Open access

Epigenetic Changes in Melanoma and the Development of Epigenetic Therapy for Melanoma

Written By

Duc P. Do and Syed A.A. Rizvi

Submitted: 01 November 2010 Published: 12 September 2011

DOI: 10.5772/19088

From the Edited Volume

Research on Melanoma - A Glimpse into Current Directions and Future Trends

Edited by Mandi Murph

Chapter metrics overview

1,859 Chapter Downloads

View Full Metrics

1. Introduction

Melanoma is responsible for 80% of all skin cancer deaths (Miller & Mihm, 2006), and it is the most common cause of cancer deaths between the age of 20 and 35 years old (Houghton & Polsky, 2002). Melanoma is a highly heterogeneous cancer that is caused by the accumulation of genetic and epigenetic defects allowing the cell to escape normal cellular controls. Genetic changes are caused by irreversible alterations in the DNA sequence, including chromosomal amplification or deletions and gene mutations that culminate in aberrant cellular functions, such as activation of oncogenes and inactivation of tumor suppressors. However, recent progress in cancer research has shown that epigenetic events may play a major role in establishing the correct program of gene expression. Epigenetics is defined as heritable changes in gene expression that are not due to any changes in the DNA sequence. Currently, four epigenetic drugs, vorinostat (Marks & Breslow, 2007), romidepsin (Campas-Moya, 2009), azacitidine (Mani & Herceg, 2010; Wijermans et al., 2005), and decitabine (Mani & Herceg, 2010), have been approved for the treatment of hematologic malignancies in the United States by the Food and Drug Administration (U.S. FDA).

In a human cell, there are approximately two meters of diploid DNA that are packaged inside the nucleus with a volume of about 1000 μm3 (Kamakaka & Biggins, 2005). This packaging of DNA is facilitated by histones. Histones are a group of highly conserved, basic (positively-charged) proteins that are rich in arginine and lysine residues. This DNA-protein complex is called the chromatin. In chromatin, proteins account for more than half of the weight, from which, histone proteins being the most abundant. There are five distinct families of histones, each with numerous variants or individual genes. DNA is packaged into nucleomes comprising a histone octamer of two copies of each core histones (H2A, H2B, H3, and H4) (Luger et al., 1997). The core histones interact in pairs. Two H3:H4 dimers interact together forming a tetramer, and two H2A:H2B dimers associate with the H3:H4 tetramer to form a nucleosome. About 146 bp of DNA is wrapped around a histone octamer. One molecule of histone H1 associates at the position where the DNA enters and exits the nucleosome core, thus sealing the two turns of DNA (Luger et al., 1997).

These core histones contain a conserved C-terminal histone fold domain and unique N-terminal tails. The histone N-terminal tails protrude from the nucleosome core and provide sites for posttranslational modifications, including acetylation, methylation, phosphorylation, and ubiquitination (Jenuwein & Allis, 2001). These distinct patterns of posttranslational modifications make up the histone code that is read by multiprotein chromatin remodelling complexes to determine the transcriptional status of the target gene (Strahl & Allis, 2000).

Advertisement

2. Histone acetylation and histone deacetylases

Epigenetic phenomena can be viewed as changes in the packaging and modifications of the DNA. In the case of DNA, it is modified only by methylation. Changes in the packaging of DNA include both histone modifications and chromatin remodeling. Histones can be modified by methylation, acetylation, phosphorylation, biotinylation, ubiquitination, sumoylation, and ADP-ribosylation. Lysine residues in the histone tails can be acetylated or methylated. Arginine residues can be methylated (Howell et al., 2009).

Among all of the posttranslational modifications on histone tails, histone acetylation is among the most extensively studied. In normal cells, acetylation and deacetylation exist in equilibrium. Acetylation is a reaction that is catalyzed by histone acetyltransferases (HATs), and the deacetylation reaction is catalyzed by histone deacetylases (HDACs). These two families of enzymes regulate the delicate balance needed for maintaining the states of chromatin and chromatin dynamics (Figure 1).

Acetylation is a reversible reaction occurring on lysine residues within the N-terminal tails of core histones H3 and H4. For histone acetylation, one of the hydrogens in the free amino group of internal lysine is substituted with an acetyl (CH3CO) group. The addition of an acetyl group removes the positive charge from the NH3 + group on lysine, thus neutralizing the basic charge of the histone tails. This modification is suggested to reduce the affinity between histones and DNA, which, in turn, correlates with active gene expression. Acetylated histone is usually associated with transcriptionally active chromatin (Hebbes et al., 1992; Kouzarides, 2007; Turner, 1993). In addition, it is involved in many processes, such as replication, nucleosome assembly, higher-order chromatin packing and interactions of nonhistone proteins (Grant & Berger, 1999). Lysine at amino acid positions 9, 14, 18, and 23 for histone H3 and at amino acid positions 5, 8, 12, 16 for histone H4 are frequent targets for acetylation. These histone modifications facilitate access and binding of transcription factors.

Histone deacetylation is associated with an inactive (closed) state of chromatin and transcriptional repression (Kouzarides, 2007; Strahl & Allis, 2000). Deacetylation is catalyzed by histone deacetylases (HDAC). HDACs catalyze the removal of acetyl groups from lysine residues. HDACs and HATs are either part of a multiprotein transcriptional complex or interact with DNA binding proteins (Haberland et al., 2009; Jenuwein & Allis, 2001). Deregulation in the activity of HDACs and HATs may lead to alterations in gene expression and has been linked to diseases, particularly cancers. Fraga et al (2005) found that the loss of acetylation of histone H4 at K16 and K20 is a common hallmark of human cancer. Recently, Kondo et al. (2008) found, 5% of the genes are silenced by trimethylation of H3K27 independent of DNA methylation.

Figure 1.

The states of chromatin and regulation of gene expression. HAT: Histone acetyltransferase; HDAC: Histone deacetylase.

