Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Feb 1.
Published in final edited form as: Gynecol Oncol. 2009 Oct 24;116(2):195. doi: 10.1016/j.ygyno.2009.09.043

Epigenetic Therapies for Chemoresensitization of Epithelial Ovarian Cancer

Daniela E Matei 1,2,3,*, Kenneth P Nephew 2,3,4,5,*
PMCID: PMC2813995  NIHMSID: NIHMS151583  PMID: 19854495

Summary

Epigenetic drugs have been shown to enhance gene expression and drug sensitivity in ovarian cancer cell lines and animal models. Based on promising pre-clinical studies, DNA methylation inhibitors in combination with existing chemotherapeutic agents have the potential for overcoming acquired drug resistance, laying the foundation for this specific class of epigenetic drug in ovarian cancer clinical trials. The recent completion of phase I trials of decitabine have yielded important information on dosing schedules and biological endpoints for evaluating patient responses. In addition, epigenetic drug effects on pharmacodyamic targets are beginning to emerge, and predictive epigenetic biomarkers and next generation epigenome therapeutics are being developed for application in clinical settings for ovarian cancer patients.

Introduction

Epithelial ovarian cancer is the most lethal gynecologic malignancy [1] causing an estimated 15,520 U.S. deaths in 2008 [2]. Most patients are diagnosed with advanced disease, and the five-year survival rate is below 25% for patients diagnosed with stage III - IV epithelial ovarian cancer [1, 3]. Most advanced stage patients respond to cytoreductive surgery and platinum-based chemotherapy, however over 70% of women relapse, and platinum-resistant epithelial ovarian cancer is uniformly fatal [4, 5].

Altered epigenetic states are a hallmark of all cancers [6, 7], including ovarian [8, 9]. Epigenetics is defined as a heritable change in gene expression without alteration of the DNA sequence itself and includes DNA methylation, histone modification, nucleosome repositioning, and most recently posttranscriptional gene regulation by micro-RNAs (miRNAs) [7, 10-12]. The transfer of a methyl group to the carbon-5 position of cytosines, almost always within the context of cytosine-guanine (CpG) dinucleotides, is the best-studied epigenetic mark and only known covalent modification of DNA itself in mammalian cells. DNA-associated histone proteins are subject to extensive modifications that mediate the assembly of transcriptionally permissive or repressive (i.e. open or closed) chromatin, and it is now recognized that DNA methylation and histone modifications are intimately linked [7]. The overall epigenetic state (e.g. DNA methylation, histone modification, and miRNA expression) corresponding to a specific cell phenotype is now referred to as the epigenome [13].

Altered epigenetic states in ovarian cancer

The first described epigenetic change in ovarian epithelial cancer was loss of DNA methylation (14). Global DNA hypomethylation in cancer is largely due to decreased methylation of repeat DNA (non-gene sequences), including centromeric satellite α DNA and juxtacentromeric satellite DNA (classical satellite 2), Alu repeats, and LINE-1 repeats [reviewed in 15]. In ovarian carcinogenesis, the extent of global and satellite DNA hypomethylation was significantly associated with the degree of malignancy [16-18]. Satellite DNA hypomethylation was shown to increase with advanced ovarian tumors and serve as an independent marker of poor prognosis [20]. Hypomethylation may contribute to ovarian carcinogenesis by promoting tumor formation or progression in a number of possible ways, including affecting transposable element activation, DNA/chromosomal rearrangements, tumor suppressor gene or oncogene copy number, and/or altered chromosome conformation [reviewed in 21]. In addition to repetitive elements and DNA satellites, promoter CpG island hypomethylation and gene overexpression has been reported in ovarian cancer. CpG islands are DNA sequences containing an atypically high frequency CpG sites [22]. CpG islands generally lack DNA methylation and are usually but not exclusively associated with gene promoters [6, 13, 22]. In normal ovarian surface epithelial cells, some CpG islands are methylated and do not express the associated containing genes. Hypomethylation and re-expression of a number of those protein encoding genes in ovarian cancer has been associated with chemoresistance, including MCJ [23], SNCG [24], and BORIS [25]. Hypomethylation of IGF2, an imprinted gene [26], and claudin-4, whose overexpression leads to disrupted tight junctions between epithelial ovarian cancer cells [27], has also been reported in ovarian cancer.

Increased methylation of CpG islands is a common occurrence in epithelial ovarian cancer [8, 9], and CpG island hypermethylation is associated with epigenetic silencing during all phases of the cancer process, including tumor initiation, progression and drug resistance [6, 7]. CpG islands aberrantly methylated in ovarian tumors are associated with silencing of genes involved in control of the cell cycle, apoptosis and drug sensitivity, as well as tumor suppressor genes [8]. A number of genes are hypermethylated and down-regulated in ovarian cancer, including the classical tumor suppressors BRCA1 (breast cancer susceptibility gene-1) [28, 29], p16 [30], and MLH1 [29, 31], putative/candidate tumor suppressor (RASSF1A, OPCML, SPARC, ANGPTL2, CTGF) [32-36] imprinted genes (ARH1 and PEG3) [37] and proapoptotic genes (LOT1, DAPK, TMS1/ASC, and PAR-4) [38-40], as well as genes associated with cell adhesion (ICAM-1, CDH1 [41, 42], cell signaling (HSulf-1) [43, 44], genome stability (PALB2) [45], and taxane resistance (TUBB3) [46], and embryonic development (HOXA10, HOXA11 [47].

Methylation microarrays have been used to globally examine DNA methylation in ovarian cancer cell lines and patient samples [48-53]. Those studies have demonstrated that ovarian tumors contain a large number of hypermethylated loci and that the degree of aberrant methylation (i.e., the total number of methylated genes) is directly correlated with ovarian tumor progression and recurrence. Methylation profiling combined with bioinformatic approaches have also identified specific methylated loci statistically associated with poor progression-free survival in ovarian cancer. As such, “methylation signatures” may contribute to the further classification of ovarian tumors and identification of altered biological pathways for individualizing treatment strategies. With the objective of using methylated loci as biomarkers for monitoring epigenetic therapies [54], we recently developed a model system to examine DNA methylation changes associated with the onset of drug resistance in ovarian cancer [53]. By integrating DNA methylation and gene expression profiles and applying bioinformatic approaches, our pathway analyses suggested significant disruption of tumor-suppressive functions by hypermethylation and upregulation of tumor-promoting cascades by hypomethylation [53]. Such experimental and computational approaches may be highly valuable for identifying key mediators of chemotherapy resistance as potential biomarkers or therapeutic targets.

Altered methylation profiles may be associated with increased or altered activity of the DNA methyltransferases (DNMTs), the family of enzymes that catalyze the transfer of a methyl group to DNA, using S-adenosyl methionine (SAM) as the methyl donor [7]. Although the relationship between gene hypermethylation and altered DNMT RNA levels in ovarian cancer cells is not straightforward [18, 55], significant upregulation of DNMTs was observed during acquired cisplatin resistance in ovarian cancer [53], suggesting that aberrant methylation patterns may be associated with increased or altered DNMT activity. Furthermore, targeted downregulaton of DNMTs using small interfering RNAs (siRNAs) resulted in loss of CpG hypermethylation and growth inhibition [56], providing functional validation of promoter DNA hypermethylation in ovarian cancer.

BRCA1 is a well-studied gene in both inherited and sporadic ovarian cancer [57]. With regard to tumors, BRCA1 hypermethylation and subsequent gene silencing occurs in 10-15% cases of sporadic disease [58] and correlates with poor clinical outcome [57, 58]. However, methylation of BRCA1 and BRCA2 rarely occurs in hereditary ovarian cancer [59], further indicating that promoter methylation is a not a frequent “second-hit” in tumors from BRCA1 or BRCA2 carriers [59]. Hypermethylation of BRCA1 has recently been detected in the serum of ovarian cancer patients [32] and serum detection of epigenetic methylation of specific genes (e.g., BRCA1, MLH1 and others) [32, 60, 61] may serve as predictive markers for patient response to standard therapies, epigenetic therapies [62], or targeted therapies for ovarian cancer [63, 64].

