Skip to main content
  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • First Disclosures
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Journals
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Molecular Cancer Therapeutics
Molecular Cancer Therapeutics
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Radiation Oncology
      • Novel Combinations
      • Reviews
      • Editors' Picks
      • "Best of" Collection
  • First Disclosures
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Research Articles: Therapeutics, Targets, and Development

Suberoylanilide hydroxamic acid (vorinostat) represses androgen receptor expression and acts synergistically with an androgen receptor antagonist to inhibit prostate cancer cell proliferation

Deborah L. Marrocco, Wayne D. Tilley, Tina Bianco-Miotto, Andreas Evdokiou, Howard I. Scher, Richard A. Rifkind, Paul A. Marks, Victoria M. Richon and Lisa M. Butler
Deborah L. Marrocco
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wayne D. Tilley
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tina Bianco-Miotto
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Andreas Evdokiou
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Howard I. Scher
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Richard A. Rifkind
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Paul A. Marks
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Victoria M. Richon
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lisa M. Butler
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1535-7163.MCT-06-0144 Published January 2007
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Growth of prostate cancer cells is initially dependent on androgens, and androgen ablation therapy is used to control tumor growth. Unfortunately, resistance to androgen ablation therapy inevitably occurs, and there is an urgent need for better treatments for advanced prostate cancer. Histone deacetylase inhibitors, such as suberoylanilide hydroxamic acid (SAHA; vorinostat), are promising agents for the treatment of a range of malignancies, including prostate cancer. SAHA inhibited growth of the androgen-responsive LNCaP prostate cancer cell line at low micromolar concentrations and induced caspase-dependent apoptosis associated with chromatin condensation, DNA fragmentation, and mitochondrial membrane depolarization at higher concentrations (≥5 μmol/L). Gene profiling and immunoblot analyses showed a decrease in androgen receptor (AR) mRNA and protein in LNCaP cells cultured with SAHA compared with control cells, with a corresponding decrease in levels of the AR-regulated gene, prostate-specific antigen. Culture of LNCaP cells in steroid-free medium markedly sensitized the cells to SAHA. Moreover, a combination of low, subeffective doses of SAHA and the AR antagonist bicalutamide resulted in a synergistic reduction in cell proliferation and increase in caspase-dependent cell death. Addition of exogenous androgen prevented the induction of cell death, indicating that suppression of androgen signaling was required for synergy. At the subeffective concentrations, these agents had no effect, alone or in combination, on proliferation or death of AR-negative PC-3 prostate cancer cells. Our findings indicate that SAHA is effective in targeting the AR signaling axis and that androgen deprivation sensitizes prostate cancer cells to SAHA. Consequently, combinatorial treatments that target different components of the AR pathway may afford a more effective strategy to control the growth of prostate cancer cells. [Mol Cancer Ther 2007;6(1):51–60]

Keywords:
  • Prostate cancer
  • histone deacetylase inhibitors
  • androgen receptor
  • antagonist
  • combination

Introduction

Prostate cancer is a serious health concern in Western countries, being the second most common cause of deaths from cancer in men (1). For patients with biochemical relapse following primary treatment with surgery and/or radiation, or who present with metastatic disease, androgen ablation is the standard first-line treatment. Although the majority of these patients respond initially to androgen ablation, virtually all tumors eventually progress with castration-resistant disease (2). Although there have been recent reports of significant responses with the use of cytotoxic agents in the treatment of castration-resistant disease (3), there remains an urgent need for new therapeutic strategies in both an adjuvant setting and following failure of androgen ablation therapy. Histone deacetylase (HDAC) inhibitors are promising novel agents for the treatment of solid tumors (4–8). Antiproliferative and proapoptotic effects of several HDAC inhibitors, including trichostatin A, depsipeptide, MS-275, sodium butyrate, valproic acid, pyroxamide, and suberoylanilide hydroxamic acid (SAHA), have been reported for prostate cancer cell lines (9–12). These agents also have efficacy in xenograft models (13–16) and are currently being investigated in clinical trials for the treatment of solid tumors (17–19).

We have shown that SAHA can inhibit the growth of prostate cancer cell lines in vitro and suppresses the growth of the androgen-dependent human prostate cancer xenograft, CWR22, at high doses (16). Moreover, we found that androgen-independent cell lines were less sensitive to growth inhibition and cell death induced by SAHA than the androgen-responsive LNCaP cells (16). Therefore, in this study, we investigated whether the sensitivity of the LNCaP prostate cancer cell line to SAHA is dependent on the presence of a functional androgen signaling axis and whether the combination of low doses of both SAHA and a specific androgen receptor (AR) antagonist would result in enhanced growth suppression and/or apoptosis.

Materials and Methods

Cells and Reagents

LNCaP and PC-3 human prostate carcinoma cells were obtained from the American Type Culture Collection (Rockville, MD). Cells were maintained in RPMI 1640 supplemented with 10% or 5% fetal bovine serum, respectively. SAHA (now designated by the generic name vorinostat) was synthesized as described previously (20) and dissolved and DMSO. Bicalutamide was obtained from Sigma (St. Louis, MO) and dissolved in ethanol. Anti-AR (C-19), anti–prostate-specific antigen (PSA; C-19), anti–cyclin D1 (A-12), and anti-p21WAF1 (AB-3) antibodies were obtained from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). Anti-calnexin (MA3-027) antibody was obtained from Affinity Bioreagents (Golden, CO). Anti-Her2/neu (Ab-15) was obtained from Lab Vision (Fremont, CA). Horseradish peroxidase–conjugated secondary antirabbit and antimouse antibodies were obtained from DAKO (Carpinteria, CA), and secondary antisheep antibody was obtained from Chemicon (Melbourne, Victoria, Australia). The tetrapeptide caspase inhibitor z-VAD-fmk was obtained from Calbiochem (Alexandria, New South Wales, Australia).

