Abstract
Histone deacetylases (HDACs) remove acetyl groups from lysine residues of histones and the deacetylation allows for tighter electrostatic interactions between DNA and histones, leading to a more compact chromatin conformation with limited access for transactivators and the suppression of transcription. HDAC mRNA and protein overexpression was observed in endometrial and ovarian cancers. Numerous in vitro studies have shown that HDAC inhibitors, through their actions on histone and nonhistone proteins, are able to reactivate the tumor suppressor genes, inhibit cell cycle progression and induce cell apoptosis in endometrial and ovarian cancer cell cultures. Results from mou se xenograft models also demonstrated the potency of HDAC inhibitors as anticancer reagents when used as single agent or in combination with classical chemotherapy drugs.
Keywords: chemotherapy, gynecologic cancer, HDAC, HDAC inhibitor
It has been well established that coordinated epigenetic modifications in DNA methylation and histone modifications play a key role in the control of gene expression [1,2]. In mammalian cells, histones occupy the backbone of chromatin and undergo several post-translational modifications, including acetylation, methylation and phosphoryl ation [3]. The dynamic interplay between histone modifications affects the intrinsic properties of histones and modulates the interactions between DNA and transcription activators [4,5]. Among all the epigenetic modifications, acetylation is one of the better characterized. High levels of acetylation of histones in local chromatins are directly correlated with active gene expression [6–8].
Aberrant transcriptional silencing of tumor-suppressor genes via epigenetic modifications, such as DNA methylation and covalent modifications of histones, is a hallmark of cancer cells [5,9]. Histone acetyltransferases (HAT) and histone deacetylases (HDACs), catalyzing the addition and removal of acetyl groups, respectively, are the key enzymes controlling the extent of histone acetylation. The mammalian HDAC family comprises of 18 members and three major classes, and the expression, regulation and function of HDACs in cancer cells have been extensively studied [2]. Given their critical roles in cell cycling, apoptosis and differentiation, HDACs are considered important therapeutic targets for the treatment of gynecologic cancer and other malignancies [10].
It has been shown that transcriptional gene silencing can be effectively reversed by epigenetic derepression using HDAC inhibitors [11,12]. Because cancer cells contain multiple gene-expression alterations, targeting a single abnormality is often inefficient [4]. The simultaneous actions of HDAC inhibitors on multiple genes represent a major advantage for this class of drugs, which perhaps explains their versatility of effect and remarkable potency in different types of cancer cells [10,11]. Vorinostat was the first reagent to be approved by the US FDA for clinical use in 2006 [13,14]. Currently, a variety of established HDAC inhibitors and new compounds are being tested in cell culture and clinical trials of all phases [4,15–17]. While most reagents are pan-HDAC inhibitors with unselective activities, several compounds are able to target a specific HDAC class or isoform. Here, we provide an update in the following areas:
▪Epigenetic alterations in gynecologic cancers;
▪Mechanisms and actions of HDAC inhibitors in endometrial and ovarian cancer cell lines;
▪Laboratory studies on the combined use of HDAC inhibitors and other anticancer reagents.
Anticancer mechanisms of HDAC inhibitors
The demonstration that small-molecule compounds with the capacity to interfere with HDAC activity have antitumor potential has led to the development of a series of structurally different HDAC inhibitors designed to release transcriptional repression of various targets, ultimately promoting cell cycle stability and/or inducing cell apoptosis [11–13,18]. Several actions by HDAC inhibitors have been suggested, including cell cycle arrest, activation of apoptotic pathways, induction of autophagy, reactive oxygen species (ROS) generation, Hsp90 inhibition and disruption of the aggresome pathway [19]. The HDAC inhibitor induces p21 expression, leading to G1 cell cycle arrest, and frequently downregulates cyclin D and c-Myc [20]. HDAC6 has been shown to be a deacetylase of both tubulin and Hsp9046. Treatment with romidepsin, vorinostat, panobinostat or valproic acid (VPA) resulted in increased histone H3 acetylation or tubulin acetylation, depending on the cell lines [21]. Several in vitro and in vivo studies have suggested that the generation of ROS is a key event in cell death induced by HDAC inhibitors. ROS generated by HDAC inhibitors lead to DNA damage and the addition of N-acetyl cysteine and free radical scavenger has been shown to result in decreased ROS generation and decreased HDAC inhibitor-mediated cell death [22]. HDAC inhibitors have been shown to facilitate death by inducing the expression of TRAIL, DR-4, DR-5, Fas and FasL, as well as a decrease in c-FLIP, a protein associated with resistance to TRAIL-mediated apoptosis [23,24]. Moreover, the miRNA expression profiles of different cell lines have been shown to rapidly change upon treatment with HDAC inhibitors [25].
Romidepsin, a natural product obtained from the bacteria Chromobacterium violaceum, is able to block HDAC activities and induces apoptosis in tumor cells [201]. In 2009, the FDA approved romidespin as an anticancer agent, and clinical trials have been performed in cutaneous T-cell lymphoma, peripheral T-cell lymphoma and a variety of tumors including prostate cancer, multiple myeloma, pancreatic cancer [26], breast cancer [202], ovarian cancer [203], melanoma [204], neuroendocrine tumors [205] and leukemias [206]. The most dramatic results were found in the treatment of cutaneous T-cell lymphoma and other peripheral T-cell lymphomas [26]. Moreover, romidepsin is one of the first HDAC inhibitors found to induce Hsp90 acetylation and cause degradation of the Hsp90 client p roteins EGFR, Her2 and Raf1 [27].
Furthermore, the original model used to explain the antiproliferative effects of these agents suggested that chromatin remodeling resulting from hyperacetylation of core histones altered gene expression [11,12]. It is now clear that HDAC inhibitors induce a wide spectrum of biologic changes resulting from their impact on both transcription and nontranscription levels, as well as their impact on histone and nonhistone proteins (see Figure 1 of [10]). Remarkably, the anticancer effects of HDAC inhibitors were first suggested as a result of their ability to induce differentiation of erythroleukemia cells [10].
To date, several classes of HDAC inhibitors have been proven to have potent and specific anticancer activities in preclinical studies. These data have justified their introduction into clinical trials for the treatment of both hematological malignancies and solid tumors [12,16,28]. Several natural and synthetic small molecular compounds have been employed as bioactive reagents in an attempt to elucidate the mechanism by which HDACs influence cell proliferation [29]. Trichostatin (TSA) is a natural HDAC inhibitor that inhibits almost all class I and II HDACs. It binds to HDACs by its long aliphatic side chain and inhibits enzyme activity by interacting with a Zn motif and other active-site residues. Compounds with HDAC inhibitory effects can be divided into six groups based on their structures:
▪Hydroxamic acids (e.g., TSA) and suberoylanilide hydroxamic acids (e.g., SAHA)
▪Cyclic tetrapeptides (e.g., trapoxin, apicidin and HC-toxin)
▪Depsipeptides (e.g., FK228)
▪Short chain fatty acids (e.g., butyrate and VPA)
▪Synthetic pyridyl carbamate (e.g., MS-275)
▪Synthetic benzamide derivatives (e.g., tacedinaline) and trifluoromethyl ketone
These reagents exhibit varied potencies in terms of upregulating the expression of tumor suppressor genes and blocking cell cycle p rogression [1,10,30].
HDAC inhibitors are small molecules capable of inducing differentiation and preventing cell cycle progression in transformed cells of varying tissue morphology, including acute myeloid leukemia, erythroleukemia and neuroblastoma carcinomas [31]. In recent years, the importance of epigenetics in cancer development has become increasingly appreciated, as is evident by its recent rising popularity in the literature. HDAC inhibitors are currently being investigated in Phase I/II trials for patients with hematologic and solid malignancies, and most data indicate generally tolerable toxicity profiles [2,16]. In regards to endometrial carcinoma (EC), epigenetic defects have been documented for multiple genes, including hMLH1, progesterone receptor-B and PTEN [32]. Silencing of hMLH1 and/or MSH2 by epigenetic mechanisms has been associated with microsatellite instability, invasive growth and acquired resistance to cisplatin [33,34]. Epigenetic reactivation of MLH1 gene expression restores normal DNA repair function [31]. Similarly, progesterone receptor-B silencing occurs commonly in high-grade EC, rendering these tumors recalcitrant to progestational therapy. Treatment with epigenetic-modifying reagents results in re-expression of progesterone receptor-B and, potentially, r esensitization of EC to hormonal therapy [35].
The effect of HDAC inhibitors on ovarian carcinoma (OC) has not been examined as extensively as it has in EC. One study indicated that sodium butyrate (NaB) had a significant growth-suppressing effect on human OC cells, irrespective of their p53 gene status [36]. The authors examined the effects of a wide array of HDAC inhibitors (SAHA, VPA, TSA and NaB) on nine OC cell lines (SK-OV-3, OVCAR-3, TOV-21G, OV-90, TOV-112D, OVCA420, OVCA429, OVCA432 and OVCA433) and found that HDAC inhibitors were able to reduce the non-functional form of the p53 tumor-suppressor protein. The molecular pathways were not investigated. Takai et al. observed that HDAC inhibitors exhibit antiproliferative activity and potently induces apoptosis in human OC cells [37]. These events are accompanied by the induction of p21WAF1 and p27KIP1, and the downregulation of several antiapoptotic and cell cycle-related proteins (Bcl2, cyclin D1 and cyclin D2). VPA also significantly inhibited tumor growth in nude mice without any apparent toxicity. The tumor sections from VPA-treated mice exhibited necrosis and histological changes suggestive of apoptosis, including the formation of apoptotic bodies. Fibrosis accounted for approximately 30% of the tumor area. Moreover, VPA-treated mice showed expression of p21WAF1 using immuno-histochemical ana lysis. These findings suggest that HDAC inhibitors may be effective in the treatment of OC.
