Basic Research Open Access
Copyright ©2007 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Sep 7, 2007; 13(33): 4458-4466
Published online Sep 7, 2007. doi: 10.3748/wjg.v13.i33.4458
Histone deacetylase inhibitor MS-275 alone or combined with bortezomib or sorafenib exhibits strong antiproliferative action in human cholangiocarcinoma cells
Viola Baradari, Michael Höpfner, Alexander Huether, Institute of Physiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
Detlef Schuppan, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States
Hans Scherübl, Klinik für Gastroenterologie und Gastrointestinale Onkologie, Klinikum Am Urban, Vivantes Netzwerk für Gesundheit, Berlin, Germany
Author contributions: All authors contributed equally to the work.
Supported by a scholarship from the Sonnenfeld-Stiftung, Berlin, Germany for Viola Baradari
Correspondence to: Professor Hans Scherübl, Klinik für Gastroenterologie und Gastrointestinale Onkologie, Klinikum Am Urban, Vivantes Netzwerk für Gesundheit, Dieffenbachstrasse 1, 10967 Berlin, Germany. hans.scheruebl@vivantes.de
Telephone: +49-30-69725201 Fax: +49-30-69725205
Received: February 28, 2007
Revised: April 13, 2007
Accepted: April 18, 2007
Published online: September 7, 2007

Abstract

AIM: To investigate the antiproliferative effect of the histone deacetylase (HDAC) inhibitor MS-275 on cholangiocarcinoma cells alone and in combination with conventional cytostatic drugs (gemcitabine or doxorubicin) or the novel anticancer agents sorafenib or bortezomib.

METHODS: Two human bile duct adenocarcinoma cell lines (EGI-1 and TFK-1) were studied. Crystal violet staining was used for detection of cell number changes. Cytotoxicity was determined by measuring the release of the cytoplasmic enzyme lactate dehydrogenase (LDH). Apoptosis was determined by measuring the enzyme activity of caspase-3. Cell cycle status reflected by the DNA content was detected by flow cytometry.

RESULTS: MS-275 treatment potently inhibited the proliferation of EGI-1 and TFK-1 cholangiocarcinoma cells by inducing apoptosis and cell cycle arrest. MS-275-induced apoptosis was characterized by activation of caspase-3, up-regulation of Bax and down-regulation of Bcl-2. Cell cycle was predominantly arrested at the G1/S checkpoint, which was associated with induction of the cyclin-dependent kinase inhibitor p21Waf/CIP1. Furthermore, additive anti-neoplastic effects were observed when MS-275 treatment was combined with gemcitabine or doxorubicin, while combination with the multi-kinase inhibitor sorafenib or the proteasome inhibitor bortezomib resulted in overadditive anti-neoplastic effects.

CONCLUSION: The growth of human cholangiocarcinoma cells can be potently inhibited by MS-275 alone or in combination with conventional cytostatic drugs or new, targeted anticancer agents.

Key Words: Apoptosis, Cholangiocarcinoma, Bortezomib, Combination treatment, Histone deacetylase inhibitor, MS-275, Proteasome inhibitor, Sorafenib



INTRODUCTION

Cholangiocarcinoma (CC) accounts for 3% of all gastrointestinal cancers[1] and is the second commonest primary hepatic tumor[1,2]. It is characterized by the malignant proliferation of cholangiocytes that line intra-hepatic and extra-hepatic bile ducts and ductules. Biliary tumors are highly malignant and have a poor prognosis[3]. The incidence of intra-hepatic CC is rising in North America, Australia and Asia[2,4,5]. In addition to the well described risk factors, such as primary sclerosing cholangitis, liver fluke infestations or hepatolithiasis[6], recent studies suggest that chronic hepatitis B and C infections, HIV, non-alcoholic steatohepatitis, especially when combined with cirrhosis, also contribute to intra-hepatic CC risk[7]. On the other hand, the incidence of extra-hepatic CC is declining[2,4,5,8] most likely as a result of increasing rates of cholecystectomy over the past decades[2,5].

Treatment options for cholangiocarcinoma are limited. Unfortunately, the majority of patients suffer from advanced CC at presentation. Therefore, curative surgical resection or liver transplantation can only be offered to a minority of CC patients, leaving biliary drainage, radiotherapy or conventional chemotherapy as unsatisfactory palliative treatment options for advanced CC[6], with marginal effect on survival or quality of life[9].

Histone deacetylase (HDAC) inhibitors receive growing interest as cancer therapeutics due to their ability to induce cell differentiation, growth arrest and apoptosis[10]. Acetylation and deacetylation of histones play an important role in the regulation of gene transcription and in the modulation of chromatin structure[11,12]. The steady state of histone acetylation is tightly controlled by antagonistic effects of histone acetyltransferases (HAT) and HDAC. Aberrant gene expression resulting in functional inactivation of HAT activity or over-expression of HDAC can promote tumor cell proliferation and survival[13]. Moreover, deregulation of HDAC recruitment to transcriptional promoters is a mechanism by which these enzymes contribute to tumorigenesis[14].

HDAC inhibitor monotherapy can inhibit the growth of various tumors in vitro and in vivo[11,15,17]. Importantly, HDAC inhibitors are relatively non-toxic to non-transformed cells[18,19], leading to their evaluation in phaseI/II clinical cancer trials[14,15,20].

