Abstract
Epigenetic inactivation of pivotal genes involved in correct cell growth is a hallmark of human pathologies, in particular cancer. These epigenetic mechanisms, including crosstalk between DNA methylation, histone modifications and non‐coding RNAs, affect gene expression and are associated with disease progression. In contrast to genetic mutations, epigenetic changes are potentially reversible. Re‐expression of genes epigenetically inactivated can result in the suppression of disease state or sensitization to specific therapies. Small molecules that reverse epigenetic inactivation, so‐called epi‐drugs, are now undergoing clinical trials. Accordingly, the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for cancer treatment have approved some of these drugs. Here, we focus on the biological features of epigenetic molecules, analyzing the mechanism(s) of action and their current use in clinical practice.
Keywords: Epigenetics, Epi-drugs, Clinical trials, Cancer
Highlights
Epi‐drugs play a key role in pathogenesis and tumorigenesis.
Epi‐drugs in clinical trials provide exciting results in human diseases therapy.
Epi‐drugs may provide an opportunity to develop tailor‐made therapies.
Epi‐drugs enhance the effects of combination therapy.
Epi‐drugs increase sensitivity to classical anti‐cancer agents.
1. Introduction
It is well established that both genetic and epigenetic changes contribute to the development of human diseases, such as cancer (Hahn and Weinberg, 2002; Jones and Baylin, 2002).
The term “epigenetics” refers to all heritable alterations in gene expression and chromatin structure due to chemical modifications that do not involve changes in the primary gene nucleotide sequence (Nightingale et al., 2006). The correct regulation of these alterations ensures appropriate cell growth (Bernstein et al., 2007; Jenuwein and Allis, 2001; Strahl and Allis, 2000). In contrast, deregulation of epigenetic patterns leads to induction and propagation of disease state (Feinberg et al., 2006; Hahn and Weinberg, 2002; Jones and Baylin, 2002).
Epigenetic inheritance can be classified into three distinct types: DNA methylation, histone modifications, and non‐coding RNAs. All of these are crucial mechanisms that ensure the stable propagation of gene activity from one generation of cells to the next (Feinberg et al., 2006; Jenuwein and Allis, 2001). Disruption of any of these three distinct and mutually reinforcing epigenetic mechanisms leads to inappropriate gene expression, resulting in cancer development and other “epigenetic diseases” (Egger et al., 2004; Feinberg et al., 2006; Feinberg and Tycko, 2004; Jones and Baylin, 2002).
Although precise underlying mechanisms are not yet clear, in recent years, scientific interest in epigenetics has increased insofar as it represents an important tool to advance our understanding of pathogenesis, in particular tumorigenesis, and to help in the development (Egger et al., 2004; Strahl and Allis, 2000) of strategies for cancer treatment and prevention.
2. Chromatin structure and regulation
In eukaryotic cells, nuclear DNA associates with histones to form a compact complex called chromatin. Histones constitute a family of basic proteins, which are generally highly conserved across eukaryotic species. The core histones, termed H2A, H2B, H3, and H4, associate to form an octamer structure comprising two molecules each of H2A, H2B, H3 and H4 histones. DNA winds around this protein core, with the basic amino acids of the histones interacting with negatively charged phosphate groups of the DNA. Approximately 146 bp of DNA wrap around a histone core to make up a nucleosome particle, the repeating structural motif of chromatin. Histone H1 assembles such nucleosomes into a higher ordered structure. The N‐terminal tails of nucleosomal histones are subject to different modifications including acetylation, methylation, phosphorylation, ubiquitination and ADP‐ribosylation. Of these, acetylation has been the most extensively investigated (Shukla et al., 2008). Recent studies in the field of chromatin research provided experimental evidence that led to a concept known as the “histone code” (Jenuwein and Allis, 2001; Strahl and Allis, 2000). It postulates that different histone modifications are combinatorial and consistent, generating a code that is read by cellular machineries to dictate functional outcomes. Different histone modifications were shown to be essential for normal cellular processes because crucially involved in transcriptional regulation. In view of this fact, attempts have been made to regulate the transcription of disease‐related genes (e.g., oncogenes and tumor suppressor genes (TSGs)) by using epigenetic regulators to treat cancers and other diseases.
3. Histone modificators in clinical trials
Epimutations play a role in the etiology of human cancers. In contrast to DNA mutations, which are passively inherited through DNA replication, epimutations must be actively maintained because they are reversible. The reversibility of epimutations by small‐molecule inhibitors provides the foundation for their use in novel cancer therapy strategies. Among the compounds that inhibit epigenetic processes, the most extensively studied are DNA methyltransferase inhibitors and HDAC inhibitors (HDACi). Here, we focus on epigenetic modulators used in clinical trials (either completed or terminated) to treat human diseases (www.clinicaltrials.gov).
3.1. HDAC inhibitors
Histone deacetylases are known to play a key role in the transcriptional machinery for regulating gene expression, to induce histone hyperacetylation and to affect gene expression. Therefore, they represent the target of therapeutic or prophylactic agents, HDACis, for diseases caused by abnormal gene expression such as inflammatory disorders, diabetes, diabetic complications, homozygous thalassemia, fibrosis, cirrhosis, acute promyelocytic leukemia (APL), autoimmune diseases and tumors as well as organ transplant rejections and protozoal infections.
Acetylation and deacetylation of histones are carried out by “writers” (histone acetyl transferases (HATs)) and “erasers” (histone deacetylases (HDAC)) enzymes. The state of acetylation of histones is an important determinant of gene transcription. Deacetylation is generally associated with reduced transcription of genes whereas increased acetylation of histones induced by the action of HDACi results in greater transcription of genes. Thus, HDACi affect multiple processes in the cell, which are likely to depend upon its dynamic state with respect to its capabilities of replication and differentiation.
The term “HDAC inhibitors” is commonly used for compounds that target the classical class I (HDAC1, 2, 3 and 8), II (HDAC4, 5, 6, 7, 8, 9 and 10), and IV (HDAC11) HDACs and are currently being evaluated in clinical trials. A number of structurally diverse HDACi have been identified, many of which are or derive from natural products. These can be classified, according to their chemical structure, into the following categories:
-
(a)
hydroxamic acids (such as trichostatin A (TSA) (Yoshida et al., 1990) and hydroxamic acid‐based hybrid polar compounds e.g., suberoylanilide hydroxamic acid (SAHA) (Richon et al., 1998)and pyroxamide (Butler et al., 2001));
-
(b)
cyclic tetrapeptides with the epoxyketone‐containing amino acid (2S,9S)‐2‐amino‐8‐oxo‐9,10‐epoxy‐decanoyl (Aoe) (such as trapoxin A and B) (Kijima et al., 1993);
-
(c)
cyclic tetrapeptides without Aoe (such as apicidin (Darkin‐Rattray et al., 1996) and the depsipeptide FR‐901228 (Romidepsin) (Nakajima et al., 1998));
-
(d)
short‐chain and aromatic fatty acids (such as butyrate (Newmark et al., 1994), 4‐phenybutyrate (Warrell et al., 1998) and valproic acid (Phiel et al., 2001));
-
(e)
benzamides (such as MS‐275 (Entinostat)) (Saito et al., 1999) and MGCD0103 (Mocetinostat) (Fournel et al., 2008);
-
(f)
miscellaneous compounds (such as depudecin (Kwon et al., 1998)).
Increasing evidence supports distinct biological roles for each of the mammalian HDACs and it is probable that inhibition of specific members of the HDAC family will have specific functional consequences such as on gene expression, cell cycle regulation, proliferation, differentiation and apoptosis. A number of HDAC isoform‐selective or ‐specific inhibitors are currently in development (Butler and Kozikowski, 2008; Estiu et al., 2008; Haggarty et al., 2003; Jones et al., 2008; Khan et al., 2008; Kozikowski et al., 2007; Moradei et al., 2008; Rasheed et al., 2007; Somoza et al., 2004). One question in the field of HDACi development, which remains unanswered, is whether selective inhibition of HDACs would be advantageous over broader‐acting HDACi as anti‐cancer agents.
HDACi have a plethora of biologic effects resulting from alterations in patterns of acetylation of histones and many non‐histone proteins, which include proteins involved in regulation of gene expression, pathways of extrinsic and intrinsic apoptosis, cell cycle progression, redox pathways, mitotic division, DNA repair, cell migration, and angiogenesis (Blackwell et al., 2008; Bolden et al., 2006; Dokmanovic et al., 2007; Glozak and Seto, 2007; Jones and Baylin, 2007; Minucci and Pelicci, 2006; Shankar and Srivastava, 2008; Xu et al., 2007). HDACi also have immunomodulatory activity that may contribute to mediating their anti‐cancer effects. Furthermore, in contrast to most cancer‐therapy agents, HDACi can induce the death of transformed cells in both proliferative and non‐proliferative phases of cell cycle (Burgess et al., 2004). The mechanism(s) of action of HDACi are complex and not completely clear.
Mechanism(s) of resistance to HDACi are not well elucidated. Resistance to HDACi may reflect drug efflux, epigenetic alterations, stress response mechanisms, and anti‐apoptotic and pro‐survival mechanisms (Rosato et al., 2006).
According to results of pre‐clinical studies and early clinical trials, HDACi in combination therapy with targeted anti‐cancer drugs, cytotoxic agents, anti‐angiogenesis drugs or radiation is potentially very useful (Bolden et al., 2006; Dokmanovic et al., 2007; Glozak and Seto, 2007; Jones and Baylin, 2007; Minucci and Pelicci, 2006; Nolan et al., 2008; Xu et al., 2007). In pre‐clinical studies, HDACi have been shown to be synergistic with diverse chemical and biological therapeutic agents. HDACi will likely be most effective therapeutically when used in combination with other anti‐cancer agents (Carew et al., 2008; Nolan et al., 2008).
Based on published reports, there are at least 20 structurally different HDACi currently in clinical trials as monotherapy and combination therapy for hematologic and solid cancers.
3.1.1. Short‐chain fatty acids
Their mechanism of action of short‐chain fatty acids is based on the carboxylic group, occupying the acetate escaping tunnel, that can have a zinc‐binding function or compete with the acetate group released in the deacetylation reaction (Mai and Altucci, 2009).
Two short‐chain fatty acids, valproic acid (VPA) and sodium phenylbutyrate, are in clinical trials (Table 1).
Table 1.