Expression of HDACs in endometrial & ovarian cancer
HDACs regulate gene transcription through the removal of acetyl groups from histone tails and DNA sequence-specific transcription factors [4,5]. Examples of nonhistone targets include tumor-suppressor protein p53, E2F and Sp3, where HDAC-based deacetylation has been linked to reduced DNA binding and transcriptional activity [38,39]. Through these mechanisms, HDACs are emerging as critical regulators of such pathways as apoptosis, cell growth and differentiation. Indeed, HDAC inhibitors including NaB, TSA, SAHA and VPA, induce cell cycle arrest, differentiation and apoptosis in colon cancer cell lines in vitro [40]. Takai et al. [37] and Dowdy et al. [41] have independently shown that treatment with HDAC inhibitors dramatically increased the number of apoptotic cells in ovarian and EC cell lines, respectively. These observations suggest a physiologic role for transcriptional repression mediated by HDACs in maintaining cell proliferation and survival. Conversely, aberrant changes in the HDAC-mediated transcriptional repression function may be related to t umorigenesis [3,42].
Normal ovarian surface epithelium showed weak nuclear expression of HDAC-1, -2 and -3 proteins [43,44]. The transcriptional co-repressors mSin3, NCoR, NuRD and SMRT are multiprotein complexes that recruit HDACs to specific chromatin domains [45]. HDAC3 is a component of the NCoR–SMRT corepressor complex, which is distinct from corepressor complexes that typically contain HDAC1 and HDAC2 [46]. On the other hand, HDAC2 and HDAC1 appear to co-exist together in multiprotein complexes and many transcription activators target HDAC1 and HDAC2 to specific p romoters to repress the transcription machinery [47].
High levels of class I HDACs were observed in OC in one study, with HDAC-1, -2 and -3 being positive in 61, 93 and 84% of the cases, respectively [44]. HDAC-1 and -2 are primarily found in the nucleus, whereas HDAC3 is found in the nucleus, cytoplasm and cell membrane [48,49]. Khabele and coauthors also reported an overexpression of all three isoforms of class I HDACs in OC [43]. As in most other entities, class I HDAC expression levels were correlated with each other. Expression levels were significantly different in specific tumor subtypes, with the following values in a decreasing order: mucinous (71%) > serous (64%) > clear cell (54%) > endometrioid (36%). In addition, expression levels were usually higher in highly proliferative tumors [43,44,50]. Class I HDAC protein expression had no statistically significant correlation with patient survival in cases of mucinous, clear cell and serous entities. By contrast, in endometrioid ovarian cancer, stronger class I HDAC expression is associated with shorter patient s urvival (Table 1) [43].
Table 1.
Gynecologic entity | Isoform | HR | 95% CI | p-value | End point | Material | Ref. |
---|---|---|---|---|---|---|---|
Endometrial endometrioid carcinoma (n = 123) |
HDAC1 | 2.19 | 0.71–6.83 | 0.18 | DSS | Protein | [44] |
HDAC2 | 1.14 | 0.25–5.10 | 0.87 | DSS | Protein | ||
HDAC3 | 1.53 | 0.35–6.80 | 0.58 | DSS | Protein | ||
Ovarian serous carcinoma (n = 176) |
HDAC1 | 0.93 | 0.59–1.48 | 0.81 | DFS | Protein | [44] |
HDAC2 | 1.00 | 0.05–20.00 | 0.02 | DFS | Protein | ||
HDAC3 | 1.07 | 0.46–2.47 | 0.09 | DFS | Protein | ||
Ovarian endometrioid carcinoma (n = 114) |
HDAC1 | 6.10 | 1.69–21.98 | <0.01 | DSS | Protein | [44] |
HDAC2 | 1.20 | 0.14–10.47 | 0.87 | DSS | Protein | ||
HDAC3 | 4.13 | 0.64–26.85 | 0.14 | DSS | Protein |
Data were calculated on the basis of the cohorts given in the respective references. HRs are given for high isoform expression; to match this, data from references were partially transformed. p-values < 0.05 were considered significant.
DFS: Disease-free survival; DSS: Disease-specific survival; HDAC: Histone deacetylase; HR: Hazard ratio.
Expression of class I HDAC-1, -2 and -3 proteins in normal endometrium varies with the cell cycle [51]. Weichert and coworkers have observed that the majority of EC showed elevated expression of class I HDAC isoforms in the nuclei of tumor cells, with the following levels of expression in decreasing order: HDAC2 (95%) > HDAC3 (83%) > HDAC1 (61%) [44]. However, a loss of HDAC1 protein expression in EC has also been reported [51]. Similar to OC, clear cell and serous subtypes showed significantly higher expression rates for all three HDACs when compared with endometrioid carcinomas [44]. The authors correlated the expression of HDAC-1, -2 and -3 within individual cells and also between cells of varying proliferative capacity. Strong HDAC1 protein expression was associated with poor prognosis in endometrioid endometrial carcinoma, analogous to its ovarian counterpart [50,51]. Notably, other isoforms that were studied failed to d emonstrate such a relationship between endometrioid s ubtype and poor prognosis [44].
Changes in HDAC expression levels may play a role in the underlying mechanisms involved in cell cycle dysregulation [52–54]. Kawai et al. reported that HDAC1 may affect breast cancer progression by promoting cell proliferation through inhibition of ER-α expression [52]. Silencing of HDAC3 expression in colonic cell lines resulted in growth inhibition, decreased cell survival and increased apoptosis. Similar effects were observed for HDAC-1 and -2 [46]. Jin et al. reported increased expression of HDCA1, HDCA2 and HDCA3 mRNA in 83, 67 and 83%, respectively, and overexpression of HDAC-1, -2 and -3 proteins in 94, 72 and 83%, respectively, in ovarian cancer tissue samples, compared with normal tissue samples [55]. The relative densities of HDAC1 and HDAC3 mRNA in serous, mucinous and endometrioid cancer tissues and HDAC2 mRNA in serous cancer subtypes were significantly higher than those found in benign tissues [55]. These findings suggest that class I HDAC-1, -2 and -3 are upregulated in OC and may play a significant role in ovarian carcinogenesis.
The class I HDACs play an important role in steroid hormone-dependent gene expression by directly interacting with proteins recruited to the steroid hormone receptor complex after ligand binding [56,57]. Recently, Hrzenjak et al. studied the expression of HDAC-1 and -2 in endometriotic and endometrial cell lines using TaqMan® gene expression assays [58]. The expression of both HDAC1 and HDAC2 genes is significantly higher in diseased cells when compared with normal endometrial cells. Steroid hormone treatment induced an upregulation of HDAC-1 and -2 in endometrial stromal cells. Moreover, HDAC1 expression was increased by progesterone, whereas HDAC2 expression was increased by both estrogen and progesterone.
Preclinical studies on HDAC inhibitors in gynecologic cancer cells
Research demonstrating that inappropriate recruitment of HDACs contributes to tumorigenesis has provided a strong mechanistic rationale for applying HDAC inhibitors to cancer therapy regimens (Table 2) [10,16,41]. Acetylation of histones may enhance or inhibit the function of transcription factors, as well as chaperone proteins such as p53, GATA1, E2F, BCL6, Ku70, Hsp90, RelA, c-Jun and STATs. Therefore, enhancing the degree of acetylation by cell treatment with an HDAC inhibitor can either increase or repress gene expression [59]. It has been found that structurally diverse compounds can bind to and inhibit HDAC catalytic activity. Currently, more than 50 naturally occurring or synthetic HDAC inhibitors have been developed [10,60]. Initial clinical trials indicate that HDAC inhibitors from several different structural classes are well tolerated and exhibit clinical efficacy against a variety of human malignancies [59].
Table 2.
Gene | Function on angiogenesis |
Target cell | Direction of modulation |
Ref. |
---|---|---|---|---|
Tie2 | Promotes | Endothelial | ↓ | [61] |
Ang2 | Promotes | Endothelial | ↓ | [61] |
Nrp1 | Promotes | Endothelial | ↓ | [62] |
SEMA3 | Suppresses | Endothelial | ↑ | [62] |
VEGFR1 | Promotes | Endothelial | ↓ | [62] |
VEGFR2 | Promotes | Endothelial | ↓ | [62] |
eNOS | Promotes | Endothelial | ↓ | [63] |
VEGFD P | Promotes | Endothelial | ↓ | [64] |
In proliferating endothelial cells, HDAC inhibitors upregulate gene expression of p21WAF1/CIP1 , which induces cell cycle arrest and downregulates gene expression of survivin, an inhibitor of apoptosis [61]. TSA inhibits the VEGF-induced expression of VEGF receptors VEGFR1, VEGFR2 and Nrp1 (Table 2). Furthermore, TSA and SAHA upregulate the expression of SEMA3, a VEGF protein competitor, at both mRNA and protein levels [62]. Downregulation of endothelial nitric oxide synthase (eNOS) in endothelial cells has also been shown to be critical for the antiangiogenic activity of VPA. Moreover, VPA was also shown to inhibit angiogenesis both in vitro and in vivo via a mechanism involving diminished expression of eNOS [63]. Depsipeptide was shown to suppress the expression of proangiogenic factors, including VEGF and bFGF [61]. Consistently, TSA and SAHA directly inhibit VEGF family member D (VEGFD) and bFGF-stimulated endothelial cell proliferation, migration, invasion, vascular sprouting and n eovascular f ormation [64].