DNA methylation and histone modifications regulate numerous normal ovarian functions [65], and altered expression of chromatin-modifying proteins has recently been reported in ovarian cancer cells [66]. Gene silencing by histone modifications, in the absence of DNA methylation, has been reported for GATA4 and GATA6 [67], cyclinB1 [68] and p21WAF1/CIP1 [69]. Similarly, repressive histone modifications (trimethyl-H3K27 and dimethyl-H3K9) together with histone deacetylase (HDAC) enzymes act to down-regulate ADAM19 without CpG island methylation in TGF-β1-refractory ovarian cancer cells [70], demonstrating that aberrant TGF-β1 signaling can result in formation of a repressive chromatin environment, without DNA methylation. In addition, genome-wide loss of the repressive trimethyl-H3K27me3 mark was associated with reduced global DNA methylation, allowing platinum resensitization of chemoresistant ovarian cancer cells, and allowing for identification of direct target genes of H3K27 methylation-mediated silencing [71]. Complex DNA methylation and histone modification patterns almost certainly contribute to ovarian cancer progression and drug resistance in patients, as loss of H3K27 trimethylation has recently been associated with poor prognosis in ovarian and other malignancies [72], and gene promoter DNA methylation can be maintained in the absence of this repressive mark [71].

Preclinical epigenetic drugs studies in ovarian cancer

Unlike cancer-associated gene mutations, amplifications, and deletions, DNA methylation and other epigenetic modifications are potentially reversible. Based on extensive findings of aberrant DNA methylation in malignancy, inhibitors of DNA methyltransferases (DNMTIs) have been examined as a means of inducing re-expression of tumor suppressor genes and reversal of malignant phenotypes [73]. These drugs are analogues of deoxycytosine possessing various substitutions at their 5-carbons, and effectively prevent transfer of the methyl group. Consequently, upon phosphorylation and incorporation into DNA, DNMTIs irreversibly “trap” the methyltransferase enzyme in a transition state complex, which is subsequently eliminated from the cell [73]. Numerous cytosine analogs that covalently and irreversibly bind to the active site of DNMTs are being investigated for their ability to clinically reverse CpG island methylation in cancer and derepress epigenetically silenced genes [54].

Recent preclinical studies have suggested that hypomethylating agents are more effective in combination with conventional chemotherapy, by promoting resensitization of chemoresistant tumor cells. It was hypothesized that this effect is due to re-expression of tumor suppressor genes by promoter demethylation; activation of those genes may then restore drug response apoptotic pathways. Several preclinical models support this hypothesis [74-78]. For instance, in an ovarian cancer model, decitabine and a related epigenetic modulator, zebularine, mediated resensitization of cisplatin-resistant epithelial ovarian cancer cells to platinum [78]. This occurred as a consequence of upregulation of tumor suppressor genes (RASSF1A, hMLH1) [78]. In another study, treatment with decitabine allowed re-expression of the DNA repair gene hMLH1 in platinum resistant A2780/CP70 ovarian cancer cells, and xenograft tumors derived from these cells were sensitized by decitabine to cisplatin, carboplatin, temozolomide, and epirubicin [74].

Histone deacetylation is another transcriptional silencing mechanism in ovarian cancer and anticancer effects of HDAC inhibitors (HDACIs) are due to inhibition of deacetylation of nonhistone proteins and subsequent release from epigenetic gene repression [79]. Preclinical studies in ovarian cancer include resensitization of ovarian cancer cells and platinum-resistant xenografts in mice by the HDACIs (belinostat; CuraGen Corp., Branford, CT) [80]; AR-42 (Arno Therapeutics, Parsippany, NJ) [81]), supporting the use of these HDACIs in ovarian cancer clinical trials. Moreover, additive or synergistic effects of HDACI and DNMTI combinations on silenced gene reexpression have been demonstrated [82], suggesting that combining these two classes of epigenetic drugs with conventional therapies may be the most effective approach to use in the clinic [62]. Toward this possibility, one preclinical study showed that a combination of decitabine with belinostat elicited greater platinum resensitization of resistant ovarian cancer xenografts than decitabine alone [83].

Summary

Due to the high levels of recurrence associated with ovarian cancer, there is a need for new treatment options for platinum resistant disease. Therapeutic agents currently under investigation include anti-angiogenesis targeted therapies, antibody-directed therapy, DNA topoisomerase inhibitors, and intraperitoneal administration of chemotherapy [3]. As discussed above, ovarian cancer cells harbor a significantly altered epigenome. Hypermethylation of promoter CpG islands, alterations in histone methylation, and interplay between DNA methylation and histone modifications result in aberrant silencing of tumor suppressor genes in ovarian cancer. Furthermore, these repressive epigenetic alterations are associated with drug resistant disease. Promising pre-clinical results using DNMT and HDAC inhibitors for chemotherapy resensitization in ovarian cancer cell lines and animal models have been reported, laying the foundation for effective epigenetic drugs in combination with platinum-based agents for overcoming resistance in women with recurrent ovarian cancer.

Clinical experience with hypomethylating agents in ovarian cancer

Based on the success of these inhibitors in the L1210 mouse leukemia model [84], early studies have focused on hematologic malignancies and specifically, leukemias and myelodyspastic syndromes (MDS) [85]. The first DNMTIs studied were 5-azacytidine (5-aza-C, Vidaza) and its deoxyribose analog, 5-aza-2′-deoxycytidine (5-aza-dC, decitabine); both were subsequently approved by the FDA for the treatment of MDS [86-91]. Their effects have been attributed to induction of cellular differentiation, directly related to reversal of epigenetic alterations [92-95]. Early studies with these agents followed the traditional model of a drug studied at or near its maximal tolerated dose (MTD) and utilized high doses of azacitadine and decitabine. Consequently, these trials were limited by toxicity and, particularly, by myelosupression [96, 97]. However, preclinical models showing that low doses of decitabine or azacitadine induce DNA demethylation, have fueled the redesign of clinical trials using regimens targeting a “biologically effective” dose of hypomethylating agents, rather than the MTD. Subsequent trials emulating these in vitro findings demonstrated that doses as low as 1/10 of MTD preserved clinical effectiveness, while improving tolerability [98-100].

Hypomethylating agents have been explored in solid tumors both as single agents or in combination regimens [101-102]. In a phase I study in patients with thoracic malignancies, decitabine was administered as a continuous infusion and was dose escalated in a typical phase I schema [103]. The MTD was 60-75mg/m2 given over 72 hours, with myelosupression as the most relevant toxicity. Tumor biopsies obtained before and 24 hours after completion of the infusion demonstrated induction of NY-ESO-1, MAGE-3 and p16 expression in roughly a third of treated patients. That trial provided proof-of-concept that decitabine modulates gene expression in solid tumors, by inducing DNA demethylation. In another phase I study, DNA hypomethylation of a panel of 19 genes was induced by continuous infusion of decitabine, although no single gene showed consistent demethylation among patients [101]. Interestingly, in that study DNA hypomethylation was documented seven days after completion of the decitabine infusion [101], which is consistent with the concept that epigenetic modulation is time-dependent, necessitating progression through several cell cycles. A single study evaluated the activity and tolerability of a demethylating agent in patients with ovarian cancer. Fazarabine (Ara-AC), a nucleoside analogue that consists of the arabinoside ring of 1-beta-D-arabinofuranosylcytosine and the pyrimidine base of 5-azacytidine was administered at a dose of 30 mg/m2/day for 5 consecutive days, on a 28 day cycle to patients with recurrent OC. At this dose the major toxicity was hematologic, with four patients exhibiting grade 4 neutropenia. No complete or partial responses were observed among 19 patients evaluated in this study however 44% of patients had stable disease [104].

To date three clinical trials examined combinations of hypomethylating agents with chemotherapy in patients with ovarian cancer (Table 1). These studies were preceded by a phase I trial demonstrating tolerability of the combination of decitabine and carboplatin in patients with solid tumors [105]. In that trial, decitabine was given as a 6-hour infusion on Day 1 and carboplatin was given as an i.v. bolus on Day 8. DLT was myelosupression, and the maximum tolerated dose of decitabine was 90mg/m2. At that dose, DNA demethylation of a commonly hypermethylated gene (MAGE1) was documented in PBMC and in 2 of 6 tumor biopsies obtained before and after treatment [105]. The trial also demonstrated that DNA demethylation was highest between days 8 and 12 after treatment with decitabine, supporting administration of the cytotoxic agent at a later time point. Subsequently, a randomized phase II trial of the UK Cancer Research Group compared the combination decitabine and carboplatin to single agent carboplatin in patients with ovarian cancer recurring within 6-12 months after first line treatment containing a platinum regimen [106]. Decitabine was given as a 6 hour infusion at 90mg/m2 on day 1 and carboplatin was administered at an AUC of 6 on day 8. However due to dose delays caused by neutropenia in patients receiving the combination regimen, the dose of decitabine was de-escalated to 45mg/m2. An increased rate of adverse events was noted on the combination arm, with more carboplatin hypersensitivity reactions (64% vs 21%) and more treatment delays for neutropenia (36% vs 10%) compared to patients receiving single agent carboplatin. Less clinical activity was noted in patients receiving the combination regimen (0 objective responses of 11 patients treated by RECIST criteria) compared to patients receiving carboplatin (7 objective responders of 14 patients treated). Biological effects on DNA methylation were not reported.