To date, 18 HDACs have been identified in humans. They are divided into four classes based on their homology to yeast HDACs (Table 1). Class I enzymes, which included HDACs 1, 2, 3, and 8, are related to the yeast RPD3 (de Ruijter et al., 2003; Paris et al., 2008). Class I HDACs 1, 2, and 3 are ubiquitously expressed and are almost exclusively found in the nuclei of cells in various cell lines and tissues (de Ruijter et al., 2003; Paris et al., 2008). Unlike HDACs 1-3, HDAC 8 is found only in cells with smooth muscle/myoepithelial differentiation. HDAC8 expression was found in smooth muscle cells where its expression is suggested to play a role in regulating the dynamics of smooth muscle cytoskeleton (Waltregny et al., 2004). These class I HDACs are involved in the regulation of proliferation, apoptosis, cardiac morphogenesis, and interferon (INF) expression through regulating gene expressions (Bernstein et al., 2000; Foglietti et al., 2006; Zupkovitz et al., 2006). Class II proteins, which included HDACs 4, 5, 6, 7, 9, and 10, share domains with the yeast HDAC-1 (de Ruijter et al., 2003; Paris et al., 2008). Class II HDACs can shuttle between the nucleus and the cytoplasm (Paris et al., 2008). Class II HDAC 6 is not seen in lymphocytes, stromal cells, and vascular endothelial cells (Yoshida et al., 2004; Zhang et al., 2004). It is localized mainly in the cytoplasm. This HDAC6 enzyme is also found on the perinuclear and leading-edge subcellular regions of cells. It is a microtubule-associated deacetylase (Hubbert et al., 2002). HDAC 7 inhibits the expression of Nur77, which is involved in the regulation of apoptosis and negative selection during developing thymocytes (Dequiedt et al., 2005). Unlike class I HDACs, class II HDACs are found only in some tissues. The recently described class IV, comprised solely of HDAC 11 enzyme, shares features of classes I and II HDACs, such as the dependence on zinc for their enzymatic activity. Classes I, II and IV are zinc dependent proteases (de Ruijter et al., 2003; Gao et al., 2002; Glozak & Seto, 2007). Class III HDACs (sirtuins) have been identified based on sequence homology with the yeast transcription repressor Sir2. To date, seven different sirtuins have been identified, and all of the enzymes of class III require NAD+ for their activity. This class of enzymes is localized in the nucleus (de Ruijter et al., 2003; Glozak & Seto, 2007). HDACs can deacetylase non-histone proteins, such as tumor suppressors (e.g., p53), and signaling molecules (e.g., STAT1 and STAT3) (Minucci & Pelicci, 2006).

HDAC Example of Biological Functions Tissue Distribution Localization Reference
Class I HDAC1 essential in cell survival and proliferation ubiquitous nucleus (Bernstein et al., 2000; Paris et al., 2008; Sun & Hampsey, 1999)
HDAC2 (Foglietti et al., 2006; Paris et al., 2008)
HDAC3 (Lagger et al., 2002; Paris et al., 2008; Zupkovitz et al., 2006)
HDAC8 (Montgomery et al., 2007; Paris et al., 2008)
Class IIa HDAC4 mediator of neuronal death heart; brain; skeletal muscle nucleus/ cytoplasm (Paris et al., 2008; Waltregny et al., 2004; Waltregny et al., 2005)
HDAC5 cardiac development heart; brain; skeletal muscle (Bolger & Yao, 2005; Paris et al., 2008)
HDAC7 regulation of apoptosis in developing thymocytes heart; skeletal muscle; pancreas; spleen (Paris et al., 2008; Vega et al., 2004)
HDAC9 cardiac development brain; skeletal muscle (Bolger & Yao, 2005; Paris et al., 2008)
Class IIb HDAC6 regulation of tubulin and Hsp90 acetylation heart; liver; kidney; pancreas mainly cytoplasm (Chang et al., 2004; Dequiedt et al., 2003; Paris et al., 2008; Zhang et al., 2002)
HDAC10 regulation of thioredoxin-interacting protein expression spleen; liver; kidney (Lee et al., 2010)
Class V HDAC11 regulation of immune function heart; brain; skeletal muscle; kidney nucleus/ cytoplasm (Villagra et al., 2009)

Table 1.

Histone deacetylases.

2.1. Clinical applications of histone modifications

Given the association between HDAC enzymes and cancers, there is growing interest in using HDAC inhibitors (HDACI) as antitumor agents. Inhibition of HDAC activity should lead to chromatin decondensation and an increase in gene transcription (Figure 1) (Karagiannis & El-Osta, 2006). HDACIs have been shown to have pleiotropic effects, including cell cycle arrest, growth inhibition and chromatin decondensation. They interfere directly with the mitotic spindle checkpoint, differentiation, and apoptosis in cancer cell types (Choi et al., 2007; Marchion & Munster, 2007; Stearns et al., 2007; Xu et al., 2005). Imre et al. (2006) showed that HDACIs reduce the responsiveness of tumor cells to the tumor necrosis factor-α (TNF-α) mediated activation of the nuclear factor-kappa B (NF-kappa B). All HDACIs upregulate p21, an important mediator of growth arrest (Richon et al., 2000). Studies in clinical trials have attempted to use HDACIs in combination therapy with some successes (Johnstone, 2002). This combined strategy has shown promise in some malignancies (Bishton et al., 2007).