The HDAC inhibitor NVPLAQ824 blocks expression of proangiogenic tyrosine kinase receptors Tie-2, Tie-2 ligand and Ang2, at both mRNA and protein levels (Table 2). However, HDAC inhibitor NVPLAQ824 exerts no effect on the Tie-1 receptor [61]. Butyrates upregulate endothelial cell adhesion molecules, including ICAM-1 and E-selectin. Downregulation of eNOS in endothelial cells has also been shown to be critical for the anti-angiogenic activity of VPA [63]. A study by Takai et al. demonstrated both antiproliferative and proapoptotic activities of HDAC inhibitors in human OC cells [65]. Furthermore, soft agar colony formation assays and 3-4,5-dimethylthiazol-2-yl-2,5-diphenyl-tetrazolium bromide MTT assays showed that many endometrial and ovarian cancer cell lines were sensitive to the growth inhibitory effect of HDAC inhibitors, although normal endometrial epithelia were viable after the treatment with the same concentrations of HDAC inhibitors. VPA significantly inhibited human endometrial and ovarian tumor growth without toxic side effects [36,66]. Terao et al. indicated that NaB had a significant growth-suppressing effect on human EC and OC cells, irrespective of their p53 status [36].
Takai et al. used six EC cell lines to investigate the antiproliferative effects of SAHA, VPA, TSA and NaB [37]. All cancer cell lines were sensitive to the growth inhibitory effect of HDAC inhibitors. Cell cycle ana lysis indicated that treatment with HDAC inhibitors decreased the proportion of cells in S phase and increased the proportion of cells in the G0–G1 and/or G2–M phases of the cell cycle. These effects were accompanied by the altered expression of genes related to malignant phenotypes, including an increase in p21Waf1, p27Kip7 and E-cadherin, as well as a decrease in Bcl2, cyclin D1 and cyclin D2 (Table 3). Similarly, apicidin, CBHA [66] and scriptaid [67] were also reported to increase the proportion of cells in G0/G1 and/or G2/M phases and to decrease the proportion of cells in S phase. Several studies indicated that HDAC inhibitor-induced apoptosis is associated with the loss of mitochondrial transmembrane potential. Accompanying cell apoptosis is the altered expression of p21WAF1, p27KIP1, p16, cyclin A and E-cadherin [68]. It was confirmed that apicidin, CBHA and scriptaid treatments indeed increased the acetylation of H3 and H4 histone tails [66–68]. These results suggest that HDAC inhibitors exert antiproliferative effects through selective induction of genes influencing cell growth, malignant phenotype and apoptosis [68]. The observation of a remarkable increase in acetylated histones associated with the p21 promoter after suberoyl anilide bishydroxamine treatment has provided evidence for the decreased HDAC activity following HDAC inhibitor administration [31]. Besides cell cycle inhibition and induction of apoptosis, an anti-inflammatory action could be an additional activity contributing to the overall anticancer effects of HDAC inhibitors. It was reported that HDAC inhibitors can suppress the expression of key adhesion molecules, such as VCAM-1, thus leading to a reduction in the number of activated monocytes recruited to inflamed endothelium [69].
Table 3.
HDACi | Class | Entity | Function | Ref. |
---|---|---|---|---|
TSA | Hydroxamic | Endometrial cancer | Inhibit VEGF receptors VEGFR1, VEGFR2 and Nrp1 Induce glycodelin expression |
[31,100] |
Valproic acid/ valproate |
Short-chain fatty acids |
Endometrial cancer | Downregulation of eNOS in endothelial cells Inhibit G1/S transition and influence expression of p21WAF1 and cyclin D1 |
[31,63] |
SAHA (vorinostat, zolinza) |
Hydroxamic | Endometrial and ovarian cancer |
Decrease the proportion of cells in S phase and increase the proportion of cells in the G0/G1 and/or G2/M phases Induce glycodelin expression |
[77] |
Ovarian cancer | Had minimal activity in a Phase II study Induce apoptosis |
[93,101] | ||
R306465 | Hydroxamic | Ovarian cancer | Induce apoptosis and inhibit angiogenesis | [102] |
Butyrates | Short-chain fatty acids |
Endometrial and ovarian cancer |
Regulate ICAM-1 and E-selectin | [102] |
PsA | A phenolic natural product isolated from marine sponges |
Endometrial and ovarian cancer |
Induce the expression of acetylated H3 and H4 histone proteins Upregulate the expression of cyclin-dependent kinase inhibitor, p21WAF1 and downregulate the expression of pRb, cyclins and cyclin-dependent kinases |
[28,71] |
PsA | Marine sponge compound |
Endometrial cancer | Induce apoptosis in endometrial carcinoma cells | [15] |
NVP-LAQ824 (dacinostat) |
Hydroxamic | Endometrial cancer | Block tyrosine kinase receptors Tie2 and Tie2 ligand, Ang2, mRNA and protein expression |
[61] |
Butyrate | Short-chain fatty acids |
Endometrial and ovarian cancers |
Induce p53 expression | [36,37] |
Oxamflatin | Hydroxamic | Endometrial cancer I and II |
Loss of mitochondrial membrane potentials consistent with the induction of apoptosis |
[76] |
HDACi-1, HDAC-I1 | Endometrial cancer I and II |
Induction of apoptosis | [76] | |
CI-958 | Benzamide | Ovarian cancer | Potently inhibit DNA and RNA synthesis | [103] |
PXD101(Belinostat) | Hydroxamic | Epithelial ovarian cancer and micropapillary/ borderline ovarian tumors |
Platinum-resistant epithelial ovarian cancer and micropapillary/borderline Histone H4 hyperacetylation |
[104,105] |
Scriptaid | Hydroxamic | Endometrial and ovarian cancers |
Induce apoptosis and increase acetylation of histone tails Decrease the proportion of cells in the S phase and increase the proportion in the G0/G1 and/or G2/M phases |
[67] |
FK228 (romidepsin/ depsipeptide) |
Endometrial cancer | Modulation of angiogenesis-related genes Inhibition of endothelial cell migration and proliferation |
[63] | |
Apicidin | Tetrapeptides | Endometrial and ovarian cancers |
Increase acetylation of H3 and H4 histone tails Induce apoptosis |
[68] |
Biphenyl-4-yl- acrylohydroxamic acid derivatives |
Hydroxamic | Ovarian cancer | Induce acetylation of p53 and tubulin | [106] |
CBHA | Hydroxamic | Endometrial and ovarian cancers |
Increase acetylation of histone proteins and induce apoptosis | [66] |
LBH589 (panobinostat) |
Hydroxamic | Endometrial cancer | Endothelial tube formation and Matrigel™ invasion | [83] |
M344 | Hydroxamic | Endometrial and ovarian cancers |
Acetylation of histones, induction of apoptosis and increase the proportion of cells in the G0/G1 and/or G2/M phases |
[107] |
Valproate | Short-chain fatty acids |
Endometrial cancer | Inhibit G1/S transition and influence expression of p21WAF1 and cyclin D1 |
[58] |
eNOS: Endothelial nitric oxide synthase; HDACi: Histone deacetylase inhibitor; SAHA: Suberoylanilide hydroxamic acid; TSA: Trichostatin.
In addition to synthetic HDAC inhibitors, natural HDAC inhibitors have also been shown to induce cell cycle arrest in various cancer cell lines. PsA is a phenolic natural product that has been isolated from marine sponges and may be a promising HDAC inhibitor [70,71]. Ahn et al. reported that PsA induces cell cycle arrest and apoptosis and increases the proportion of human EC cells in the G1 phase and G2/M phases of the cell cycle (Table 3) [71]. This PsA-induced cell cycle arrest was associated with the downregulation of cyclins/CDKs and pRb, as well as the induction of p21WAF1 through a p53-independent pathway. Recently, it was reported that PsA has antibacterial and antitumor properties and inhibits various enzymes including FPT, topoisomerase II, leucine amino chitinase and peptidase [28,70,72]. There is evidence that PsA inhibits both HDACs and DNMTs, although the molecular mechanism for the dual inhibitory effects is not clear [70]. Recently, Berry et al. proposed that PsA induces apoptosis in endometrial cancer cells through increased expression of FOXO1 [15], a member of the Forkhead/winged helix family that plays a role in cell survival, cell cycle progression and oxidative-stress resistance [73]. These observations show diversified and dynamic activities of HDAC inhibitors on a broad spectrum of regulatory factors/pathways. Sulforaphane (SFN), a compound found in cruciferous vegetables, inhibits HDAC activity in human colorectal and prostate cancer cells [74]. Myzak et al. demonstrated that SFN acted as an HDAC inhibitor in the prostate, causing enhanced histone acetylation, derepression of p21 and Bax and induction of cell cycle arrest/ apoptosis, leading to cancer prevention [75]. The ability of SFN to target aberrant acetylation patterns, in addition to effects on phase 2 enzymes, may make it an effective c hemoprevention agent.
Takai and coworkers discovered two putative tumor-suppressor genes, Tig1 and CCAAT/ enhancer binding protein-α (c/ebp-α), that are stimulated by demethylating agents and/or HDAC inhibitors in endometrial cancers [31]. In additional studies, HDAC inhibitors were found to inhibit the growth of human endometrial cancer cells both in vitro and in a mouse xenograft model [31]. Jiang et al. examined the effects of HDAC inhibitors oxamflatin and HDAC inhibitor-1 in endometrial cancer cells and observed significant growth inhibition and morphologic changes (Table 3) [76]. Sensitivity to individual agents appears to be cell type specific, with oxamflatin having a stronger growth inhibitory effect than HDAC inhibitor-1 in the Ark2 cell line, while the reverse is true in the AN3 cell line. With respect to the specific apoptotic pathways involved, both caspase-8 and caspase-9 are activated by oxamflatin in the Ark2 cell line. Furthermore, loss of mitochondrial membrane potential occurs upon treatment with the agents. These results suggest that the intrinsic apoptotic pathway may play an important role in the induction of cell death by oxamflatin [76]. Clinically speaking, these findings suggest that HDAC inhibitors could have an important impact on the treatment of the most aggressive subset of EC.