Drug | Conditions | Recruitment | NCT number | Phases |
---|---|---|---|---|
Valproic acid & fludarabine | Chronic lymphocytic leukemia | Terminated | NCT00524667 | Phase 2 |
Azacitidine, valproic acid | Advanced cancers | Completed | NCT00496444 | Phase 1 |
Temozolomide + valproic acid | Brain metastases | Terminated | NCT00437957 | Phase 1 |
5‐Azacytidine + valproic acid + ATRA | Myelodysplastic syndromes | Completed | NCT00439673 | Phase 2 |
5 azacytidine + valproic acid + retinoic acid | AML, MDS | Completed | NCT00339196 | Phase 2 |
Enfuvirtide + valproic acid | HIV infections | Terminated | NCT00312546 | Phase 1 |
Valproic acid | HTLV‐I‐associated myelopathy | Terminated | NCT00519181 | N/A |
Karenitecin + valproic acid | Malignant melanoma | Terminated | NCT00358319 | Phase 1, 2 |
Valproic acid | ALPS|hypersplenism|lymphadenopathy | Completed | NCT00605657 | Phase 1, 2 |
Raltegravir + valproic acid | HIV infections | Terminated | NCT00614458 | Phase 2 |
Hydralazine + magnesium valproate | Cervical cancer | Completed | NCT00404326 | Phase 2 |
Hydralazine + magnesium valproate | Locally advanced breast cancer | Terminated | NCT00395655 | Phase 2 |
Hydralazine + magnesium valproate | Refractory solid tumors | Completed | NCT00404508 | Phase 2 |
Sodium phenylbutyrate | Huntington's disease | Completed | NCT00212316 | Phase 2 |
Sodium phenylbutyrate | Amyotrophic lateral sclerosis | Completed | NCT00107770 | Phase 1, 2 |
Pivanex | Leukemia, lymphocytic, lymphoma, small lymphocytic | Terminated | NCT00083473 | Phase 2 |
Pivanex | Malignant melanoma | Terminated | NCT00087477 | Phase 1, 2 |
Pivanex/docetaxel | Carcinoma, non‐small‐cell lung | Completed | NCT00073385 | Phase 2 |
VPA, first used as an anticonvulsant and mood‐stabilizing agent, is a pan‐HDACi. Phase 1–2 clinical trials tested VPA alone or in combination treatment for lymphocytic leukemia (Bouzar et al., 2009; Lagneaux et al., 2007; Stamatopoulos et al., 2009), AML and myelodysplastic syndromes (MDS) in combination with 5‐azacytidine (Kuendgen et al., 2011), melanoma (Daud et al., 2009), HIV infection (Archin et al., 2010), autoimmune lymphoproliferative syndrome (ALPS) (Dowdell et al., 2009), human T‐lymphotropic virus type‐1‐associated myelopathy/tropical spastic paraparesis (HAM/TSP) (Olindo et al., 2011).
Some phase 2 clinical trials involved the combination therapy of hydralazine, a DNA demethylating agent, and magnesium valproate for treatment of cervical cancer (Candelaria et al., 2010), breast cancer (Arce et al., 2006) and refractory solid tumors (Candelaria et al., 2007).
There are also two clinical trials with sodium phenylbutyrate: one in phase 2 for treatment of Huntington's disease (Gardian et al., 2005; Hogarth et al., 2007) and one in phase 1–2 for amyotrophic lateral sclerosis (ALS) (Cudkowicz et al., 2009; Ryu et al., 2005).
Pivaloyloxymethyl butyrate (Pivanex, AN‐9) is in phase 1–2 clinical trials for chronic lymphocytic leukemia and lymphoma (Rabizadeh et al., 2007), malignant melanoma (Tarasenko et al., 2008), and non‐small cell lung carcinoma (NSCLC) (Reid et al., 2004).
3.1.2. Hydroxamic acids
Hydroxamic acids include the majority of HDACi currently in clinical trials for the treatment of several human diseases such as cancer. Peculiarities of hydroxamate‐based molecules are the polar hydroximic group, a 4–6 carbon hydrophobic methylene spacer (CU, polar connection unit), a second polar site, and a terminal hydrophobic group. In the majority of HDACi, the CU can interact with amino acids in the tunnel, and a linker of 4 or 6 units of carbon causing the following zinc‐binding group to bind the zinc ion inhibiting the enzyme (Mai et al., 2005).
The HDACi Vorinostat is at the most advanced stage in the clinical development (2, 3) (Siegel et al., 2009). Vorinostat, a second‐generation polar‐planar compound, binds to the catalytic domain of histone deacetylases (HDACs). This allows the hydroxamic moiety to chelate zinc ion located in the catalytic pockets of HDACs, thereby inhibiting deacetylation and leading to an accumulation of both hyperacetylated histones and transcription factors. Vorinostat was the first HDACi approved by the Federal Drug Administration in 2006 for clinical use in treating patients with advanced cutaneous T‐cell lymphoma (Duvic and Vu, 2007; Marks and Breslow, 2007). 2, 3 show about 60 Vorinostat clinical trials, either alone or in combination, completed or terminated against multiple myeloma (MM) (Badros et al., 2009; Mazumder et al., 2010; Richardson et al., 2008), head and neck cancer (Borbone et al., 2010; Gillenwater et al., 2007), pelvic cancer (Bratland et al., 2011; Ree et al., 2010), lymphoma (Dummer et al., 2012; Kirschbaum et al., 2011; Stathis et al., 2011), leukemia and myelodysplastic syndromes (MDS) (Garcia‐Manero et al., 2008b; Prebet and Vey, 2011), breast cancer (Munster et al., 2011; Shi et al., 2010), small cell lung cancer (SCLC) (Gray et al., 2012), brain and central nervous system tumors (Friday et al., 2012), prostate and urothelial cancers (Schneider et al., 2012), colorectal cancer (Fakih et al., 2012), kidney cancer (Sato et al., 2010), pancreatic cancer (Tinari et al., 2012), and ovarian cancer (Modesitt et al., 2008).
Table 2.
Drug | Conditions | Status | NCT number | Phases |
---|---|---|---|---|
Vorinostat | Glioblastoma, gliosarcoma, brain tumor | Completed | NCT00641706 | Phase 2 |
Vorinostat | Ovarian cancer, primary peritoneal cavity cancer | Completed | NCT00132067 | Phase 2 |
Vorinostat | Lymphoma | Completed | NCT00771472 | Phase 1 |
Vorinostat | Leukemia, myelodysplastic syndromes | Completed | NCT00305773 | Phase 2 |
Vorinostat | Advanced cancer relapsed and refractory | Completed | NCT00632931 | Phase 1 |
Vorinostat | Carcinoma, non‐small‐cell lung | Terminated with result | NCT00251589 | Phase 1, 2 |
Vorinostat | Glioblastoma, gliosarcoma, brain tumor | Completed | NCT01647100 | Phase 2 |
Vorinostat | Mesothelioma|lung cancer | Completed | NCT00128102 | Phase 3 |
Vorinostat | Breast cancer | Completed | NCT00132002 | Phase 2 |
Vorinostat | Lung cancer | Completed | NCT00138203 | Phase 2 |
Vorinostat | Brain and central nervous system tumors | Completed | NCT00238303 | Phase 2 |
Vorinostat | Myelodysplastic syndromes | Completed | NCT00776503 | Phase 1, 2 |
Vorinostat | Cancer | Completed | NCT00045006 | Phase 1 |
Vorinostat | Head and neck cancer | Completed | NCT00134043 | Phase 2 |
Vorinostat | Pelvic cancer|radiotherapy | Completed | NCT00455351 | Phase 1 |
Vorinostat | Hematologic and solid cancer | Terminated | NCT01116154 | Phase 1 |
Vorinostat | Tumors | Completed | NCT00127127 | Phase 1 |
Vorinostat | Brain, CNS, intestine tumors, lymphoma | Completed | NCT00499811 | Phase 1 |
Vorinostat | MDS, blood and bone marrow disease | Terminated | NCT00486720 | Phase 2 |
Vorinostat | Advanced cancer | Completed | NCT00907738 | Phase 2 |
Vorinostat | Lymphoma | Completed | NCT00127140 | Phase 1 |
Vorinostat | Colorectal cancer | Completed | NCT00336141 | Phase 1 |
Vorinostat | Hematologia and solid cancer | Completed | NCT00005634 | Phase 1 |
Vorinostat | Kidney cancer | Completed | NCT00354250 | Phase 2 |
Table 3.
Drug | Conditions | Status | NCT number | Phases |
---|---|---|---|---|
Isotretinoi + vorinostat | Brain, neuroblastoma, CNS, hematologic tumors | Completed | NCT00217412 | Phase 1 |
Idarubicin + vorinostat | Leukemia, myelodysplastic syndromes | Completed | NCT00331513 | Phase 1 |
Vorinostat + gemcitabine + platinum | NSCL cancer | Completed | NCT00423449 | Phase 1 |
Vorinostat + pemetrexed + cisplatin | Advanced cancer | Completed | NCT00106626 | Phase 1 |
MK0683 + vorinostat | Breast cancer, colorectal cancer, NSCL | Terminated | NCT00126451 | Phase 2 |
Vorinostat + docetaxel | NSCL, prostate, bladder, urothelial cancers | Terminated | NCT00565227 | Phase 1 |
Vorinostat + tamoxifen | Breast cancer | Terminated | NCT01194427 | Phase 2 |
MK0683 + vorinostat | B‐cell lymphoma | Completed | NCT00097929 | Phase 2 |
Carboplatin + paclitaxel + vorinostat | Unspecified adult solid tumor, protocol specific | Completed | NCT00287937 | Phase 1 |
Vorinostat, vinorelbine | Malignant solid tumor | Terminated | NCT00801151 | Phase 1 |
Azacitidine + vorinostat | Leukemia, myelodysplastic syndromes | Suspended | NCT00392353 | Phase 1, 2 |
Gemtuzumab ozogamicin + vorinostat | Hematologic cancer | Terminated | NCT00673153 | Phase 2 |
MK0683, vorinostat | Cutaneous T‐cell lymphoma, mycosis fungoides | Completed | NCT00091559 | Phase 2 |
Vorinostat + decitabine | Leukemia, acute myelodysplastic syndromes | Completed | NCT00479232 | Phase 1 |
Fluorouracil, leucovorin calcium, oxaliplatin, vorinostat | Colorectal cancer | Completed | NCT00138177 | Phase 1 |
Carboplatin + paclitaxel + vorinostat | Lung cancer | Completed | NCT00481078 | Phase 2 |
Vorinostat + paclitaxel + carboplatin | Stage IIIB or IV non‐small cell lung cancer | Terminated | NCT00473889 | Phase 2, 3 |
Chemoradiation + vorinostat | Pancreatic cancer | Terminated | NCT00831493 | Phase 1, 2 |
Vorinostat – bexarotene | Lymphoma | Terminated with with result | NCT00127101 | Phase 1 |
Vorinostat + tamoxifen | Breast cancer | Completed with result | NCT00365599 | Phase 2 |
Gemcitabine + vorinostat | Unspecified adult solid tumor, protocol specific | Completed | NCT00243100 | Phase 1 |
Vorinostat + alvocidib | Unspecified adult solid tumor, protocol specific | Completed | NCT01645514 | Phase 1 |
Vorinostat + alvocidib | Leukemia, myelodysplastic syndromes | Completed | NCT00278330 | Phase 1 |
Topotecan + vorinostat | Small cell lung cancer | Terminated | NCT00697476 | Phase 1, 2 |
Vorinostat + alvocidib | Unspecified adult solid tumor, protocol specific | Completed | NCT00324480 | Phase 1 |
Fluorouracil + leucovorin calcium + vorinostat | Hematologic and solid cancer | Completed | NCT00942266 | Phase 2 |
MK0683, vorinostat | Neoplasms | Terminated | NCT00424775 | Phase 1 |
MK0683, vorinostat | Cancer, advanced | Completed | NCT00750178 | Phase 1 |
Vorinostat + bortezomib + dexamethasone | Relapsed or refractory multiple myeloma | Completed | NCT00773838 | Phase 2 |
Vorinostat + bortezomib | Lymphoma, sarcoma, Wilms tumor, neuroblastoma | Completed | NCT01132911 | Phase 1 |
Vorinostat + bortezomib | Unspecified adult solid tumor, protocol specific | Completed | NCT00227513 | Phase 1 |
Vorinostat + bortezomib | Multiple myeloma | Completed | NCT00111813 | Phase 1 |
Bortezomib + dexamethasone + vorinostat | Multiple myeloma and plasma cell neoplasm | Completed | NCT00310024 | Phase 1 |
Vorinostat + bortezomib | Multiple myeloma | Completed | NCT00858234 | Phase 1 |
Vorinostat + bortezomib | Leukemia, myelodysplastic syndromes | Terminated | NCT00818649 | Phase 2 |
Vorinostat + bortezomib | Lymphoma | Terminated | NCT00810576 | Phase 2 |
Dasatinib + vorinostat | Leukemia | Completed | NCT00816283 | Phase 1 |
Panobinostat (LBH589) is currently in phase 1 and 2 clinical trials as monotherapy and combination therapy for hematologic tumors such as non‐Hodgkin's lymphoma (Ellis et al., 2008; Shao et al., 2010), acute lymphoblastic or acute myeloid leukemia (Fiskus et al., 2009; George et al., 2005; Rosato et al., 2012; Scuto et al., 2008), MM (Maiso et al., 2006), for advanced solid tumors (Fukutomi et al., 2012), breast cancer (Tate et al., 2012; Zhou et al., 2007) and lung cancer (Crisanti et al., 2009) (Table 4).