The synthetic orally available HDAC inhibitor, MS-275, potently inhibits histone deacetylases of several human tumor cells[21]. With a benzamide backbone, MS-275 is structurally unrelated to previous HDAC inhibitors, while showing a 30-fold stronger HDAC inhibitory activity than other natural HDAC inhibitors like sodium butyrate[22]. Recently, we and others demonstrated strong anti-proliferative activity of MS-275 towards several human cancer cells in vitro and in vivo[21,23,24]. MS-275 has now entered clinical trials both for single and combination therapy in solid and haematological malignancies. Since HDAC inhibition has not yet been evaluated for its anti-neoplastic effects on cholangiocarcinoma, we characterized the anti-neoplastic potency of the HDAC inhibitor MS-275 in human CC cells. We showed that MS-275 potently inhibited growth of CC cells, especially in combination with conventional cytostatic drugs or new, targeted anticancer agents, such as sorafenib (NexavarTM) or bortezomib (VelcadeTM). Furthermore, we provided an insight into major underlying mechanisms of MS-275-induced growth inhibition of CC cells.

MATERIALS AND METHODS
Cell lines and drugs

The poorly differentiated human bile duct adenocarcinoma cell line EGI-1[25] (DSMZ # ACC385) and the human papillary bile duct adenocarcinoma cell line TFK-1[26] (DSMZ # ACC344) were derived from patient cells prior to any exposure to chemotherapy or radiotherapy. Both cell lines were cultured in RPMI 1640 medium supplemented with 100 mL/L fetal calf serum (FCS), 100 kU/L penicillin and 100 mg/L streptomycin (Biochrom, Berlin, Germany) and kept at 37°C in a humidified atmosphere containing 50 mL/L CO2 in air.

MS-275 (N-(2-Aminophenyl)-4-[N-(3-pyridineyl-methoxycarbonyl)aminomethyl]-benzamide) was purchased from ALEXIS Biochemicals (Lausen, Switzerland). The 26S proteasome inhibitor bortezomib (VelcadeTM) was bought from Millennium Pharmaceuticals, Inc. (Cambridge, MA, USA). The multi-kinase inhibitor sorafenib tosylate (NexavarTM) was a kind gift from Bayer Health Care (West Haven, CT, USA). Stock solutions were prepared in dimethyl sulfoxide (DMSO) and stored at -20°C until use. Gemcitabine hydrochloride (GemzarTM) was bought from Lilly Pharma (Gieβen, Germany). Doxorubicin hydrochloride was from Sigma (Deisenhofen, Germany) and also prepared in DMSO and stored at -20°C. All drugs were diluted in fresh medium before each experiment. In all experiments, the final DMSO concentration was ≤ 5 g/L, not affecting cell growth. To evaluate the effects of the drugs, cells were incubated with either control medium or medium containing rising concentrations of the respective drug(s). Cell culture material was from Biochrom (Berlin, Germany). All other chemicals were from Sigma if not stated otherwise.

Measurement of growth inhibition

Drug-induced changes in cell numbers of EGI-1 and TFK-1 cells were evaluated by crystal violet staining as previously described[27]. In brief, cells in 96-well microtiter plates were fixed with 10 g/L glutaraldehyde. Then cells were stained with 1 g/L crystal violet in phosphate buffer solution (PBS). The unbound dye was removed by washing with water. Bound crystal violet was solubilized with 2 mL/L Triton-X-100 in PBS. Light extinction which increases linearly with the cell number was analyzed at 570 nm using an ELISA-Reader. To check for possible overadditive anti-proliferative effects, combination treatments of MS-275 plus conventional cytostatics (gemcitabine or doxorubicin) or plus sorafenib or plus bortezomib were performed. Increasing concentrations of the respective drug were combined with 0.25 and/or 0.5 μmol/L MS-275. The anti-neoplastic effects of the combination therapies were compared to those of each drug alone. Concentration range and effectiveness of the respective drugs have been determined in prior experiments.

Determination of cytotoxicity

Cells were seeded at a density of 8000 cells/well into 96-well microtiter plates and incubated with rising concentrations of MS-275 for 1, 2.5, 5 or 24 h. Release of the cytoplasmic enzyme lactate dehydrogenase (LDH), indicating cytotoxicity, was measured by using a colorimetric kit from Roche Diagnostics (Mannheim, Germany) as described elsewhere[28].

Detection of apoptosis

Preparation of cell lysates and determination of caspase-3 activity were performed as described previously[27]. The activity of caspase-3 was calculated from cleavage of the fluorogenic substrate Ac-DEVD-AMC (Calbiochem, Bad Soden, Germany). Cell lysates were incubated with substrate solution (caspase-3 substrate Ac-DEVD-AMC 20 mg/L, HEPES 20 mmol/L, glycerol 100 mL/L, DTT 2 mmol/L, pH 7.5) for 1 h at 37°C, and substrate cleavage was measured with a VersaFluor fluorometer (excitation: 360 nm emission: 460 nm) from Biorad (Munich, Germany).

Cell cycle analysis

Cell cycle analysis was performed by the method of Vindelov and Christensen as described previously[29]. Cells were trypsinized, washed and the nuclei were isolated using the CycleTest PLUS DNA Reagent Kit (Becton Dickinson, Heidelberg, Germany). DNA was stained with propidium iodide according to the manufacturers’ instructions. The DNA content of the nuclei was measured by flow cytometry and analyzed using ModFit software (Becton Dickinson, Heidelberg, Germany).