TABLE 1.

Clinical trials in ovarian cancer testing combinations of demethylating agents and carboplatin

Study Type Demethylating Agent Carboplatin Toxicity Efficacy
UK
(Ref. 106)
Phase II randomized
(n=29)
platinum sensitive
Decitabine 90mg/m2
Day 1
AUC 6
Day 8
Neutropenia
Platinum allergy
0 responses
IUSCC
(Ref. 107)
Phase I (n=10)
Platinum resistant
Decitabine 10mg/m2 qd X
5 days
AUC 5
Day 8
Neutropenia
Platinum Allergy
1 CR
MD Anderson
(Ref. 109)
Phase II (n=30)
Platinum resistant
Azacitadine 75mg/m2 qd
X 5 days
AUC 5
Day 2
neutropenia
Fatigue
1 CR, 3 PR

CR, complete response; PR, partial response

Concomitantly, a phase I-II trial at Indiana University Simon Cancer Center (IUSCC) investigated the decitabine and carboplatin combination in patients with platinum-resistant or refractory ovarian cancer [107]. To minimize toxicity and enhance the demethylating properties of decitabine, the regimen studied in this trial used low daily doses of decitabine for five days prior to carboplatin. A similar schema of low dose decitabine as single agent had been used for elderly leukemic patients, was well tolerated and induced responses in 54% of treated patients [85]. In that leukemia study there was a gradual and slow time-to-response, consistent with the concept that DNA hypomethylation is time-dependent and requires 2-3 cell cycles to be effective, DNA demethylation being maximal between days 7 and 14. Ten patients were enrolled in the phase I component of the IUSCC trial [107], with nine patients completing at least one cycle of therapy and two dose levels were tested: dose level 1 (10 mg/m2) and 2 (20 mg/m2). DLT consisted of grade 4 neutropenia recorded in 2 of 3 patients treated at dose level 2, therefore dose level 1 was recommended for the phase II component of the trial. The most common toxicities were nausea (80%), allergic reactions (60%), neutropenia (70%), fatigue (50%), anorexia (50%), vomiting (40%), and abdominal pain (40%), the majority being grades 1-2. Grade 3-4 toxicities affecting more than one patient included neutropenia (n=4) and carboplatin allergic reaction (n=3). Efficacy analysis had only an exploratory intent in this part of the study. Patients enrolled on this protocol were heavily pre-treated, with a median number of prior regimens of 5 (range 2-9), had measurable disease and were assessed by RECIST. One complete response (10%) was observed and six patients (60%) had stable disease as their best response. At six months, four (40%) patients were without disease progression. Exploratory biomarker analyses in this study utilized plasma or peripheral blood mononuclear cells (PBMCs) collected at baseline and serially during treatment (on days 5, 8 and 15). Global DNA methylation levels were assessed by MethyLight assay of LINE-1 (long-interspersed) repetitive elements [108] in PBMCs and were reduced in all patients on days 8 and 15, as compared to day 1. Interestingly, no dose effect was observed, suggesting that low dose decitabine (e.g. 10mg/m2 daily) was sufficient to induce DNA demethylation, while avoiding excessive toxicity. Additionally, demethylation of five ovarian cancer specific genes (BRCA1, RASSF1A, WWOX, HOX A10 and HOX A11) was examined by using Methylight in plasma of patients treated on this protocol. Demethylation of BRCA1 and of HOXA11 was recorded in plasma collected on days 8 and 15 compared to baseline. The phase II trial examining this combination regimen is ongoing.

Another phase II trial conducted at M.D. Anderson Cancer Center tested a combination regimen consisting of azacitidine given iv at a dose of 75 mg/m2/day for 5 days and carboplatin administered at an AUC of 5 on day 2 on a 28 day cycle [109]. Thirty patients with platinum-resistant or refractory OC were treated on this study. Most prominent side effects were myelosupression, fatigue and nausea. In this cohort there were 4 objective responses (RR of 14%), of which one was a complete response. The median duration of response was 7.5 months with two patients continuing treatment beyond one year. The long duration of response observed in this study and the proportion of patients without progression recorded in the IUSCC phase I trial suggest that demethylation by decitabine may play a role in re-sensitizing platinum resistant ovarian tumors to platinum. Future trials testing this concept should incorporate measuring progression free survival as a primary endpoint.

Clinical experience with histone deacetylase inhibitors in ovarian cancer

The dynamic equilibrium between histone acetylation and deacetylation is tightly regulated by histone acetyltransferases (HATs) and histone deacetylase (HDAC). HDAC effects on nucleosomal histones leads to tight coiling of chromatin and silencing of genes implicated in regulation of cell survival and differentiation [110]. Because aberrant HDAC activity has been implicated in cancer, HDACIs are being investigated as anticancer therapeutics [111]. Several HDACIs have been studied, including trichostatin and butyric acid which are active in preclinical models [112], but demonstrated limited clinical activity [113]. Depsipeptide was the first HDACI with demonstrated clinical efficacy [114, 115], however this agent was not tested in ovarian cancer. Vorinostat (suberoylanilide hydroxamic acid, SAHA) is a small molecule that binds directly in the HDAC’s active site in the presence of zinc. Vorinostat can be administered orally and has excellent bioavailability, major dose-limiting toxicities in phase I trials being anorexia, dehydration, diarrhea, and fatigue [116, 117]. Accumulation of acetylated histones post-therapy was demonstrated in PBMCs in patients receiving 200 to 600 mg of oral vorinostat [116, 117]. A phase II trial of vorinostat as a single agent in patients with recurrent ovarian cancer relapsing within 12 months after platinum based therapy was performed through the Gynecologic Oncology Group (GOG) [118]. The most significant toxicities included two grade 4 toxicities (leukopenia and neutropenia), 3 constitutional grade 3 toxicities and 3 grade 3 gastrointestinal events. Among 27 women treated on this trial, two survived progression-free for over 6 months and one partial response was recorded. This level of activity was deemed insufficient for patients with recurrent OC and its further investigation as single agent was stopped. Another HDACI that recently entered the clinical arena is belinostatt (PDX101), a novel hydroxamic acid HDACI with potent antiproliferative and HDAC inhibitory activities in vitro and in xenograft ovarian and colorectal cancer models [119]. In a phase I trial, belinostat was administered i.v. as a 30-min infusion daily on days 1 to 5 of a 21-day cycle to patients with advanced solid tumors, starting with a dose of 150 mg/m2 [120]. Dose limiting toxicities were grade 3 fatigue, diarrhea and cardiac arrhythmia at 1200 mg/m2, concluding that the maximum tolerated dose was 1000 mg/m2 daily for 5 days. Dose-dependent increase in histone H4 acetylation was observed in peripheral blood mononuclear cells, with maximal effects observed at the 900 mg/m2 dose, indicating that belinostat in biologically active in vivo. Disease stabilization was observed in patients with different malignancies, including sarcoma, renal cancer, thymoma and melanoma, positioning belinostat as an interesting agent for further development in cancer. As many preclinical models suggest that HDACIs exert enhanced anti-cancer activity in combination with demethylating agents [121], chemotherapy [122], or other biological agents [123, 124], we believe that further development of HDACIs should include rationally designed combinations.

Figure 1.

Figure 1

Resensitization of chemoresistant ovarian tumors to carboplatin by epigenetic drug therapy. A. DNA methylation inhibitors (DNMTIs) or histone deacetylase inhibitors (HDACIs), alone or in combination, can reverse platinum resistance in chemoresistant ovarian cancer Additive or synergistic effects of DNMTI and HDACI combinations on silenced gene reexpression have been demonstrated. B. This chemosensitization is hypothesized to be due to the derepression of tumor suppressor genes (TSG) that were previously silenced by promoter DNA methylation or a transcriptionally repressed (closed) chromatin environment. Epigenetic therapies restore TSG gene expression by creating a more active (open) chromatin environment, reestablishing chemotherapy drug response cascades. See text for details. Black circles, methylated DNA; white circles, unmethylated DNA; green circles, repressive histone modification; stars, activating histone modifications;

Acknowledgments

Financial support: This work was funded by National Cancer Institute Awards CA133877-01 (to DM), CA085289 and CA113001(to KPN), and awards from the Phi Beta Psi Sorority (Brownsburg, IN) (to KPN).