Table 4.
Drug | Conditions | Recruitment | NCT number | Phases |
---|---|---|---|---|
Panobinostat (LBH589) | Breast cancer | Terminated | NCT00993642 | Phase 0 |
Panobinostat (LBH589) | Cutaneous T‐cell lymphoma | Completed | NCT00490776 | Phase 2, 3 |
Panobinostat (LBH589) | Non‐Hodgkin's lymphoma | Terminated | NCT01090973 | Phase 2 |
Panobinostat (LBH589) | Leukemia | Completed | NCT00723203 | Phase 2 |
Panobinostat (LBH589) | Cancer, advanced solid tumor | Completed | NCT00739414 | Phase 1 |
Panobinostat (LBH589) | Lymphoma, leukemia, multiple myeloma | Completed | NCT00621244 | Phase 1, 2 |
LBH589 + trastuzumab + paclitaxel | HER‐2 positive breast cancer, metastatic breast cancer | Completed | NCT00788931 | Phase 1 |
Panobinostat + trastuzumab | Breast cancer | Completed | NCT00567879 | Phase 1 |
Panobinostat (LBH589) | Cancer | Completed | NCT00570284 | Phase 1 |
Panobinostat (LBH589) | Cutaneous T‐cell lymphoma, leukemia–lymphoma | Terminated | NCT00699296 | Phase 2 |
Panobinostat (LBH589) | Tumors, cutaneous T‐cell lymphoma | Completed | NCT00412997 | Phase 1 |
Panobinostat (LBH589) | Recurrent malignant gliomas | Terminated | NCT00848523 | Phase 2 |
Panobinostat (LBH589) | Non‐Hodgkin lymphoma, neoplasms | Completed | NCT00503451 | Phase 1 |
Panobinostat (LBH589) | Small cell lung carcinoma | Completed | NCT01222936 | Phase 2 |
CHR‐3996 | Solid tumor | Completed | NCT00697879 | Phase 1 |
CHR‐2845 | Hematological disease, lymphoid malignancies | Completed | NCT00820508 | Phase 1 |
ITF2357 (givinostat) | Hodgkin's lymphoma | Terminated | NCT00496431 | Phase 1, 2 |
ITF2357 (givinostat) | Myeloproliferative diseases | Completed | NCT00606307 | Phase 2 |
ITF2357 (givinostat) | Chronic lymphocytic leukemia | Terminated | NCT00792831 | Phase 2 |
ITF2357 (givinostat) | Hodgkin's lymphoma | Completed | NCT00792467 | Phase 1, 2 |
ITF2357 (givinostat) | Polycythemia vera | Completed | NCT00928707 | Phase 2 |
Belinostat or PXD101 | De novo myelodysplastic syndromes | Completed | NCT01647009 | Phase 2 |
Belinostat | Myelodysplastic syndromes | Completed | NCT00357162 | Phase 2 |
PXD101|dexamethasone | Multiple myeloma | Completed | NCT00131261 | Phase 2 |
Belinostat, carboplatin, paclitaxel and bevacizumab | Non‐small‐cell lung carcinoma | Terminated | NCT01090830 | Phase 1, 2 |
Belinostat + erlotinib | Non small cell lung cancer | Terminated | NCT01188707 | Phase 1, 2 |
JNJ‐26481585 | Hematologic cancer | Terminated | NCT00676728 | Phase 1 |
SB939 | Solid tumor | Completed | NCT00504296 | Phase 1 |
SB939 + azacitidine | Solid tumors and hematologic malignancies | Completed | NCT00741234 | Phase 1 |
CHR‐3996 is a second‐generation hydroxamic acid‐based HDACi. Specifically, it is a selective class I HDAC inhibitor with potential antineoplastic activity (Moffat et al., 2010), currently in clinical trials for the treatment of advanced tumors (Banerji et al., 2012) (Table 4).
CHR‐2845 is a novel hydroxamic acid derivative HDACi, which is a selective substrate for the intracellular carboxylesterase hCE‐1, whose expression is restricted to cells of the monocyte–macrophage lineage. One phase 1 clinical trial has been completed for this drug in treatment of hematological diseases and lymphoid malignancies (Table 4).
SB939 is a new hydroxamic acid‐based HDACi with improved physico‐chemical, pharmaceutical and pharmacokinetic properties. In vitro, SB939 inhibits class I, II, and IV HDACs, with no effects on other zinc‐binding enzymes, and shows significant antiproliferative activity against a wide variety of tumor cell lines (Novotny‐Diermayr et al., 2010). It is in phase 1 clinical trials for treatment of solid tumors (Razak et al., 2011), hematologic malignancies and MDS (Novotny‐Diermayr et al., 2012) (Table 4).
ITF2357 (Givinostat) is a novel hydroxamic acid‐based HDACi that inhibits both class I and II HDACs. It has been used in five phase 1–2 clinical trials for the treatment of hematological diseases (Galli et al., 2010; Rambaldi et al., 2010) (Table 4).
PXD101 (Belinostat) is another inhibitor of class I and II HDACs that has been included in phase 2 clinical trials for MDS (Cashen et al., 2012), MM (Feng et al., 2007; Gimsing et al., 2008) and phase 1–2 for NSLC (Force et al., 2011; Luchenko et al., 2011) (Table 4).
JHJ‐26481585 is a pyrimidyl‐hydroxamic acid analog showing a pan‐HDACi activity and is in phase 1 for leukemia and MDS (Arts et al., 2009; Stuhmer et al., 2010; Tong et al., 2010) (Table 4).
3.1.3. Benzamides
This class is composed of HDACi containing a characteristic 2′‐aminoanilide moiety able to contact specific aminoacids in the tube‐like active site of the HDAC core, with or without coordination/chelation of zinc ion (Pontiki and Hadjipavlou‐Litina, 2012).
At least two benzamides, MS‐275 (SNDX‐275, Entinostat) and MGCD0103 (Mocetinostat) are in clinical development. These agents are in trials as single agents and in combination with other drugs.
Clinical trials with MS‐275, a class I selective inhibitor, include patients with a wide variety of hematologic and solid neoplasms (Knipstein and Gore, 2011) such as leukemia (Gojo et al., 2007; Lucas et al., 2004), melanoma (Gore et al., 2008), MDS (Fandy et al., 2009) and colorectal cancer (Bracker et al., 2009) (Table 5).
Table 5.
Drug | Condition | Status | NCT number | Phases |
---|---|---|---|---|
Entinostat | Melanoma | Completed | NCT00185302 | Phase 2 |
Entinostat | Cancer | Completed | NCT00020579 | Phase 1 |
Entinostat | Leukemia, multiple myeloma and plasma cell neoplasm, MDS | Completed | NCT00015925 | Phase 1 |
Entinostat + isotretinoin | Lymphoma, small intestine cancer | Completed | NCT00098891 | Phase 1 |
Azacitidine + entinostat | Leukemia, myelodysplastic syndromes | Completed | NCT00101179 | Phase 1 |
Azacitidine + entinostat | Colorectal cancer | Completed | NCT01105377 | Phase 2 |
MGCD0103 + docetaxel | Solid cancer | Terminated | NCT00511576 | Phase 1 |
Azacitidine + MGCD0103 | Acute myeloid leukemia (AML)|myelodysplastic syndrome (MDS) | Terminated | NCT00666497 | Phase 2 |
MGCD0103 | Hodgkin's lymphoma | Terminated | NCT00358982 | Phase 2 |
MGCD0103 | Myelodysplastic syndrome, acute myelogenous leukemia | Completed | NCT00324220 | Phase 1, 2 |
MGCD0103 | Leukemia, myelodysplastic syndromes | Completed | NCT00324129 | Phase 1 |
MGCD0103 | Tumors, non‐Hodgkin's lymphoma | Completed | NCT00323934 | Phase 1 |
MGCD0103 | Lymphocytic leukemia, chronic | Completed | NCT00431873 | Phase 2 |
MGCD0103 | Lymphoma | Completed | NCT00359086 | Phase 2 |
MGCD0103 | Leukemia, myelodysplastic syndromes | Completed | NCT00324194 | Phase 1 |
MGCD0103 | Myelogenous leukemia, acute, myelodysplastic syndromes | Terminated | NCT00374296 | Phase 2 |
MGCD0103 + gemcitabine | Tumors | Completed | NCT00372437 | Phase 1, 2 |
MGCD0103 + azacitidine | Hodgkin lymphoma | Terminated | NCT00543582 | Phase 2 |
MGCD0103 is an isotype‐selective HDACi that potently targets human HDAC1 but also exerts inhibitory activity against HDAC2, HDAC3, and HDAC11 in vitro (Fournel et al., 2008). Some phase 1–2 clinical trials are for treatment of hematological diseases such as leukemia (Blum et al., 2009; El‐Khoury et al., 2010; Garcia‐Manero et al., 2008a), lymphoma (Buglio et al., 2010; Younes et al., 2011) and also for solid cancers (Siu et al., 2008) (Table 5).
3.1.4. Cyclic peptides
Romidepsin (Depsipeptide or FK228) acts as an HDACi with the disulfide bond undergoing reduction within the cell to release a zinc‐binding thiol. Thiol interacts with a zinc atom in the binding pocket of zinc‐dependent HDAC to block its activity. Romidepsin is a natural product obtained from the bacteria Chromobacterium violaceum. On November 5, 2009, it was approved by the FDA for the treatment of cutaneous T‐cell lymphoma (CTCL). Terminated clinical trials show the activity of this compound in treating lymphoma (Coiffier et al., 2012), MM (Khan et al., 2004; Kikuchi et al., 2010; Niesvizky et al., 2011), CTCL (Piekarz et al., 2011; Prince and Dickinson, 2012), MDS (Klimek et al., 2008), and solid tumors such as pancreatic (Sato et al., 2004), colorectal (Whitehead et al., 2009), lung (Otterson et al., 2010; Schrump et al., 2008), prostate (Lai et al., 2008), renal (Stadler et al., 2006), bladder (Karam et al., 2007), brain (Iwamoto et al., 2011), thyroid (Kitazono et al., 2001) and ovarian cancers (Son et al., 2010) (Table 6).
Table 6.