Measurement of Cell Viability

Cells were seeded in triplicate in 24-well dishes at a density of 2.5 × 104 per well. Cells were allowed to attach for 48 h before incubation with fresh medium containing SAHA (0, 0.5, 1, 2.5, 5, 7.5, or 10 μmol/L) or bicalutamide (0, 1.25, or 2.5 μmol/L). Cells were counted using a hemocytometer at 24- or 48-h intervals after initiation of treatment, and cell viability was assessed by trypan blue dye exclusion as described previously (16). Data are expressed as the mean ± the SE of triplicate wells and are representative of at least three independent experiments.

4′,6-Diamidino-2-Phenylindole Staining of Nuclei

Cells were seeded on plastic chamber slides and treated as indicated. After two washes with PBS, cells were fixed in methanol for 5 min, washed again with PBS, and incubated with 0.8 mg/mL 4′,6-diamidino-2-phenylindole (DAPI, Roche Diagnostics, Castle Hill, New South Wales, Australia) in PBS for 15 min at 37°C. After several washes in PBS, the coverslips were mounted on PBS/glycerin. DAPI staining was visualized by fluorescence microscopy.

Measurement of Mitochondrial Membrane Potential

Cells (5 × 105) seeded in 10-cm plates were treated as indicated for up to 48 h. Cells were harvested by trypsinization and resuspended in complete tissue culture medium containing 2 μg/mL rhodamine 123 dye (kindly provided by Prof. Sharad Kumar, Hanson Institute, Adelaide, South Australia, Australia). Cells were incubated at 37°C for 20 min in the dark and then washed twice in PBS. Cells were resuspended in 1 mL PBS containing the viability dye 7-aminoactinomycin D (2 μg/mL) and incubated at room temperature for 10 min. Cell fluorescence was analyzed by flow cytometry, with green fluorescence (rhodamine 123) measured in cells with intact cellular membranes (i.e., 7-aminoactinomycin D negative).

Cell Cycle Analysis

Fluorescence-activated cell sorting analysis of cell cycle distribution and sub-G1 fraction was done on untreated and SAHA-treated cells after 24 to 96 h of culture, using standard histogram analysis as described previously (12).

Measurement of DEVD-Caspase Activity

DEVD-caspase activity was assayed by cleavage of z-asp-glu-val-asp-7-amino-4-trifluoro-methyl-coumarin, a fluorogenic substrate based on the peptide sequence at the caspase-3 cleavage site of poly(ADP-ribose) polymerase. Cells (1.5 × 104) seeded in 96-well plates were cultured with SAHA (0, 2.5, 5, or 7.5 μmol/L). After 48 h, the cells were lysed in 50 μL NP40 lysis buffer [5 mmol/L Tris-HCl, 5 mmol/L EDTA, 0.5% NP40 (pH 7.5)] and stored at −70°C until assayed. Cell lysates (20 μL) were added to each assay tube containing 8 μmol/L substrate in 200 μL fluorometric caspase assay buffer [50 mmol/L HEPES, 10% sucrose of substrate in 1 mL protease buffer (50 mmol/L HEPES, 10% sucrose, 10 mmol/L DTT, 0.1% CHAPS (pH 7.4))]. After incubation for 4 h at room temperature, fluorescence was quantified (excitation of 400 nm and emission of 505 nm) in a Perkin-Elmer LS50 spectrofluorometer (Wellesley, MA). One unit of caspase activity was taken as one fluorescence unit (at slit widths of 10 nm) per 4 h of incubation with substrate. The tetrapeptide caspase inhibitor z-VAD-fmk was dissolved in DMSO and added to cells at a final concentration of 50 μmol/L, 30 min before addition of SAHA. Control cells were incubated with DMSO at the same concentration.

Microarray Analysis

LNCaP cells were cultured with 2.5 or 7.5 μmol/L SAHA for 6 h. Total RNA was isolated from the cells using Trizol reagent (Invitrogen, Carlsbad, CA). Poly(A)+ mRNA was isolated from the total RNA using Oligotex columns (Qiagen, Valencia, CA). Poly(A)+ mRNA from cells cultured with SAHA was compared with mRNA from cells cultured in the absence of SAHA, using the UniGEM human cDNA version 2.0 array, which contained 9,182 cDNA probes representing 8,372 individual genes/expressed sequence tags and 192 internal controls (Incyte, St. Louis, MO). The results were analyzed using GEM Tools image and data analysis software, and a 2-fold change was considered as a threshold for regulation of gene expression.

Immunoblotting

Cells were lysed in radioimmunoprecipitation assay lysis buffer (10 mmol/L Tris-HCl, 150 mmol/L NaCl, 1 mmol/L EDTA, 1% Triton X-100) containing mini-complete protease inhibitor pellets (Roche, Mannheim, Germany). Lysates (20 μg) were electrophoresed through 7.5% or 15% polyacrylamide gels, electrophoretically transferred to nitrocellulose membranes (Amersham Biosciences, Buckinghamshire, England), and blocked in 3% nonfat milk powder in TBS containing 0.05% Tween 20 overnight. Immunodetection was done for 1 h at room temperature in 1% nonfat milk powder in TBS containing 0.05% Tween 20, using anti-AR, anti-PSA, anti-Her2/neu, anti-p21WAF1, and anti–cyclin D1 antibodies diluted according to the manufacturer's instructions. Membranes were incubated with anti-calnexin as a loading control. Immune complexes were detected with horseradish peroxidase–conjugated secondary antibodies and visualized on X-ray film using the enhanced chemiluminescence detection system (Amersham Biosciences) as described previously (21).