Uchida et al. found that HDAC inhibitors, in particular SAHA, enhanced single and collective cell migration, and HDAC inhibitor-induced glycodelin expression played an essential role in promotion of Ishikawa cell migration (Table 3) [77]. Glycodelin is the progesterone-induced glycoprotein secreted into the uterine luminal cavity by secretory/decidualized endometrial glands [78]. A study by Mandelin et al. showed that chemotherapy-treated serous OC patients with glycodelin-expressing tumors have longer survival time than those with glycodelin-negative tumors [79]. Thus, HDAC inhibitor treatment appears to alter some of the malignant phenotypes and improve OC patient survival. However, it is not clear how the glycodelin-induced cell migration is related to the longer survival and the HDAC inhibitor therapeutic effects or if the anticancer effects of HDAC inhibitors may rely on different pathways. It is also noteworthy that not all the diversified effects by HDAC inhibitors are solely therapeutic. Rather, it is highly possible that some effects may be antitherapeutic or even constitute a source of toxicity, as frequently observed in HDAC inhibitors.
Tumor growth and metastasis depend on the development of a neovasculature within and around the tumor [80]. Angiogenesis is regulated by the balance between inhibitory factors, for example, angiostatin, IL-10 and interferon, as well as stimulatory factors, for example, bFGF, IL-8, MMP-2, MMP-9, TGF-β1 and VEGF, that are released by tumor cells and surrounding stroma [81]. HDAC inhibitors modulate angiogenesis in a manner that has potential therapeutic implications. HDAC1 downregulates the expression of p53 and the von Hippel–Lindau tumor suppressor gene and stimulates angiogenesis of human endothelial cells. Correspondingly, HDAC inhibitors inhibit endothelial cell proliferation and angiogenesis by downregulating angiogenesis-related gene expression [63,82]. Phenyl butyrate, LBH589, LAQ824 and TSA are able to exert antiangiogenic activity both in vitro and in vivo [61,83]. Other HDAC inhibitors, such as SAHA, FK228, VPA and apicidin, have also been shown to possess antiangiogenic activities [63,84,85]. Angiogenesis inhibition, induced by HDAC inhibitors, was associated with modulation of angiogenesis-related genes in both neoplastic cells; for example, inhibition of HIF-1α and VEGF, and benign endothelial cells; for example, inhibition of Tie-2 and survivin. Downregulation of endothelial cell migration and proliferation [85] may also contribute to the angio-suppressive effect. Furthermore, LBH589 was shown to inhibit human umbilical vein endothelial cell Matrigel™ invasion and endothelial tube formation in vitro [83].
Studies on combination therapy for gynecologic cancers
Many investigators feel that HDAC inhibitors are more likely to be effective in solid tumors if used in combination with other cytotoxic reagents. Conceivably, a HDAC inhibitor-mediated increase in histone acetylation produces a more open chromatin conformation that facilitates the re-expression of silenced tumor-suppressor genes. This may result in restoration of apoptotic and/or cell cycle control mechanisms, enhancing the chemo-sensitivity of tumor cells [86,87]. For example, SAHA, depsipeptide, MS-275 and TSA enhance the activity of carboplatin, docetaxel gemcitabine, cisplatin, etoposide, doxorubicin and paclitaxel in ovarian as well as endometrial cancer cells (Table 4). HDAC inhibitor-induced apoptosis was greatly enhanced in the presence of the DNA methyltransferase inhibitor 5-aza-2′-deoxycytidine, suggesting that DNA methylation status plays an important role in the efficacy of HDAC inhibitors [88]. Interestingly, HDAC inhibitors enhanced paclitaxel-induced cell death in ovarian cancer cell lines, independent of p53 status [89]. While these results indicate clinical value for the combined administration of HDAC inhibitors with other treatment, one precaution for HDAC inhibitor application is that pretreatment of tumor cells with HDAC inhibitors may also cause initial cell damage, resulting in increased side effects.
Table 4.
HDACi | Class | Other agents | Disease | Function | Ref. |
---|---|---|---|---|---|
TSA | Hydroxamic | 5-aza-2′- deoxycytidine |
Endometrial cancer | Inhibit VEGF receptors VEGFR1, VEGFR2 and Nrp1-1.85 Downregulate DNMT3B mRNA and protein expression |
[31,62,108] |
PXD101 (belinostat) |
Hydroxamic | Carboplatin Paclitaxel |
Ovarian, epithelial and fallopian tube cancers |
Histone H4 hyperacetylation | [12,104] |
TSA, NaB | Paclitaxel | Ovarian cancer | Induction of p53 protein | [89] | |
TSA | Hydroxamic | Paclitaxel | Endometrial cancer | Induction of p21 Increase acetylation of tubulin and microtubule stabilization |
[41] |
SAHA (vorinostat), NaB |
Hydroxamic, short-chain fatty acids |
Aspirin | Ovarian cancer | Induction of apoptosis | [97] |
PXD101 (belinostat) |
Hydroxamic | Docetaxel Paclitaxel Carboplatin |
Ovarian cancer | Enhance carboplatin anticancer effect in xenograft Increase the phosphorylation of H2AX induced by carboplatin Enhance induction of histone H4 hyperacetylation |
[83] |
SAHA | Hydroxamic | Paclitaxel | Ovarian cancer | Slightly induce apoptosis | [103 |
HDACi: Histone deacetylase inhibitor; SAHA: Suberoylanilide hydroxamic acid; TSA: Trichostatin.
Uterine papillary serous carcinomas, with their propensity for metastasis and recurrence, present difficult therapeutic challenges for clinicians. Paclitaxel showed limited efficacy for patients suffering from this type of diseases, with a median progression-free interval of only 7.3 months [90]. Improved results come from a combined treatment with carboplatin and paclitaxel. In the adjuvant setting, this treatment increased the median progression-free survival to 30 months [91]. Dowdy et al. performed preclinical studies in type II endometrial cancer cells and found that while treatment of Ark2 and KLE cells with a single reagent, paclitaxel, doxorubicin, carboplatin or the HDAC inhibitor TSA all inhibited cell proliferation, significant synergistic action was detected in the combination of TSA/paclitaxel [41]. Cell death was accompanied by the activation of the cell apoptotic cascade. Dramatic morphological changes were also observed following treatment with TSA/paclitaxel [41]. The synergism most likely resulted from a common pathway involving the inhibition of HDAC activity since similar effects were detected when TSA was substituted by diversified HDAC inhibitors such as HDAC inhibitor-1 and oxamflatin. Indeed, further experiments demonstrated that tubulin acetylation plays a central role for the synergistic actions of the two drugs. While treatment with a single drug led to an increase in acetylated tubulin and microtubule stabilization, combined treatment with TSA/paclitaxel resulted in much enhanced effects. Moreover, in a mouse xenograft model, TSA/paclitaxel treatment resulted in a greater tumor weight reduction than the single agent regimens. Thus, the TSA/paclitaxel combination seems to hold promise for the treatment of EC and other malignancies that are resistant to paclitaxel [41].
Sonnemann et al. investigated the sensitivity of ovarian cancer cells to SAHA and paclitaxel in a pilot study employing three established cell lines (OVCAR-3, SK-OV-3 and A2780) as well as cancer cell primary cultures [92]. All five isolates were sensitive to SAHA, whereas four out of five were resistant to paclitaxel. These ex vivo findings support the idea that SAHA might be effective in the treatment of paclitaxel-resistant ovarian cancer in vivo. Cooper et al. examined the effects of the SAHA/paclitaxel combination in ovarian cancer cell line 2774 using in vitro culture and mouse xenograft models [93]. They found that while a combination of the two did not generate significantly different results as compared with single agents, paclitaxel followed by SAHA and paclitaxel alone increased survival compared with SAHA alone or SAHA followed by paclitaxel. The authors concluded that adding SAHA to ovarian cancer chemotherapy regimens could increase the drug efficacy and that sequential administration of the drugs is critical to achieving synergism.
PXD101 is a novel hydroxamate-type HDAC inhibitor [94]. A Phase I trial of PXD101 was performed in patients with advanced solid tumors [95]. Phase II study results of PXD101 were reported by Ramalingam et al. for the treatment of advanced malignant pleural mesothelioma [96]. In this study, 30 patients with metastatic or recurrent and refractory pleural mesothelioma were enrolled. Two out of the 30 patients had stable diseases. While recruitment was still ongoing, initial results appear to be promising. No reported data regarding PXD101 effects for gynecologic cancer are currently available. It will be of great interests to examine how effective this potent reagent may be when applied to treat gynecologic cancers.
Qian et al. examined the anticancer effect of the combined application of PXD101 with docetaxel, paclitaxel and carboplatin [83]. In the A2780 ovarian cancer xenograft model, PXD101 as a single agent showed moderate antitumor activity. In the same system, the combination of PXD101/carboplatin resulted in much increased effects. Interestingly, it was found that PXD101 was able to increase the acetylation of α-tubulin induced by docetaxel as well as the phosphorylation of H2AX induced by carboplatin. The efficacy of PXD101 alone or in combination therapy for the treatment of OC requires c linical evaluation [83].
The nonsteroidal anti-inflammatory drug aspirin has shown promise as an antineoplastic agent. Sonnemann et al. found that HDAC inhibitors SAHA, NaB and aspirin cooperated synergistically to induce cell death in the OC cell line A2780 [97]. However, histone acetylation was not affected by aspirin neither in the absence or presence of HDAC inhibitors. The exact molecular pathway responsible for the observed synergism could not be fully explained by the currently available data.