Drug | Conditions | Recruitment | NCT number | Phases |
---|---|---|---|---|
Romidepsin + bortezomib | Multiple myeloma | Terminated | NCT00765102 | Phase 2 |
Romidepsin + ketoconazole | Hematologic cancer | Completed | NCT01324310 | Phase 1 |
Romidepsin (FR901228) | Mycosis fungoides, CTCL, neoplasms | Terminated | NCT01445340 | Phase 1 |
Romidepsin | Peritoneal, epithelial, ovarian cancer | Completed | NCT01645670 | Phase 2 |
Romidepsin + rifampin | Hematologic cancer | Completed | NCT01324323 | Phase 1 |
Romidepsin (FR901228) | Lymphoma | Completed | NCT00383565 | Phase 2 |
Romidepsin + gemcitabine | Pancreatic cancer | Completed | NCT00379639 | Phase 1, 2 |
Romidepsin (depsipeptide, FK228) | Cutaneous T‐cell lymphoma | Completed | NCT00106431 | Phase 2 |
Depsipeptide, FR901228, FK228 | Neoplasms | Completed | NCT00048334 | Phase 1 |
Romidepsin + rituximab + fludarabine | Lymphoma | Completed | NCT00079443 | Phase 2 |
Romidepsin | Brain and nervous system tumors | Completed | NCT00085540 | Phase 2 |
Romidepsin | Lung cancer | Completed | NCT00086827 | Phase 2 |
Romidepsin | Colorectal cancer | Completed | NCT00077337 | Phase 2 |
Romidepsin | Bladder, renal pelvis and urethral cancer | Terminated | NCT00087295 | Phase 2 |
Romidepsin | Leukemia, lymphoma | Completed | NCT00024180 | Phase 1 |
Romidepsin | Lymphoma | Completed | NCT00019318 | Phase 1 |
Romidepsin | Leukemia, lymphoma, MDS syndrome | Completed | NCT00042822 | Phase 2 |
Romidepsin + decitabine | Leukemia, lymphoma, MDS syndrome | Completed | NCT00114257 | Phase 1 |
Romidepsin | Breast cancer | Completed | NCT00098397 | Phase 2 |
Romidepsin | Esophageal cancer, gastric cancer | Suspended | NCT00098527 | Phase 2 |
Romidepsin | Ovarian, primary peritoneal cavity cancer | Completed | NCT00091195 | Phase 2 |
Romidepsin | Lymphoma | Completed | NCT00077194 | Phase 2 |
Romidepsin | Head and neck cancer | Completed | NCT00098813 | Phase 2 |
Decitabine + depsipeptide | Small cell lung carcinoma | Completed | NCT00037817 | Phase 1 |
Romidepsin | Nervous system tumors, leukemia | Completed | NCT00053963 | Phase 1 |
Romidepsin (depsipeptide, FK228) | Prostate cancer, metastases | Completed | NCT00106418 | Phase 2 |
Romidepsin (FR901228) | Small cell lung carcinoma | Completed | NCT00020202 | Phase 2 |
Romidepsin (depsipeptide, FK228) | Renal cell carcinoma, metastases | Completed | NCT00106613 | Phase 2 |
3.2. Sirtuins
Class III HDACs, also known as sirtuins, are the silent information regulator 2 (Sir2) family of proteins. Distinct from other HDACs, sirtuins are NAD+‐dependent protein deacetylases, not modulated by HDACi. The mammalian sirtuin family comprises seven proteins (SirT1‐7), which differ widely in their cellular localization, activity and functions, and are subdivided into 4 classes (Carafa et al., 2012). The deacetylase activity of sirtuins is controlled by the cellular [NAD+]/[NADH] ratio; NAD+ works as an activator, whereas nicotinamide and NADH inhibit their activity. Two reactions are catalyzed by sirtuins: deacetylation and ADP‐ribosylation. In both, the cleavage of NAD+ is the initial chemical step. SirT1, 2, 3, 5 and 7 catalyze a deacetylation reaction, in which they deacetylate lysine residues of target proteins using NAD+ as cofactor and releasing nicotinamide with the production of 2′‐O‐acetyl‐ADP ribose. In contrast, SirT4 and 6, catalyze ADP‐ribosylation reaction, during which ADP‐ribosyl moiety is transferred to the protein substrate (Yamamoto et al., 2007).
Expressed from bacteria to humans (Vaquero, 2009), sirtuins seem to preferentially target non‐histone proteins, though little is known about target specificity and selectivity. Sirtuins are connected to chromatin regulation as they are responsible for controlling two post‐translational histone modifications crucial for chromatin structure: H4K16ac and H3K9ac. SirT1, 2, 3 and 6 are involved in chromatin regulation (McGuinness et al., 2011). SirT1, the best‐studied family member, is responsible for heterochromatin formation by a deacetylating process.
In the last decade interest for sirtuins has grown, mainly due to their critical role in several biological processes, such as regulation of gene expression, control of metabolic processes, apoptosis and cell survival, DNA repair, development, neuroprotection and inflammation. Sirtuins control many vital functions and are involved in many disorders such as metabolic diseases, neurodegenerative diseases and cancer (Stunkel and Campbell, 2011). SirT1 displays contradictory roles, and has been suggested either as tumor suppressor or tumor promoter (Bosch‐Presegue and Vaquero, 2011; Deng, 2009). The initial evidence of SirT1 as a tumor promoter derived from its repressive effect on the tumor suppressor p53. However, a potential tumor suppressor role has also been proposed for other human sirtuins (McGuinness et al., 2011). This hypothesis is mainly sustained by reduction of SirT2 in a large number of human brain tumor cell lines, and its involvement in cell cycle progression. SirT3 is the only mitochondrial sirtuin implicated in tumorigenesis. Its reduction in several cancers leads to an increase in reactive oxygen species (ROS) production, which results in enhanced tumor growth. SirT5 overexpression has been implicated in a study of pancreatic cancer (Kim et al., 2010). Recently, the role of SirT6 and SirT7 in tumorigenesis has been demonstrated. SirT6 controls NF‐kB pathway and plays a role in DNA double‐strand repair, indicating that this sirtuin has a key function in tumorigenesis. However, very little is known about the specific correlation with cancer. mRNA levels of SirT7 have been inversely correlated with the ability to undergo tumorigenesis in mouse cell lines, and levels of SirT7 have been found elevated in some forms of breast cancer (Ashraf et al., 2006).
Sirtuins are involved in aging diseases such as metabolic disease, neurodegeneration and aging itself. It is well known that overexpression of Sir2 (or its orthologs) can extend organism lifespan in a wide range of lower eukaryotes (Bosch‐Presegue and Vaquero, 2011; Vaquero, 2009). Sir2 functions are often correlated to calorie restriction (CR). The link between the role of sirtuins, CR and longevity was first described in Saccharomyces cerevisiae. In yeast, CR leads to increased replicative lifespan. Lifespan extension was not observed in yeast lacking the Sir2 gene. Currently, the role of sirtuins in the regulation of mammalian lifespan is not clear. However, starting from the premise that sirtuins are an evolutionary conserved protein family, it is fair to assume that they play a role in the modulation of aging‐related processes in higher organisms (Brooks and Gu, 2009; Westphal et al., 2007). In humans, the aging process is associated to telomere erosion. SirT1 and 6 are involved in the maintenance of telomeres and telomeric function, and are implicated in the aging process. Recent studies have demonstrated that reduction or removal of SirT6 results in telomere dysfunction and end‐to‐end chromosomal fusions. In terms of symptoms, the absence of SirT6 is similar to a disease characterized by premature aging, Werner's syndrome. Although very little is known about other sirtuins, no evidence suggests their involvement with telomere function, formation and stability (McGuinness et al., 2011).
Given that SirT1 has been reported to increase tumorigenesis, and despite its role as tumor suppressor or promoter, it is important to identify small chemical compounds that inhibit or activate SirT1. To date, a number of specific inhibitors of SirT1 have been proposed for cancer therapy (Alcain and Villalba, 2009). Moreover, both activators and inhibitors of sirtuins might act beneficially against different types of neurodegenerations. Thus, in addition to nicotinamide, the physiological inhibitor, some specific inhibitors have been characterized, including splitomicin and its analogs, tenovins, AGK2, sirtinol, suramin, the indole derivative EX‐257, salermide and UVI5008.
SIRT activators are being studied in the hope of providing benefit to patients with neurodegenerative, inflammatory, metabolic and autoimmune diseases, and some tumor types.
Phenol derivates, including quercetin, piceatannol, and resveratrol, have been shown to posses SirT1‐activating properties (Alcain and Villalba, 2009). The most potent SirT activator, and the first to be characterized, is resveratrol, a polyphenol found in grapes, and grape products. Subsequently, much more potent and efficacious SirT1 activators were reported as potential therapeutics for treatment of metabolic diseases (SRT1720, SRT2183, and SRT1460) (Saunders and Verdin, 2007). However, their activity is still debated. The question is whether they are SIRT activators or assay artifacts. Given that the activation of SirT1 by activators requires the use of fluorescently labeled substrate in a fluorescence assay, it was demonstrated that they do not activate SirT1 when using native peptide or protein substrate conjugated with a non‐physiological fluorophore (Borra et al., 2005). To establish their effective modulation of SirT1, other technical approaches are necessary. Despite these issues, resveratrol is now in clinical trials (Table 7). Resveratrol can exhibit benefits against cardiovascular diseases (CVDs) or in its prevention, although its cardioprotective role as part of the human diet is not yet clear (Ruana et al., 2012). CR is a low calorie diet (about 30% fewer calories than the American Dietetic Association (ADA) recommends). CR has also been linked to health benefits (enhanced cardiovascular and metabolic health) and an extended lifespan. Many studies have compared the health benefits of both resveratrol and CR to determine whether resveratrol mimics some of the health benefits shown with CR. The consumption of this polyphenol could modulate cerebral blood flow and this in turn could influence cognitive performance by increasing access to blood‐borne metabolic fuel. Research shows that resveratrol is able to induce vasodilation (and therefore blood flow) by interacting with nitric oxide (NO). Because tumors develop resistance to chemotherapeutic agents, the aim of cancer research is to discover effective chemosensitizers. One promising possibility is to use dietary agents, such as resveratrol, that sensitize tumors to chemotherapeutics (Aggarwal et al., 2004). Through its ability to modulate multiple cell‐signaling molecules such as cell survival proteins, members of NF‐kB and STAT3 pathways, resveratrol is able to prevent cancer (Gupta et al., 2011). Tumors shown to be sensitized by resveratrol include lung and breast cancer, AML, promyelocitic leukemia, MM, prostate, pancreatic and epidermoid cancers (Fulda and Debatin, 2004). Patients with colon cancer received treatment with resveratrol, and correlative laboratory studies examined its effects directly on colon cancer and normal colonic mucosa. Resveratrol may stop the growth of tumor cells by blocking some of the enzymes required for cell growth, and by inducing cell cycle arrest, apoptosis, inhibition of cell proliferation, stimulation of antiangiogenic responses and increased antioxidant and anti‐inflammatory activity (Talero et al., 2012). The activity of resveratrol shows great potential in the prevention and therapy of a wide variety of human diseases.
Table 7.