Footnotes

Conflict of Interest Statement: D.E.M. received research support from EISAI (Dacogen) for conduct of the clinical trial discussed in this manuscript. K.P.N. has nothing to disclose.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Bukowski RM, Ozols RF, Markman M. The management of recurrent ovarian cancer. Semin Oncol. 2007;34(2 Suppl 2):S1–15. doi: 10.1053/j.seminoncol.2007.03.012. [DOI] [PubMed] [Google Scholar]
  • 2.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ. Cancer statistics, 2008. CA Cancer J Clin. 2008;58:71. doi: 10.3322/CA.2007.0010. [DOI] [PubMed] [Google Scholar]
  • 3.Yap TA, Carden CP, Kaye SB. Beyond chemotherapy: targeted therapies in ovarian cancer. Nat Rev Cancer. 2009;9(3):167–81. doi: 10.1038/nrc2583. [DOI] [PubMed] [Google Scholar]
  • 4.Sandercock J, Parmar MK, Torri V, Qian W. First-line treatment for advanced ovarian cancer: paclitaxel, platinum and the evidence. Br J Cancer. 2002;87(8):815–24. doi: 10.1038/sj.bjc.6600567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Liu CM. Cancer of the ovary. N Engl J Med. 2005;352(12):1268–9. [PubMed] [Google Scholar]
  • 6.Toyota M, Issa JP. Epigenetic changes in solid and hematopoietic tumors. Semin Oncol. 2005;32:521–30. doi: 10.1053/j.seminoncol.2005.07.003. [DOI] [PubMed] [Google Scholar]
  • 7.Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683–6. doi: 10.1016/j.cell.2007.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Balch C, Huang TH, Brown R, Nephew KP. The epigenetics of ovarian cancer drug resistance and resensitization. Am J Obstet Gynecol. 2004;191(5):1552–72. doi: 10.1016/j.ajog.2004.05.025. [DOI] [PubMed] [Google Scholar]
  • 9.Barton CA, Hacker NF, Clark SJ, O’Brien PM. DNA methylation changes in ovarian cancer: implications for early diagnosis, prognosis and treatment. Gynecol Oncol. 2008;109(1):129–39. doi: 10.1016/j.ygyno.2007.12.017. [DOI] [PubMed] [Google Scholar]
  • 10.Esteller M. Epigenetics in cancer. N Engl J Med. 2008;358:1148–1159. doi: 10.1056/NEJMra072067. [DOI] [PubMed] [Google Scholar]
  • 11.Schickel R, Boyerinas B, Park SM, Peter ME. MicroRNAs: key players in the immune system, differentiation, tumorigenesis and cell death. Oncogene. 2008;27:5959–5974. doi: 10.1038/onc.2008.274. [DOI] [PubMed] [Google Scholar]
  • 12.Iorio MV, Visone R, Di Leva G, Donati V, Petrocca F, Casalini P, Taccioli C, Volinia S, Liu CG, Alder H, Calin GA, Menard S, Croce CM. MicroRNA signatures in human ovarian cancer. Cancer Res. 2007;67:8699–8707. doi: 10.1158/0008-5472.CAN-07-1936. [DOI] [PubMed] [Google Scholar]
  • 13.Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell. 2007;128:669–68. doi: 10.1016/j.cell.2007.01.033. [DOI] [PubMed] [Google Scholar]
  • 14.Gama-Sosa MA, Slagel VA, Trewyn RW, Oxenhandler R, Kuo KC, Gehrke CW, Ehrlich M. The 5-methylcytosine content of DNA from human tumors. Nucleic Acids Res. 1983;11:6883–6894. doi: 10.1093/nar/11.19.6883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ehrlich M. DNA methylation in cancer: too much, but also too little. Oncogene. 2002;21:5400–5413. doi: 10.1038/sj.onc.1205651. [DOI] [PubMed] [Google Scholar]
  • 16.Qu G, Dubeau L, Narayan A, Yu M, Ehrlich M. Satellite DNA hypomethylation vs. overall genomic hypomethylation in ovarian epithelial tumors of different malignant potential. Mut. Res. 1999;423:91–101. doi: 10.1016/s0027-5107(98)00229-2. [DOI] [PubMed] [Google Scholar]
  • 17.Cheng P, Schmutte C, Cofer KF, Felix JC, Yu MC, Dubeau L. Alterations in DNA methylation are early, but not initial, events in ovarian tumorigenesis. Br. J. Cancer. 1997;75:396–402. doi: 10.1038/bjc.1997.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ehrlich M, Woods C, Yu M, Dubeau L, Yang F, Campan M, Weisenberger D, Long TI, Youn B, Fiala E, Laird P. Quantitative analysis of association between DNA hypermethylation, hypomethylation, and DNMT RNA levels in ovarian tumors. Oncogene. 2006;25:2636–2645. doi: 10.1038/sj.onc.1209145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Pattamadilok J, Huapai N, Rattanatanyong P, Vasurattana A, Triratanachat S, Tresukosol D, Mutirangura A. LINE-1 hypomethylation level as a potential prognostic factor for epithelial ovarian cancer. Int J Gynecol Cancer. 2008;18:711–7. doi: 10.1111/j.1525-1438.2007.01117.x. [DOI] [PubMed] [Google Scholar]
  • 20.Widschwendter M, Jiang G, Woods C, Muller HM, Fiegl H, Goebel G, Marth C, Holzner EM, Zeimet AG, Laird PW, Ehrlich M. DNA hypomethylation and ovarian cancer biology. Cancer Res. 2004;64:4472–4480. doi: 10.1158/0008-5472.CAN-04-0238. [DOI] [PubMed] [Google Scholar]
  • 21.Ehrlich M. DNA methylation and cancer-associated genetic instability. Adv Exp Med Biol. 2005;570:363–92. doi: 10.1007/1-4020-3764-3_13. [DOI] [PubMed] [Google Scholar]
  • 22.Illingworth RS, Bird AP. CpG islands-a rough guide. FEBS Lett. 2009;583:1713–20. doi: 10.1016/j.febslet.2009.04.012. [DOI] [PubMed] [Google Scholar]
  • 23.Strathdee G, Vass JK, Oien KA, Siddiqui N, Curto-Garcia J, Brown R. Demethylation of the MCJ gene in stage III/IV epithelial ovarian cancer and response to chemotherapy. Gynecol Oncol. 2005;97:898–903. doi: 10.1016/j.ygyno.2005.03.023. [DOI] [PubMed] [Google Scholar]
  • 24.Czekierdowski A, Czekierdowska S, Wielgos M, Smolen A, Kaminski P, Kotarski J. The role of CpG islands hypomethylation and abnormal expression of neuronal protein synuclein-gamma (SNCG) in ovarian cancer. Neuro Endocrinol Lett. 2006;27:381–386. [PubMed] [Google Scholar]
  • 25.Woloszynska-Read A, James SR, Link PA, Yu J, Odunsi K, Karpf AR. DNA methylation-dependent regulation of BORIS/CTCFL expression in ovarian cancer. Cancer Immun. 2007;7:21. [PMC free article] [PubMed] [Google Scholar]
  • 26.Murphy SK, Huang Z, Wen Y, Spillman MA, Whitaker RS, Simel LR, Nichols TD, Marks JR, Berchuck A. Frequent IGF2/H19 domain epigenetic alterations and elevated IGF2 expression in epithelial ovarian cancer. Mol Cancer Res. 2006;4:283–292. doi: 10.1158/1541-7786.MCR-05-0138. [DOI] [PubMed] [Google Scholar]