Evaluation of Interaction between SAHA and the Anti-Androgen Bicalutamide

Dose-response curves were generated for bicalutamide and SAHA, both independently and in combination. The effect of combining the two drugs was assessed by the isobole method (22), using the equation Ac / Ae + Bc / Be = D, to determine if their activity was synergistic. Ac and Bc represent the concentration of drug A and drug B used in the combination, and Ae and Be represent the conentration of drug A and B that produced the same magnitude of effect when administered alone. If D, the combination index, is <1, then the drugs are considered to act synergistically. If the combination index is ≥1, then the drugs act in an antagonistic or additive manner, respectively.

Results

SAHA Suppresses Growth and Induces Cell Cycle Arrest and Death of LNCaP Prostate Cancer Cells

A dose-dependent inhibition of LNCaP cell growth was observed with concentrations of SAHA ranging from 0.5 to 10 μmol/L (Fig. 1A ). Although growth inhibition was observed with 0.5 and 1 μmol/L doses of SAHA over the 7 days of experimental period (Fig. 1A), maximal reduction of cell number was achieved with 2.5 μmol/L and greater concentrations of SAHA. Minimal cell death was observed with 0.5 to 2.5 μmol/L of SAHA, but 5 to 10 μmol/L markedly induced LNCaP cell death at 7 days posttreatment (Fig. 1B). Whereas a dose-dependent inhibition of PC-3 cell growth was also observed with SAHA (Fig. 1C), there was little effect on PC-3 cell death, even with the highest concentration of SAHA (10 μmol/L; Fig. 1D).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Inhibition of LNCaP and PC-3 prostate cancer cell growth by SAHA. LNCaP and PC-3 cells (2.5 × 104 per well in 24-well plates) were cultured with SAHA (0, 0.5, 1, 2.5, 5, 7.5, or 10 μmol/L) in RPMI 1640 containing 10% FCS, for up to 7 d. Cells were counted every day (PC-3) or every second day (LNCaP) using a hemocytometer, and cell viability was assessed by trypan blue dye exclusion (A and C). The number of dead cells is expressed as a percentage of total cells counted (B and D). Representative of at least three independent experiments. Points, mean of triplicate wells; bars, SE.

To further investigate the mechanism of SAHA-induced suppression of cell growth, we analyzed cell cycle distribution in control- and SAHA-treated cells. After 24-h culture with 2.5 μmol/L SAHA, LNCaP cells were arrested in the G1 phase of the cell cycle (Fig. 2A ). This block in cell cycle progression was maintained for at least 72 h (data not shown). In contrast, LNCaP cells cultured with 7.5 μmol/L SAHA for 24 h showed a shift in cell cycle from S phase to the G2-M phase, with no change in the proportion of cells in G1 (Fig. 2A and B). Whereas there was no significant change in the sub-G1 nuclear fraction in cells cultured with 7.5 μmol/L SAHA for 24 h (Fig. 2B, top), an increased sub-G1 fraction, indicative of DNA fragmentation, was observed in cells cultured with 7.5 μmol/L SAHA for 4 days (Fig. 2B, bottom).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Effect of SAHA on cell cycle distribution in LNCaP cells. A, cells cultured with SAHA (0, 2.5, or 7.5 μmol/L) for 24 h were harvested by trypsinization, fixed in 70% ethanol, and stained with propidium iodide. Cell cycle distribution was determined by flow cytometry. B, cells cultured in the absence or presence of SAHA (7.5 μmol/L) for 24 h or 4 d were harvested and fixed as described above and stained with propidium iodide. The fraction of hypodiploid (sub-G1) nuclei was measured by flow cytometry using standard histogram analysis.

Cell Death Induced by SAHA Is Characteristic of Apoptosis

SAHA induced caspase-3 activity in LNCaP cells in a dose-dependent manner (Fig. 3A ). Caspase-3 activity was blocked when cells were cocultured with the z-VAD-fmk caspase inhibitor. Moreover, addition of the caspase inhibitor prevented cell death induced by either 2.5 or 7.5 μmol/L SAHA (Fig. 3B). Staining of cellular DNA with DAPI showed that LNCaP cells cultured with SAHA (7.5 μmol/L) for 24 h exhibited chromatin condensation and nuclear fragmentation (Fig. 3C), characteristic of apoptosis. Mitochondrial membrane potential was measured in cells cultured in the presence or absence of SAHA (7.5 μmol/L), using the lipophilic, green fluorescent dye rhodamine 123. A 3-fold increase in rhodamine-negative cells, indicating loss of mitochondrial membrane potential, was observed in cells cultured with SAHA for either 24 h (data not shown) or 48 h compared with cells cultured with vehicle alone (vehicle control, 6.4%, versus SAHA, 19.7%; Fig. 3D).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Induction of LNCaP cell death by SAHA. A, LNCaP cells (2.5 × 104 per well in 24-well plates) were cultured with SAHA (0, 2.5, or 7.5 μmol/L) with or without the z-VAD-fmk caspase inhibitor for 48 h and assayed for caspase-3 activity. B, LNCaP cells (2.5 × 104) were cultured with SAHA (0, 2.5, or 7.5 μmol/L) with or without the z-VAD-fmk caspase inhibitor for 5 d. Cells were counted at day 5 using a hemocytometer, and cell viability was assessed by trypan blue exclusion. The number of dead cells expressed as a percentage of total cells was counted. Columns, mean of triplicate wells in a representative experiment; bars, SE. C, DAPI staining of cell nuclei. LNCaP cells were seeded onto plastic coverslips and cultured with vehicle alone (DMSO) or SAHA (7.5 μmol/L) for 24 h. Cells were fixed with methanol and incubated with DAPI, before washing in PBS and mounting on PBS/glycerin. DAPI staining was visualized by fluorescence microscopy. D, effect of SAHA on mitochondrial membrane potential in LNCaP cells. Cells were cultured with vehicle alone (DMSO) or SAHA (7.5 μmol/L) for 48 h. Cells were harvested by trypsinization, resuspended in RPMI 1640 containing 10% FCS, and incubated with the mitochondrial dye rhodamine 123 (Rho123; 2 μg/mL) for 20 min at 37°C. Cells were washed and incubated in PBS containing 2 μg/mL of the viability dye 7-aminoactinomycin D at room temperature for 10 min and then analyzed by flow cytometry. Results shown indicate the level of rhodamine 123 fluorescence in the 7-aminoactinomycin D–negative population.