Drug | Conditions | Status | NCT number | Phases |
---|---|---|---|---|
Resveratrol and midazolam | Healthy | Completed | NCT01173640 | |
Resveratrol | Cognitive and cerebral blood flow | Completed | NCT01010009 | |
Resveratrol | Cardiovascular diseases | Completed | NCT01449110 | Phase 2 |
Resveratrol | Colon cancer | Completed | NCT00256334 | Phase 1 |
Resveratrol | Chronic subclinic inflammation | Completed | NCT01492114 | Phase 3 |
Resveratrol | Type 2 diabetes mellitus | Completed | NCT01038089 | |
Resveratrol | Healthy | Completed | NCT01331382 | |
Resveratrol | Sports concussion | Completed | NCT01321151 | Phase 1 |
Resveratrol | Neoplasms, colorectal cancer | Completed | NCT00920803 | Phase 1 |
Resveratrol | Sedentary lifestyle | Completed | NCT01615445 | Phase 2 |
Resveratrol | Melanoma (skin) | Completed | NCT00721877 | Phase 1 |
Resveratrol | Metabolic syndrome, obesity | Completed | NCT01150955 | |
Resveratrol | Multiple myeloma | Terminated | NCT00920556 | Phase 2 |
Resveratrol | Healthy | Completed | NCT01640197 | Phase 1 |
Resveratrol | Colorectal cancer | Completed | NCT00433576 | Phase 1 |
Resveratrol | Unspecified adult solid tumor | Completed | NCT00098969 | Phase 1 |
Resveratrol | Obesity, metabolic syndrome, diabetes | Completed | NCT00823381 | |
Resveratrol | Obesity | Completed | NCT00998504 | |
Resveratrol | Cardiovascular disease | Completed | NCT01199549 | |
Resveratrol | Cellulite (orange peel skin) | Terminated | NCT01321268 | |
Resveratrol | Type 2 diabetes | Completed | NCT00937222 |
3.3. HAT inhibitors
Of the several known covalent histone modifications, the reversible acetylation of key lysine residues in histones holds a pivotal position in transcriptional regulation. Histone acetylation is a distinctive feature of transcriptionally active genes, whereas deacetylation indicates the repressed state of a gene. The balance between the acetylation and deacetylation states of histones regulates transcription. Dysfunction of enzymes involved in these events is often associated with the manifestation of several diseases, including cancer, cardiac hypertrophy and asthma (Kramer et al., 2001; McKinsey and Olson, 2004; Yang, 2004). These enzymes are therefore potential new targets for therapy. Acetyl transferases (HATs) modulate gene expression by catalyzing targeted acetylation of the ϵ‐amino group of lysine residues on histone and non‐histone proteins. HATs can be classified into several families on the basis of number of highly conserved structural motifs. These include the GNAT family (Gcn5‐related N‐acetyltransferase, e.g., Gcn5, PCAF), the MYST group (MOZ, YBF2/SAS3 and TIP60) and the p300/CBP family (Kramer et al., 2001; Sterner and Berger, 2000; Yang, 2004).
Although a wide number of transcriptional co‐activator proteins are now recognized to possess HAT activity, very few HAT inhibitors (HATi) have been identified to date. Availability of recombinant HATs made it possible to synthesize and test more target‐specific inhibitors: Lys‐CoA for p300 and H3‐CoA‐20 for PCAF (Lau et al., 2000). Though Lys‐CoA has been extensively employed for in vitro transcription studies, it is unable to permeate cells (Cebrat et al., 2003). In the early 2000s, two important HATis were isolated: anacardic acid from cashew nut shell liquid and garcinol from Garcinia indica, which are both non‐specific inhibitors of p300/CBP and PCAF but are capable of easily permeating cells in culture (Balasubramanyam et al., 2003, 2004). Different chemical modifications of these inhibitors were attempted to identify enzyme‐specific inhibitors, but it serendipitously led to the synthesis of the only known p300‐specific activator, N‐(4‐chloro‐3‐trifluoromethyl‐phenyl)‐2‐ethoxy‐6‐pentadecyl‐benzamide (CTPB) (Balasubramanyam et al., 2003) (Souto et al., 2010, 2008). None of the above is in clinical trials.
Curcumin (diferuloylmethane) is the major curcuminoid of turmeric, Curcuma longa, a characteristically orange‐yellow colored spice often found in curry powder. In recent years, considerable interest has been focused on this substance due to its use in treating a wide variety of disorders without any side effects. It was used in ancient times to treat various illnesses such as rheumatism, body ache, skin diseases, intestinal worms, diarrhea, intermittent fevers, hepatic disorders, biliousness, urinary discharges, dyspepsia, inflammation, constipation, leukoderma, amenorrhea, and colic. Curcumin has the potential to treat a wide variety of inflammatory diseases including cancer, diabetes, cardiovascular diseases, arthritis, Alzheimer's disease and psoriasis through modulation of numerous molecular targets. Curcumin was identified as the first p300/CBP‐specific cell permeable HATi (Balasubramanyam et al., 2004b). It does not affect the HAT activity of PCAF or histone deacetylase and methyltransferase activities. However, p300 HAT activity‐dependent chromatin transcription is efficiently repressed by curcumin but not transcription from DNA template. Curcumin could also inhibit histone acetylation in vivo. It is the only HATi in clinical trials (Table 8) and exhibits great promise as a therapeutic agent. Its applications include atopic asthma (Kobayashi et al., 1997), chronic obstructive pulmonary disease (Rennolds et al., 2012), multiple myeloma (Ghoneum and Gollapudi, 2011), irritable bowel syndrome (Binion et al., 2008; Rapin and Wiernsperger, 2010), ulcerative colitis (Baliga et al., 2012), Crohn's disease (Mouzaoui et al., 2012), breast cancer (Nagaraju et al., 2012), Alzheimer's disease (Darvesh et al., 2012; Huang et al., 2012), pancreatic cancer (Dandawate et al., 2012; Veeraraghavan et al., 2011), colorectal cancer (Guo et al., 2012; Lin et al., 2011), diabetes (Abdel Aziz et al., 2012), and psoriasis (Kurd et al., 2008).
Table 8.
Drugs | Conditions | Recruitment | NCT number | Phases |
---|---|---|---|---|
Curcumin | Atopic asthma | Completed | NCT01179256 | N/A |
Curcumin + bioperine | Multiple myeloma | Completed | NCT00113841 | N/A |
Curcumin | Irritable bowel syndrome | Completed | NCT00779493 | Phase 4 |
Curcumin | Healthy | Completed | NCT00181662 | N/A |
Curcumin | Healthy | Completed | NCT00895167 | Phase 1 |
Curcumin | Breast cancer | Completed | NCT01042938 | Phase 2 |
Curcumin | Alzheimer's disease | Completed | NCT00164749 | Phase 1 and 2 |
Curcumin | Inflammatory bowel disease|ulcerative colitis|Crohn's disease | Completed | NCT00889161 | Phase 1 |
Curcumin + bioperine | Chronic obstructive pulmonary disease | Completed | NCT01514266 | N/A |
Curcumol | Chemotherapy induced mucositis | Completed | NCT00475683 | Phase 3 |
Curcumin + bioperine | Mild cognitive impairment | Completed | NCT00595582 | N/A |
Curcumin C3 complex | Alzheimer's disease | Completed | NCT00099710 | Phase 2 |
Curcumin + fluoxetine | Major depressive disorder | Completed | NCT01022632 | N/A |
Curcumin + gemcitabine | Pancreatic cancer | Completed | NCT00192842 | Phase 2 |
Curcumin | Familial adenomatous polyposis | Terminated | NCT00248053 | Phase 2 |
Curcumin | Colorectal cancer | Completed | NCT00027495 | Phase 1 |
Curcumin | Aberrant crypt foci | Terminated | NCT00176618 | N/A |
Curcumin | Healthy, no evidence of disease | Completed | NCT00768118 | N/A |
Curcumin + quercitin + sulindac | Colorectal cancer | Terminated | NCT00003365 | N/A |
Observational study | Diabetes | Completed | NCT01029327 | N/A |
Curcumin C3 complex | Psoriasis | Completed | NCT00235625 | Phase 2 |
Curcuminoids | Oral lichen planus | Completed | NCT00525421 | Phase 2 |
Standardized turmeric root extract | Cystic fibrosis | Completed | NCT00219882 | Phase 1 |
Carbohydrate drink | Nutrition processes | Completed | NCT00799630 | N/A |
3.4. Histone methyltransferases
Histone methylation has been shown to play a key function in the regulation of gene‐expression patterns and DNA repair; this kind of post‐transcriptional (epigenetically controlled) modification can affect lysine (K) or arginine (R) residues of histone tails (Greer and Shi, 2012). In contrast to histone acetylation, histone methylation does not alter the charge of the histone tail, but influences the basicity, hydrophobicity of histones and their affinity to certain proteins such as transcription factors (Rice and Allis, 2001). A methyl group is relatively small and its addition to lysine or arginine residues does not neutralize their charge, and it is therefore unlikely that methylation alone will significantly affect the chromatin structure (Bannister and Kouzarides, 2011). Lysine side chains may be mono‐, di‐ or tri‐methylated, whereas the arginine side chain may be mono‐methylated or (symmetrically or asymmetrically) di‐methylated (Smith and Denu, 2009).
On the basis of target residue for methylation, histone methyltransferases (HMTase) can be grouped into two different enzymatic classes: lysine methyltransferases and arginine methyltransferases.
Arginine methylation of histones H3 (Arg2, 17, 26) and H4 (Arg3) promotes transcriptional activation and is mediated by the family of protein arginine methyltransferases (PRMTs), including the co‐activators PRMT1 and CARM1 (PRMT4). In contrast, a more diverse set of histone lysine methyltransferases has been identified, all but one of which contain a conserved catalytic SET domain originally identified in the Drosophila Su[var]3–9, enhancer of zeste, and Trithorax proteins. Lysine methylation has been implicated in both transcriptional activation (H3 Lys4, 36, 79) and silencing (H3 Lys9, 27, H4 Lys20).
At present, there are many well‐known methylation sites on histones. Taking into consideration all three possible methylation states of lysine and arginine, an enormous number of methylation states of histones exist. This might explain the difficulty in studying the methylation pattern on histones.
Conversely, due to the complex pattern of histone methylation, it may be possible to interfere with this enzyme in a promising manner.
Although attempts to interfere with DNA methylation (e.g., by DNMTIs) and histone deacetylation (e.g., by HDACIs) have received the bulk of attention, recent efforts have begun to focus on pharmacologic disruption of other epigenetic regulatory processes. Histone methylation represents one such target, but to date studies on specific HMTi classes are far from the clinic.
3.4.1. Histone methyltransferase inhibitors
The first applied inhibitor used as an anticancer drug was S‐adenosylmethionine (SAM) and its analogs (e.g., SAH). However, these compounds target not only HMTs but also other enzymatic classes using AdoMet as methyl‐donor (such as DNMTs). Their use is therefore limited by low specificity (Spannhoff et al., 2009a).
As a lysine methyltransferase, chaetocin, a fungal mycotoxin, has been reported to act against G9a at low concentration without inhibition of other KMT enzymes (such as EZH2 or SET7/9) (Copeland et al., 2009; Greiner et al., 2005; Spannhoff et al., 2009b). Inhibition mediated by chaetocin is competitive against the co‐substrate SAM. Chaetocin potently induces cellular oxidative stress, selectively killing cancer cells and rapidly proliferating primary cells (Isham et al., 2007). The effects of chaetocin on oxidative stress are at least in part due to its capacity to act as a competitive and selective substrate for theoredoxin reductase‐1 (K m = 4.6 μM) (Tibodeau et al., 2009).
Another specific inhibitor of G9a is BIX‐01294 (and its derivative BIX‐01338, both hydrochloride hydrates), which is effective in vitro at a concentration of 2.7 μM with no effects on SUV39H1 and PRMT1 (Kubicek et al., 2007; Shi et al., 2008). Cell lines treated with BIX‐01294 showed a reduction in histone H3 lysine 9 (H3K9) dimethylation, while the mono‐ or tri‐methyl levels were unaffected. Other lysine methylation sites such as H3K27 or H4K20 were not altered. Based on a kinetic inhibition model, BIX‐01294 showed an uncompetitive pattern compared to the co‐substrate SAM, suggesting that it only binds to G9a complexed with SAM. BIX01294 has been used in combination with the calcium channel activator BayK8644 to facilitate the generation of induced pluripotent stem cells from somatic cells in vitro.
UNC0224 is another potent and relatively selective G9a HMTi, exhibiting an IC50 value of 15 nM. Isothermal titration calorimetry revealed that UNC0224 binds to G9a with a K d value of 29 nM. UNC0224 also inhibits G9a‐like protein (GLP), a closely related H3K9 HMTase, with assay‐dependent IC50 values of 20–58 nM, but is over 1000‐fold selective against SET7/9 (a H3K4 HMTase) and SET8 (a H4K20 HMTase) (Liu et al., 2009).