  • 27.Litkouhi B, Kwong J, Lo CM, Smedley JG, 3rd, McClane BA, Aponte M, Gao Z, Sarno JL, Hinners J, Welch WR, Berkowitz RS, Mok SC, Garner EI. Claudin-4 overexpression in epithelial ovarian cancer is associated with hypomethylation and is a potential target for modulation of tight junction barrier function using a C-terminal fragment of Clostridium perfringens enterotoxin. Neoplasia. 2007;9:304–314. doi: 10.1593/neo.07118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Press JZ, De Luca A, Boyd N, Young S, Troussard A, Ridge Y, Kaurah P, Kalloger SE, Blood KA, Smith M, Spellman PT, Wang Y, Miller DM, Horsman D, Faham M, Gilks CB, Gray J, Huntsman DG. Ovarian carcinomas with genetic and epigenetic BRCA1 loss have distinct molecular abnormalities. BMC Cancer. 2008;8:17. doi: 10.1186/1471-2407-8-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Strathdee G, Appleton K, Illand M, Millan DW, Sargent J, Paul J, Brown R. Primary ovarian carcinomas display multiple methylator phenotypes involving known tumor suppressor genes. Am J Pathol. 2001;58(3):1121–7. doi: 10.1016/S0002-9440(10)64059-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Milde-Langosch K, Ocon E, Becker G, Loning T. p16/MTS1 inactivation in ovarian carcinomas: high frequency of reduced protein expression associated with hypermethylation or mutation in endometrioid and mucinous tumors. Int J Cancer. 1998;79:61–65. doi: 10.1002/(sici)1097-0215(19980220)79:1<61::aid-ijc12>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  • 31.Strathdee G, MacKean MJ, Illand M, Brown R. A role for methylation of the hMLH1 promoter in loss of hMLH1 expression and drug resistance in ovarian cancer. Oncogene. 1999;18:2335–41. doi: 10.1038/sj.onc.1202540. [DOI] [PubMed] [Google Scholar]
  • 32.Ibanez de Caceres I, Battagli C, Esteller M, Herman JG, Dulaimi E, Edelson MI, Bergman C, Ehya H, Eisenberg BL, Cairns P. Tumor cell-specific BRCA1 and RASSF1A hypermethylation in serum, plasma, and peritoneal fluid from ovarian cancer patients. Cancer Res. 2004;64:6476–6481. doi: 10.1158/0008-5472.CAN-04-1529. [DOI] [PubMed] [Google Scholar]
  • 33.Sellar GC, Watt KP, Rabiasz GJ, Stronach EA, Li L, Miller EP, Massie CE, Miller J, Contreras-Moreira B, Scott D, Brown I, Williams AR, Bates PA, Smyth JF, Gabra H. OPCML at 11q25 is epigenetically inactivated and has tumor-suppressor function in epithelial ovarian cancer. Nat Genet. 2003;34:337–34. doi: 10.1038/ng1183. [DOI] [PubMed] [Google Scholar]
  • 34.Socha MJ, Said N, Dai Y, Kwong J, Ramalingam P, Trieu V, Desai N, Mok SC, Motamed K. Aberrant promoter methylation of sparc in ovarian cancer. Neoplasia. 2009;11:126–135. doi: 10.1593/neo.81146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kikuchi R, Tsuda H, Kozaki K, Kanai Y, Kasamatsu T, Sengoku K, Hirohashi S, Inazawa J, Imoto I. Frequent inactivation of a putative tumor suppressor, angiopoietin-like protein 2, in ovarian cancer. Cancer Res. 2008;68:5067–5075. doi: 10.1158/0008-5472.CAN-08-0062. [DOI] [PubMed] [Google Scholar]
  • 36.Kikuchi R, Tsuda H, Kanai Y, Kasamatsu T, Sengoku K, Hirohashi S, Inazawa J, Imoto I. Promoter hypermethylation contributes to frequent inactivation of a putative conditional tumor suppressor gene connective tissue growth factor in ovarian cancer. Cancer Res. 2007;67:7095–710. doi: 10.1158/0008-5472.CAN-06-4567. [DOI] [PubMed] [Google Scholar]
  • 37.Feng W, Marquez RT, Lu Z, Liu J, Lu KH, Issa JP, Fishman DM, Yu Y, Bast RC., Jr. Imprinted tumor suppressor genes ARHI and PEG3 are the most frequently down-regulated in human ovarian cancers by loss of heterozygosity and promoter methylation. Cancer. 2008;112:1489–1502. doi: 10.1002/cncr.23323. [DOI] [PubMed] [Google Scholar]
  • 38.Cvetkovic D, Pisarcik D, Lee C, Hamilton TC, Abdollahi A. Altered expression and loss of heterozygosity of the LOT1 gene in ovarian cancer. Gynecol Oncol. 2004;95:449–455. doi: 10.1016/j.ygyno.2004.08.051. [DOI] [PubMed] [Google Scholar]
  • 39.Pruitt K, Ulku AS, Frantz K, Rojas RJ, Muniz-Medina VM, Rangnekar VM, Der CJ, Shields JM. Ras-mediated loss of the pro-apoptotic response protein Par-4 is mediated by DNA hypermethylation through Raf-independent and Raf-dependent signaling cascades in epithelial cells. J Biol Chem. 2005;280:23363–23370. doi: 10.1074/jbc.M503083200. [DOI] [PubMed] [Google Scholar]
  • 40.Terasawa K, Sagae S, Toyota M, Tsukada K, Ogi K, Satoh A, Mita H, Imai K, Tokino T, Kudo R. Epigenetic inactivation of TMS1/ASC in ovarian cancer. Clin Cancer Res. 2004;10:2000–2006. doi: 10.1158/1078-0432.ccr-0932-03. [DOI] [PubMed] [Google Scholar]
  • 41.Arnold JM, Cummings M, Purdie D, Chenevix-Trench G. Reduced expression of intercellular adhesion molecule-1 in ovarian adenocarcinomas. Br J Cancer. 2001;85:1351–1358. doi: 10.1054/bjoc.2001.2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Yuecheng Y, Hongmei L, Xiaoyan X. Clinical evaluation of E-cadherin expression and its regulation mechanism in epithelial ovarian cancer. Clin Exp Metastasis. 2006;23:65–74. doi: 10.1007/s10585-006-9020-3. [DOI] [PubMed] [Google Scholar]
  • 43.Backen AC, Cole CL, Lau SC, Clamp AR, McVey R, Gallagher JT, Jayson GC. Heparan sulphate synthetic and editing enzymes in ovarian cancer. Br J Cancer. 2007;96:1544–1548. doi: 10.1038/sj.bjc.6603747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Staub J, Chien J, Pan Y, Qian X, Narita K, Aletti G, Scheerer M, Roberts LR, Molina J, Shridhar V. Epigenetic silencing of HSulf-1 in ovarian cancer:implications in chemoresistance. Oncogene. 2007;26:4969–4978. doi: 10.1038/sj.onc.1210300. [DOI] [PubMed] [Google Scholar]
  • 45.Potapova A, Hoffman AM, Godwin AK, Al-Saleem T, Cairns P. Promoter hypermethylation of the PALB2 susceptibility gene in inherited and sporadic breast and ovarian cancer. Cancer Res. 2008;68:998–1002. doi: 10.1158/0008-5472.CAN-07-2418. [DOI] [PubMed] [Google Scholar]
  • 46.Izutsu N, Maesawa C, Shibazaki M, Oikawa H, Shoji T, Sugiyama T, Masuda T. Epigenetic modification is involved in aberrant expression of class III beta-tubulin, TUBB3, in ovarian cancer cells. Int J Oncol. 2008;32:1227–1235. doi: 10.3892/ijo_32_6_1227. [DOI] [PubMed] [Google Scholar]
  • 47.Fiegl H, Windbichler G, Mueller-Holzner E, Goebel G, Lechner M, Jacobs IJ, Widschwendter M. HOXA11 DNA methylation--a novel prognostic biomarker in ovarian cancer. Int J Cancer. 2008;123:725–729. doi: 10.1002/ijc.23563. [DOI] [PubMed] [Google Scholar]
  • 48.Ahluwalia A, Yan P, Hurteau JA, Bigsby RM, Jung SH, Huang TH, Nephew KP. DNA methylation and ovarian cancer. I. Analysis of CpG island hypermethylation in human ovarian cancer using differential methylation hybridization. Gynecol Oncol. 2001;82:261–8. doi: 10.1006/gyno.2001.6291. [DOI] [PubMed] [Google Scholar]
  • 49.Wei SH, Chen CM, Strathdee G, Harnsomburana J, Shyu CR, Rahmatpanah F, Shi H, Ng SW, Yan PS, Nephew KP, Brown R, Huang TH. Methylation microarray analysis of late-stage ovarian carcinomas distinguishes progression-free survival in patients and identifies candidate epigenetic markers. Clin Cancer Res. 2002;8:2246–225. [PubMed] [Google Scholar]
  • 50.Barton CA, Hacker NF, Clark SJ, O’Brien PM. DNA methylation changes in ovarian cancer: implications for early diagnosis, prognosis and treatment. Gynecol Oncol. 2008;109:129–139. doi: 10.1016/j.ygyno.2007.12.017. [DOI] [PubMed] [Google Scholar]