It was thought that the antitumor effects of HDAC inhibitors are mediated by the reactivation of silenced tumor-suppressor genes. However, it is clear that a variety of nonhistone proteins are acetylation targets. The regulation mechanisms and cellular effects of nonhistone acetylation are not fully understood. As described above, one HDAC target is tubulin. Tubulin acetylation modulates microtubule assembly and disassembly and the disturbance of the balanced actions has destructive effects on cellular functions, ultimately leading to cell death. Zhang et al. reported that HDAC6 is capable of interacting with purified tubulin and microtubules in vitro and HDAC6 colocalizes with microtubules in NIH-3T3 cells [98]. Tubulin acetylation is increased following TSA treatment. Moreover, knockout of the HDAC6 gene also led to elevated levels of α-tubulin acetylation. Blagosklonny et al. demonstrated an immediate acetylation of the Lys40 residue in α-tubulin following TSA treatment, and that this process was also accompanied by the initiation of cell apoptosis [20]. Paclitaxel has also been shown to bind to and stabilize microtubules, eventually precipitating apoptosis [99]. As discussed above, Dowdy et al. observed a pronounced synergistic effect of the TSA/paclitaxel combination on α-tubulin acetylation, microtubule stabilization and endometrial cancer cell apoptosis in cell culture and mouse xenograft models [41], thereby suggesting a significant role for nonhistone p roteins in HDAC-mediated cell function.
Conclusion
Accumulated evidence supports the idea that acetylation of histone and nonhistone proteins plays a critical role in a variety of cell functions, including gene expression, microtubule assembly, cell apoptosis and cell cycle regulation. The significant alterations of HDAC expression levels and frequent epigenetic silencing of tumor-suppressor genes in ovarian and endometrial cancers further corroborates the value of HDACs as therapeutic targets for the treatment of gynecologic malignancies. Numerous in vitro, ex vivo and clinical trials have been performed to investigate the effects, regimen designs, molecular mechanisms, therapeutic values and side effects of HDAC inhibitors as a new class of chemotherapy reagents. Relatively high response rates and low toxicity profiles of available HDAC inhibitors have been observed in several studies. Despite the promising develop ment, there are several issues that need to be addressed before moving forward. A better understanding of the molecular basis behind the antiendometrial and antiovarian cancer activities of HDAC inhibitors is essential. Translational studies are also required to correlate protein acetylation and gene transcription to tumor response and patient survival for more objective data interpretation and improvement of regimen arrangement in terms of dosing, scheduling and formulations. Owing to the need for constant drug exposure to achieve in vivo tumor mass reduction, a more detailed study and comparison of the pharmacokinetic profiles for various HDAC inhibitor is needed. Rationally designed combinations of HDAC inhibitors with conventional chemotherapy drugs for treatment of gynecologic cancers are showing encouraging results in tumor cell culture and animal models. However, the efficacy of these combination therapies has yet to be proven in clinical trials. Despite these difficulties, through coordinated efforts in basic, translational and clinical fields, the use of HDAC inhibitors is expected to open up a new avenue in mechanism-based therapy for gynecologic cancers.
Future perspective
The FDA has approved vorinostat (SAHA) and romidepsin (Istodax®) for the treatment of cutaneous lymphoma. These drugs are actively being tested for application in solid tumors. Representing a new class of anticancer reagents, HDAC inhibitors will be more extensively investigated, setting the stage for the discovery and development of novel compounds with improved inhibitory potency or HDAC isoform specificity. In 5–10 years, additional HDAC inhibitors will be subjected to clinical trials. It is also expected that the combination treatment strategy will allow for a significant decrease in the doses of both drugs without sacrificing efficacy. Future clinical trials may provide evidence for the efficacy of HDAC inhibitors in sensitizing hormone- and drug-resistant gynecologic cancers to radiation and chemotherapy.
While the major cellular effects of HDAC inhibitors, including those affecting cell cycle progression and apoptosis, have been well recognized, the detailed molecular mechanisms remain to be elucidated. Further characterization of tissue-specific control, hormonal regulation and the oncogenic alterations in HDAC gene expression will provide insight into how HDAC inhibitors affect cell function. Studies on the regulation of nonhistone protein acetylation will enrich our knowledge on the diverse acetylation targets and pathways. The development of new proteomics and DNA sequencing technologies has opened the door for in-depth analysis of the mechanistic aspects attributable to the observed effects. These studies may lead to a better understanding of the cellular response to HDAC inhibitors and, ultimately, to an i ndividualized regimen for gynecologic cancer patients.
Executive summary.
Expression of histone deacetylases in endometrial & ovarian cancer
▪Overexpression of class I histone deacetylases (HDACs) is detected in ovarian carcinoma (OC) and endometrial carcinoma (EC); increased HDAC expression levels correlate with a poor prognosis of OC and EC patients.
▪HDACs participate in regulation of the cell cycle and may also play a role in the control of steroid hormone-dependent gene expression.
Preclinical studies on HDAC inhibitors in gynecologic cancer cells
▪HDAC inhibitors (HDACi) induce dramatic changes in gene expression. HDACi are potent inhibitors for the proliferation of gynecologic cancer cells.
▪HDACi inhibits angiogenesis by downregulating the expression of angiogenesis-related genes.
Studies on combination therapy for gynecologic cancers
▪Combination approaches using HDACi and conventional reagents have been tested in cell culture, as well as mouse xenografts. Improved drug effects have been observed in many experiments.
▪The HDAC inhibitor/taxol combination has been shown to be very effective for EC and OC cells.
Acknowledgments
This work is partially funded by the Georgia Cancer Coalition Distinguished Cancer Scholarship (funding to S-W Jiang), NIH R01 HD 41577 (funding to S-W Jiang), NIH/National Cancer Institute MD Anderson Uterine Cancer SPORE (funding to J Li and S-W Jiang), The Pancreatic Cancer Program of Anderson Cancer Institute/Memorial Health University Center (funding to J Li to S-W Jiang) and Research Supplement from the Mercer University School of Medicine (funding to J Li and S-W Jiang).
Footnotes
Financial & competing interests disclosure The authors have no other relevant affiliations or financial involvements with any organization or entity with a financial i nterest in or financial conflict with the subject manner or materials discussed in the manuscript apart from those disclosed.
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as:
▪ of interest
▪▪ of considerable interest
- 1.Khan N, Jeffers M, Kumar S, et al. Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors. Biochem. J. 2008;409(2):581–589. doi: 10.1042/BJ20070779. [DOI] [PubMed] [Google Scholar]
- 2.Cang S, Ma Y, Liu D. New clinical developments in histone deacetylase inhibitors for epigenetic therapy of cancer. J. Hematol. Oncol. 2009;2:22. doi: 10.1186/1756-8722-2-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kouzarides T. Chromatin modifications and their function. Cell. 2007;128(4):693–705. doi: 10.1016/j.cell.2007.02.005. [DOI] [PubMed] [Google Scholar]
- 4.Smith BC, Hallows WC, Denu JM. Mechanisms and molecular probes of sirtuins. Chem. Biol. 2008;15(10):1002–1013. doi: 10.1016/j.chembiol.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Spange S, Wagner T, Heinzel T, et al. Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int. J. Biochem. Cell Biol. 2009;41(1):185–198. doi: 10.1016/j.biocel.2008.08.027. [DOI] [PubMed] [Google Scholar]
- 6.Fullgrabe J, Kavanagh E, Joseph B. Histone onco-modifications. Oncogene. 2011;30(31):3391–3403. doi: 10.1038/onc.2011.121. [DOI] [PubMed] [Google Scholar]
- 7.Sawan C, Herceg Z. Histone modifications and cancer. Adv. Genet. 2010;70:57–85. doi: 10.1016/B978-0-12-380866-0.60003-4. [DOI] [PubMed] [Google Scholar]
- 8.Ellis L, Atadja PW, Johnstone RW. Epigenetics in cancer: targeting chromatin modifications. Mol. Cancer Ther. 2009;8(6):1409–1420. doi: 10.1158/1535-7163.MCT-08-0860. [DOI] [PubMed] [Google Scholar]
- 9.Gallinari P, Di Marco S, Jones P, et al. HDACs, histone deacetylation and gene transcription: from molecular biology to cancer therapeutics. Cell Res. 2007;17(3):195–211. doi: 10.1038/sj.cr.7310149. ▪▪ Broad review covering histone deacetylase (HDAC) actions: tightly controlled through targeted recruitment, protein– protein interactions and post-translational modifications. Describes the important role of these HDAC enzymes: control of cell cycle progression, cell survival and differentiation. Since these processes are affected by malignant transformation, HDAC inhibitors were developed as antineoplastic drugs.
- 10.Buchwald M, Kramer OH, Heinzel T. HDACi – targets beyond chromatin. Cancer Lett. 2009;280(2):160–167. doi: 10.1016/j.canlet.2009.02.028. ▪ Describes HDAC inhibitor-mediated depletion of oncoproteins via modulation of the ubiquitin–proteasome pathway. It highlights the cytokine- and HDAC inhibitor-stimulated acetylation-dependent cross-talks between signaling pathways. It also describes how the reagents could be beneficial for the treatment and prevention of human ailments, such as cancer and unbalanced immune functions.
- 11.Marks PA, Xu WS. Histone deacetylase inhibitors: potential in cancer therapy. J. Cell Biochem. 2009;107(4):600–608. doi: 10.1002/jcb.22185. ▪▪ Detailed review on the development of HDAC inhibitors. Targeted inhibitors of HDACs may lead to anticancer drugs that are safer and more effective than nonselective agents.
- 12.Ma X, Ezzeldin HH, Diasio RB. Histone deacetylase inhibitors: current status and overview of recent clinical trials. Drugs. 2009;69(14):1911–1934. doi: 10.2165/11315680-000000000-00000. ▪▪ Provides an overview on recent clinical trials with HDAC inhibitors. The combination of HDAC inhibitors with other anticancer agents demonstrated favorable clinical outcome.