The lysine methyltransferase EZH2 (KMT6), part of the polycomb repressive complex 2, catalyzes trimethylation of lysine 27 on histone H3 and is involved in proliferation and aggressive cell growth associated with neoplastic cells. Given its importance in cell proliferation and homeostasis maintenance, intense efforts have been directed toward discovering specific EZH2 inhibitors (Simon and Lange, 2008). 3‐Deazaneplanocin A (DZnep) is a cyclopentenyl analog of 3‐deazaadenosine, originally synthesized as an inhibitor of S‐adenosyl‐l‐homocysteine hydrolase (Tseng et al., 1989). It has been shown to deplete EZH2 levels and to inhibit trimethylation of lysine 27 on histone H3 in cultured human acute myeloid leukemia (AML) HL‐60 and OCI‐AML3 cells and in primary AML cells in a dose‐dependent manner (0.2–1 μM) (Fiskus et al., 2009). DZnep treatment of cultured human AML cells induces increased expression of the cell‐cycle regulators p21, p27, and FBXO32, leading to cell cycle arrest and apoptosis. When used in combination with the pan‐HDACi Panobinostat (10 mg/kg), the antileukemic effects of DZnep (1 mg/kg) are synergistically enhanced in mice implanted with AML cells.
The arginine methyltransferase enzyme AMI‐1 (and subsequently its derivatives AMI‐2 to AMI‐6) is the first and to date best‐known inhibitor. It is cell‐permeable, symmetrical sulfonated urea compound that acts as a potent, specific and non‐AdoMet (SAM)‐competitive inhibitor of protein arginine N‐methyltransferases (PRMTs; IC50 = 8.81 μM for PRMT1 and 3.03 μM for yeast‐RMT1p) with minimal effect on lysine methyltransferases. It inhibits nuclear receptor reporter gene activation in MCF‐7 cells, and HIV‐1 RT polymerase (IC50 = 5 μM).
In summary, although many of these inhibitors are able to reduce or abolish HMT activity in vitro or in cell‐based assays, they are still at pre‐clinical stage due to the low specificity and toxicity observed in different cell lines.
3.5. Histone demethylase enzymes and their inhibitors
Two families of histone demethylating enzymes (HDs) have recently been discovered. Lysine‐specific demethylase 1 (LSD1) is a flavin‐dependent monoamine oxidase which can demethylate mono‐ and di‐methylated lysines, specifically histone 3, lysines 4 and 9 (H3K4 and H3K9) (Forneris et al., 2005). Jumonji domain‐containing (JmjC) histone demethylases are able to demethylate mono‐, di‐, or tri‐methylated lysines. Two specific JmjC HDs are PHF8 and JHDM1D.
LSD1 shares similar catalytic sites with monoamine oxidases (MAO) A and B, the inhibition of which is used clinically to treat depression, anxiety, and Parkinson's disease (Lee et al., 2006).
2‐PCPA (Tranylcypromine) is an inhibitor of LSD1 with an IC50 value of 20.7 μM and a K i value of 242.7 μM, which effectively inhibits histone demethylation in vivo. Although not as selective, 2‐PCPA also irreversibly inhibits MAO A and MAO B with IC50 values of 2.3 and 0.95 μM and K i values of 101.9 and 16 μM, respectively (Schmidt and McCafferty, 2007). Given these collateral effects, Tranylcypromine has not yet entered clinical trials.
4. DNA methyltransferases enzymes and their inhibitors
DNA methylation refers to a covalent modification of the cytosine base (C), localized at 5′ of a guanidine base (G) in a CpG dinucleotide (Das and Singal, 2004; Robertson, 2001; Robertson and Wolffe, 2000). DNA methylation is involved in the control of gene expression, regulation of parental imprinting and stabilization of X chromosome inactivation as well as maintenance of genome integrity. It is also implicated in the development of immune system, cellular reprogramming, brain function and behavior (Di Croce et al., 2002).
The transfer of methyl groups from SAM to cytosine in CpGs is catalyzed by a family of enzymes called DNA methyltransferase (DNMTs) (Iyer et al., 2011; Jurkowska et al., 2011; Peedicayil, 2012; Xu et al., 2010). In mammals, three DNMTs have so far been identified, including two “de novo” methyltransferases (DNMT3A and DNMT3B) and “maintenance” methyltransferase (DNMT1), generally the most abundant and active of the three (Goll and Bestor, 2005). The protein DNMT2 can also be found in mammalian cells. Although the structure of DNMT2 is very similar to other DNMTs, its role is less understood (Schaefer and Lyko, 2010). It has been reported that DNMT2 does not methylate DNA but instead methylates aspartic acid transfer RNA (tRNAAsp) (Jurkowski and Jeltsch, 2011). Recent evidence suggests that DNMT2 activity is not limited to tRNAAsp and that DNMT2 represents a non‐canonical enzyme of the DNMT family (Schaefer et al., 2010). Given the critical role of DNMTs, intense interest has focused on developing drugs able to interfere with aberrant DNMT activities, and using them to correct epigenetic defects such as tumor suppression gene (TSG) silencing (Rajendran et al., 2011).
DNMTs modulators represent a useful tool in epigenetic therapies. Several epi‐drugs interfering with DNMT activity are currently in pre‐clinical and clinical trials (Foulks et al., 2012; McGovern et al., 2012). Most of these trials have involved various types of cancer, such as solid and hematological tumors (Chaib et al., 2011; Fandy, 2009; Ren et al., 2011; Song et al., 2011). Currently, however, the main challenge in using epigenetic modulators for therapy, especially for interfering with DNMT enzymes is their specificity (Veeck and Esteller, 2010).
It is well known that abnormal patterns of DNA methylation are often displayed in human neoplasms (Chin et al., 2011; Fonseca et al., 2012; Lokk et al., 2012; Peedicayil, 2012). A combination of regional promoter hypermethylation with the concomitant silencing of important genes involved in cell death, surveillance and proliferation is always present. However, demethylation per se is not sufficient to induce gene expression, as it is regulated by a combination of different epi‐mutations (Choo, 2011; Eglen and Reisine, 2011). DNA methylation and histone modifications are tightly correlated because inactive chromatin is enriched with hypermethylated DNA and active chromatin is associated with hypomethylated DNA. In contrast, genomic hypomethylation mediates tumorigenesis (via chromosomal instability) and supports the metastatic process (Hatziapostolou and Iliopoulos, 2011; Kulis and Esteller, 2010; Plitta et al., 2011; Watanabe and Maekawa, 2010). Consequently, induction of global DNA hypomethylation using DNMTi may help speed tumor progression from cancer cells surviving DNA demethylation therapy (Guz et al., 2010; Villa et al., 2004; Wild and Flanagan, 2010).
Moreover, the interrelation between drugs targeting chromatin and those targeting DNA methylation could be utilized therapeutically by combining different epigenetic drugs against different epigenetic effectors to increase the efficacy of a monotherapy.
Several clinical trials testing different DNMTi are terminated and ongoing.
4.1. Nucleoside analogs
This class of DNMTi includes nucleoside analogs, which are phosphorylated by cellular kinases. Once incorporated into DNA, they form a covalent bond with the DNMT trapping the enzyme and making it unavailable for further methylation, thus resulting in demethylation of replicating nascent DNA (Szyf, 2009). Two agents have been developed clinically: 5‐aza‐2′‐deoxycytidine (decitabine) and 5‐azacytidine (azacitidine or Vidaza). These agents have been tested in numerous trials (Goffin and Eisenhauer, 2002; Sorm et al., 1964) (9, 10, 11, 12).
Table 9.
Drug | Conditions | Status | NCT number | Phases |
---|---|---|---|---|
Decitabine | MDS|CML | Completed | NCT00067808 | Phase 2 |
Decitabine | Thalassemia | Completed | NCT00661726 | Phase 2 |
Decitabine | Lymphoma, intestinal neoplasms | Completed | NCT00089089 | Phase 1 |
Decitabine | AML | Completed | NCT00866073 | Phase 2 |
Decitabine | AML, MDS | Completed | NCT00760084 | Phase 2 |
Decitabine | MDS, CML | Terminated | NCT00113321 | Phase 2 |
Decitabine | Leukemia | Completed | NCT01378416 | Phase 1 |
Decitabine | CML | Completed | NCT01098084 | Phase 2 |
Decitabine | AML | Completed | NCT00398983 | Phase 2|3 |
Decitabine | MDS | Completed | NCT00043381 | Phase 3 |
Decitabine | AML | Completed | NCT00538876 | Phase 1 |
Decitabine | MDS | Terminated | NCT00282399 | Phase 1|2 |
Decitabine | CML | Completed | NCT00042003 | Phase 2 |
Decitabine | CML | Completed | NCT00042016 | Phase 2 |
Decitabine | CML | Completed | NCT00041990 | Phase 2 |
Decitabine | MDS | Completed | NCT00260065 | Phase 2 |
Decitabine | MDS | Completed | NCT01041846 | N/A |
Decitabine | MDS | Completed | NCT00796003 | Phase 1 |
Decitabine | AML | Completed | NCT00358644 | Phase 2 |
Decitabine | Leukemia, MDS | Completed | NCT00003361 | Phase 2 |
Decitabine | Leukemia, MDS | Completed | NCT00049582 | Phase 1 |
Decitabine | MDS, secondary myelofibrosis | Terminated | NCT00630994 | Phase 2 |
Decitabine | Bladder cancer, breast cancer, melanoma | Completed | NCT00030615 | Phase 1 |
Decitabine | Esophageal, lung cancer, malignant mesothelioma | Completed | NCT00019825 | Phase 1 |
Decitabine | Leukemia | Completed | NCT00042796 | Phase 1 |
Decitabine | MDS, leukemia, lymphoma, MM | Completed | NCT00002980 | Phase 1 |
Table 10.
Drug | Conditions | Status | NCT number | Phases |
---|---|---|---|---|
Decitabine + vorinostat | Myeloproliferative disorders, leukemia | Completed | NCT00357708 | Phase 1 |
Decitabine + interferon alfa‐2b | Renal cell carcinoma | Terminated | NCT00561912 | Phase 2 |
Decitabine + filgrastim + cyclosporine | Leukemia, MDS | Completed | NCT00002832 | Phase 1|2 |
Decitabine + VPA | Leukemia, MDS | Completed | NCT00075010 | Phase 1|2 |
Decitabine + cytarabine or supportive care | AML | Completed | NCT00260832 | Phase 3 |
Decitabine + carboplatin | Fallopian tube, ovarian, peritoneal cavity cancer | Terminated | NCT00748527 | Phase 2 |
Decitabine + azacitidine | MDS | Terminated | NCT01011283 | Phase 4 |
Decitabine + arsenic trioxide + ascorbic acid | MDS | Completed | NCT00621023 | Phase 2 |
Decitabine + PEG‐interferon alfa‐2b | Unspecified adult solid tumor | Completed | NCT00701298 | Phase 1 |
Decitabine + pegylated interferon‐alfa 2b | Cancer | Terminated | NCT00886457 | Phase 1 |
Decitabine + chemotherapy | AML | Terminated | NCT00943553 | Phase 2 |
Decitabine + VPA | Lymphoma | Completed | NCT00109824 | Phase 1 |
Decitabine + romidepsin | SCC|mesothelioma|NSCLC | Completed | NCT00037817 | Phase 1 |
Decitabine + vorinostat | Leukemia|MDS | Completed | NCT00479232 | Phase 1 |
Decitabine + filgrastim + pegfilgrastim + cyclophosphamide + doxorubicin hydrochloride | Neuroblastoma, childhood solid tumor | Completed | NCT00075634 | Phase 1 |
Decitabine + imatinib mesylate | Leukemia | Completed | NCT00054431 | Phase 2 |
Decitabine + romidepsin | Leukemia, MDS | Completed | NCT00114257 | Phase 1 |
Decitabine + filgrastim + busulfan + cyclophosphamide + cyclosporine + methotrexate + methylprednisolon + tacrolimus | Leukemia | Completed | NCT00002831 | Phase 1|2 |
Decitabine + AMG 531 (romiplostim) + azacitidine | MDS, thrombocytopenia | Completed | NCT00321711 | Phase 2 |
Decitabine deferoxamine + deferiprone + arginine + sildenafil | Hematologic diseases, osteoporosis, hypertension | Completed | NCT00000623 | N/A |
Table 11.