Western blot analysis

Western blotting was performed as described previously[30]. In brief, whole-cell extracts were prepared by lysing cells. Lysates containing 30 μg protein were subjected to gel electrophoresis. Proteins were then transferred onto PVDF membranes by electroblotting for 90 min. Blots were blocked with 50 g/L non-fat dry milk in TBS-Tween-solution for 1 h at room temperature, and then incubated at 4°C overnight with antibodies directed against anti-human Bax (1:1000), p27Kip1 (1:200) (both from Santa Cruz Biotechnology, CA, USA), Bcl-2 (1:1000), or p21Waf-1/CIP1 (1:1000) (both from Cell Signaling, MA USA). Anti-β-actin (1:5000) from Sigma (Deisenhofen, Germany) served as loading control. After incubation with horseradish peroxidase-coupled anti-IgG antibodies (1:10 000, GE Healthcare, Uppsala, Sweden) at room temperature for at least 1 h, the blot was developed using enhanced chemiluminescent detection (GE Healthcare) and subsequently exposed to Hyperfilm ECL film (GE Healthcare, Uppsala, Sweden).

Statistical analysis

If not stated otherwise, data were expressed as means of at least three independent experiments ± SEM. Significance between controls and treated samples was calculated by Student’s two-sided t-test. Caspase-3 measurements were evaluated using the two-sided Welch t-test. P < 0.05 was considered statistically significant.

RESULTS
Growth inhibitory effects of the HDAC inhibitor MS-275

The growth inhibitory effects of MS-275 on CC cells were studied by crystal violet staining. Treating EGI-1 and TFK-1 cells with 0.25-2.5 μmol/L MS-275 for 3 d reduced the growth of both cell lines in a dose-dependent manner by up to 100% (Figure 1). The IC50 value of MS-275, determined after 72 h of incubation, was 0.48 ± 0.02 μmol/L for EGI-1 cells and 0.46 ± 0.02 μmol/L for TFK-1 cells. Determination of growth inhibition after 24 h and 48 h of incubation revealed that MS-275 inhibited cell proliferation in a time-dependent manner (data not shown).

Figure 1
Figure 1 Anti-proliferative effects of MS-275 on human EGI-1 and TFK-1 cholangiocarcinoma cells. mean ± SEM, n > 5, aP < 0.05 vs controls.
LDH release after MS-275 treatment

Cytotoxicity was evaluated by LDH release of the cells into the culture medium. Incubating EGI-1 and TFK-1 cells with 0.1-1 μmol/L MS-275 for up to 24 h did not result in a significant increase of LDH release (Figure 2), indicating that MS-275 does not directly affect cell membrane integrity and does not have immediate toxic effects even at higher concentrations.

Figure 2
Figure 2 MS-275-induced LDH release into the supernatant of EGI-1 and TFK-1 cells. mean ± SEM, n > 3 independent experiments; No significant increase in LDH release was observed.
Induction of apoptosis by MS-275

To determine the contribution of apoptosis to the observed growth inhibitory effect of MS-275, the activation of caspase-3, one of the key enzymes in the apoptotic signaling cascade, was determined after 24 h and 48 h of incubation. In both cell lines, HDAC inhibition by MS-275 resulted in a time- and dose-dependent increase of caspase-3 enzyme activity (Figure 3). After 48 h, 1 μmol/L MS-275 led to a 3-fold increase of caspase-3 activity in TFK-1 cells and even 7-fold increase in EGI-1 cells.

Figure 3
Figure 3 MS-275-induced apoptosis-specific caspase-3 activity increase in EGI-1 and TFK-1 cells. mean ± SEM, n > 3 independent experiments, aP < 0.05 vs controls.
Cell cycle regulation

To test whether an induction of cell cycle arrest contributed to the antiproliferative potency of MS-275 in cholangiocarcinoma cells, we performed flow cytometric cell cycle analyses. Incubating EGI-1 and TFK-1 cells with 0.1-1 μmol/L MS-275 for 24 h resulted in a dose-dependent arrest in the G0/G1 phase of the cell cycle, whereas the proportion of cells in the S phase significantly decreased (Figure 4). The G0/G1-phase arrest by MS-275 was significant above concentrations of 0.1 µmol/L in TFK-1 and 0.25 μmol/L in EGI-1 cells. Moreover, the proportion of cells in the G2/M phase also increased, indicating an additional block in the G2/M phase. The G2/M-phase arrest was more pronounced in TFK-1 cells.

Figure 4
Figure 4 Induction of cell cycle arrest by MS-275 on EGI-1 and TFK-1 cells. means of > 3 independent experiments, aP < 0.05 vs controls.
Bax, Bcl-2, p21Waf-1/CIP1 and p27Kip1 expression

To further characterize the apoptotic and cell cycle arresting effects of the HDAC inhibition, we performed Western blotting. Treating EGI-1 and TFK-1 cells for 48 h with MS-275 (0.1-1 μmol/L) resulted in a dose-dependent decrease of anti-apoptotic Bcl-2, whereas the expression of the pro-apoptotic Bax was increased in both cell lines (Figure 5A). Moreover, upon MS-275 treatment, a dose-dependent increase in the expression of p21Waf-1/CIP1 was observed. By contrast, no regulation of the cyclin-dependent kinase inhibitor (CDKI) p27Kip1 was detected (Figure 5B).

Figure 5
Figure 5 MS-275-induced modulation of apoptosis- and cell cycle-relevant proteins (n = 3 independent experiments).
Antineoplastic potency of MS-275 in combination with other drugs

To test potential (over-) additive anti-proliferative effects of MS-275-based combination treatment, EGI-1 or TFK-1 cells were treated with combinations of 0.5 μmol/L MS-275 plus either gemcitabine (0-500 nmol/L) or doxorubicin (0-100 nmol/L) for 2 d. In both combinations, an augmentation of the anti-proliferative effect was observed (Figure 6). Although the anti-proliferative efficacy of gemcitabine monotherapy differed in EGI-1 and TFK-1 cells, the addition of MS-275 caused an enhanced anti-proliferative efficacy in either cell line (Figure 6A and B). Similar results were obtained when co-administering MS-275 and doxorubicin, with additive growth inhibitory effects on both cell lines (Figure 6C and D).