Gene Expression Profiles in LNCaP Prostate Cancer Cells Cultured with SAHA

Gene expression profiles were examined following culture of LNCaP cells for 6 h with low (2.5 μmol/L) or high (7.5 μmol/L) concentrations of SAHA. Scatter plots of control hybridization intensity versus SAHA hybridization intensity showed that the expression of the majority of genes either was not altered or was altered by <2-fold (Fig. 4A ). As shown by the Venn diagrams (Fig. 4B), a >2-fold change in gene expression was observed for only ∼1% of the total number of genes analyzed following culture with 2.5 or 7.5 μmol/L SAHA. The majority of genes identified as being modulated by SAHA in LNCaP cells are involved in signal transduction, transcription, or translation (see Supplementary Tables). The expression levels of the known SAHA target genes, cyclin D1 and thioredoxin-binding protein-2/VDUP1, were decreased and increased, respectively (see Supplementary Tables).6 Interestingly, a subset of genes involved in AR signaling was reduced in expression by at least 2-fold following culture with SAHA (Fig. 4C; refs. 23–28). Of particular interest, the mRNA levels of the AR and the AR-regulated genes, PSA (kallikrein 3) and kallikrein 2, were reduced in cells cultured with SAHA. Whereas expression of AR was decreased by both 2.5 and 7.5 μmol/L SAHA, the expression of PSA and kallikrein 2 was only decreased by 7.5 μmol/L SAHA. Real-time reverse transcription PCR confirmed that the levels of AR, PSA, and kallikrein 2 mRNA were reduced in cells cultured with 7.5 μmol/L SAHA for 2 h (Fig. 4D).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Summary of gene expression changes in LNCaP cells following SAHA treatment. A, scatter plot of control intensity versus SAHA (2.5 μmol/L) intensity for all elements on the UniGEM version 2.0 human cDNA microarray, which contains 8,372 unique gene sequences. B, Venn diagrams depicting the number of genes with ≥2-fold changes in mRNA levels. Top, the number of genes induced; bottom, the number of genes repressed by 2.5 and/or 7.5 μmol/L SAHA. C, summary of SAHA-induced alterations in expression of genes involved in androgen signaling. D, real-time PCR analysis of AR, PSA, and kallikrein 2 (KLK2) mRNA levels 2 h following treatment with SAHA.

To determine whether the gene expression data identified by the microarray and mRNA expression analyses resulted in biologically meaningful changes in protein expression in the cell, the effects of SAHA on AR and PSA expression were evaluated by immunoblot analysis (Fig. 5 ). AR steady-state protein levels were markedly reduced in a dose-dependent manner for up to 48 h, with a corresponding decrease in the steady-state level of PSA (Fig. 5A). In addition, steady-state protein levels of the heat shock protein 90 (hsp90) client protein, Her2/neu, were reduced 48 h following SAHA treatment (Fig. 5B). Protein levels of cyclin D1 and p21WAF1, which were used as controls for SAHA activity (29, 30), were decreased and increased, respectively, in a dose-dependent manner by SAHA (Fig. 5C).

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Protein expression changes in LNCaP cells following culture with SAHA. Lysates from cells cultured in the absence or presence of SAHA (2.5, 5, or 7.5 μmol/L) for 12, 24, or 48 h were analyzed by immunoblotting for expression of AR and PSA (A), Her2/neu (B), and cyclin D1 and p21WAF1(C). For each immunoblot, detection of calnexin was used as a loading control (bottom).

Effects of SAHA on AR Expression and Cell Viability Are Enhanced by Culture in Steroid-Free Medium

Culture of LNCaP cells in charcoal-stripped serum (i.e., under conditions of androgen withdrawal) increased the sensitivity of the cells to SAHA. AR levels were markedly reduced in cells cultured with either 2.5 or 7.5 μmol/L SAHA for 24 h in medium containing charcoal-stripped serum (Fig. 6A ). In cells cultured with SAHA in medium with normal serum, only 7.5 μmol/L SAHA caused an equivalent reduction in AR levels (Fig. 6A). Irrespective of whether the cells were grown in medium containing normal or stripped FCS, pretreatment with 5α-dihydrotestosterone (1 nmol/L) had no effect on the ability of SAHA to reduce AR levels (Fig. 6A), indicating that activation and/or stabilization of the AR by 5α-dihydrotestosterone did not prevent the reduction in AR levels caused by SAHA. When cultured in medium containing charcoal-stripped serum, cell growth was completely suppressed by 0.5 μmol/L SAHA (Fig. 6B), which is substantially lower than was observed in cells cultured in normal medium (2.5 μmol/L; Fig. 1A). An increased percentage of dead cells was also seen with all doses of SAHA, with a maximum cell death of 60% being attained with 10 μmol/L SAHA (Fig. 6B).