To date, more than 18 HDACIs have been tested in clinical trials for cancer therapy (Carew et al., 2008; Paris et al., 2008). In the United States, two histone deacetylase inhibitors, namely vorinostat (Zolinza) and romidepsin (Istodax), have been approved for the treatment of cutaneous T-cell lymphoma. HDACIs are usually classified into various groups based on their structures, including hydroxamic acids, cyclic peptides, short chain fatty acids, and benzamides. Hydroxamic acid derived compounds (trichostatin A, oxamflatin) have been used in clinical trials for treating both hematologic malignancies and solid tumors. These compounds contain an acid moiety that can fit into the catalytic site and bind to the zinc atom, thus inhibiting the HDAC enzyme (Marchion & Munster, 2007; Marks et al., 2000). For cyclic peptide group (depsipeptide, trapoxin), HDACIs are effective in nanomolar range. On the other hand, short chain fatty acid compounds (butyrate, trybutyrin) require relatively high concentrations for their action. A member of this group, valproic acid has been used in antiepileptic treatment. The use of valproic acid as an anti-epileptic underlines the wide functional distribution of HDACs, contributing to problems targeting the cancer treatments using histone deacetylase inhibitors. The benzamide group molecules (MS-275, CI-994) exert their action at micromolar concentrations. Since the enzymatic pocket is highly conserved in nature, most HDACIs do not selectively inhibit individual HDAC enzymes. Rather, HDACIs inhibit several HDAC enzymes simultaneously. They target mainly classes I and II HDACs (Marks & Xu, 2009; Paris et al., 2008). Table 2 shows the histone deacetylase inhibitors.

Histone deacetylase inhibitors have been investigated in clinical trials for melanoma (Table 3). A multicenter, phase II clinical trial was conducted to evaluate the efficacy, safety, and pharmacokinetics of the histone deacetylase inhibitor, pyridylmethyl-N-{4-[(2-aminophenyl)-carbamoyl]-benzyl}-carbamate (MS-275) in 28 patients with pretreated metastatic melanoma. MS-275 is an oral benzamide HDACI. In the study, patients with unresectable American Joint Committee on Cancer (AJCC) stage IV melanoma, refractory to at least one earlier systemic therapy, were randomized to receive MS-275 3 mg bi-weekly or 7 mg weekly on a 28-day cycle. The primary endpoint of the study was objective tumor response, and the secondary study endpoints were safety and time-to-progression. No objective responses were observed in pretreated metastatic melanoma patients. The median time-to-progession was comparable in both arms of the study. MS-275 was well tolearted, with nausea, diarrhea, and hypophosphatemia as the most frequently reported adverse events (Hauschild et al., 2008).

Compound Chemical Group [Range] HDAC Specificity In Vitro Effect Reference
Sodium butyrate Short-chain fatty acid mM Class I, IIa Apoptosis
Cell-cycle arrest
(Bhalla, 2005; Bolden et al., 2006; Johnstone, 2002; Schwabe & Lubbert, 2007)
Valproic acid (VPA) Short-chain fatty acid mM Class I, IIa Apoptosis
Differentiation
(Bhalla, 2005; Bolden et al., 2006; Johnstone, 2002; Schwabe & Lubbert, 2007)
Trichostatin A (TSA) Hydroxamic acid nM Class I, IIa Apoptosis
Cell-cycle arrest
Differentiation
(Bhalla, 2005; Bolden et al., 2006; Johnstone, 2002; Schwabe & Lubbert, 2007)
Suberoylanilide hydroxamic acid (SAHA) or vorinostat Hydroxamic acid µM Class I, II Apoptosis
Cell-cycle arrest
(Bhalla, 2005; Bolden et al., 2006; Johnstone, 2002; Schwabe & Lubbert, 2007)
Depsipeptide (FK 228) Cyclic tetrapeptide nM Class I Apoptosis
Cell-cycle arrest
(Bhalla, 2005; Bolden et al., 2006; Johnstone, 2002; Schwabe & Lubbert, 2007)
Apicidin Cyclic tetrapeptide nM HDACs 1 and 3 Apoptosis
Cell-cycle arrest
(Bolden et al., 2006; Johnstone, 2002; Schwabe & Lubbert, 2007; Vannini et al., 2004)
MS-275 Benzamide µM Class I Cell-cycle arrest (Bolden et al., 2006; Hu et al., 2003; Johnstone, 2002; Schwabe & Lubbert, 2007)

Table 2.

Histone deacetylase inhibitors

Due to the low response rates of HDACIs as single-agent therapies, HDACIs have also been investigated in combination with other therapeutic agents (Table 3). In a phase I/II clinical trial for patients with stage IV melanoma, the combination of valproic acid and the topoisomerase I inhibitor karenitecin associated with disease stabilization in 47% of patients. The median overall survival and time-to-progression were 32.8 and 10.2 weeks, respectively. In addition, histone hyperacetylation was observed in peripheral blood mononuclear cells (Daud et al., 2009).

HDACIs have also been investigated in combination with other treatment modalities. A phase I/II study of HDACI valproic acid with standard chemoimmunotherapy in patients with advanced melanoma was conducted to evaluate its clinical activity and to assess toxicity. In the study, patients were treated initialy with valproic acid alone for 6 weeks. After the treatment with valproic acid alone, dacarbazine plus interferon-α therapy was started in combination with the valproic acid. However, the results showed that the combination of valproic acid and chemoimmunotherapy did not produce superior results as compared to standard therapy (Rocca et al., 2009).

Epigenetic Agent Combination Malignancy Phase Reference
Histone Deacetylase Inhibitors (HDACIs)
Valproic acid Karenitecin (topoisomerase I inhibitor) Melanoma I, II (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)
Vorinostat (Zolinza) NPI-0052 (proteasome inhibitor) Melanoma I (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)
Vorinostat (Zolinza) Unresectable Metastatic Melanoma (Stage IV) II (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)
MS-275 Melanoma II (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)
Romidepsin Nonresectable Intraocular Melanoma or Unresectable Stage III or Stage IV Melanoma II (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)
DNA Methyltransferase Inhibitors (DNA Hypomethylating Agents) (DNMTIs)
5-azacytidine (Vidaza) Recombinant Interferon α-2b Melanoma I (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)
5-aza-2-deoxycytidine (Dacogen) Temozolomide Melanoma I, II (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)
Pegylated Interferon α-2b Melanoma I, II (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)
Panobinostat, Temozolomide Melanoma I, II (Cang et al., 2009; Howell et al., 2009; Sigalotti et al., 2010)

Table 3.