Drug | Conditions | Status | NCT number | Phases |
---|---|---|---|---|
Azacitidine | MDS|AML | Completed | NCT00795548 | Phase 2 |
Azacitidine | Ovarian cancer | Terminated | NCT00842582 | Phase 1 |
Azacitidine | MDS|leukemia | Completed | NCT00350818 | Phase 1 |
Azacitidine | Myelofibrosis | Completed | NCT00569660 | Phase 2 |
Azacitidine | MDSs | Completed | NCT00118287 | Phase 1|2 |
Azacitidine | Leukemia | Completed | NCT00739388 | Phase 2 |
Azacitidine | MDSs | Completed | NCT01186939 | Phase 3 |
Azacitidine | AML|MDSs | Terminated | NCT00446303 | Phase 2 |
Azacitidine | MDSs | Completed | NCT00102687 | Phase 2 |
Azacitidine | MDSs | Completed | NCT00071799 | Phase 3 |
Azacitidine for injectable suspension | Prostate cancer | Completed | NCT00384839 | Phase 2 |
Azacitidine | Beta thalassemia | Completed | NCT00005934 | Phase 2 |
Azacitidine | CMPD, secondary myelofibrosis | Terminated | NCT00381693 | Phase 2 |
Table 12.
Drug | Conditions | Status | NCT number | Phases |
---|---|---|---|---|
Azacitidine + VPA + ATRA | AML|MDS | Completed | NCT00339196 | Phase 2 |
Azacitidine + VPA + ATRA | MDSs | Completed | NCT00439673 | Phase 2 |
Azacitidine + Ara‐C | AML, MDS, leukemia | Completed | NCT00569010 | Phase 1|2 |
Azacitidine + sodium phenylbutyrate | Leukemia, MDSs | Completed | NCT00004871 | Phase 1 |
Azacitidine + arsenic trioxide | Leukemia, MDSs | Terminated | NCT00234000 | Phase 1 |
Azacitidine + sodium phenylbutyrate | Lymphoma, small intestine cancer | Completed | NCT00005639 | Phase 1 |
Azacitidine + entinostat | Colorectal cancer | Completed | NCT01105377 | Phase 2 |
Azacitidine + arsenic trioxide | Leukemia, MDSs | Completed | NCT00118196 | Phase 2 |
Azacitidine + amifostine trihydrate | MDSs | Completed | NCT00005598 | Phase 2 |
Azacitidine + sodium phenylbutyrate | Hematologic and solid tumors | Completed | NCT00006019 | Phase 2 |
Azacitidine + liothyronine sodium + radiation: iodine I 131 | Head and neck cancer | Completed | NCT00004062 | Phase 1 |
Azacitidine + sodium phenylbutyrate | Thalassemia major | Terminated | NCT00007072 | Phase 2 |
Azacitidine + AMG 531 (romiplostim) + decitabine | MDS, thrombocytopenia | Completed | NCT00321711 | Phase 2 |
Azacitadine and hematopoietic growth factors | Leukemia, MDSs | Terminated | NCT00398047 | N/A |
Intravenous azacitidine | MDSs | Completed | NCT00384956 | Phase 2 |
Azacitidine | AML, MDS | Completed | NCT00422890 | Phase 3 |
Azacitidine + VPA + ATRA | MDS, AML | Completed | NCT01575691 | Phase 1 |
Azacitidine + MGCD0103 | AML, MDS | Terminated | NCT00666497 | Phase 2 |
Azacitidine + cisplatin | Squamous cell carcinoma | Terminated | NCT00443261 | Phase 1 |
Azacitidine + entinostat | Lung cancer | Suspended | NCT00387465 | Phase 1|2 |
Azacitidine + erythropoietin | MDSs | Terminated | NCT00379912 | Phase 2 |
Azacitidine + VPA + ATRA | MDS, AML | Completed | NCT00326170 | Phase 2 |
Azacitidine + midostaurin | Hematologic cancers | Suspended | NCT01093573 | Phase 1|2 |
Azacitidine + VPA | Advanced cancers | Completed | NCT00496444 | Phase 1 |
Azacitidine + Ara‐C + VPA | AML, MDS, leukemia | Completed | NCT00382590 | Phase 2 |
Azacitidine, erlotinib | Advanced solid tumor malignancies | Completed | NCT00996515 | Phase 1 |
Azacitidine, decitabine | MDSs | Terminated | NCT01011283 | Phase 4 |
Azacitidine, MGCD0103 | Hodgkin and non‐Hodgkin lymphoma | Terminated | NCT00543582 | Phase 2 |
Azacitidine, entinostat | Leukemia, MDSs | Completed | NCT00101179 | Phase 1 |
Decitabine is incorporated into DNA, while azacitidine is incorporated preferentially into RNA (Leone et al., 2002). Demethylation by decitabine has been shown to allow re‐expression of silenced genes and cellular differentiation. Additionally, incorporation of these agents into RNA causes ribosomal disassembly, defective tRNA function and inhibited protein production. However, azacitidine exhibits greater cytotoxicity during S‐phase, supporting the greater importance of its DNA effects. Both these drugs are inactivated via deamination by cytidine deaminase (Galmarini et al., 2001) and have been approved by the FDA for the treatment of myelodysplastic syndromes (Kadia et al., 2011; Marks, 2012).
To date, decitabine and azacitidine have been studied as single drug treatment (9, 11) for many malignancies, though the best results have been obtained for MDS and various types of leukemia.
These DNMTis have been in clinical trial for hematological diseases, such as MDS and leukemias (Chen et al., 2012; Garcia‐Manero, 2012; Keating, 2012; Kimura et al., 2012; Oki et al., 2012; Platzbecker et al., 2012; Ritchie, 2012), thalassemia (Mabaera et al., 2008; Olivieri et al., 2011; Rose et al., 2011; Saunthararajah et al., 2003) and solid tumors, such as prostate (Gravina et al., 2011; Shabbeer et al., 2012), colon (Al‐Salihi et al., 2011; Hagemann et al., 2011; Ikehata et al., 2012; Yang et al., 2012), bladder (Shang et al., 2008), breast (Mirza et al., 2010), melanoma (Alcazar et al., 2012; Liu et al., 2011), esophageal (Dong et al., 2012; Liu et al., 2005; Meng et al., 2012; Schrump et al., 2006), lung cancers (Kaminskyy et al., 2011; Wu and Hu, 2011).
The most promising results have been obtained with combination therapy, especially with HDACi (10, 12).
4.2. Small molecules
Blocking the enzymatic activity of DNMTs by using small molecule inhibitors is another strategy to achieve gene demethylation (Datta et al., 2009). Hydralazine and procainamide are FDA approved for the treatment of hypertension and cardiac arrhythmia, respectively (Peng et al., 2010). Recently, their ability to inhibit DNMT activity has been discovered and is associated with direct binding to CpG‐rich sequences (Amatori et al., 2010). More precisely, these molecules act as partial competitive inhibitors of DNMT1, decreasing the affinity of DNMT for its substrates (DNA and S‐adenosyl‐l‐methionine), reducing the processivity of the enzyme and favoring the dissociation of DNMT1 from hemimethylated DNA. No data are currently available for their use in clinical trails as DNMT inhibitors; however, they are in pre‐clinical studies.
The antihypertensive (Singh et al., 2009) hydralazine was tested as a DNMT inhibitor due to its capability to induce (as a side effect) a lupus‐like syndrome known to be related to disorders associated with DNA methylation (Candelaria et al., 2011; Lu et al., 2005). Although its mechanism of action is still under investigation, some evidence indicates that hydralazine, similar to procaine and procainamide, binds to CpG‐rich sequences and interferes with translocation of DNMTs along the DNA strand. A phase 1 study has shown that hydralazine is able to induce re‐expression of various tumor suppressor genes, including p16 and RAR‐β, in cervical cancer patients, even at lower doses than those considered safe for the treatment of cardiovascular disorders. On the basis of promising data arising from phase 1, hydralazine is currently under phase 2 and 3 investigations. Table 1 reports three clinical trials in which hydralazine is used in combination with magnesium valproate against solid tumors (discussed in “ Short‐chain fatty acid”).
A major drawback of these drugs is the high concentrations required for their demethylating activity, which can elicit undesired toxic effects if administered clinically.
Rational design of DNMTi that interact noncovalently with the active catalytic site of DNMTs, utilizing a three‐dimensional model of the human DNMT1 catalytic pocket, is a sound alternative approach to silence the DNA methylation machinery. RG108 [2‐(1,3‐dioxo‐1,3‐dihydro‐2H‐isoindol‐2‐yl)‐3‐(1H‐indol‐3‐yl)propanoic acid] is the first rationally designed DNMT1i that demonstrates demethylating activity both in vivo and in vitro (Braun et al., 2010; Brueckner et al., 2005; Chestnut et al., 2011; Schirrmacher et al., 2006; Suzuki et al., 2010).
This molecule was found to induce the re‐expression of different hypermethylation‐silenced genes, such as p16 and the putative tumor suppressor genes SRFP1 and TIMP‐3, in colon cancer cells. Despite its encouraging activity, RG108 is still in pre‐clinical phase.
4.3. Natural molecules
The use of natural products in cancer chemoprevention is currently receiving much attention.
Psammaplins, for example, are bisulfide bromotyrosines derived from a marine sponge and are able to inhibit both DNMT1 and HDAC in vitro. Although psammaplin A administration at low doses was found to exert a strong cytotoxic effect in human tumor cell lines and to limit tumor cell growth in a A549 lung xenograft mouse model, DNMT inhibition was not followed by DNA demethylation and re‐expression of tumor suppressor genes, suggesting that an intracellular target different from DNMT1 is responsible for the cytotoxic effect of the molecule (Baud et al., 2012).
Tea polyphenols are strong antioxidants and tea preparations demonstrate inhibitory activity against carcinogenesis. (−)‐Epigallocatechin‐3‐gallate (EGCG), the major polyphenol from green tea, is a potent inhibitor of catechol‐O‐methyltransferase activity (COMT) (Fang et al., 2007; Lee et al., 2005; Nandakumar et al., 2011). The structural similarity between DNMTs and COMT suggests possible inhibition of DNMTs by EGCG. EGCG inhibits DNMT activity in a dose‐dependent manner and induces re‐expression of hypermethylated genes such as CDKN2A, RARbeta and MGMT (Nandakumar et al., 2011). In many pre‐clinical studies, EGCG exhibits a very strong DNMT inhibitory action and this natural molecule is able to induce cell death and apoptosis in many cancer types (Gu et al., 2009).
Interestingly, other dietary catechol‐containing polyphenols, such as different tea catechins (catechin, epicatechin) and bioflavonoids (quercetin, genistein, fisetin), were also found to inhibit DNMT activity in vitro through mechanisms different to that of EGCG (Fang et al., 2007; Lee et al., 2005). The results obtained from the use of natural compounds are intriguing, especially considering their ease of use and low costs. However, further investigation is necessary to establish their real efficacy as DNMTi, as well as to evaluate the toxicity induced by their administration at pharmacological doses.