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

Effect of SAHA in combination with androgen withdrawal on AR expression, cell proliferation, and cell death. A, LNCaP cells (5 × 105 per well in six-well plates) were cultured for 24 h with SAHA (0, 2.5, or 7.5 μmol/L) in the presence or absence of 5α-dihydrotestosterone (DHT; 1 nmol/L), in either regular RPMI 1640 with 10% FCS (left) or phenol red–free RPMI 1640 with 10% charcoal-stripped FCS (right). Cells were lysed and analyzed by immunoblotting for expression of AR, and calnexin was used as a loading control. B and C, LNCaP cells (2.5 × 104 per well in 24-well plates) were cultured in medium containing charcoal-stripped FCS in the absence or presence of SAHA for up to 7 d. B, cells were counted every 2nd day using a hemocytometer, and cell viability was assessed by trypan blue exclusion. C, the number of dead cells is expressed as a percentage of total cells counted. Points, mean of triplicate wells in a representative experiment; bars, SE.

SAHA and the AR Antagonist Bicalutamide Act Synergistically to Suppress LNCaP Cell Proliferation and Increase Apoptosis

The enhanced sensitivity of LNCaP cells to SAHA in steroid-free medium suggested that combining SAHA with other inhibitors of androgen signaling may result in increased cell lethality. Culture of LNCaP prostate cancer cells with low, subeffective doses of SAHA (0.5 μmol/L) or the specific AR antagonist bicalutamide (1.25 μmol/L) alone, in normal medium supplemented with 10% serum, had minimal effects on LNCaP cell growth (Fig. 7A ) and the percentage of dead cells (Fig. 7B). However, when combined, at the lower doses, these agents caused complete suppression of LNCaP cell growth and induction of cell death (Fig. 7A and B). The action of SAHA and bicalutamide on LNCaP cells was synergistic according to the isobole method of Berenbaum (22), the D values for cell proliferation and cell death being 0.2 and 0.8, respectively. An equivalent effect on cell viability was only observed with 50 μmol/L bicalutamide treatment or 7.5 μmol/L SAHA when these agents were used individually. Addition of the z-VAD-fmk caspase inhibitor (Fig. 7C), or excess exogenous androgen (Fig. 7D), completely prevented the synergistic effect of 0.5 μmol/L SAHA and 1.25 μmol/L bicalutamide on cell death. There was no difference in the levels of AR protein in LNCaP cells treated with 0.5 μmol/L SAHA alone or in combination with 1.25 μmol/L bicalutamide (Fig. 7E), indicating that this synergy was not due to altered AR levels. Although there was a modest decrease in PSA expression at 24 and 48 h posttreatment with bicalutamide, this was not enhanced with the addition of SAHA (Fig. 7E). Consistent with a specific effect of this combination of agents at the subeffective doses on AR-dependent cells, there was no effect of either agent alone or in combination on cellular viability of androgen-independent, AR-negative PC-3 prostate cancer cells (Fig. 8A and B ).

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

Effect of SAHA in combination with the AR antagonist bicalutamide on LNCaP cell proliferation and cell death. A and B, cells (2.5 × 104 per well in 24-well plates) were cultured in the absence or presence of 0.5 μmol/L SAHA and 1.25 μmol/L bicalutamide, either alone or in combination. A, cells were counted every 2nd day using a hemocytometer, and cell viability was assessed by trypan blue exclusion. B, the number of dead cells is expressed as a percentage of total cells counted. Points, mean of triplicate wells in a representative experiment; bars, SE. C and D, LNCaP cells (2.5 × 104 per well in 24-well plates) were cultured with 0.5 μmol/L SAHA and 1.25 μmol/L bicalutamide, alone or in combination, in the presence or absence of (C) the z-VAD-fmk caspase inhibitor or (D) the androgen 5α-dihydrotestosterone (10 nmol/L) for 5 d. Cell viability was assessed as described above. E, lysates from untreated LNCaP cells (U) and cells cultured with 0.5 μmol/L SAHA (S), 1.25 μmol/L bicalutamide (B), or SAHA and bicalutamide (S+B) were analyzed by immunoblotting for expression of AR and PSA. Calnexin was used as a loading control (bottom).

Figure 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 8.

Effect of SAHA in combination with bicalutamide on PC-3 cell proliferation and cell death. PC-3 cells (2.5 × 104 per well in 24-well plates) were cultured in the absence or presence of 0.5 μmol/L SAHA and 1.25 μmol/L bicalutamide, alone or in combination. A, cell viability was assessed as described above. B, the number of dead cells is expressed as a percentage of total cells counted. Points, mean of triplicate wells in a representative experiment; bars, SE.

Discussion

SAHA is a promising agent currently in clinical trials for treatment of hematologic malignancies and solid tumors. Although previous studies suggest that SAHA can effectively inhibit the growth of prostate cancer cells (16), the mechanism of growth inhibition is not well understood. Moreover, it was evident from our previous studies that SAHA is more efficacious in terms of growth inhibition and induction of cell death in androgen-responsive cells (e.g., LNCaP and CWR22) than in cells that lack AR (PC-3; ref. 16), suggesting that a component of the activity of SAHA in prostate cancer cells relates to the presence of a functional androgen signaling axis. Our current data, showing the modulation of the AR and genes involved in AR signaling, including its direct target genes PSA and kallikrein 2, as well as genes reported to be androgen regulated, such as transmembrane serine protease and NEDD4L, provide direct evidence of an effect of SAHA on AR signaling. In addition, our demonstration that both androgen withdrawal (steroid-free cell culture) and blocking the effects of androgens with an AR antagonist, bicalutamide, result in markedly enhanced sensitivity to SAHA, suggests that HDAC inhibitors, in combination with agents that target androgen signaling, may be a more effective strategy for the treatment of prostate cancer.