Figure 6
Figure 6 Anti-proliferative effects of MS-275 plus cytostatics on EGI-1 and TFK-1 cells. mean ± SEM, n = 3-4 independent experiments.

Recently, we could demonstrate that the multi-kinase inhibitor sorafenib potently inhibited the proliferation of cholangiocarcinoma cells[31]. To check out the potency of inhibiting multiple targets of mitogenic signaling pathways for enhanced inhibition of cholangiocarcinoma cell growth, we treated EGI-1 and TFK-1 cells with MS-275 and sorafenib. After two days of incubation, a pronounced (over-)additive growth inhibitory effect of the combination was observed on both cell lines (Figure 7A and B).

Figure 7
Figure 7 Anti-proliferative effects of MS-275 plus sorafenib or bortezomib on EGI-1 and TFK-1. mean ± SEM, n = 3-4 independent experiments.

Applying the proteasome inhibitor bortezomib alone (1-10 nmol/L) for two days reduced the growth of TFK-1 and EGI-1 cells by up to 65% and 84%, respectively. Again, additive and overadditive growth inhibitory effects were observed, when bortezomib was combined with MS-275 (Figure 7C and D).

DISCUSSION

Treatment options of advanced cholangiocarcinoma (CC) are unsatisfactory, and the prognosis of patients suffering from advanced CC is poor. New, effective and well-tolerated therapy strategies are urgently needed.

Aberrant gene expressions resulting in functional inactivation of histone acetyltransferase (HAT) activity or over-expression of histone deacetylases (HDAC) have been shown to contribute to tumor cell proliferation[13]. For this reason, HDAC inhibition has been regarded as a promising anticancer strategy to inhibit the multiple cellular processes that are dysregulated in various neoplastic cells[14].

The novel synthetic benzamide derivative MS-275 is an orally available HDAC inhibitor which has been shown to exert strong anti-proliferative effects on a variety of human cancers[21], but its potential suitability for the treatment of cholangiocarcinomas have not been tested so far. Here, we provide evidence that inhibition of HDAC activity by MS-275, alone or in combination with conventional chemotherapeutics or new, targeted anticancer drugs, may be a promising approach for the treatment of cholangiocarcinoma.

MS-275 potently inhibited cell growth of the cholangiocarcinoma cells EGI-1 and TFK-1 in a time- and dose-dependent manner. Submicromolar concentrations of MS-275 were already sufficient to significantly inhibit the proliferation of EGI-1 and TFK-1 cells, and the concentration needed to induce halfmaximal anti-neoplastic effects (IC50 value) was approximately 0.5 μmol/L in both cell lines. This is comparable to our previous findings on the effects of MS-275 on the growth of gastrointestinal neuroendocrine tumor cells[24].

Recent studies have shown that inhibition of HDAC activity induces apoptosis in a variety of cancers, including breast and prostate cancer, neuroblastoma, hepatoma, gastrointestinal neuroendocrine tumor cells and some types of hematologic malignancies[24,32-34].

The mechanisms involved in the HDAC inhibitor-induced apoptosis are complex and differ among cell types[35]. They can involve both intrinsic pathways and extrinsic pathways of apoptosis. MS-275 can induce mitochondrial permeability transition with a subsequent release of pro-apoptotic cytochrome c into the cytosol, resulting in the activation of caspase-9 and caspase-3, thus triggering the intrinsic apoptotic pathway[36-38]. Additionally, other HDAC inhibitors have been shown to upregulate pro-apoptotic Fas, a member of the tumor necrosis factor receptor superfamily and the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL-) receptors/death receptors DR4 and DR5, thereby triggering the extrinsic pathway, paralleled by down-regulation of the caspase-8 inhibitor c-FLIP, leading to caspase-8 and subsequently to caspase-3 activation[39,40]. Moreover, an altered expression of several pro- and anti-apoptotic intracellular genes by HDAC inhibitors has been reported. Up-regulation of pro-apoptotic Bak and induction of conformational changes of the pro-apoptotic protein Bax are some of the HDAC inhibitor-induced upstream events that trigger the mitochondrial pathway of apoptosis[33,41,42]. HDAC inhibitors can also down-regulate anti-apoptotic proteins, such as Bcl-2, Bcl-XL, XIAP, Mcl-1, and survivin[37,43], further tilting the balance in favor of apoptosis. Here, we demonstrated that the pro-apoptotic effect of HDAC inhibition by MS-275 in CC cells is mediated by the activation of caspase-3 and a shift in the balance of pro-apoptotic Bax over anti-apoptotic Bcl-2.

HDAC-inhibition-induced changes in the expression pattern of apoptosis-related proteins seem to be cell type-specific. For example, in hepatoma and breast cancer cells, HDAC inhibition decreased the expression of Bcl-2, while expression of pro-apoptotic Bax was increased[33,44], in line with the present study. On the other hand, no changes in the expression pattern of Bax and Bcl-2 were found in HDAC-inhibitor-treated glioma cells[45], and in gastrointestinal neuroendocrine tumor cells, Bcl-2 expression was down-regulated upon MS-275 treatment, while Bax remained unchanged[24]. Especially for an estimation of suitable combination treatment regimens being based on the enhanced induction of apoptosis, it is necessary to check out cell type-specific changes in the expression pattern of HDAC-induced pro- and anti-apoptotic proteins.