Current epigenetic agents used in clinical trials for melanoma patients

Advertisement

3. DNA methylation

DNA methylation is carried out by different DNA methyltransferases (DNMT). DNMT1 involves in the maintenance of established methylation patterns. DNMT3a and DNMT3b are implicated in de novo DNA methylation (Bestor, 2000; Okano et al., 1999; Rothhammer & Bosserhoff, 2007). This epigenetic event takes place at the C5 position of cytosine on the CpG dinucleotide rich regions (CpG islands) that are distributed throughout the genome. Proper DNA methylation patterns are essential for human development and normal functioning. In normal cells, CpG islands located in the promoter regions are mainly unmethylated; however, in melanoma cancer cells, aberrant hypermethylation occur. In addition, genome-wide hypomethylation occurs in melanoma cancer cells (Jones & Baylin, 2007). This epigenetic modification results in silencing the transcription of selected tumor suppressor genes (Robertson, 2005; Rothhammer & Bosserhoff, 2007). Aberrant DNA hypermethylation of promoter regions has been shown to result in the silencing of at least 50 genes (Fulda et al., 2001; Gallagher et al., 2005; Mori et al., 2005; Muthusamy et al., 2006; Paz et al., 2003; Rothhammer & Bosserhoff, 2007; Soengas et al., 2001; van der Velden et al., 2003). Table 4 shows some genes affected by promoter DNA hypermethylation in melanoma. For example, CDKN2A is a major gene involved in the pathogenesis of melanoma. It is the most frequently mutated gene inherited in familial cutaneous melanoma (Palmieri et al., 2009; Sigalotti et al., 2010). Freedberg et al (2008) showed that aberrant promoter DNA hypermethylation at CDKN2A locus independently affects the tumor suppressors p16INK4A and p14ARF, which function in the pRB and p53 pathways, respectively. In human melanoma, p16INK4A and p14ARF are methylated.

Gene Pathway Reference
APAF1(apoptotic protease activating factor 1) Apoptosis (Soengas et al., 2001)
MT2A (methallothionein 2A) Apoptosis (Gallagher et al., 2005)
HSPB1 (heat shock 27 kDa protein) Apoptosis (Gallagher et al., 2005)
MAGE-A1(melanoma antigen, family A1) Immune recognition (De Smet et al., 1996; Karpf et al., 2004; Sigalotti et al., 2010)
ER-α (estrogen receptor alpha) Signaling (Mori et al., 2006)
WFDC1 (wap 4-disulfide core domain 1) Proliferation (Muthusamy et al., 2006)
CDKN1B (cyclin-dependent kinase inhibitor 1B Cell cycle (Worm et al., 2000)
CDKN1C (cyclin-dependent kinase inhibitor 1C) Cell cycle (Shen et al., 2007)
APC (adenomatous polyposis coli gene) Cell fate determination (Worm et al., 2004)
GDF15 (growth/differentiation factor 15) Differentiation (Muthusamy et al., 2006)
TPM1 (tropomyosin 1) Anchorage-independent growth (Liu et al., 2008)
MIB2 (skeletrophin) Cell fate determination (Takeuchi et al., 2006)
MGMT (06-methylguanine-DNA-methyltransferase) DNA repair (Hoon et al., 2004)
CDH1 (E-cadherin) Invasion/metastasis (Liu et al., 2008)
CDH8 (cadherin 8) Invasion/metastasis (Muthusamy et al., 2006)

Table 4.

Genes with an altered DNA methylation status in melanoma

3.1. Clinical applications of DNA methylation

DNA methylation is a reversible epigenetic event and can be nullified by specific DNA demethylating agents (DNA methyltransferase inhibitors). Several ongoing clinical trials are conducted to investigate their clinical effectiveness and safety in melanoma patients (Table 3). In these studies, DNA demethylating agents 5-azacytidine (Vidaza) and 5-aza-2-deoxycytidine (decitabine, Dacogen) are the most intensively studied. Azacytidine is a pyrimidine nucleoside analog of cytidine, and decitabine is a cytosine nucleoside (cytidine) analog. These epigenetic agents were approved by the FDA for the treatment of myelodysplastic syndromes and acute myeloid leukemia. Agents that inhibit DNA methyltransferases can reactivate silenced genes and induce apoptosis of cancerous cells (Howell et al., 2009). Since epigenetic modifications affect cellular pathways, epigenetic agents also display pleiotropic activities (Howell et al., 2009). In a phase I trial, Gollob et al. (2006) found that a low dose of 5-aza-2'-deoxycytidine (decitabine) can be safely administered with high-dose interleukin to cancer patients and has antitumor activity in melanoma. The inclusion of decitabine resulted in DNA hypomethylation. In addition, Appleton et al. (2007) showed that decitabine reduces DNA methylation and can be combined safely with carboplatin for the treatment of melanoma.

In addition to therapeutic applications, modifications of DNA methylation may serve as biomarkers in clinical use for melanoma (Howell et al., 2009). Mori et al. (2006) showed that methylated ER-α can be detected in paraffin-embedded primary and metastatic melanoma tumors. In addition, methylated ER-α DNA was detected in the serum of melanoma patients with AJCC stage I to IV disease. Methylated ER-α was detected in 42% of stage III and 86% of stage IV metastatic melanomas. Serum methylated ER-α is an unfavorable prognostic factor. Liu et al (2008) found that SOCS1, SOCS2, RARβ2, DcR1, and DcR2 genes were the most frequently methylated genes in melanoma. The investigators also found that RECK, IRF7, PAWR, DR5, and Rb were not methylated in melanoma although these genes were found to be highly methylated in other cancers (Howell et al., 2009; Liu et al., 2008), suggesting that different cancers have distinct methylated genes. This is important since biomarkers must be specific and be able to differentiate between different forms of malignancies.

Advertisement

4. Conclusions and future perspectives

Melanoma is a complex disease that is caused by aberrant genetic and epigenetic events.