- 13.Marks PA. Discovery and development of SAHA as an anticancer agent. Oncogene. 2007;26(9):1351–1356. doi: 10.1038/sj.onc.1210204. [DOI] [PubMed] [Google Scholar]
- 14.Jabbour E, Issa JP, Garcia-Manero G, et al. Evolution of decitabine development: accomplishments, ongoing investigations, and future strategies. Cancer. 2008;112(11):2341–2351. doi: 10.1002/cncr.23463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Berry E, Hardt JL, Clardy J, et al. Induction of apoptosis in endometrial cancer cells by psammaplysene A involves FOXO1. Gynecol. Oncol. 2009;112(2):331–336. doi: 10.1016/j.ygyno.2008.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Garcia-Manero G, Issa JP. Histone deacetylase inhibitors: a review of their clinical status as antineoplastic agents. Cancer Invest. 2005;23(7):635–642. doi: 10.1080/07357900500283119. ▪ A comprehensive introduction to the field of histone acetylation and its inhibitors; reviews the most relevant clinical data.
- 17.Camphausen K, Tofilon PJ. Inhibition of histone deacetylation: a strategy for tumor radiosensitization. J. Clin. Oncol. 2007;25(26):4051–4056. doi: 10.1200/JCO.2007.11.6202. [DOI] [PubMed] [Google Scholar]
- 18.Johnstone RW, Licht JD. Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell. 2003;4(1):13–18. doi: 10.1016/s1535-6108(03)00165-x. [DOI] [PubMed] [Google Scholar]
- 19.Lane AA, Chabner BA. Histone deacetylase inhibitors in cancer therapy. J. Clin. Oncol. 2009;27(32):5459–5468. doi: 10.1200/JCO.2009.22.1291. [DOI] [PubMed] [Google Scholar]
- 20.Blagosklonny MV, Robey R, Sackett DL, et al. Histone deacetylase inhibitors all induce p21 but differentially cause tubulin acetylation, mitotic arrest, and cytotoxicity. Mol. Cancer Ther. 2002;1(11):937–941. [PubMed] [Google Scholar]
- 21.Nishioka C, Ikezoe T, Yang J, et al. MS-275, a novel histone deacetylase inhibitor with selectivity against HDAC1, induces degradation of FLT3 via inhibition of chaperone function of heat shock protein 90 in AML cells. Leuk. Res. 2008;32(9):1382–1392. doi: 10.1016/j.leukres.2008.02.018. [DOI] [PubMed] [Google Scholar]
- 22.Lee JH, Choy ML, Ngo L, et al. Histone deacetylase inhibitor induces DNA damage, which normal but not transformed cells can repair. Proc. Natl Acad. Sci. USA. 2010;107(33):14639–14644. doi: 10.1073/pnas.1008522107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kauh J, Fan S, Xia M, et al. c-FLIP degradation mediates sensitization of pancreatic cancer cells to TRAIL-induced apoptosis by the histone deacetylase inhibitor LBH589. PLoS ONE. 2010;5(4):E10376. doi: 10.1371/journal.pone.0010376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Yeh CC, Deng YT, Sha DY, et al. Suberoylanilide hydroxamic acid sensitizes human oral cancer cells to TRAIL-induced apoptosis through increase DR5 expression. Mol. Cancer Ther. 2009;8(9):2718–2725. doi: 10.1158/1535-7163.MCT-09-0211. [DOI] [PubMed] [Google Scholar]
- 25.Scott GK, Mattie MD, Berger CE, et al. Rapid alteration of microRNA levels by histone deacetylase inhibition. Cancer Res. 2006;66(3):1277–1281. doi: 10.1158/0008-5472.CAN-05-3632. [DOI] [PubMed] [Google Scholar]
- 26.Masuoka Y, Shindoh N, Inamura N. Histone deacetylase inhibitors from microorganisms: the Astellas experience. Prog. Drug Res. 2008;66(335):337–359. doi: 10.1007/978-3-7643-8595-8_7. [DOI] [PubMed] [Google Scholar]
- 27.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(7):504–513. doi: 10.1093/jnci/94.7.504. [DOI] [PubMed] [Google Scholar]
- 28.Cho JW, Jeong YW, Han SW, et al. Aberrant p16INK4A RNA transcripts expressed in hepatocellular carcinoma cell lines regulate pRb phosphorylation by binding with CDK4, resulting in delayed cell cycle progression. Liver Int. 2003;23(3):194–200. doi: 10.1034/j.1600-0676.2003.00821.x. [DOI] [PubMed] [Google Scholar]
- 29.Leder A, Orkin S, Leder P. Differentiation of erythroleukemic cells in the presence of inhibitors of DNA synthesis. Science. 1975;190(4217):893–894. doi: 10.1126/science.1059262. [DOI] [PubMed] [Google Scholar]
- 30.Schneider-Stock R, Ocker M. Epigenetic therapy in cancer: molecular background and clinical development of histone deacetylase and DNA methyltransferase inhibitors. IDrugs. 2007;10(8):557–561. [PubMed] [Google Scholar]
- 31.Takai N, Desmond JC, Kumagai T, et al. Histone deacetylase inhibitors have a profound antigrowth activity in endometrial cancer cells. Clin. Cancer Res. 2004;10(3):1141–1149. doi: 10.1158/1078-0432.ccr-03-0100. [DOI] [PubMed] [Google Scholar]
- 32.Vigushin DM, Ali S, Pace PE, et al. Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo. Clin. Cancer Res. 2001;7(4):971–976. [PubMed] [Google Scholar]
- 33.Kwon HJ, Owa T, Hassig CA, et al. Depudecin induces morphological reversion of transformed fibroblasts via the inhibition of histone deacetylase. Proc. Natl Acad. Sci. USA. 1998;95(7):3356–3361. doi: 10.1073/pnas.95.7.3356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Marks P, Rifkind RA, Richon VM, et al. Histone deacetylases and cancer: causes and therapies. Nature Rev. 2001;1(3):194–202. doi: 10.1038/35106079. [DOI] [PubMed] [Google Scholar]
- 35.Zhou XC, Dowdy SC, Podratz KC, et al. Epigenetic considerations for endometrial cancer prevention, diagnosis and treatment. Gynecol. Oncol. 2007;107(1):143–153. doi: 10.1016/j.ygyno.2007.06.019. [DOI] [PubMed] [Google Scholar]
- 36.Terao Y, Nishida J, Horiuchi S, et al. Sodium butyrate induces growth arrest and senescence-like phenotypes in gynecologic cancer cells. Int. J. Cancer. 2001;94(2):257–267. doi: 10.1002/ijc.1448. [DOI] [PubMed] [Google Scholar]
- 37.Takai N, Kawamata N, Gui D, et al. Human ovarian carcinoma cells: histone deacetylase inhibitors exhibit antiproliferative activity and potently induce apoptosis. Cancer. 2004;101(12):2760–2770. doi: 10.1002/cncr.20709. [DOI] [PubMed] [Google Scholar]
- 38.Marzio G, Wagener C, Gutierrez MI, et al. E2F family members are differentially regulated by reversible acetylation. J. Biol. Chem. 2000;275(15):10887–10892. doi: 10.1074/jbc.275.15.10887. [DOI] [PubMed] [Google Scholar]
- 39.Ammanamanchi S, Freeman JW, Brattain MG. Acetylated SP3 is a transcriptional activator. J. Biol. Chem. 2003;278(37):35775–35780. doi: 10.1074/jbc.M305961200. [DOI] [PubMed] [Google Scholar]
- 40.Mariadason JM, Velcich A, Wilson AJ, et al. Resistance to butyrate-induced cell differentiation and apoptosis during spontaneous Caco-2 cell differentiation. Gastroenterology. 2001;120(4):889–899. doi: 10.1053/gast.2001.22472. [DOI] [PubMed] [Google Scholar]
- 41.Dowdy SC, Jiang S, Zhou XC, et al. Histone deacetylase inhibitors and paclitaxel cause synergistic effects on apoptosis and microtubule stabilization in papillary serous endometrial cancer cells. Mol. Cancer Ther. 2006;5(11):2767–2776. doi: 10.1158/1535-7163.MCT-06-0209. [DOI] [PubMed] [Google Scholar]
- 42.Marks PA, Richon VM, Rifkind RA. Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J. Natl Cancer Inst. 2000;92(15):1210–1216. doi: 10.1093/jnci/92.15.1210. ▪▪ Inhibition of HDAC activity leads to the modulation of expression of a specific set of genes, which in turn results in growth arrest, differentiation and/or cell death. The hydroxamic acid-based hybrid polar compounds are potentially effective agents for cancer therapy and, possibly, cancer chemoprevention.
- 43.Khabele D, Son DS, Parl AK, et al. Drug-induced inactivation or gene silencing of class I histone deacetylases suppresses ovarian cancer cell growth: implications for therapy. Cancer Biol. Ther. 2007;6(5):795–801. doi: 10.4161/cbt.6.5.4007. [DOI] [PubMed] [Google Scholar]
- 44.Weichert W. HDAC expression and clinical prognosis in human malignancies. Cancer Lett. 2009;280(2):168–176. doi: 10.1016/j.canlet.2008.10.047. ▪ Points to a potential biological role of altered HDAC expression in human tumors. Future translational studies will focus on such question as whether HDAC expression patterns could be prognostic for response to treatment with HDAC inhibitors.