  • 51.Wei SH, Balch C, Paik HH, Kim YS, Baldwin RL, Liyanarachchi S, Li L, Wang Z, Wan JC, Davuluri RV, Karlan BY, Gifford G, Brown R, Kim S, Huang TH, Nephew KP. Prognostic DNA methylation biomarkers in ovarian cancer. Clin Cancer Res. 2006;12:2788–2794. doi: 10.1158/1078-0432.CCR-05-1551. [DOI] [PubMed] [Google Scholar]
  • 52.Watts GS, Futscher BW, Holtan N, Degeest K, Domann FE, Rose SL. DNA methylation changes in ovarian cancer are cumulative with disease progression and identify tumor stage. BMC Med Genomics. 2008;1:47. doi: 10.1186/1755-8794-1-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Li M, Balch C, Montgomery JS, Jeong M, Chung JH, Yan P, Huang TH, Kim S, Nephew KP. Integrated analysis of DNA methylation and gene expression reveals specific signaling pathways associated with platinum resistance in ovarian cancer. BMC Med Genomics. 2009 doi: 10.1186/1755-8794-2-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Balch C, Montgomery JS, Paik HI, Kim S, Kim S, Huang TH, Nephew KP. New anti-cancer strategies: epigenetic therapies and biomarkers. Front Biosci. 2005 May 1;10:1897–931. doi: 10.2741/1668. [DOI] [PubMed] [Google Scholar]
  • 55.Ahluwalia A, Hurteau JA, Bigsby RM, Nephew KP. DNA methylation in ovarian cancer. II. Expression of DNA methyltransferases in ovarian cancer cell lines and normal ovarian epithelial cells. Gynecol Oncol. 2001;82:299–304. doi: 10.1006/gyno.2001.6284. [DOI] [PubMed] [Google Scholar]
  • 56.Leu YW, Rahmatpanah F, Shi H, Wei SH, Liu JC, Yan PS, Huang TH. Double RNA interference of DNMT3b and DNMT1 enhances DNA demethylation and gene reactivation. Cancer Res. 2003;63:6110–6115. [PubMed] [Google Scholar]
  • 57.Baldwin RL, Nemeth E, Tran H, Shvartsman H, Cass I, Narod S, Karlan BY. BRCA1 promoter region hypermethylation in ovarian carcinoma: a population-based study. Cancer Res. 2000;60:5329–5333. [PubMed] [Google Scholar]
  • 58.Hilton JL, Geisler JP, Rathe JA, Hattermann-Zogg MA, DeYoung B, Buller RE. Inactivation of BRCA1 and BRCA2 in ovarian cancer. J Natl Cancer Inst. 2002;94:1396–1406. doi: 10.1093/jnci/94.18.1396. [DOI] [PubMed] [Google Scholar]
  • 59.Dworkin AM, Spearman AD, Tseng SY, Sweet K, Toland AE. Methylation not a frequent “second hit” in tumors with germline BRCA mutations. Fam Cancer. 2009 doi: 10.1007/s10689-009-9240-1. [DOI] [PubMed] [Google Scholar]
  • 60.Gifford G, Paul J, Vasey PA, Kaye SB, Brown R. The acquisition of hMLH1 methylation in plasma DNA after chemotherapy predicts poor survival for ovarian cancer patients. Clin Cancer Res. 2004;10:4420–6. doi: 10.1158/1078-0432.CCR-03-0732. [DOI] [PubMed] [Google Scholar]
  • 61.Su HY, Lai HC, Lin YW, Chou YC, Liu CY, Yu MH. An epigenetic marker panel for screening and prognostic prediction of ovarian cancer. Int J Cancer. 2009;124:387–93. doi: 10.1002/ijc.23957. [DOI] [PubMed] [Google Scholar]
  • 62.Graham JS, Kaye SB, Brown R. The promises and pitfalls of epigenetic therapies in solid tumours. Eur J Cancer. 2009;45:1129–36. doi: 10.1016/j.ejca.2009.01.003. [DOI] [PubMed] [Google Scholar]
  • 63.Yap TA, Carden CP, Kaye SB. Beyond chemotherapy: targeted therapies in ovarian cancer. Nat Rev Cancer. 2009;9(3):167–81. doi: 10.1038/nrc2583. [DOI] [PubMed] [Google Scholar]
  • 64.Bast RC, Jr, Hennessy B, Mills GB. The biology of ovarian cancer: new opportunities for translation. Nat Rev Cancer. 2009;9:415–28. doi: 10.1038/nrc2644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.LaVoie HA. Epigenetic control of ovarian function: the emerging role of histone modifications. Mol Cell Endocrinol. 2005;243:12–18. doi: 10.1016/j.mce.2005.09.005. [DOI] [PubMed] [Google Scholar]
  • 66.Ozdag H, Teschendorff AE, Ahmed AA, Hyland SJ, Blenkiron C, Bobrow L, Veerakumarasivam A, Burtt G, Subkhankulova T, Arends MJ, Collins VP, Bowtell D, Kouzarides T, Brenton JD, Caldas C. Differential expression of selected histone modifier genes in human solid cancers. BMC genomics. 2006;7:90. doi: 10.1186/1471-2164-7-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Caslini C, Capo-chichi CD, Roland IH, Nicolas E, Yeung AT, Xu XX. Histone modifications silence the GATA transcription factor genes in ovarian cancer. Oncogene. 2006;25:5446–5461. doi: 10.1038/sj.onc.1209533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Valls E, Sanchez-Molina S, Martinez-Balbas MA. Role of histone modifications in marking and activating genes through mitosis. J Biol Chem. 2005;280:42592–42600. doi: 10.1074/jbc.M507407200. [DOI] [PubMed] [Google Scholar]
  • 69.Richon VM, Sandhoff TW, Rifkind RA, Marks PA. Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:10014–10019. doi: 10.1073/pnas.180316197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chan MW, Huang YW, Hartman-Frey C, Kuo CT, Deatherage D, Qin H, Cheng AS, Yan PS, Davuluri RV, Huang TH, Nephew KP, Lin HJ. Aberrant transforming growth factor beta1 signaling and SMAD4 nuclear translocation confer epigenetic repression of ADAM19 in ovarian cancer. Neoplasia. 2008;10:908–919. doi: 10.1593/neo.08540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Abbosh PH, Montgomery JS, Starkey JA, Novotny M, Zuhowski EG, Egorin MJ, Moseman AP, Golas A, Brannon KM, Balch C, Huang TH, Nephew KP. Dominant-negative histone H3 lysine 27 mutant derepresses silenced tumor suppressor genes and reverses the drug-resistant phenotype in cancer cells. Cancer Res. 2006;66:5582–5591. doi: 10.1158/0008-5472.CAN-05-3575. [DOI] [PubMed] [Google Scholar]
  • 72.Wei Y, Xia W, Zhang Z, Liu J, Wang H, Adsay NV, Albarracin C, Yu D, Abbruzzese JL, Mills GB, Bast RC, Jr., Hortobagyi GN, Hung MC. Loss of trimethylation at lysine 27 of histone H3 is a predictor of poor outcome in breast, ovarian, and pancreatic cancers. Mol Carcinog Mol Carcinog. 2008;47:701–6. doi: 10.1002/mc.20413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lyko F, Brown R. DNA methyltransferase inhibitors and the development of epigenetic cancer therapies. J Natl Cancer Inst. 2005;97:1498–1506. doi: 10.1093/jnci/dji311. [DOI] [PubMed] [Google Scholar]
  • 74.Plumb JA, Strathdee G, Sludden J, Kaye SB, Brown R. Reversal of drug resistance in human tumor xenografts by 2′-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter. Cancer Res. 2000;60(21):6039–44. [PubMed] [Google Scholar]
  • 75.Li Y, Hu W, Shen DY, Kavanagh JJ, Fu S. Azacitidine enhances sensitivity of platinum-resistant ovarian cancer cells to carboplatin through induction of apoptosis. Am J Obstet Gynecol. 2009;200(2):177 e1–9. doi: 10.1016/j.ajog.2008.08.030. [DOI] [PubMed] [Google Scholar]
  • 76.Gomyo Y, Sasaki J, Branch C, Roth JA, Mukhopadhyay T. 5-aza-2′-deoxycytidine upregulates caspase-9 expression cooperating with p53-induced apoptosis in human lung cancer cells. Oncogene. 2004;23(40):6779–87. doi: 10.1038/sj.onc.1207381. [DOI] [PubMed] [Google Scholar]