Due to the dependence of prostate cells on androgens for growth, current hormonal therapies for advanced prostate cancer aim to inhibit androgen signaling by removing or blocking the effect of bioavailable androgens (31). However, the AR in prostate cancer cells may continue to signal even in the presence of low levels of androgens due to up-regulation of AR expression or activation by alternate mechanisms, including nonclassic ligands or ligand-independent mechanisms (32). The consequence of this continued androgen signaling in prostate cancer is patient relapse with castration-resistant disease (31). Agents that are able to reduce AR expression or function are therefore potentially effective therapies for prostate cancer, especially in conjunction with other agents that target different components of the AR signaling pathway. Previous microarray studies have shown that culture of prostate cancer cells with the AR antagonist bicalutamide results in a reduction in levels of the AR-regulated genes, PSA and kallikrein 2 (33). In the present study, we found that treatment with SAHA also reduced expression of these AR-regulated genes but there was minimal overlap in any other genes regulated by SAHA or bicalutamide in microarray studies. Decreasing the level of AR within prostate cancer cells using HDAC inhibitors could enhance the ability of hormonal agents, such as AR antagonists, to inhibit the growth of prostate cancer and possibly delay the emergence of castration-resistant disease. Alternatively, the reduction in AR signaling caused by bicalutamide or androgen withdrawal may be sufficient to sensitize the LNCaP cells to SAHA-induced apoptosis. Taken together with the observation that the androgen-independent, AR-negative PC-3 prostate cancer cell line was not affected by the combination of SAHA and bicalutamide and that addition of exogenous androgen could prevent cell death induced by this combination, our data strongly suggest that a functional androgen signaling axis is required for this synergistic effect on LNCaP cell death.

Our data suggest that the reduction in AR level induced by SAHA in LNCaP cells may involve translational as well as transcriptional mechanisms. Acetylation of histones in the AR gene promoter could result in direct repression of the AR gene through changes in chromatin structure, analogous to the regulation of the cyclin D1 gene. An AR transcriptional repressor complex has been identified in LNCaP prostate cancer cells that can be activated by treatment with a HDAC inhibitor, in turn decreasing AR levels (34). Although our microarray and real-time RT-PCR analyses indicate that expression of AR is reduced by SAHA at the level of mRNA, acetylation of non-histone proteins by SAHA (4), such as the chaperone protein hsp90, or the AR itself, could also influence steady-state AR protein levels. Inhibition of hsp90 activity causes degradation of its client proteins, including the AR (35–37). The HDAC inhibitors depsipeptide and LAQ824 have been shown to cause hsp90 acetylation, thereby inhibiting normal protein-protein interactions and leading to degradation of client proteins (including the AR), in a similar manner to that seen following treatment with the specific hsp90 inhibitor 17-allylaminogeldanamycin (38–40). This mechanism may explain the synergistic activity of HDAC inhibitors and the hsp90 inhibitor 17-allylaminogeldanamycin for inhibition of cancer cell growth (41–43). Our data, showing a reduction in the hsp90 client protein Her2/neu in LNCaP cells cultured with SAHA, are consistent with an effect of SAHA on hsp90-dependent protein maturation, which may contribute to the reduction in AR levels.

In summary, we have shown that SAHA decreases AR protein levels in prostate cancer cells and results in cell cycle arrest and apoptosis. Furthermore, when used at a low dose that has no effect on prostate cancer cell growth, SAHA acts synergistically with a traditional hormonal agent, bicalutamide, to induce apoptosis. Taken together, our results suggest that inhibition of androgen signaling is an important component of SAHA activity in prostate cancer cells and that androgen withdrawal or blockade may sensitize prostate cancer cells to undergo cell death in response to HDAC inhibitors. Given that the majority of clinical prostate tumors express the AR, including those that fail hormonal therapy, further investigation into the use of SAHA for the treatment of prostate cancer is warranted, particularly in the context of combination therapy with conventional androgen ablation therapies.

Acknowledgments

We thank Melissa Lee, Bronwyn Cambareri, Ben Copeland, and Shelley Hay for technical assistance and Dr. Fares Al-Ejeh for providing expertise in the mitochondrial membrane potential analyses.

Footnotes

  • ↵6 Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/).

  • Grant support: Burke Foundation (L.M. Butler and H.I. Scher); the Japan Foundation for Cancer Research, the DeWitt Wallace Fund for Memorial Sloan-Kettering Cancer Center, the David H. Koch Prostate Cancer Research Award, and NIH grant CA-0974823 (P.A. Marks and R.A. Rifkind); the Prostate Cancer Foundation of Australia (L.M. Butler); and the National Health and Medical Research Council of Australia grant 299048 (L.M. Butler and W.D. Tilley).

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • Disclosure statement: Memorial Sloan-Kettering Cancer Center and Columbia University jointly hold the patents on the hydroxamic acid–based hybrid polar compounds, including suberoylanilide hydroxamic acid, which are exclusively licensed to Aton Pharmaceuticals, Inc. of which Richard A. Rifkind, Paul A. Marks, and Victoria M. Richon are founders. Aton Pharma is a wholly-owned subsidiary of Merck & Co. Both Memorial Sloan-Kettering Cancer Center, Columbia University, and the founders had an equity position in Aton Pharmaceuticals.