- 45.Glass CK, Rosenfeld MG. The coregulator exchange in transcriptional functions of nuclear receptors. Genes Dev. 2000;14(2):121–141. [PubMed] [Google Scholar]
- 46.Wilson AJ, Byun DS, Popova N, et al. Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. J. Biol. Chem. 2006;281(19):13548–13558. doi: 10.1074/jbc.M510023200. [DOI] [PubMed] [Google Scholar]
- 47.Ayer DE. Histone deacetylases: transcriptional repression with SINers and NuRDs. Trends Cell. Biol. 1999;9(5):193–198. doi: 10.1016/s0962-8924(99)01536-6. [DOI] [PubMed] [Google Scholar]
- 48.Yang XJ, Gregoire S. Class II histone deacetylases: from sequence to function, regulation, and clinical implication. Mol. Cell Biol. 2005;25(8):2873–2884. doi: 10.1128/MCB.25.8.2873-2884.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yang XJ, Seto E. The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men. Nat. Rev. Mol. Cell Biol. 2008;9(3):206–218. doi: 10.1038/nrm2346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Weichert W, Denkert C, Noske A, et al. Expression of class I histone deacetylases indicates poor prognosis in endometrioid subtypes of ovarian and endometrial carcinomas. Neoplasia. 2008;10(9):1021–1027. doi: 10.1593/neo.08474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Krusche CA, Vloet AJ, Classen-Linke I, et al. Class I histone deacetylase expression in the human cyclic endometrium and endometrial adenocarcinomas. Hum. Reprod. 2007;22(11):2956–2966. doi: 10.1093/humrep/dem241. [DOI] [PubMed] [Google Scholar]
- 52.Kawai H, Li H, Avraham S, et al. Overexpression of histone deacetylase HDAC1 modulates breast cancer progression by negative regulation of estrogen receptor alpha. Int. J. Cancer. 2003;107(3):353–358. doi: 10.1002/ijc.11403. [DOI] [PubMed] [Google Scholar]
- 53.Zhu P, Martin E, Mengwasser J, et al. Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell. 2004;5(5):455–463. doi: 10.1016/s1535-6108(04)00114-x. [DOI] [PubMed] [Google Scholar]
- 54.Rampalli S, Pavithra L, Bhatt A, et al. Tumor suppressor SMAR1 mediates cyclin D1 repression by recruitment of the SIN3/histone deacetylase 1 complex. Mol. Cell Biol. 2005;25(19):8415–8429. doi: 10.1128/MCB.25.19.8415-8429.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jin KL, Pak JH, Park JY, et al. Expression profile of histone deacetylases 1, 2 and 3 in ovarian cancer tissues. J. Gynecol. Oncol. 2008;19(3):185–190. doi: 10.3802/jgo.2008.19.3.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Shang Y, Hu X, DiRenzo J, et al. Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription. Cell. 2000;103(6):843–852. doi: 10.1016/s0092-8674(00)00188-4. [DOI] [PubMed] [Google Scholar]
- 57.Liu XF, Bagchi MK. Recruitment of distinct chromatin-modifying complexes by tamoxifen-complexed estrogen receptor at natural target gene promoters in vivo. J. Biol. Chem. 2004;279(15):15050–15058. doi: 10.1074/jbc.M311932200. [DOI] [PubMed] [Google Scholar]
- 58.Hrzenjak A, Moinfar F, Kremser ML, et al. Valproate inhibition of histone deacetylase 2 affects differentiation and decreases proliferation of endometrial stromal sarcoma cells. Mol. Cancer Ther. 2006;5(9):2203–2210. doi: 10.1158/1535-7163.MCT-05-0480. [DOI] [PubMed] [Google Scholar]
- 59.Drummond DC, Noble CO, Kirpotin DB, et al. Clinical development of histone deacetylase inhibitors as anticancer agents. Annu. Rev. Pharmacol. Toxicol. 2005;45:495–528. doi: 10.1146/annurev.pharmtox.45.120403.095825. ▪▪ Improved understanding of the mechanistic role of specific HDACs in human tumorigenesis, as well as the identification of more specific HDAC inhibitors, will likely accelerate the clinical development and broaden the future scope and utility of HDAC inhibitors for cancer treatment.
- 60.Botrugno OA, Santoro F, Minucci S. Histone deacetylase inhibitors as a new weapon in the arsenal of differentiation therapies of cancer. Cancer Lett. 2009;280(2):134–144. doi: 10.1016/j.canlet.2009.02.027. ▪▪ HDAC inhibitors can act both on cancer stem cells and the rest of the tumor cell mass, leading to complex biological outputs. As a note of caution, when used as single agent, HDAC inhibitors show only a moderate and limited biological response, which is augmented in combinatorial therapies with drugs designed against other epigenetic targets. This approach will target a set of enzymes responsible for the set-up and maintenance of the epigenome.
- 61.Qian DZ, Wang X, Kachhap SK, et al. The histone deacetylase inhibitor NVP-LAQ824 inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584. Cancer Res. 2004;64(18):6626–6634. doi: 10.1158/0008-5472.CAN-04-0540. [DOI] [PubMed] [Google Scholar]
- 62.Deroanne CF, Bonjean K, Servotte S, et al. Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene. 2002;21(3):427–436. doi: 10.1038/sj.onc.1205108. [DOI] [PubMed] [Google Scholar]
- 63.Michaelis M, Michaelis UR, Fleming I, et al. Valproic acid inhibits angiogenesis in vitro and in vivo. Mol. Pharmacol. 2004;65(3):520–527. doi: 10.1124/mol.65.3.520. [DOI] [PubMed] [Google Scholar]
- 64.Sawa H, Murakami H, Ohshima Y, et al. Histone deacetylase inhibitors such as sodium butyrate and trichostatin A inhibit vascular endothelial growth factor (VEGF) secretion from human glioblastoma cells. Brain Tumor Pathol. 2002;19(2):77–81. doi: 10.1007/BF02478931. [DOI] [PubMed] [Google Scholar]
- 65.Takai N, Ueda T, Nishida M, et al. Histone deacetylase inhibitors induce growth inhibition, cell cycle arrest and apoptosis in human choriocarcinoma cells. Int. J. Mol. Med. 2008;21(1):109–115. [PubMed] [Google Scholar]
- 66.Takai N, Narahara H. Human endometrial and ovarian cancer cells: histone deacetylase inhibitors exhibit antiproliferative activity, potently induce cell cycle arrest, and stimulate apoptosis. Curr. Med. Chem. 2007;14(24):2548–2553. doi: 10.2174/092986707782023299. [DOI] [PubMed] [Google Scholar]
- 67.Takai N, Ueda T, Nishida M, et al. CBHA is a family of hybrid polar compounds that inhibit histone deacetylase, and induces growth inhibition, cell cycle arrest and apoptosis in human endometrial and ovarian cancer cells. Oncology. 2006;70(2):97–105. doi: 10.1159/000092585. [DOI] [PubMed] [Google Scholar]
- 68.Ueda T, Takai N, Nishida M, et al. Apicidin, a novel histone deacetylase inhibitor, has profound anti-growth activity in human endometrial and ovarian cancer cells. Int. J. Mol. Med. 2007;19(2):301–308. [PubMed] [Google Scholar]
- 69.Inoue K, Kobayashi M, Yano K, et al. Histone deacetylase inhibitor reduces monocyte adhesion to endothelium through the suppression of vascular cell adhesion molecule-1 expression. Arterioscler. Thromb. Vasc. Biol. 2006;26(12):2652–2659. doi: 10.1161/01.ATV.0000247247.89787.e7. [DOI] [PubMed] [Google Scholar]
- 70.Pina IC, Gautschi JT, Wang GY, et al. Psammaplins from the sponge Pseudoceratina purpurea: inhibition of both histone deacetylase and DNA methyltransferase. J. Org. Chem. 2003;68(10):3866–3873. doi: 10.1021/jo034248t. [DOI] [PubMed] [Google Scholar]
- 71.Ahn MY, Jung JH, Na YJ, et al. A natural histone deacetylase inhibitor, Psammaplin A, induces cell cycle arrest and apoptosis in human endometrial cancer cells. Gynecol. Oncol. 2008;108(1):27–33. doi: 10.1016/j.ygyno.2007.08.098. [DOI] [PubMed] [Google Scholar]
- 72.Tabudravu JN, Eijsink VG, Gooday GW, et al. Psammaplin A, a chitinase inhibitor isolated from the Fijian marine sponge Aplysinella rhax. Bioorg. Med. Chem. 2002;10(4):1123–1128. doi: 10.1016/s0968-0896(01)00372-8. [DOI] [PubMed] [Google Scholar]
- 73.Huang H, Tindall DJ. FOXO factors: a matter of life and death. Future Oncol. 2006;2(1):83–89. doi: 10.2217/14796694.2.1.83. [DOI] [PubMed] [Google Scholar]
- 74.Myzak MC, Karplus PA, Chung FL, et al. A novel mechanism of chemoprotection by sulforaphane: inhibition of histone deacetylase. Cancer Res. 2004;64(16):5767–5774. doi: 10.1158/0008-5472.CAN-04-1326. [DOI] [PubMed] [Google Scholar]
- 75.Myzak MC, Hardin K, Wang R, et al. Sulforaphane inhibits histone deacetylase activity in BPH-1, LnCaP and PC-3 prostate epithelial cells. Carcinogenesis. 2006;27(4):811–819. doi: 10.1093/carcin/bgi265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Jiang S, Dowdy SC, Meng XW, et al. Histone deacetylase inhibitors induce apoptosis in both type I and type II endometrial cancer cells. Gynecol. Oncol. 2007;105(2):493–500. doi: 10.1016/j.ygyno.2007.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Uchida H, Maruyama T, Ono M, et al. Histone deacetylase inhibitors stimulate cell migration in human endometrial adenocarcinoma cells through up-regulation of glycodelin. Endocrinology. 2007;148(2):896–902. doi: 10.1210/en.2006-0896. [DOI] [PubMed] [Google Scholar]