To further characterize the growth inhibitory activity of MS-275, we performed cell cycle analyses. Upon treatment with MS-275 we found that cell cycle progression was blocked both in the G0/G1 and the G2/M phase. The induction of cell cycle arrest was associated with an increase in the expression of the cyclin-dependent kinase inhibitor (CDKI) p21Waf-1/Cip1. p21Waf-1/CIP1 is a key component of cell cycle checkpoints, i.e., the G1/S[46] and the G2/M checkpoints[47,48]. Accordingly, HDAC inhibitors were found to inhibit both the G1/S and G2/M transition[49-51].

The combination of HDAC inhibitors with existing chemotherapeutic agents appears to be a promising approach to reduce the dose of other anti-neoplastic drugs and to overcome drug resistance. For non-cholangiocarcinoma tumors (e.g. glioblastoma and breast cancer cells), a potentiation of anti-neoplastic effects has already been described for HDAC inhibitors combined with cytostatic drugs[52]. However, except for the interaction of HDAC and topoisomerase inhibitors, in which HDAC-inhibitor-mediated increases in topoisomerase II levels appear to contribute to lethality[53], little is known about the mechanisms by which HDAC inhibitors increase the anti-neoplastic efficacy of cytostatic agents. Maggio et al[37] reported that combination of MS-275 with nucleoside analogues (e.g. Fludarabine) produced high synergy in triggering mitochondrial dysfunction and caspase activation in human leukemia cells. In our study, the nucleoside analogue gemcitabine and the topoisomerase-II-inhibitor doxorubicin were chosen for the evaluation of synergy with MS-275 in CC cells. While both combinations resulted in additive anti-proliferative effects, further studies are needed to clarify the exact mechanisms underlying the observed potentiation by MS-275. Nevertheless, the fact that MS-275-based combination therapies with cytostatics lead to an enhanced growth inhibition of CC cells gives rise to the hope that a HDAC-inhibitor-based combination therapy may hold a promise for more effective treatment of CC.

The same holds true for the combination of HDAC inhibition and multi-kinase inhibition by sorafenib, which mainly targets raf and ras kinases that are members of the MAPK (mitogen-activated protein kinase) pathway. The MAPK pathway is centrally involved in the regulation of multiple cellular functions, such as cell growth, apoptosis and cell cycle progression[54]. Recently, we could demonstrate a potent growth inhibition of CC cells by sorafenib monotherapy[31]. HDAC inhibitors can also down-regulate the MAPK-signaling pathway, as exemplified by HDAC-induced growth inhibition of transformed hepatocytes due to the suppression of oncogenic ras and ERK1/2 (extracellular regulated kinase)[55]. Here we found that coapplication of MS-275 and sorafenib resulted in an additive to overadditive growth inhibition of CC cells, indicating that HDAC-based dual-targeting of MAPK and pro-apoptotic pathways may have promising clinical implications for the future treatment of CC.

Finally, we also studied how far combination treatment of MS-275 together with the proteasome inhibitor bortezomib could enhance the anti-neoplastic effects of the monotherapies. Bortezomib has recently been shown to enhance the anti-proliferative effects of HDAC inhibitors on several non-CC cancer cells[56-58]. The sensitizing effect of bortezomib has been attributed to a blockade of the cytoprotective nuclear factor kappa B (NFκB)[59,60]. There is accumulating evidence that NFκB activation status plays an important role in regulating the response of cells to HDAC inhibitors[61]. Thus, in non-small cell lung cancer, HDAC inhibitors are only modestly effective due to high steady state levels of NFκB[11,57], but addition of bortezomib dramatically increased the pro-apoptotic effect of the HDAC inhibitor sodium butyrate[57]. Our findings are in line with these data in clearly demonstrating that bortezomib overadditively enhanced the anti-proliferative effect of MS-275 on CC cells. Thus, the combined treatment with HDAC inhibitors and bortezomib (and possibly sorafenib as a third agent) appears to be a promising approach for the treatment of cholangiocarcinomas.

In conclusion, our study provides first evidence that the HDAC inhibitor MS-275 potently inhibits the growth of human CC cells by inducing both cell cycle arrest and apoptosis. Importantly, we could demonstrate that combination treatment of MS-275 with gemcitabine, doxorubicin, and especially sorafenib or bortezomib leads to (over-)additive growth inhibitory effects. Our study may provide a rationale for future in vivo evaluations of MS-275 in combination therapies for growth control of advanced cholangiocarcinomas.

ACKNOWLEDGMENTS

We thank André Bosch and Björn Hoffmann for expert technical assistance, and Bayer Health Care (West Haven, CT, USA) for providing us with sorafenib.

COMMENTS
Background

Cholangiocarcinomas (CC) are highly malignant and have a poor prognosis. Since there is no satisfactory therapy for advanced CC, we studied the effects of the histone deacetylase inhibitor MS-275 alone or in combination with bortezomib or sorafenib on the proliferation of cholangiocarcinoma cells.

Research frontiers

Histone modifications have been identified to play prominent roles in the epigenetic regulation of gene transcription. Accumulating evidence indicates that dysregulation of epigenetic processes causes transcriptional repression of a subset of genes, contributing to the pathogenesis of many human diseases, including cancer. In the past decade, substantial progress has been made in understanding the relationship between aberrant epigenetic changes and tumorigenesis. Enzymes involved in these epigenetic events include histone acetyltransferases (HAT) and histone deacetylases (HDAC), which tightly control the steady state of histone acetylation. Recently, histone deacetylase inhibitors receive growing interest as cancer therapeutics, due to their ability to induce growth arrest, differentiation and/or apoptosis.

Innovations and breakthroughs

Previous studies already demonstrated that targeting the histone deacetylase (HDAC) activity by specific HDAC inhibitors is an effective treatment option for cell growth control of haematological malignancies and of several solid tumors. Especially the novel, orally available HDAC inhibitor MS-275 has been shown to exert strong anti-proliferative effects on a variety of cancers.