    • Accepted November 16, 2006.
    • Received March 17, 2006.
    • Revision received October 6, 2006.
  • American Association for Cancer Research

References

  1. ↵
    Greenlee RT, Murray T, Bolden S, Wingo PA. Cancer statistics, 2000. CA Cancer J Clin 2000;50:7–33.
    OpenUrlCrossRefPubMed
  2. ↵
    Scher HI, Isaacs JT, Zelefsky M, Scardino PT. Prostate Cancer. 2nd ed. In: Abeloff M, Armitage J, Lichter, Niederhuber J, editors. Clinical Oncology. New York: Churchill Livingston; 2000. p. 1823–84.
  3. ↵
    Morris MJ, Scher HI. Novel strategies and therapeutics for the treatment of prostate carcinoma. Cancer 2000;89:1329–48.
    OpenUrlCrossRefPubMed
  4. ↵
    Richon VM, O'Brien JP. Histone deacetylase inhibitors: a new class of potential therapeutic agents for cancer treatment. Clin Cancer Res 2002;8:662–4.
    OpenUrlFREE Full Text
  5. Marks PA, Richon VM, Miller T, Kelly WK. Histone deacetylase inhibitors. Adv Cancer Res 2004;91:137–68.
    OpenUrlCrossRefPubMed
  6. Lindemann RK, Gabrielli B, Johnstone RW. Histone-deacetylase inhibitors for the treatment of cancer. Cell Cycle 2004;3:779–88.
    OpenUrlPubMed
  7. Drummond DC, Noble CO, Kirpotin DB, Guo Z, Scott GK, Benz CC. Clinical development of histone deacetylase inhibitors as anticancer agents. Annu Rev Pharmacol Toxicol 2005;45:495–528.
  8. ↵
    Rosato RR, Grant S. Histone deacetylase inhibitors in clinical development. Expert Opin Investig Drugs 2004;13:21–38.
    OpenUrlCrossRefPubMed
  9. ↵
    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 2005;12:482–91.
    OpenUrlCrossRefPubMed
  10. Fronsdal K, Saatcioglu F. Histone deacetylase inhibitors differentially mediate apoptosis in prostate cancer cells. Prostate 2005;62:299–306.
    OpenUrlCrossRefPubMed
  11. Thelen P, Schweyer S, Hemmerlein B, Wuttke W, Seseke F, Ringert RH. Expressional changes after histone deacetylase inhibition by valproic acid in LNCaP human prostate cancer cells. Int J Oncol 2004;24:25–31.
    OpenUrlPubMed
  12. ↵
    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 2001;7:962–70.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Gleave ME, Sato N, Sadar M, Yago V, Bruchovsky N, Sullivan L. Butyrate analogue, isobutyramide, inhibits tumor growth and time to androgen-independent progression in the human prostate LNCaP tumor model. J Cell Biochem 1998;69:271–81.
    OpenUrlCrossRefPubMed
  14. 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 Cancer 2004;90:535–41.
    OpenUrlCrossRefPubMed
  15. 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 2004;10:6066–71.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    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 2000;60:5165–70.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Sandor V, Bakke S, Robey RW, et al. Phase I trial of the histone deacetylase inhibitor, depsipeptide (FR901228, NSC 630176), in patients with refractory neoplasms. Clin Cancer Res 2002;8:718–28.
    OpenUrlAbstract/FREE Full Text
  18. Reid T, Valone F, Lipera W, et al. Phase II trial of the histone deacetylase inhibitor pivaloyloxymethyl butyrate (Pivanex, AN-9) in advanced non-small cell lung cancer. Lung Cancer 2004;45:381–6.
    OpenUrlCrossRefPubMed
  19. ↵
    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;23:3923–31.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Richon VM, Webb Y, Merger R, et al. Second generation hybrid polar compounds are potent inducers of transformed cell differentiation. Proc Natl Acad Sci U S A 1996;93:5705–8.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Buchanan G, Yang M, Harris JM, et al. Mutations at the boundary of the hinge and ligand binding domain of the androgen receptor confer increased transactivation function. Mol Endocrinol 2001;15:46–56.
    OpenUrlCrossRefPubMed
  22. ↵
    Berenbaum KC. Criteria for analysing interaction between biologically active agents. Adv Cancer Res 1981;25:269–335.
    OpenUrl
  23. ↵
    Manin M, Baron S, Goossens K, et al. Androgen receptor expression is regulated by the phosphoinositide 3-kinase/Akt pathway in normal and tumoral epithelial cells. Biochem J 2002;366:729–36.
    OpenUrlPubMed
  24. DeFranco DB, Ramakrishnan C, Tang Y. Molecular chaperones and subcellular trafficking of steroid receptors. J Steroid Biochem Mol Biol 1998;65:51–8.
    OpenUrlCrossRefPubMed
  25. Pestell RG, Albanese C, Reutens AT, Segall JE, Lee RJ, Arnold A. The cyclins and cyclin-dependent kinase inhibitors in hormonal regulation of proliferation and differentiation. Endocr Rev 1999;20:501–34.
    OpenUrlCrossRefPubMed
  26. Sun Z, Pan J, Balk SP. Androgen receptor-associated protein complex binds upstream of the androgen-responsive elements in the promoters of human prostate-specific antigen and kallikrein 2 genes. Nucleic Acids Res 1997;25:3318–25.
    OpenUrlAbstract/FREE Full Text
  27. Qi H, Grenier J, Fournier A, Labrie C. Androgens differentially regulate the expression of NEDD4L transcripts in LNCaP human prostate cancer cells. Mol Cell Endocrinol 2003;210:51–62.
    OpenUrlCrossRefPubMed
  28. ↵