  • 77.Frost P, Abbruzzese JL, Hunt B, Lee D, Ellis M. Synergistic cytotoxicity using 2′-deoxy-5-azacytidine and cisplatin or 4-hydroperoxycyclophosphamide with human tumor cells. Cancer Res. 1990;50(15):4572–7. [PubMed] [Google Scholar]
  • 78.Balch C, Yan P, Craft T, Young S, Skalnik DG, Huang TH, et al. Antimitogenic and chemosensitizing effects of the methylation inhibitor zebularine in ovarian cancer. Mol Cancer Ther. 2005;4(10):1505–14. doi: 10.1158/1535-7163.MCT-05-0216. [DOI] [PubMed] [Google Scholar]
  • 79.Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer. 2006;6:38–51. doi: 10.1038/nrc1779. [DOI] [PubMed] [Google Scholar]
  • 80.Qian X, LaRochelle WJ, Ara G, Wu F, Petersen KD, Thougaard A, Sehested M, Lichenstein HS, Jeffers M. Activity of PXD101, a histone deacetylase inhibitor, in preclinical ovarian cancer studies. Mol Cancer Ther. 2006;5:2086–2095. doi: 10.1158/1535-7163.MCT-06-0111. [DOI] [PubMed] [Google Scholar]
  • 81.Yang Y-T, Balch C, Kulp S, Mand MR, Nephew KP, Chen C-S. A rationally designed histone deacetylase inhibitor with distinct antitumor activity against ovarian cancer. Neoplasia. 2009;11:552–63. doi: 10.1593/neo.09204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Karpf AR, Jones DA. Reactivating the expression of methylation silenced genes in human cancer. Oncogene. 2002;21:5496–5503. doi: 10.1038/sj.onc.1205602. [DOI] [PubMed] [Google Scholar]
  • 83.Steele N, Finn P, Brown R, Plumb JA. Combined inhibition of DNA methylation and histone acetylation enhances gene re-expression and drug sensitivity in vivo. Br J Cancer. 2009;100:758–7631. doi: 10.1038/sj.bjc.6604932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kim IK, Li CC, Young HA, Lee JH, Kim HS, Pardhasaradhi K, Garcia GE, Chiang PK. Apoptosis of L1210 Leukemia Cells Induced by 3-Deazaadenosine Analogs: Differential Expression of c-myc, NF-Kappa B and Molecular Events. J Biomed Sci. 1997;4:83–90. doi: 10.1007/BF02255598. [DOI] [PubMed] [Google Scholar]
  • 85.Issa JP, Gharibyan V, Cortes J, Jelinek J, Morris G, Verstovsek S, Talpaz M, Garcia-Manero G, Kantarjian HM. Phase II study of low-dose decitabine in patients with chronic myelogenous leukemia resistant to imatinib mesylate. J Clin Oncol. 2005;23:3948–3956. doi: 10.1200/JCO.2005.11.981. [DOI] [PubMed] [Google Scholar]
  • 86.Kantarjian HM. Treatment of myelodysplastic syndrome: questions raised by the azacitidine experience. J Clin Oncol. 2002;20:2415–2416. doi: 10.1200/JCO.2002.20.10.2415. [DOI] [PubMed] [Google Scholar]
  • 87.Kornblith AB, Herndon JE, 2nd, Silverman LR, Demakos EP, Odchimar-Reissig R, Holland JF, Powell BL, DeCastro C, Ellerton J, Larson RA, Schiffer CA, Holland JC. Impact of azacytidine on the quality of life of patients with myelodysplastic syndrome treated in a randomized phase III trial: a Cancer and Leukemia Group B study. J Clin Oncol. 2002;20:2441–2452. doi: 10.1200/JCO.2002.04.044. [DOI] [PubMed] [Google Scholar]
  • 88.Silverman LR, Demakos EP, Peterson BL, Kornblith AB, Holland JC, Odchimar-Reissig R, Stone RM, Nelson D, Powell BL, DeCastro CM, Ellerton J, Larson RA, Schiffer CA, Holland JF. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol. 2002;20:2429–2440. doi: 10.1200/JCO.2002.04.117. [DOI] [PubMed] [Google Scholar]
  • 89.Silverman LR, McKenzie DR, Peterson BL, Holland JF, Backstrom JT, Beach CL, Larson RA. Further analysis of trials with azacitidine in patients with myelodysplastic syndrome: studies 8421, 8921, and 9221 by the Cancer and Leukemia Group B. J Clin Oncol. 2006;24:3895–3903. doi: 10.1200/JCO.2005.05.4346. [DOI] [PubMed] [Google Scholar]
  • 90.Kuykendall JR. 5-azacytidine and decitabine monotherapies of myelodysplastic disorders. Ann Pharmacother. 2005;39:1700–1709. doi: 10.1345/aph.1E612. [DOI] [PubMed] [Google Scholar]
  • 91.de Vos D, van Overveld W. Decitabine: a historical review of the development of an epigenetic drug. Ann Hematol. 2005;84(Suppl 13):3–8. doi: 10.1007/s00277-005-0008-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kantarjian H, Issa JP, Rosenfeld CS, Bennett JM, Albitar M, DiPersio J, Klimek V, Slack J, de Castro C, Ravandi F, Helmer R, 3rd, Shen L, Nimer SD, Leavitt R, Raza A, Saba H. Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study. Cancer. 2006;106:1794–1803. doi: 10.1002/cncr.21792. [DOI] [PubMed] [Google Scholar]
  • 93.Attadia V. Effects of 5-aza-2′-deoxycytidine on differentiation and oncogene expression in the human monoblastic leukemia cell line U-937. Leukemia. 1993;7(Suppl 1):9–16. [PubMed] [Google Scholar]
  • 94.Pinto A, Attadia V, Fusco A, Ferrara F, Spada OA, Di Fiore PP. 5-Aza-2′-deoxycytidine induces terminal differentiation of leukemic blasts from patients with acute myeloid leukemias. Blood. 1984;64:922–929. [PubMed] [Google Scholar]
  • 95.Jones PA, Taylor SM. Cellular differentiation, cytidine analogs and DNA methylation. Cell. 1980;20:85–93. doi: 10.1016/0092-8674(80)90237-8. [DOI] [PubMed] [Google Scholar]
  • 96.Willemze R, Archimbaud E, Muus P, The EORTC Leukemia Cooperative Group Preliminary results with 5-aza-2′-deoxycytidine (DAC)-containing chemotherapy in patients with relapsed or refractory acute leukemia. Leukemia. 1993;7(Suppl 1):49–50. [PubMed] [Google Scholar]
  • 97.Kantarjian HM, Issa JP. Decitabine dosing schedules. Semin Hematol. 2005;42:S17–22. doi: 10.1053/j.seminhematol.2005.05.006. [DOI] [PubMed] [Google Scholar]
  • 98.O’Brien SM, R.-K F, Giles S, et al. Decitabine low dose schedule in myelodysplastic syndrome, comparison of three different dose schedules. J Clinical Oncology. 2005;(suppl 16) [Google Scholar]
  • 99.Wijermans P, Lubbert M, Verhoef G, Bosly A, Ravoet C, Andre M, Ferrant A. Low-dose 5-aza-2′-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase II study in elderly patients. J Clin Oncol. 2000;18:956–962. doi: 10.1200/JCO.2000.18.5.956. [DOI] [PubMed] [Google Scholar]
  • 100.Samlowski WE, Leachman SA, Wade M, Cassidy P, Porter-Gill P, Busby L, Wheeler R, Boucher K, Fitzpatrick F, Jones DA, Karpf AR. Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. J Clin Oncol. 2005;23:3897–3905. doi: 10.1200/JCO.2005.06.118. [DOI] [PubMed] [Google Scholar]
  • 101.Aparicio A, Eads CA, Leong LA, Laird PW, Newman EM, Synold TW, Baker SD, Zhao M, Weber JS. Phase I trial of continuous infusion 5-aza-2′-deoxycytidine. Cancer Chemother Pharmacol. 2003;51:231–239. doi: 10.1007/s00280-002-0563-y. [DOI] [PubMed] [Google Scholar]
  • 102.Aparicio A, Weber JS. Review of the clinical experience with 5-azacytidine and 5-aza-2′-deoxycytidine in solid tumors. Curr Opin Investig Drugs. 2002;3:627–633. [PubMed] [Google Scholar]