Applications

In this paper, we studied the anti-proliferative effect of the potent histone deacetylase inhibitor MS-275 on cholangiocarcinoma cells for the first time. MS-275 significantly inhibits the proliferation of cholangiocarcinoma cells by inducing apoptosis and cell cycle arrest. Importantly, we could further demonstrate that combination treatment using MS-275 together with doxorubicin, gemcitabine, sorafenib or bortezomib, respectively, leads to (over-)additive growth inhibitory effects, thus providing a rationale for future in vivo evaluations.

Terminology

Histone acetyltransferases (HAT) and histone deacetylases (HDAC): HAT and HDAC are two sets of histone-modifying enzymes, which control the steady state of histone acetylation. The equilibrium of acetylation (HAT) and deacetylation (HDAC) of histones plays an important role in the modulation of chromatin structure and in the regulation of gene transcription.

Peer review

This manuscript is a good suggestion to be considered and it is true that the treatment options for advanced cholangiocarcinomas are still unsatisfactory. New therapeutic approaches are urgently needed.

Footnotes

S- Editor Ma N L- Editor Kumar M E- Editor Wang HF

References
1.  Vauthey JN, Blumgart LH. Recent advances in the management of cholangiocarcinomas. Semin Liver Dis. 1994;14:109-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 227]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
2.  Khan SA, Taylor-Robinson SD, Toledano MB, Beck A, Elliott P, Thomas HC. Changing international trends in mortality rates for liver, biliary and pancreatic tumours. J Hepatol. 2002;37:806-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 399]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
3.  Gores GJ. Cholangiocarcinoma: current concepts and insights. Hepatology. 2003;37:961-969.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 195]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
4.  Patel T. Increasing incidence and mortality of primary intrahepatic cholangiocarcinoma in the United States. Hepatology. 2001;33:1353-1357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 773]  [Cited by in F6Publishing: 756]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]
5.  Taylor-Robinson SD, Toledano MB, Arora S, Keegan TJ, Hargreaves S, Beck A, Khan SA, Elliott P, Thomas HC. Increase in mortality rates from intrahepatic cholangiocarcinoma in England and Wales 1968-1998. Gut. 2001;48:816-820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 298]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
6.  Khan SA, Thomas HC, Davidson BR, Taylor-Robinson SD. Cholangiocarcinoma. Lancet. 2005;366:1303-1314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 872]  [Cited by in F6Publishing: 867]  [Article Influence: 45.6]  [Reference Citation Analysis (0)]
7.  Shaib YH, El-Serag HB, Davila JA, Morgan R, McGlynn KA. Risk factors of intrahepatic cholangiocarcinoma in the United States: a case-control study. Gastroenterology. 2005;128:620-626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 401]  [Cited by in F6Publishing: 364]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
8.  Shaib Y, El-Serag HB. The epidemiology of cholangiocarcinoma. Semin Liver Dis. 2004;24:115-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 839]  [Cited by in F6Publishing: 815]  [Article Influence: 40.8]  [Reference Citation Analysis (0)]
9.  Patel T. Cholangiocarcinoma. Nat Clin Pract Gastroenterol Hepatol. 2006;3:33-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 217]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
10.  Pandolfi PP. Histone deacetylases and transcriptional therapy with their inhibitors. Cancer Chemother Pharmacol. 2001;48 Suppl 1:S17-S19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 55]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
11.  Marks P, Rifkind RA, Richon VM, Breslow R, Miller T, Kelly WK. Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer. 2001;1:194-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1425]  [Cited by in F6Publishing: 1473]  [Article Influence: 64.0]  [Reference Citation Analysis (0)]
12.  Kouraklis G, Theocharis S. Histone deacetylase inhibitors and anticancer therapy. Curr Med Chem Anticancer Agents. 2002;2:477-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 60]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
13.  Kouzarides T. Histone acetylases and deacetylases in cell proliferation. Curr Opin Genet Dev. 1999;9:40-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 456]  [Cited by in F6Publishing: 457]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
14.  Monneret C. Histone deacetylase inhibitors. Eur J Med Chem. 2005;40:1-13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 286]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
15.  Krämer OH, Göttlicher M, Heinzel T. Histone deacetylase as a therapeutic target. Trends Endocrinol Metab. 2001;12:294-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 202]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
16.  Vigushin DM, Ali S, Pace PE, Mirsaidi N, Ito K, Adcock I, Coombes RC. Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo. Clin Cancer Res. 2001;7:971-976.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Johnstone RW. Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov. 2002;1:287-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1146]  [Cited by in F6Publishing: 1119]  [Article Influence: 50.9]  [Reference Citation Analysis (0)]
18.  Byrd JC, Shinn C, Ravi R, Willis CR, Waselenko JK, Flinn IW, Dawson NA, Grever MR. Depsipeptide (FR901228): a novel therapeutic agent with selective, in vitro activity against human B-cell chronic lymphocytic leukemia cells. Blood. 1999;94:1401-1408.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Rosato RR, Grant S. Histone deacetylase inhibitors in cancer therapy. Cancer Biol Ther. 2003;2:30-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 116]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
20.  Acharya MR, Sparreboom A, Venitz J, Figg WD. Rational development of histone deacetylase inhibitors as anticancer agents: a review. Mol Pharmacol. 2005;68:917-932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 180]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