Epigenetic modifications play a significant role in the biology of melanoma, and epigenetic therapy emerges as a promising treatment modality for melanoma as well as for dignostic developments for the malignancy. A major difference between the two events is that epigenetic changes can be reversed by chemical and/or environmental modalities. Histone modifications and DNA methylation are extensively studied epigenetic events that affect the expression of genes. Currently, four epigentic agents have been approved by the U.S. FDA for hematologic malignancies and many HDACIs and DNMTIs are being investigated in clinical trials for solid tumors, such as melanoma. However, there have not been any FDA-approved epigenetic agents for solid tumors. Consequently, further investigations are required to find successful treatment strategies or protocols involving epigenetic agents. Future developments would address the issues of systemic toxicities, nonspecific epigenetic effect, and low bioavailability. In addition, a promising strategy is combination therapy. In tumors, DNA methylation and histone acetylation can act synergistically to silence tumor suppressor genes. This approach could potentially enhance the reversal of epigenetic silencing. Although in its infancy, epigenetic therapy has been shown to be an effective treatment modality for cancers, as evident by the approval of 4 epigenetic drugs by the U.S. FDA. Encouraging results from preclinical and clinical trials prompts further investigations into designing new drugs or strategy that are more suitable for epigenetic therapies for melanoma patients, with the goal of improving patient outcomes.