  • 103.Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, Hancox A, Hong JA, Chen GA, Pishchik V, Figg WD, Murgo AJ, Steinberg SM. Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura. Clin Cancer Res. 2006;12:5777–5785. doi: 10.1158/1078-0432.CCR-06-0669. [DOI] [PubMed] [Google Scholar]
  • 104.Manetta A, Blessing JA, Look KY. A phase II study of fazarabine in patients with advanced ovarian cancer. A Gynecologic Oncology Group study. Am J Clin Oncol. 1995;18:156–157. doi: 10.1097/00000421-199504000-00012. [DOI] [PubMed] [Google Scholar]
  • 105.Appleton K, Mackay HJ, Judson I, Plumb JA, McCormick C, Strathdee G, Lee C, Barrett S, Reade S, Jadayel D, Tang A, Bellenger K, Mackay L, Setanoians A, Schatzlein A, Twelves C, Kaye SB, Brown R. Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. J Clin Oncol. 2007;25:4603–4609. doi: 10.1200/JCO.2007.10.8688. [DOI] [PubMed] [Google Scholar]
  • 106.Brown R, Plumb JA, McCormick C, Lee C, Barrett S, Tang A, Schaetzlein A, Kaye SB, Judson I, McKay HJ. Pharmacodynamic Effects in Patients Treated with 6 Hour Infusion of the Demethylating Agent 5-aza-2′deoxycytidine (decitabine) EORTC, Supplement to Clinical Cancer Research. 2005 December 1;11(23) 2005. [Google Scholar]
  • 107.Fang F, Balch C, Schilder S, Breen T, Zhang S, Shen C, Li L, Kulesavage C, Snyder AJ, Nephew KP, Matei DE. 2009 American Assoc Cancer Res. A phase I and pharmacodynamic study of decitabine in combination with carboplatin in patients with recurrent, platinum-resistant, epithelial ovarian cancer. Abstract #3593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Weisenberger DJ, Campan M, Long TI, Kim M, Woods C, Fiala E, Ehrlich M, Laird PW. Analysis of repetitive element DNA methylation by MethyLight. Nucleic Acids Res. 2005;33:6823–6836. doi: 10.1093/nar/gki987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bast RC, I RB, Hu W, Kavanagh J, Coleman RL, Levenback C, Sood AK, Wolf J, Gershenson DM, Markman M, Fu S. A phase IIa study of a sequential regimen using azacitidine to reverse platinum resistance to carboplatin in patients with platinum resistant or refractory epithelial ovarian cancer. J Clin Oncol. 2008 May 20;26(suppl) 2008. abstr 3500. [Google Scholar]
  • 110.Camphausen K, Tofilon PJ. Inhibition of histone deacetylation: a strategy for tumor radiosensitization. J Clin Oncol. 2007;25:4051–4056. doi: 10.1200/JCO.2007.11.6202. [DOI] [PubMed] [Google Scholar]
  • 111.Lin RJ, Nagy L, Inoue S, Shao W, Miller WH, Jr., Evans RM. Role of the histone deacetylase complex in acute promyelocytic leukaemia. Nature. 1998;391:811–814. doi: 10.1038/35895. [DOI] [PubMed] [Google Scholar]
  • 112.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] [Google Scholar]
  • 113.Carducci MA, Gilbert J, Bowling MK, Noe D, Eisenberger MA, Sinibaldi V, Zabelina Y, Chen TL, Grochow LB, Donehower RC. A Phase I clinical and pharmacological evaluation of sodium phenylbutyrate on an 120-h infusion schedule. Clin Cancer Res. 2001;7:3047–3055. [PubMed] [Google Scholar]
  • 114.Fouladi M, Furman WL, Chin T, Freeman BB, 3rd, Dudkin L, Stewart CF, Krailo MD, Speights R, Ingle AM, Houghton PJ, Wright J, Adamson PC, Blaney SM. Phase I study of depsipeptide in pediatric patients with refractory solid tumors: a Children’s Oncology Group report. J Clin Oncol. 2006;24:3678–3685. doi: 10.1200/JCO.2006.06.4964. [DOI] [PubMed] [Google Scholar]
  • 115.Klimek VM, Fircanis S, Maslak P, Guernah I, Baum M, Wu N, Panageas K, Wright JJ, Pandolfi PP, Nimer SD. Tolerability, pharmacodynamics, and pharmacokinetics studies of depsipeptide (romidepsin) in patients with acute myelogenous leukemia or advanced myelodysplastic syndromes. Clin Cancer Res. 2008;14:826–832. doi: 10.1158/1078-0432.CCR-07-0318. [DOI] [PubMed] [Google Scholar]
  • 116.Kelly WK, O’Connor OA, Krug LM, Chiao JH, Heaney M, Curley T, MacGregore-Cortelli B, Tong W, Secrist JP, Schwartz L, Richardson S, Chu E, Olgac S, Marks PA, Scher H, Richon VM. Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol. 2005;23:3923–3931. doi: 10.1200/JCO.2005.14.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.O’Connor OA, Heaney ML, Schwartz L, Richardson S, Willim R, MacGregor-Cortelli B, Curly T, Moskowitz C, Portlock C, Horwitz S, Zelenetz AD, Frankel S, Richon V, Marks P, Kelly WK. Clinical experience with intravenous and oral formulations of the novel histone deacetylase inhibitor suberoylanilide hydroxamic acid in patients with advanced hematologic malignancies. J Clin Oncol. 2006;24:166–173. doi: 10.1200/JCO.2005.01.9679. [DOI] [PubMed] [Google Scholar]
  • 118.Modesitt SC, Sill M, Hoffman JS, Bender DP. 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;109:182–186. doi: 10.1016/j.ygyno.2008.01.009. [DOI] [PubMed] [Google Scholar]
  • 119.Plumb JA, Finn PW, Williams RJ, Bandara MJ, Romero MR, Watkins CJ, La Thangue NB, Brown R. Pharmacodynamic response and inhibition of growth of human tumor xenografts by the novel histone deacetylase inhibitor PXD101. Mol Cancer Ther. 2003;2:721–728. [PubMed] [Google Scholar]
  • 120.Steele NL, Plumb JA, Vidal L, Tjornelund J, Knoblauch P, Rasmussen A, Ooi CE, Buhl-Jensen P, Brown R, Evans TR, DeBono JS. A phase 1 pharmacokinetic and pharmacodynamic study of the histone deacetylase inhibitor belinostat in patients with advanced solid tumors. Clin Cancer Res. 2008;14:804–810. doi: 10.1158/1078-0432.CCR-07-1786. [DOI] [PubMed] [Google Scholar]
  • 121.Venturelli S, Armeanu S, Pathil A, Hsieh CJ, Weiss TS, Vonthein R, Wehrmann M, Gregor M, Lauer UM, Bitzer M. Epigenetic combination therapy as a tumor-selective treatment approach for hepatocellular carcinoma. Cancer. 2007;109:2132–2141. doi: 10.1002/cncr.22652. [DOI] [PubMed] [Google Scholar]
  • 122.Dalgard CL, Van Quill KR, O’Brien JM. Evaluation of the in vitro and in vivo antitumor activity of histone deacetylase inhibitors for the therapy of retinoblastoma. Clin Cancer Res. 2008;14:3113–3123. doi: 10.1158/1078-0432.CCR-07-4836. [DOI] [PubMed] [Google Scholar]
  • 123.Fiskus W, Wang Y, Joshi R, Rao R, Yang Y, Chen J, Kolhe R, Balusu R, Eaton K, Lee P, Ustun C, Jillella A, Buser CA, Peiper S, Bhalla K. Cotreatment with vorinostat enhances activity of MK-0457 (VX-680) against acute and chronic myelogenous leukemia cells. Clin Cancer Res. 2008;14:6106–6115. doi: 10.1158/1078-0432.CCR-08-0721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Dasmahapatra G, Yerram N, Dai Y, Dent P, Grant S. Synergistic interactions between vorinostat and sorafenib in chronic myelogenous leukemia cells involve Mcl-1 and p21CIP1 down-regulation. Clin Cancer Res. 2007;13:4280–4290. doi: 10.1158/1078-0432.CCR-07-0835. [DOI] [PubMed] [Google Scholar]

RESOURCES