenkel E, Kabbani W, Hsieh JT. Enhanced therapeutic effect on androgen-independent prostate cancer by depsipeptide (FK228), a histone deacetylase inhibitor, in combination with docetaxel. Urology70(2), 396–401 (2007).
  • Chen CS, Wang YC, Yang HC et al. Histone deacetylase inhibitors sensitize prostate cancer cells to agents that produce DNA double-strand breaks by targeting Ku70 acetylation. Cancer Res.67(11), 5318–5327 (2007).
  • Borgstrom P, Bourdon MA, Hillan KJ, Sriramarao P, Ferrara N. Neutralizing anti-vascular endothelial growth factor antibody completely inhibits angiogenesis and growth of human prostate carcinoma micro tumors in vivo. Prostate35(1), 1–10 (1998).
  • Melnyk O, Zimmerman M, Kim KJ, Shuman M. Neutralizing anti-vascular endothelial growth factor antibody inhibits further growth of established prostate cancer and metastases in a pre-clinical model. J. Urol.161(3), 960–963 (1999).
  • Takei Y, Kadomatsu K, Yuzawa Y, Matsuo S, Muramatsu T. A small interfering RNA targeting vascular endothelial growth factor as cancer therapeutics. Cancer Res.64(10), 3365–3370 (2004).
  • Fox WD, Higgins B, Maiese KM et al. Antibody to vascular endothelial growth factor slows growth of an androgen-independent xenograft model of prostate cancer. Clin. Cancer Res.8(10), 3226–3231 (2002).
  • Verheul H, Pinedo H. Vascular endothelial growth factor and its inhibitors. Drugs Today (Barc.)39(Suppl. C), 81–93 (2003).
  • Nakamura K, Yamamoto A, Kamishohara M et al. KRN633: a selective inhibitor of vascular endothelial growth factor receptor-2 tyrosine kinase that suppresses tumor angiogenesis and growth. Mol. Cancer Ther.3(12), 1639–1649 (2004).
  • Nicholson B, Gulding K, Conaway M, Wedge SR, Theodorescu D. Combination antiangiogenic and androgen deprivation therapy for prostate cancer: a promising therapeutic approach. Clin. Cancer Res.10(24), 8728–8734 (2004).
  • Stadler WM, Cao D, Vogelzang NJ et al. A randomized Phase II trial of the antiangiogenic agent SU5416 in hormone-refractory prostate cancer. Clin. Cancer Res.10(10), 3365–3370 (2004).
  • Ellis L, Hammers H, Pili R. Targeting tumor angiogenesis with histone deacetylase inhibitors. Cancer Lett.280(2), 145–153 (2009).
  • Kimberley FC, Screaton GR. Following a TRAIL: update on a ligand and its five receptors. Cell Res.14(5), 359–372 (2004).
  • Yagita H, Takeda K, Hayakawa Y, Smyth MJ, Okumura K. TRAIL and its receptors as targets for cancer therapy. Cancer Sci.95(10), 777–783 (2004).
  • Norris JS, Hyer ML, Voelkel-Johnson C et al. The use of Fas Ligand, TRAIL and Bax in gene therapy of prostate cancer. Curr. Gene Ther.1(1), 123–136 (2001).
  • Taghiyev AF, Guseva NV, Sturm MT, Rokhlin OW, Cohen MB. Trichostatin A (TSA) sensitizes the human prostatic cancer cell line DU145 to death receptor ligands treatment. Cancer Biol. Ther.4(4), 382–390 (2005).
  • Vanoosten RL, Moore JM, Ludwig AT, Griffith TS. Depsipeptide (FR901228) enhances the cytotoxic activity of TRAIL by redistributing TRAIL receptor to membrane lipid rafts. Mol. Ther.11(4), 542–552 (2005).
  • Lakshmikanthan V, Kaddour-Djebbar I, Lewis RW, Kumar MV. SAHA-sensitized prostate cancer cells to TNFα-related apoptosis-inducing ligand (TRAIL): mechanisms leading to synergistic apoptosis. Int. J. Cancer119(1), 221–228 (2006).
  • VanOosten RL, Earel JK Jr, Griffith TS. Histone deacetylase inhibitors enhance Ad5-TRAIL killing of TRAIL-resistant prostate tumor cells through increased caspase-2 activity. Apoptosis12(3), 561–571 (2007).
  • Huggins C SRHC. Studies on prostatic cancer. II. The effect of castration on advanced carcinoma of the prostate gland. Arch. Surg.43, 209–228 (1941).
  • Mohler JL. A role for the androgen-receptor in clinically localized and advanced prostate cancer. Best Pract. Res. Clin. Endocrinol. Metab.22(2), 357–372 (2008).
  • Mehra R, Tomlins SA, Shen R et al. Comprehensive assessment of TMPRSS2 and ETS family gene aberrations in clinically localized prostate cancer. Mod. Pathol.20(5), 538–544 (2007).
  • Bradley DA, Dunn R, Rathkopf D et al. Vorinistat in hormone refractory prostate cancer (HRPC): trial results and IL-6 analysis. Program and Abstracts of the American Society of Clinical Oncology Genitourinary Cancers Symposium. San Francisco, CA, USA, 14–16 February 2008 (Absract 211).
  • Sharma S, Symanowski J, Wong B et al. A Phase II clinical trial of oral valproic acid in patients with castration-resistant prostate cancers using an intensive biomarker sampling strategy. Transl. Oncol.1(3), 141–147 (2008).
  • Molife LR, Attard G, Fong PC et al. Phase II, two-stage, single-arm trial of the histone deacetylase inhibitor (HDACi) romidepsin in metastatic castration-resistant prostate cancer (CRPC). Ann. Oncol. (2009) (Epub ahead of print).
  • Shah MH, Binkley P, Chan K et al. Cardiotoxicity of histone deacetylase inhibitor depsipeptide in patients with metastatic neuroendocrine tumors. Clin. Cancer Res.12(13), 3997–4003 (2006).
  • Molife R, Patterson S, Riggs C et al. Phase II study of FK228 in patients with hormone refractory prostate cancer (HRPC). J. Clin. Oncol. (Meeting Abstracts)24, 14554 (2006).
  • Piekarz RL, Frye AR, Wright JJ et al. Cardiac studies in patients treated with depsipeptide, FK228, in a Phase II trial for T-cell lymphoma. Clin. Cancer Res.12(12), 3762–3773 (2006).

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.