References

  1. 1. Appleton K. Mackay H. J. Judson I. Plumb J. A. Mc Cormick C. Strathdee G. Lee C. Barrett S. Reade S. Jadayel D. Tang A. Bellenger K. Mackay L. Setanoians A. Schatzlein A. Twelves C. Kaye S. B. Brown R. 2007Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. J Clin Oncol, 25 29 4603 4609
  2. 2. Bernstein B. E. Tong J. K. Schreiber S. L. 2000Genomewide studies of histone deacetylase function in yeast. Proc Natl Acad Sci U S A, 97 25 13708 13713
  3. 3. Bestor T. H. 2000The DNA methyltransferases of mammals. Hum Mol Genet, 9 16 2395 2402
  4. 4. Bhalla K. N. 2005Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies. J Clin Oncol, 23 17 3971 3993
  5. 5. Bishton M. Kenealy M. Johnstone R. Rasheed W. Prince H. M. 2007Epigenetic targets in hematological malignancies: combination therapies with HDACis and demethylating agents. Expert Rev Anticancer Ther, 7 10 1439 1449
  6. 6. Bolden J. E. Peart M. J. Johnstone R. W. 2006Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov, 5 9 769 784
  7. 7. Bolger T. A. Yao T. P. 2005Intracellular trafficking of histone deacetylase 4 regulates neuronal cell death. J Neurosci, 25 41 9544 9553
  8. 8. Campas-Moya C. 2009Romidepsin for the treatment of cutaneous T-cell lymphoma. Drugs Today (Barc), 45 11 787 795
  9. 9. Cang S. Ma Y. Liu D. 2009New clinical developments in histone deacetylase inhibitors for epigenetic therapy of cancer. J Hematol Oncol, 2 22
  10. 10. Carew J. S. Giles F. J. Nawrocki S. T. 2008Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett, 269 1 7 17
  11. 11. Chang S. Mc Kinsey T. A. Zhang C. L. Richardson J. A. Hill J. A. Olson E. N. 2004Histone deacetylases 5 and 9 govern responsiveness of the heart to a subset of stress signals and play redundant roles in heart development. Mol Cell Biol, 24 19 8467 8476
  12. 12. Choi S. Lew K. L. Xiao H. Herman-Antosiewicz A. Xiao D. Brown C. K. Singh S. V. 2007D,L-Sulforaphane-induced cell death in human prostate cancer cells is regulated by inhibitor of apoptosis family proteins and Apaf-1. Carcinogenesis, 28 1 151 162
  13. 13. Daud A. I. Dawson J. De Conti R. C. Bicaku E. Marchion D. Bastien S. Hausheer F. A. 3rd Lush R. Neuger A. Sullivan D. M. Munster P. N. 2009Potentiation of a topoisomerase I inhibitor, karenitecin, by the histone deacetylase inhibitor valproic acid in melanoma: translational and phase I/II clinical trial. Clin Cancer Res, 15 7 2479 2487
  14. 14. de Ruijter A. J. van Gennip A. H. Caron H. N. Kemp S. van Kuilenburg A. B. 2003Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J, 370No.Pt 3, 737 749
  15. 15. De Smet C. De Backer O. Faraoni I. Lurquin C. Brasseur F. Boon T. 1996The activation of human gene MAGE-1 in tumor cells is correlated with genome-wide demethylation. Proc Natl Acad Sci U S A, 93 14 7149 7153
  16. 16. Dequiedt F. Kasler H. Fischle W. Kiermer V. Weinstein M. Herndier B. G. Verdin E. 2003HDAC7, a thymus-specific class II histone deacetylase, regulates Nur77 transcription and TCR-mediated apoptosis. Immunity, 18 5 687 698
  17. 17. Dequiedt F. Van Lint J. Lecomte E. Van Duppen V. Seufferlein T. Vandenheede J. R. Wattiez R. Kettmann R. 2005Phosphorylation of histone deacetylase 7 by protein kinase D mediates T cell receptor-induced Nur77 expression and apoptosis. J Exp Med, 201 5 793 804
  18. 18. Foglietti C. Filocamo G. Cundari E. De Rinaldis E. Lahm A. Cortese R. Steinkuhler C. 2006Dissecting the biological functions of Drosophila histone deacetylases by RNA interference and transcriptional profiling. J Biol Chem, 281 26 17968 17976
  19. 19. Fraga M. F. Ballestar E. Villar-Garea A. Boix-Chornet M. Espada J. Schotta G. Bonaldi T. Haydon C. Ropero S. Petrie K. Iyer N. G. Perez-Rosado A. Calvo E. Lopez J. A. Cano A. Calasanz M. J. Colomer D. Piris M. A. Ahn N. Imhof A. Caldas C. Jenuwein T. Esteller M. 2005Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat Genet, 37 4 391 400
  20. 20. Freedberg D. E. Rigas S. H. Russak J. Gai W. Kaplow M. Osman I. Turner F. Randerson-Moor J. A. Houghton A. Busam K. Timothy Bishop. D. Bastian B. C. Newton-Bishop J. A. Polsky D. 2008Frequent p16-independent inactivation of p14ARF in human melanoma. J Natl Cancer Inst, 100 11 784 795
  21. 21. Fulda S. Kufer M. U. Meyer E. van Valen F. Dockhorn-Dworniczak B. Debatin K. M. 2001Sensitization for death receptor- or drug-induced apoptosis by re-expression of caspase-8 through demethylation or gene transfer. Oncogene, 20 41 5865 5877
  22. 22. Gallagher W. M. Bergin O. E. Rafferty M. Kelly Z. D. Nolan I. M. Fox E. J. Culhane A. C. Mc Ardle L. Fraga M. F. Hughes L. Currid C. A. O’Mahony F. Byrne A. Murphy A. A. Moss C. Mc Donnell S. Stallings R. L. Plumb J. A. Esteller M. Brown R. Dervan P. A. Easty D. J. 2005Multiple markers for melanoma progression regulated by DNA methylation: insights from transcriptomic studies. Carcinogenesis, 26 11 1856 1867
  23. 23. Gao L. Cueto M. A. Asselbergs F. Atadja P. 2002Cloning and functional characterization of HDAC11, a novel member of the human histone deacetylase family. J Biol Chem, 277 28 25748 25755
  24. 24. Glozak M. A. Seto E. 2007Histone deacetylases and cancer. Oncogene, 26 37 5420 5432
  25. 25. Gollob J. A. Sciambi C. J. Peterson B. L. Richmond T. Thoreson M. Moran K. Dressman H. K. Jelinek J. Issa J. P. 2006Phase I trial of sequential low-dose 5-aza-2’-deoxycytidine plus high-dose intravenous bolus interleukin-2 in patients with melanoma or renal cell carcinoma. Clin Cancer Res, 12 15 4619 4627
  26. 26. Grant P. A. Berger S. L. 1999Histone acetyltransferase complexes. Semin Cell Dev Biol, 10 2 169 177
  27. 27. Haberland M. Montgomery R. L. Olson E. N. 2009The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet, 10 1 32 42
  28. 28. Hauschild A. Trefzer U. Garbe C. Kaehler K. C. Ugurel S. Kiecker F. Eigentler T. Krissel H. Schott A. Schadendorf D. 2008Multicenter phase II trial of the histone deacetylase inhibitor pyridylmethyl-N-{4-[(2-aminophenyl)-carbamoyl]-benzyl}-carbamate in pretreated metastatic melanoma. Melanoma Res, 18 4 274 278