- 78.Seppala M, Taylor RN, Koistinen H, et al. Glycodelin: a major lipocalin protein of the reproductive axis with diverse actions in cell recognition and differentiation. Endocr. Rev. 2002;23(4):401–430. doi: 10.1210/er.2001-0026. [DOI] [PubMed] [Google Scholar]
- 79.Mandelin E, Lassus H, Seppala M, et al. Glycodelin in ovarian serous carcinoma: association with differentiation and survival. Cancer Res. 2003;63(19):6258–6264. [PubMed] [Google Scholar]
- 80.Folkman J. Role of angiogenesis in tumor growth and metastasis. Semin. Oncol. 2002;29(6 Suppl. 16):15–18. doi: 10.1053/sonc.2002.37263. [DOI] [PubMed] [Google Scholar]
- 81.Folkman J. Angiogenesis and proteins of the hemostatic system. J. Thromb. Haemost. 2003;1(8):1681–1682. doi: 10.1046/j.1538-7836.2003.00344.x. [DOI] [PubMed] [Google Scholar]
- 82.Michaelis M, Suhan T, Reinisch A, et al. Increased replication of human cytomegalovirus in retinal pigment epithelial cells by valproic acid depends on histone deacetylase inhibition. Invest. Ophthalmol. Vis. Sci. 2005;46(9):3451–3457. doi: 10.1167/iovs.05-0369. [DOI] [PubMed] [Google Scholar]
- 83.Qian DZ, Kato Y, Shabbeer S, et al. Targeting tumor angiogenesis with histone deacetylase inhibitors: the hydroxamic acid derivative LBH589. Clin. Cancer Res. 2006;12(2):634–642. doi: 10.1158/1078-0432.CCR-05-1132. [DOI] [PubMed] [Google Scholar]
- 84.Kim MS, Kwon HJ, Lee YM, et al. Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat. Med. 2001;7(4):437–443. doi: 10.1038/86507. [DOI] [PubMed] [Google Scholar]
- 85.Kwon HJ, Kim MS, Kim MJ, et al. Histone deacetylase inhibitor FK228 inhibits tumor angiogenesis. Int. J. Cancer. 2002;97(3):290–296. doi: 10.1002/ijc.1602. [DOI] [PubMed] [Google Scholar]
- 86.Teodoridis JM, Strathdee G, Brown R. Epigenetic silencing mediated by CpG island methylation: potential as a therapeutic target and as a biomarker. Drug Resist. Updat. 2004;7(4–5):267–278. doi: 10.1016/j.drup.2004.06.005. [DOI] [PubMed] [Google Scholar]
- 87.Graham JS, Kaye SB, Brown R. The promises and pitfalls of epigenetic therapies in solid tumours. Eur. J. Cancer. 2009;45(7):1129–1136. doi: 10.1016/j.ejca.2009.01.003. [DOI] [PubMed] [Google Scholar]
- 88.Zhu WG, Lakshmanan RR, Beal MD, et al. DNA methyltransferase inhibition enhances apoptosis induced by histone deacetylase inhibitors. Cancer Res. 2001;61(4):1327–1333. [PubMed] [Google Scholar]
- 89.Chobanian NH, Greenberg VL, Gass JM, et al. Histone deacetylase inhibitors enhance paclitaxel-induced cell death in ovarian cancer cell lines independent of p53 status. Anticancer Res. 2004;24(2B):539–545. [PubMed] [Google Scholar]
- 90.Ramondetta L, Burke TW, Levenback C, et al. Treatment of uterine papillary serous carcinoma with paclitaxel. Gynecol. Oncol. 2001;82(1):156–161. doi: 10.1006/gyno.2001.6211. [DOI] [PubMed] [Google Scholar]
- 91.Zanotti KM, Belinson JL, Kennedy AW, et al. The use of paclitaxel and platinum-based chemotherapy in uterine papillary serous carcinoma. Gynecol. Oncol. 1999;74(2):272–277. doi: 10.1006/gyno.1999.5444. [DOI] [PubMed] [Google Scholar]
- 92.Sonnemann J, Gange J, Pilz S, et al. Comparative evaluation of the treatment efficacy of suberoylanilide hydroxamic acid (SAHA) and paclitaxel in ovarian cancer cell lines and primary ovarian cancer cells from patients. /BMC cancer. 2006;6:183. doi: 10.1186/1471-2407-6-183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Cooper AL, Greenberg VL, Lancaster PS, et al. In vitro and in vivo histone deacetylase inhibitor therapy with suberoylanilide hydroxamic acid (SAHA) and paclitaxel in ovarian cancer. Gynecol. Oncol. 2007;104(3):596–601. doi: 10.1016/j.ygyno.2006.09.011. [DOI] [PubMed] [Google Scholar]
- 94.Plumb JA, Finn PW, Williams RJ, et al. Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101. Mol. Cancer Ther. 2003;2(8):721–728. [PubMed] [Google Scholar]
- 95.Gimsing P, Hansen M, Knudsen LM, et al. A Phase I clinical trial of the histone deacetylase inhibitor belinostat in patients with advanced hematological neoplasia. Eur. J. Haematol. 2008;81(3):170–176. doi: 10.1111/j.1600-0609.2008.01102.x. [DOI] [PubMed] [Google Scholar]
- 96.Ramalingam SS, Belani CP, Ruel C, et al. Phase II study of belinostat (PXD101), a histone deacetylase inhibitor, for second line therapy of advanced malignant pleural mesothelioma. J. Thorac. Oncol. 2009;4(1):97–101. doi: 10.1097/JTO.0b013e318191520c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Sonnemann J, Huls I, Sigler M, et al. Histone deacetylase inhibitors and aspirin interact synergistically to induce cell death in ovarian cancer cells. Oncol. Rep. 2008;20(1):219–224. [PubMed] [Google Scholar]
- 98.Zhang Y, Li N, Caron C, et al. HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo. EMBO J. 2003;22(5):1168–1179. doi: 10.1093/emboj/cdg115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Schiff PB, Horwitz SB. Taxol stabilizes microtubules in mouse fibroblast cells. Proc. Natl Acad. Sci. USA. 1980;77(3):1561–1565. doi: 10.1073/pnas.77.3.1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Uchida H, Maruyama T, Nagashima T, et al. Histone deacetylase inhibitors induce differentiation of human endometrial adenocarcinoma cells through up-regulation of glycodelin. Endocrinology. 2005;146(12):5365–5373. doi: 10.1210/en.2005-0359. [DOI] [PubMed] [Google Scholar]
- 101.Modesitt SC, Sill M, Hoffman JS, et al. A Phase II study of vorinostat in the treatment of persistent or recurrent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecol. Oncol. 2008;109(2):182–186. doi: 10.1016/j.ygyno.2008.01.009. [DOI] [PubMed] [Google Scholar]
- 102.Arts J, Angibaud P, Marien A, et al. R306465 is a novel potent inhibitor of class I histone deacetylases with broad-spectrum antitumoral activity against solid and haematological malignancies. Br. J. Cancer. 2007;97(10):1344–1353. doi: 10.1038/sj.bjc.6604025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Hoffman MA, Blessing JA, Morgan M. Phase II trial of CI-958 in recurrent platinum-refractory ovarian carcinoma. A Gynecologic Oncology Group Study. Gynecol. Oncol. 2000;79(3):463–465. doi: 10.1006/gyno.2000.5984. [DOI] [PubMed] [Google Scholar]
- 104.Glaser KB. HDAC inhibitors: clinical update and mechanism-based potential. Biochem. Pharmacol. 2007;74(5):659–671. doi: 10.1016/j.bcp.2007.04.007. [DOI] [PubMed] [Google Scholar]
- 105.Steele NL, Plumb JA, Vidal L, et al. A Phase 1 pharmacokinetic and pharmacodynamic study of the histone deacetylase inhibitor belinostat in patients with advanced solid tumors. Clin. Cancer Res. 2008;14(3):804–810. doi: 10.1158/1078-0432.CCR-07-1786. [DOI] [PubMed] [Google Scholar]
- 106.Dallavalle S, Cincinelli R, Nannei R, et al. Design, synthesis, and evaluation of biphenyl-4-yl-acrylohydroxamic acid derivatives as histone deacetylase (HDAC) inhibitors. Eur. J. Med. Chem. 2009;44(5):1900–1912. doi: 10.1016/j.ejmech.2008.11.005. [DOI] [PubMed] [Google Scholar]
- 107.Takai N, Ueda T, Nishida M, et al. M344 is a novel synthesized histone deacetylase inhibitor that induces growth inhibition, cell cycle arrest, and apoptosis in human endometrial cancer and ovarian cancer cells. Gynecol. Oncol. 2006;101(1):108–113. doi: 10.1016/j.ygyno.2005.09.044. [DOI] [PubMed] [Google Scholar]
- 108.Xiong Y, Dowdy SC, Podratz KC, et al. Histone deacetylase inhibitors decrease DNA methyltransferase-3B messenger RNA stability and down-regulate de novo DNA methyltransferase activity in human endometrial cells. Cancer Res. 2005;65(7):2684–2689. doi: 10.1158/0008-5472.CAN-04-2843. [DOI] [PubMed] [Google Scholar]
Websites
- 201.National Cancer Institute [Accessed 18 October 2011];NCI Drug Dictionary: romidepsin. www.cancer.gov/Templates/drugdictionary.aspx?CdrID=42600.
- 202. [Accessed 18 October 2011];NIH clinical trial: FR901228 in Treating Patients with Metastatic Breast Cancer. http://clinicaltrials.gov/ct2/show/NCT00098397.
- 203. [Accessed 18 October 2011];NIH clinical trial: FR901228 in Treating Patients with Relapsed or Refractory Advanced Ovarian Epithelial Cancer. http://clinicaltrials.gov/ct2/show/NCT00085527.
- 204. [Accessed 18 October 2011];NIH clinical trial: FR901228 in Treating Patients with Unresectable Stage III or Stage IV Malignant Melanoma. http://clinicaltrials.gov/ct2/show/NCT00104884.
- 205. [Accessed 18 October 2011];NIH clinical trial: FR901228 in Treating Patients with Locally Advanced or Metastatic Neuroendocrine Tumors. http://clinicaltrials.gov/ct2/show/NCT00084461.
- 206. [Accessed 18 October 2011];NIH clinical trial: FR901228 in Treating Patients with Relapsed or Refractory Acute Myeloid Leukemia. http://clinicaltrials.gov/ct2/show/NCT00062075.