    Lin B, Ferguson C, White JT, et al. Prostate-localized and androgen-regulated expression of the membrane-bound serine protease TMPRSS2. Cancer Res 1999;59:4180–4.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Sakajiri S, Kumagai T, Kawamata N, Saitoh T, Said JW, Koeffler HP. Histone deacetylase inhibitors profoundly decrease proliferation of human lymphoid cancer cell lines. Exp Hematol 2005;33:53–61.
    OpenUrlCrossRefPubMed
  30. ↵
    Gui C-Y, Ngo L, Xu WS, Richon VM, Marks PA. Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci U S A 2004;101:1241–6.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Scher HI, Buchanan G, Gerald W, Butler LM, Tilley WD. Targeting the androgen receptor: improving outcomes for castration-resistant prostate cancer. Endocr Relat Cancer 2004;11:459–76.
    OpenUrlAbstract/FREE Full Text
  32. ↵
    Kokontis JM, Liao S. Molecular action of androgen in the normal and neoplastic prostate. Vitam Horm 1999;55:219–307.
    OpenUrlPubMed
  33. ↵
    Bouchal J, Baumforth K, Svachova M, Murray P, von Angerer E, Kolar Z. Microarray analysis of bicalutamide action on telomerase activity, p53 pathway, and viability of prostate carcinoma cell lines. J Pharm Pharmacol 2005;57:83–92.
    OpenUrlCrossRefPubMed
  34. ↵
    Wang LG, Ossowski L, Ferrari AC. Androgen receptor level controlled by a suppressor complex lost in an androgen-independent prostate cancer cell line. Oncogene 2004;23:5175–84.
    OpenUrlCrossRefPubMed
  35. ↵
    Rosenhagen MC, Soti C, Schmidt U, et al. The heat shock protein 90-targeting drug cisplatin selectively inhibits steroid receptor activation. Mol Endocrinol 2003;17:1991–2001.
    OpenUrlCrossRefPubMed
  36. Vanaja DK, Mitchell SH, Toft DO, Young CY. Effect of geldanamycin on androgen receptor function and stability. Cell Stress Chaperones 2002;7:55–64.
    OpenUrlCrossRefPubMed
  37. ↵
    Solit DB, Zheng FF, Drobnjak M, et al. 17-Allylamino-17-demethoxygeldanamycin induces the degradation of androgen receptor and HER-2/neu and inhibits the growth of prostate cancer xenografts. Clin Cancer Res 2002;8:986–93.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    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 2002;94:504–13.
    OpenUrlAbstract/FREE Full Text
  39. 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 2003;63:5126–35.
    OpenUrlAbstract/FREE Full Text
  40. ↵
    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 2005;4:1311–9.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    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 2003;63:8420–7.
    OpenUrlAbstract/FREE Full Text
  42. Rahmani M, Reese E, Dai Y, et al. Cotreatment with suberanoylanilide hydroxamic acid and 17-allylamino 17-demethoxygeldanamycin synergistically induces apoptosis in Bcr-Abl+ cells sensitive and resistant to STI571 (imatinib mesylate) in association with down-regulation of Bcr-Abl, abrogation of signal transducer and activator of transcription 5 activity, and Bax conformational change. Mol Pharmacol 2005;67:1166–76.
    OpenUrlAbstract/FREE Full Text
  43. ↵
    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 2005;105:1768–76.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Molecular Cancer Therapeutics: 6 (1)
January 2007
Volume 6, Issue 1
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Molecular Cancer Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Suberoylanilide hydroxamic acid (vorinostat) represses androgen receptor expression and acts synergistically with an androgen receptor antagonist to inhibit prostate cancer cell proliferation
(Your Name) has forwarded a page to you from Molecular Cancer Therapeutics
(Your Name) thought you would be interested in this article in Molecular Cancer Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Suberoylanilide hydroxamic acid (vorinostat) represses androgen receptor expression and acts synergistically with an androgen receptor antagonist to inhibit prostate cancer cell proliferation
Deborah L. Marrocco, Wayne D. Tilley, Tina Bianco-Miotto, Andreas Evdokiou, Howard I. Scher, Richard A. Rifkind, Paul A. Marks, Victoria M. Richon and Lisa M. Butler
Mol Cancer Ther January 1 2007 (6) (1) 51-60; DOI: 10.1158/1535-7163.MCT-06-0144

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Suberoylanilide hydroxamic acid (vorinostat) represses androgen receptor expression and acts synergistically with an androgen receptor antagonist to inhibit prostate cancer cell proliferation
Deborah L. Marrocco, Wayne D. Tilley, Tina Bianco-Miotto, Andreas Evdokiou, Howard I. Scher, Richard A. Rifkind, Paul A. Marks, Victoria M. Richon and Lisa M. Butler
Mol Cancer Ther January 1 2007 (6) (1) 51-60; DOI: 10.1158/1535-7163.MCT-06-0144
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Dihydrofolate reductase amplification and sensitization to methotrexate of methotrexate-resistant colon cancer cells
  • MAPK-independent impairment of T-cell responses by the multikinase inhibitor sorafenib
  • Interruption of RNA processing machinery by a small compound, 1-[(4-chlorophenyl)methyl]-1H-indole-3-carboxaldehyde (oncrasin-1)
Show more Research Articles: Therapeutics, Targets, and Development
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About MCT

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Molecular Cancer Therapeutics
eISSN: 1538-8514
ISSN: 1535-7163

Advertisement