  29. 29. Hebbes T. R. Thorne A. W. Clayton A. L. Crane-Robinson C. 1992Histone acetylation and globin gene switching. Nucleic Acids Res, 20 5 1017 1022
  30. 30. Hoon D. S. Spugnardi M. Kuo C. Huang S. K. Morton D. L. Taback B. 2004Profiling epigenetic inactivation of tumor suppressor genes in tumors and plasma from cutaneous melanoma patients. Oncogene, 23 22 4014 4022
  31. 31. Houghton A. N. Polsky D. 2002Focus on melanoma. Cancer Cell, 2 4 275 278
  32. 32. Howell P. M. Jr Liu S. Ren S. Behlen C. Fodstad O. Riker A. I. 2009Epigenetics in human melanoma. Cancer Control, 16 3 200 218
  33. 33. Hu E. Dul E. Sung C. M. Chen Z. Kirkpatrick R. Zhang G. F. Johanson K. Liu R. Lago A. Hofmann G. Macarron R. de los Frailes. M. Perez P. Krawiec J. Winkler J. Jaye M. 2003Identification of novel isoform-selective inhibitors within class I histone deacetylases. J Pharmacol Exp Ther, 307 2 720 728
  34. 34. Hubbert C. Guardiola A. Shao R. Kawaguchi Y. Ito A. Nixon A. Yoshida M. Wang X. F. Yao T. P. 2002HDAC6 is a microtubule-associated deacetylase. Nature, 417 6887 455 458
  35. 35. Imre G. Gekeler V. Leja A. Beckers T. Boehm M. 2006Histone deacetylase inhibitors suppress the inducibility of nuclear factor-kappaB by tumor necrosis factor-alpha receptor-1 down-regulation. Cancer Res, 66 10 5409 5418
  36. 36. Jenuwein T. Allis C. D. 2001Translating the histone code. Science, 293 5532 1074 1080
  37. 37. Johnstone R. W. 2002Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov, 1 4 287 299
  38. 38. Jones P. A. Baylin S. B. 2007The epigenomics of cancer. Cell, 128 4 683 692
  39. 39. Kamakaka R. T. Biggins S. 2005Histone variants: deviants? Genes Dev, 19 3 295 310
  40. 40. Karagiannis T. C. El -Osta A. 2006Clinical potential of histone deacetylase inhibitors as stand alone therapeutics and in combination with other chemotherapeutics or radiotherapy for cancer. Epigenetics, 1 3 121 126
  41. 41. Karpf A. R. Lasek A. W. Ririe T. O. Hanks A. N. Grossman D. Jones D. A. 2004Limited gene activation in tumor and normal epithelial cells treated with the DNA methyltransferase inhibitor 5-aza-2’-deoxycytidine. Mol Pharmacol, 65 1 18 27
  42. 42. Kondo Y. Shen L. Cheng A. S. Ahmed S. Boumber Y. Charo C. Yamochi T. Urano T. Furukawa K. Kwabi-Addo B. Gold D. L. Sekido Y. Huang T. H. Issa J. P. 2008Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet, 40 6 741 750
  43. 43. Kouzarides T. 2007Chromatin modifications and their function. Cell, 128 4 693 705
  44. 44. Lagger G. O’Carroll D. Rembold M. Khier H. Tischler J. Weitzer G. Schuettengruber B. Hauser C. Brunmeir R. Jenuwein T. Seiser C. 2002Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression. EMBO J, 21 11 2672 2681
  45. 45. Lee J. H. Jeong E. G. Choi M. C. Kim S. H. Park J. H. Song S. H. Park J. Bang Y. J. Kim T. Y. 2010Inhibition of histone deacetylase 10 induces thioredoxin-interacting protein and causes accumulation of reactive oxygen species in SNU-620 human gastric cancer cells. Mol Cells, 30 2 107 112
  46. 46. Liu S. Ren S. Howell P. Fodstad O. Riker A. I. 2008Identification of novel epigenetically modified genes in human melanoma via promoter methylation gene profiling. Pigment Cell Melanoma Res, 21 5 545 558
  47. 47. Luger K. Mader A. W. Richmond R. K. Sargent D. F. Richmond T. J. 1997Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature, 389 6648 251 260
  48. 48. Mani S. Herceg Z. 2010DNA demethylating agents and epigenetic therapy of cancer. Adv Genet, 70 327 340
  49. 49. Marchion D. Munster P. 2007Development of histone deacetylase inhibitors for cancer treatment. Expert Rev Anticancer Ther, 7 4 583 598
  50. 50. Marks P. A. Breslow R. 2007Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol, 25 1 84 90
  51. 51. Marks P. A. Richon V. M. Rifkind R. A. 2000Histone deacetylase inhibitors: inducers of differentiation or apoptosis of transformed cells. J Natl Cancer Inst, 92 15 1210 1216
  52. 52. Marks P. A. Xu W. S. 2009Histone deacetylase inhibitors: Potential in cancer therapy. J Cell Biochem, 107 4 600 608
  53. 53. Miller A. J. Mihm M. C. Jr 2006Melanoma. N Engl J Med, 355 1 51 65
  54. 54. Minucci S. Pelicci P. G. 2006Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer, 6 1 38 51
  55. 55. Montgomery R. L. Davis C. A. Potthoff M. J. Haberland M. Fielitz J. Qi X. Hill J. A. Richardson J. A. Olson E. N. 2007Histone deacetylases 1 and 2 redundantly regulate cardiac morphogenesis, growth, and contractility. Genes Dev, 21 14 1790 1802
  56. 56. Mori T. Martinez S. R. O’Day S. J. Morton D. L. Umetani N. Kitago M. Tanemura A. Nguyen S. L. Tran A. N. Wang H. J. Hoon D. S. 2006Estrogen receptor-alpha methylation predicts melanoma progression. Cancer Res, 66 13 6692 6698
  57. 57. Mori T. O’Day S. J. Umetani N. Martinez S. R. Kitago M. Koyanagi K. Kuo C. Takeshima T. L. Milford R. Wang H. J. Vu V. D. Nguyen S. L. Hoon D. S. 2005Predictive utility of circulating methylated DNA in serum of melanoma patients receiving biochemotherapy. J Clin Oncol, 23 36 9351 9358
  58. 58. Muthusamy V. Duraisamy S. Bradbury C. M. Hobbs C. Curley D. P. Nelson B. Bosenberg M. 2006Epigenetic silencing of novel tumor suppressors in malignant melanoma. Cancer Res, 66 23 11187 11193
  59. 59. Okano M. Bell D. W. Haber D. A. Li E. 1999DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell, 99 3 247 257
  60. 60. Palmieri G. Capone M. Ascierto M. L. Gentilcore G. Stroncek D. F. Casula M. Sini M. C. Palla M. Mozzillo N. Ascierto P. A. 2009Main roads to melanoma. J Transl Med, 7 86
  61. 61. Paris M. Porcelloni M. Binaschi M. Fattori D. 2008Histone deacetylase inhibitors: from bench to clinic. J Med Chem, 51 6 1505 1529