21.  Saito A, Yamashita T, Mariko Y, Nosaka Y, Tsuchiya K, Ando T, Suzuki T, Tsuruo T, Nakanishi O. A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proc Natl Acad Sci USA. 1999;96:4592-4597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 545]  [Cited by in F6Publishing: 542]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
22.  Suzuki T, Ando T, Tsuchiya K, Fukazawa N, Saito A, Mariko Y, Yamashita T, Nakanishi O. Synthesis and histone deacetylase inhibitory activity of new benzamide derivatives. J Med Chem. 1999;42:3001-3003.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 224]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
23.  Rosato RR, Almenara JA, Grant S. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1 1. Cancer Res. 2003;63:3637-3645.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Baradari V, Huether A, Höpfner M, Schuppan D, Scherübl H. Antiproliferative and proapoptotic effects of histone deacetylase inhibitors on gastrointestinal neuroendocrine tumor cells. Endocr Relat Cancer. 2006;13:1237-1250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 56]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
25.  International Conference on Tumor Necrosis Factor and Related Cytotoxins. September 14-18, 1987, Heidelberg, Federal Republic of Germany. Abstracts. Immunobiology. 1987;175:1-143.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Saijyo S, Kudo T, Suzuki M, Katayose Y, Shinoda M, Muto T, Fukuhara K, Suzuki T, Matsuno S. Establishment of a new extrahepatic bile duct carcinoma cell line, TFK-1. Tohoku J Exp Med. 1995;177:61-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 102]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
27.  Sutter AP, Maaser K, Grabowski P, Bradacs G, Vormbrock K, Höpfner M, Krahn A, Heine B, Stein H, Somasundaram R. Peripheral benzodiazepine receptor ligands induce apoptosis and cell cycle arrest in human hepatocellular carcinoma cells and enhance chemosensitivity to paclitaxel, docetaxel, doxorubicin and the Bcl-2 inhibitor HA14-1. J Hepatol. 2004;41:799-807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 42]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
28.  Hopfner M, Sutter AP, Huether A, Baradari V, Scherubl H. Tyrosine kinase of insulin-like growth factor receptor as target for novel treatment and prevention strategies of colorectal cancer. World J Gastroenterol. 2006;12:5635-5643.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Maaser K, Höpfner M, Jansen A, Weisinger G, Gavish M, Kozikowski AP, Weizman A, Carayon P, Riecken EO, Zeitz M. Specific ligands of the peripheral benzodiazepine receptor induce apoptosis and cell cycle arrest in human colorectal cancer cells. Br J Cancer. 2001;85:1771-1780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 99]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
30.  Höpfner M, Sutter AP, Huether A, Ahnert-Hilger G, Scherübl H. A novel approach in the treatment of neuroendocrine gastrointestinal tumors: additive antiproliferative effects of interferon-gamma and meta-iodobenzylguanidine. BMC Cancer. 2004;4:23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
31.  Huether A, Höpfner M, Baradari V, Schuppan D, Scherübl H. Sorafenib alone or as combination therapy for growth control of cholangiocarcinoma. Biochem Pharmacol. 2007;73:1308-1317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 65]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
32.  Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Natl Cancer Inst. 2000;92:1210-1216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 883]  [Cited by in F6Publishing: 879]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
33.  Herold C, Ganslmayer M, Ocker M, Hermann M, Geerts A, Hahn EG, Schuppan D. The histone-deacetylase inhibitor Trichostatin A blocks proliferation and triggers apoptotic programs in hepatoma cells. J Hepatol. 2002;36:233-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 107]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
34.  Henderson C, Mizzau M, Paroni G, Maestro R, Schneider C, Brancolini C. Role of caspases, Bid, and p53 in the apoptotic response triggered by histone deacetylase inhibitors trichostatin-A (TSA) and suberoylanilide hydroxamic acid (SAHA). J Biol Chem. 2003;278:12579-12589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 127]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
35.  Duan H, Heckman CA, Boxer LM. Histone deacetylase inhibitors down-regulate bcl-2 expression and induce apoptosis in t(14; 18) lymphomas. Mol Cell Biol. 2005;25:1608-1619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 202]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
36.  Rosato RR, Wang Z, Gopalkrishnan RV, Fisher PB, Grant S. Evidence of a functional role for the cyclin-dependent kinase-inhibitor p21WAF1/CIP1/MDA6 in promoting differentiation and preventing mitochondrial dysfunction and apoptosis induced by sodium butyrate in human myelomonocytic leukemia cells (U937). Int J Oncol. 2001;19:181-191.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Maggio SC, Rosato RR, Kramer LB, Dai Y, Rahmani M, Paik DS, Czarnik AC, Payne SG, Spiegel S, Grant S. The histone deacetylase inhibitor MS-275 interacts synergistically with fludarabine to induce apoptosis in human leukemia cells. Cancer Res. 2004;64:2590-2600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 100]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
38.  Roh MS, Kim CW, Park BS, Kim GC, Jeong JH, Kwon HC, Suh DJ, Cho KH, Yee SB, Yoo YH. Mechanism of histone deacetylase inhibitor Trichostatin A induced apoptosis in human osteosarcoma cells. Apoptosis. 2004;9:583-589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
39.  Bhalla K, List A. Histone deacetylase inhibitors in myelodysplastic syndrome. Best Pract Res Clin Haematol. 2004;17:595-611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 13]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
40.  Natoni F, Diolordi L, Santoni C, Gilardini Montani MS. Sodium butyrate sensitises human pancreatic cancer cells to both the intrinsic and the extrinsic apoptotic pathways. Biochim Biophys Acta. 2005;1745:318-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