  62. 62. Paz M. F. Fraga M. F. Avila S. Guo M. Pollan M. Herman J. G. Esteller M. 2003A systematic profile of DNA methylation in human cancer cell lines. Cancer Res, 63 5 1114 1121
  63. 63. Richon V. M. Sandhoff T. W. Rifkind R. A. Marks P. A. 2000Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci U S A, 97 18 10014 10019
  64. 64. Robertson K. D. 2005DNA methylation and human disease. Nat Rev Genet, 6 8 597 610
  65. 65. Rocca A. Minucci S. Tosti G. Croci D. Contegno F. Ballarini M. Nole F. Munzone E. Salmaggi A. Goldhirsch A. Pelicci P. G. Testori A. 2009A phase I-II study of the histone deacetylase inhibitor valproic acid plus chemoimmunotherapy in patients with advanced melanoma. Br J Cancer, 100 1 28 36
  66. 66. Rothhammer T. Bosserhoff A. K. 2007Epigenetic events in malignant melanoma. Pigment Cell Res, 20 2 92 111
  67. 67. Schwabe M. Lubbert M. 2007Epigenetic lesions in malignant melanoma. Curr Pharm Biotechnol, 8 6 382 387
  68. 68. Shen L. Kondo Y. Guo Y. Zhang J. Zhang L. Ahmed S. Shu J. Chen X. Waterland R. A. Issa J. P. 2007Genome-wide profiling of DNA methylation reveals a class of normally methylated CpG island promoters. PLoS Genet, 3 10 2023 2036
  69. 69. Sigalotti L. Covre A. Fratta E. Parisi G. Colizzi F. Rizzo A. Danielli R. Nicolay H. J. Coral S. Maio M. 2010Epigenetics of human cutaneous melanoma: setting the stage for new therapeutic strategies. J Transl Med, 8 56
  70. 70. Soengas M. S. Capodieci P. Polsky D. Mora J. Esteller M. Opitz-Araya X. Mc Combie R. Herman J. G. Gerald W. L. Lazebnik Y. A. Cordon-Cardo C. Lowe S. W. 2001Inactivation of the apoptosis effector Apaf-1 in malignant melanoma. Nature, 409 6817 207 211
  71. 71. Stearns V. Zhou Q. Davidson N. E. 2007Epigenetic regulation as a new target for breast cancer therapy. Cancer Invest, 25 8 659 665
  72. 72. Strahl B. D. Allis C. D. 2000The language of covalent histone modifications. Nature, 403 6765 41 45
  73. 73. Sun Z. W. Hampsey M. 1999A general requirement for the Sin3-Rpd3 histone deacetylase complex in regulating silencing in Saccharomyces cerevisiae. Genetics, 152 3 921 932
  74. 74. Takeuchi T. Adachi Y. Sonobe H. Furihata M. Ohtsuki Y. 2006A ubiquitin ligase, skeletrophin, is a negative regulator of melanoma invasion. Oncogene, 25 53 7059 7069
  75. 75. Turner B. M. 1993Decoding the nucleosome. Cell, 75 1 5 8
  76. 76. van der Velden P. A. Zuidervaart W. Hurks M. H. Pavey S. Ksander B. R. Krijgsman E. Frants R. R. Tensen C. P. Willemze R. Jager M. J. Gruis N. A. 2003Expression profiling reveals that methylation of TIMP3 is involved in uveal melanoma development. Int J Cancer, 106 4 472 479
  77. 77. Vannini A. Volpari C. Filocamo G. Casavola E. C. Brunetti M. Renzoni D. Chakravarty P. Paolini C. De Francesco R. Gallinari P. Steinkuhler C. Di Marco S. 2004Crystal structure of a eukaryotic zinc-dependent histone deacetylase, human HDAC8, complexed with a hydroxamic acid inhibitor. Proc Natl Acad Sci U S A, 101 42 15064 15069
  78. 78. Vega R. B. Matsuda K. Oh J. Barbosa A. C. Yang X. Meadows E. Mc Anally J. Pomajzl C. Shelton J. M. Richardson J. A. Karsenty G. Olson E. N. 2004Histone deacetylase 4 controls chondrocyte hypertrophy during skeletogenesis. Cell, 119 4 555 566
  79. 79. Villagra A. Cheng F. Wang H. W. Suarez I. Glozak M. Maurin M. Nguyen D. Wright K. L. Atadja P. W. Bhalla K. Pinilla-Ibarz J. Seto E. Sotomayor E. M. 2009The histone deacetylase HDAC11 regulates the expression of interleukin 10 and immune tolerance. Nat Immunol, 10 1 92 100
  80. 80. Waltregny D. De Leval L. Glenisson W. Ly Tran. S. North B. J. Bellahcene A. Weidle U. Verdin E. Castronovo V. 2004Expression of histone deacetylase 8, a class I histone deacetylase, is restricted to cells showing smooth muscle differentiation in normal human tissues. Am J Pathol, 165 2 553 564
  81. 81. Waltregny D. Glenisson W. Tran S. L. North B. J. Verdin E. Colige A. Castronovo V. 2005Histone deacetylase HDAC8 associates with smooth muscle alpha-actin and is essential for smooth muscle cell contractility. FASEB J, 19 8 966 968
  82. 82. Wijermans P. W. Lubbert M. Verhoef G. Klimek V. Bosly A. 2005An epigenetic approach to the treatment of advanced MDS; the experience with the DNA demethylating agent 5-aza-2’-deoxycytidine (decitabine) in 177 patients. Ann Hematol, 84Suppl 1, 9 17
  83. 83. Worm J. Bartkova J. Kirkin A. F. Straten P. Zeuthen J. Bartek J. Guldberg P. 2000Aberrant p27Kip1 promoter methylation in malignant melanoma. Oncogene, 19 44 5111 5115
  84. 84. Worm J. Christensen C. Gronbaek K. Tulchinsky E. Guldberg P. 2004Genetic and epigenetic alterations of the APC gene in malignant melanoma. Oncogene, 23 30 5215 5226
  85. 85. Xu C. Shen G. Chen C. Gelinas C. Kong A. N. 2005Suppression of NF-kappaB and NF-kappaB-regulated gene expression by sulforaphane and PEITC through IkappaBalpha, IKK pathway in human prostate cancer PC-3 cells. Oncogene, 24 28 4486 4495
  86. 86. Yoshida N. Omoto Y. Inoue A. Eguchi H. Kobayashi Y. Kurosumi M. Saji S. Suemasu K. Okazaki T. Nakachi K. Fujita T. Hayashi S. 2004Prediction of prognosis of estrogen receptor-positive breast cancer with combination of selected estrogen-regulated genes. Cancer Sci, 95 6 496 502
  87. 87. Zhang C. L. Mc Kinsey T. A. Chang S. Antos C. L. Hill J. A. Olson E. N. 2002Class II histone deacetylases act as signal-responsive repressors of cardiac hypertrophy. Cell, 110 4 479 488
  88. 88. Zhang Z. Yamashita H. Toyama T. Sugiura H. Omoto Y. Ando Y. Mita K. Hamaguchi M. Hayashi S. Iwase H. 2004HDAC6 expression is correlated with better survival in breast cancer. Clin Cancer Res, 10 20 6962 6968
  89. 89. Zupkovitz G. Tischler J. Posch M. Sadzak I. Ramsauer K. Egger G. Grausenburger R. Schweifer N. Chiocca S. Decker T. Seiser C. 2006Negative and positive regulation of gene expression by mouse histone deacetylase 1. Mol Cell Biol, 26 21 7913 7928

Written By

Duc P. Do and Syed A.A. Rizvi

Submitted: 01 November 2010 Published: 12 September 2011