41.  Fuino L, Bali P, Wittmann S, Donapaty S, Guo F, Yamaguchi H, Wang HG, Atadja P, Bhalla K. Histone deacetylase inhibitor LAQ824 down-regulates Her-2 and sensitizes human breast cancer cells to trastuzumab, taxotere, gemcitabine, and epothilone B. Mol Cancer Ther. 2003;2:971-984.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Johnstone RW, Licht JD. Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell. 2003;4:13-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 364]  [Cited by in F6Publishing: 378]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
43.  Guo F, Sigua C, Tao J, Bali P, George P, Li Y, Wittmann S, Moscinski L, Atadja P, Bhalla K. Cotreatment with histone deacetylase inhibitor LAQ824 enhances Apo-2L/tumor necrosis factor-related apoptosis inducing ligand-induced death inducing signaling complex activity and apoptosis of human acute leukemia cells. Cancer Res. 2004;64:2580-2589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 162]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
44.  Singh TR, Shankar S, Srivastava RK. HDAC inhibitors enhance the apoptosis-inducing potential of TRAIL in breast carcinoma. Oncogene. 2005;24:4609-4623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 145]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
45.  Sawa H, Murakami H, Ohshima Y, Sugino T, Nakajyo T, Kisanuki T, Tamura Y, Satone A, Ide W, Hashimoto I. Histone deacetylase inhibitors such as sodium butyrate and trichostatin A induce apoptosis through an increase of the bcl-2-related protein Bad. Brain Tumor Pathol. 2001;18:109-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 65]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
46.  Yamashita Y, Shimada M, Harimoto N, Rikimaru T, Shirabe K, Tanaka S, Sugimachi K. Histone deacetylase inhibitor trichostatin A induces cell-cycle arrest/apoptosis and hepatocyte differentiation in human hepatoma cells. Int J Cancer. 2003;103:572-576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 125]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
47.  Noh EJ, Lee JS. Functional interplay between modulation of histone deacetylase activity and its regulatory role in G2-M transition. Biochem Biophys Res Commun. 2003;310:267-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
48.  Peart MJ, Tainton KM, Ruefli AA, Dear AE, Sedelies KA, O'Reilly LA, Waterhouse NJ, Trapani JA, Johnstone RW. Novel mechanisms of apoptosis induced by histone deacetylase inhibitors. Cancer Res. 2003;63:4460-4471.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Fournel M, Trachy-Bourget MC, Yan PT, Kalita A, Bonfils C, Beaulieu C, Frechette S, Leit S, Abou-Khalil E, Woo SH. Sulfonamide anilides, a novel class of histone deacetylase inhibitors, are antiproliferative against human tumors. Cancer Res. 2002;62:4325-4330.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Takai N, Desmond JC, Kumagai T, Gui D, Said JW, Whittaker S, Miyakawa I, Koeffler HP. Histone deacetylase inhibitors have a profound antigrowth activity in endometrial cancer cells. Clin Cancer Res. 2004;10:1141-1149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 171]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
51.  Ryu JK, Lee WJ, Lee KH, Hwang JH, Kim YT, Yoon YB, Kim CY. SK-7041, a new histone deacetylase inhibitor, induces G2-M cell cycle arrest and apoptosis in pancreatic cancer cell lines. Cancer Lett. 2006;237:143-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
52.  Kim MS, Blake M, Baek JH, Kohlhagen G, Pommier Y, Carrier F. Inhibition of histone deacetylase increases cytotoxicity to anticancer drugs targeting DNA. Cancer Res. 2003;63:7291-7300.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Kurz EU, Wilson SE, Leader KB, Sampey BP, Allan WP, Yalowich JC, Kroll DJ. The histone deacetylase inhibitor sodium butyrate induces DNA topoisomerase II alpha expression and confers hypersensitivity to etoposide in human leukemic cell lines. Mol Cancer Ther. 2001;1:121-131.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Chang F, Steelman LS, Shelton JG, Lee JT, Navolanic PM, Blalock WL, Franklin R, McCubrey JA. Regulation of cell cycle progression and apoptosis by the Ras/Raf/MEK/ERK pathway (Review). Int J Oncol. 2003;22:469-480.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Jung JW, Cho SD, Ahn NS, Yang SR, Park JS, Jo EH, Hwang JW, Jung JY, Kim SH, Kang KS. Ras/MAP kinase pathways are involved in Ras specific apoptosis induced by sodium butyrate. Cancer Lett. 2005;225:199-206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
56.  Yu C, Rahmani M, Conrad D, Subler M, Dent P, Grant S. The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571. Blood. 2003;102:3765-3774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 219]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
57.  Denlinger CE, Keller MD, Mayo MW, Broad RM, Jones DR. Combined proteasome and histone deacetylase inhibition in non-small cell lung cancer. J Thorac Cardiovasc Surg. 2004;127:1078-1086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 63]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
58.  Pei XY, Dai Y, Grant S. Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin Cancer Res. 2004;10:3839-3852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 297]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
59.  Cusack JC, Liu R, Houston M, Abendroth K, Elliott PJ, Adams J, Baldwin AS. Enhanced chemosensitivity to CPT-11 with proteasome inhibitor PS-341: implications for systemic nuclear factor-kappaB inhibition. Cancer Res. 2001;61:3535-3540.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Suzuki M, Shinohara F, Sato K, Taniguchi T, Takada H, Rikiishi H. Interleukin-1beta converting enzyme subfamily inhibitors prevent induction of CD86 molecules by butyrate through a CREB-dependent mechanism in HL60 cells. Immunology. 2003;108:375-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
61.  Dai Y, Rahmani M, Dent P, Grant S. Blockade of histone deacetylase inhibitor-induced RelA/p65 acetylation and NF-kappaB activation potentiates apoptosis in leukemia cells through a process mediated by oxidative damage, XIAP downregulation, and c-Jun N-terminal kinase 1 activation. Mol Cell Biol. 2005;25:5429-5444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 198]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]