A major concern associated with the use of natural products is product standardization. Multiple sources can provide extracts with different activities and therefore create discrepancies in their reported demethylating activity.
4.4. Antisense oligonucleotide inhibitors of DNMTs
Antisense oligonucleotides are short, defined sequences of nucleotides that are complementary to mRNAs. They hybridize mRNAs, making them inactive and thereby blocking translation. Antisense oligonucleotides that are complementary to mRNAs for human DNMT1 are undergoing pre‐clinical (Yan et al., 2003) as well as clinical trials (Davis et al., 2003).
The idea to target directly DNMT1 enzyme arises from the observation that, excluding some rare exceptions, tumor cells generally show increased expression levels of DNMTs.
The most interesting isotypic‐specific DNMT1 inhibitor tested in clinical trials is MG98 (Amato et al., 2012; Patutina et al., 2009; Winquist et al., 2006), a second‐generation antisense oligonucleotide that specifically targets DNMT1 mRNA. This agent eliminates the expression of DNMT1 protein resulting in the inhibition of DNA replication, triggering of damage response, and induction of TSGs. The immediate blockage of replication by DNMT1 knockdown dramatically limits demethylation induced by DNMT1 inhibition, thus avoiding the potential deleterious impact of global demethylation. The main issue with antisense oligonucleotides is delivery to solid tumors (Davis et al., 2003; Klisovic et al., 2008; Plummer et al., 2009).
Some clinical trials utilizing MG98 were stopped because of lack of objective response in the last phase 2 trials in metastatic renal cancer. Nevertheless, this strategy carries great promise. A new trial has been completed (Stewart et al., 2003) in which pharmacodynamic evaluations were performed to explore and validate the biological mechanisms of MG98 in solid tumors (Clinical trial identifier: NCT00003890).
Interesting results are provided by miR‐29b, a microRNA (miRNA) that directly targets DNMT3A and DNMT3B expression. MiR‐29b induces a decrease in methylation levels and induces the re‐expression of hypermethylated TSGs, FHIT and WWOX in lung cancer cells, as well as of p15 and ER in AML cells. Although these studies are all still at pre‐clinical level, the above findings support the possibility of miRNA‐based approaches.
5. Non‐coding RNAs
MicroRNAs (miRNAs) are short RNA molecules, 19–25 nucleotides long, that bind to complementary sequences in the 3′ UTR of multiple target mRNAs, usually resulting in their silencing. miRNAs were recently identified as key players in regulating gene expression by inhibiting translation and/or triggering degradation of their target mRNAs (Bartel, 2004). miRNAs target ∼60% of all genes, are abundantly present in all human cells and are each able to repress hundreds of targets. Most miRNAs are transcribed by RNA polymerase II and subsequently processed by multiple maturation steps resulting in mature double‐stranded miRNA duplexes. miRNAs are implicated in a wide range of basic biological processes, including development, differentiation, apoptosis and proliferation (Bartel, 2004; Harfe, 2005) and their misregulation is linked to the development of diseases in humans and other organisms. Researchers worldwide have validated the theory of “miRNA replacement therapy”, which involves introducing synthetic miRNAs or miRNA mimetics into diseased tissues in an attempt to restore normal proliferation, apoptosis, cell cycle, and other cellular functions that have been affected by the misregulation of one or more miRNAs (Raver‐Shapira et al., 2007).
Over the last 5 years, a particularly important role for miRNAs in cancer pathogenesis has emerged. Many tumor types are characterized by globally abnormal miRNA expression patterns. Up to now, 23 studies have been completed (Table 13), and describe the use of miRNAs in cancer therapy. miRNA expression profiles are highly informative for tumor classification, prognosis, and response to therapy. Moreover, recent results have documented a functional contribution of specific miRNAs to cellular transformation and tumorigenesis. The pioneering groups of specialized pharmaceutical companies have initiated studies on creating viable therapeutic candidates with miRNA inhibitors and miRNA mimetics in diverse fields such as cancer, cardiovascular diseases, neurological disorders, and viral infections.
Table 13.
Drug | Conditions | Status | NCT number | Phases |
---|---|---|---|---|
Observational study | Sepsis | Completed | NCT00862290 | N/A |
Observational study | Cutaneous melanoma | Completed | NCT01444560 | N/A |
Observational study | Aspirin‐exacerbated respiratory disease | Completed | NCT01631773 | N/A |
Observational study | Periodontal disease | Completed | NCT01399034 | N/A |
Observational study | Basal cell carcinoma | Completed | NCT01498250 | N/A |
Observational study | Cutaneous squamous cell carcinoma | Completed | NCT01500954 | N/A |
Observational study | Malignant melanoma | Completed | NCT00862914 | N/A |
Observational study | Skin cancer | Completed | NCT00849914 | N/A |
Observational study | Leukemia | Completed | NCT01057199 | N/A |
Observational study | Barrett's esophagus, esophagea, adenocarcinoma | Completed | NCT00909350 | N/A |
Observational study | Cutaneous malignant melanoma | Completed | NCT01482260 | N/A |
Observational study | Cancer of the skin | Terminated | NCT01143311 | N/A |
Observational study | Melanoma | Completed | NCT00536029 | N/A |
Observational study | Skin cancer | Completed | NCT01345760 | N/A |
Observational study | The study focused on reducing tobacco smoke exposure for child | Completed | NCT01317628 | N/A |
Observational study | Muscle anabolism | Completed | NCT00812396 | Phase 1 |
Observational study | Hepatitis C | Completed | NCT00688012 | Phase 1 |
Observational study | Lung cancer | Completed | NCT00897234 | N/A |
Observational study | Influence of genotype of drug | Completed | NCT01503658 | Phase 4 |
Observational study | Awaiting organ transplant|infection in solid organ transplant recipients | Completed | NCT01558037 | N/A |
Observational study | Melanoma (skin) | Completed | NCT01216787 | Phase 2 |
Observational study | Neuroblastoma | Completed | NCT01058798 | N/A |
Observational study | Recurrent calcic urolithiasis | Completed | NCT01022060 | Phase 3 |
6. Summary and concluding remarks
The present review highlights the enormous impact that small molecules such as HDACi and DNMTi have and will continue to have in the treatment of human diseases, especially cancer. Their key importance is due to the differentiation‐ and apoptosis‐inducing activity of this new class of anticancer drugs. By targeting histone (chromatin) modulators, epi‐drugs activate a complex transcriptional reprogramming, exemplified by reactivation of TSGs and repression of oncogenes.
Widespread changes in post‐translational modifications of histones and DNA determine diseases and represent marks that play a crucial role in chromatin packaging and gene expression. The aberrant recruitment of HDACs and/or DNMTs or the activation of miRNAs in normal cells are involved in a number of pathologies, primarily cancer. It has been reported that HDACis/HMTis or DNMTis induce strong re‐expression of TSGs and generally induce transcription‐regulating activity on various genes, cell cycle inhibitory activity and apoptosis. Hence, their importance in epigenetic therapy. The vast number of clinical trials underlines the promising use of these drugs in diagnostic therapeutics of human diseases. However, their application is more effective in combination therapy. Trials related to combination therapies are based on the recognition that certain combinations of compounds combine more effectively with each other, improve tolerance, require lower dosages of each agent, and/or reduce side effects caused by at least one of the compounds in the combination. Although many anti‐cancer therapies exist, there is a need to develop therapeutics that are safe and effective, circumvent resistance to hormonal and other therapies, do not cause the onset of other pathologies, and extend the disease‐free survival of patients.
Cells that have developed mutations within the drug‐binding pocket display a growth advantage in the presence of the drug, eventually leading to disease progression. Current clinical strategies aimed at combining these molecularly targeted drugs with standard chemotherapeutics, radiation, or other targeted agents will lead to novel strategies aimed at improving the overall response rate and increasing the number of complete remissions.
A good synergistic effect is obtained by the combination of drugs that inhibit DNMTs and HDACs.
In recent years, we have been facing the new concept of “personalized therapy”, which takes into consideration individual differences between patients, and represents an attempt to ethically and medically improve cost performance of medication treatment by administering a pharmaceutical agent to patients after verification in advance of the probability of its effect, thereby enhancing efficacy as well as avoiding toxicity of the agent, and reducing inappropriate use of drugs. In cancer treatment, the development of a method for predicting the efficacy of anti‐cancer agents is highly desirable as it may be an important way to bridge the gap between basic research and clinical application.
The clinical benefits of epi‐therapy are currently being investigated in several human diseases but better results could undoubtedly be obtained with the development of new‐generation epi‐drugs. It is therefore essential to identify a characteristic profile for successful candidates as epi‐drugs.
Acknowledgments
This work was supported by EU: Blueprint (contract no. 282510), ATLAS (contract no. 221952); Epigenomics Flagship Project EPIGEN (MIUR‐CNR); the Italian Association for Cancer Research (AIRC no. 11812); Italian Ministry of University and Research (PRIN_2009PX2T2E_004; PON002782; PON0101227). We thank Catherine Fisher for linguistic editing of the manuscript. The authors declare that they have no conflict of interest.
Nebbioso Angela, Carafa Vincenzo, Benedetti Rosaria, Altucci Lucia, (2012), Trials with ‘epigenetic’ drugs: An update, Molecular Oncology, 6, doi: 10.1016/j.molonc.2012.09.004.
References
- Abdel Aziz, M.T. , El-Asmar, M.F. , El-Ibrashy, I.N. , Rezq, A.M. , Al-Malki, A.L. , Wassef, M.A. , Fouad, H.H. , Ahmed, H.H. , Taha, F.M. , Hassouna, A.A. , Morsi, H.M. , 2012. Effect of novel water soluble curcumin derivative on experimental type-1 diabetes mellitus (short term study). Diabetology & Metabolic Syndrome. 4, 30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aggarwal, B.B. , Bhardwaj, A. , Aggarwal, R.S. , Seeram, N.P. , Shishodia, S. , Takada, Y. , 2004. Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies. Anticancer Research. 24, 2783–2840. [PubMed] [Google Scholar]
- Al-Salihi, M. , Yu, M. , Burnett, D.M. , Alexander, A. , Samlowski, W.E. , Fitzpatrick, F.A. , 2011. The depletion of DNA methyltransferase-1 and the epigenetic effects of 5-aza-2'deoxycytidine (decitabine) are differentially regulated by cell cycle progression. Epigenetics. 6, 1021–1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alcain, F.J. , Villalba, J.M. , 2009. Sirtuin inhibitors. Expert Opinion on Therapeutic Patents. 19, 283–294. [DOI] [PubMed] [Google Scholar]
- Alcazar, O. , Achberger, S. , Aldrich, W. , Hu, Z. , Negrotto, S. , Saunthararajah, Y. , Triozzi, P. , 2012. Epigenetic regulation by decitabine of melanoma differentiation in vitro and in vivo. International Journal of Cancer. 131, 18–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amato, R.J. , Stephenson, J. , Hotte, S. , Nemunaitis, J. , Belanger, K. , Reid, G. , Martell, R.E. , 2012. MG98, a second-generation DNMT1 inhibitor, in the treatment of advanced renal cell carcinoma. Cancer Investigation. 30, 415–421. [DOI] [PubMed] [Google Scholar]
- Amatori, S. , Bagaloni, I. , Donati, B. , Fanelli, M. , 2010. DNA demethylating antineoplastic strategies: a comparative point of view. Genes & Cancer. 1, 197–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arce, C. , Perez-Plasencia, C. , Gonzalez-Fierro, A. , de la Cruz-Hernandez, E. , Revilla-Vazquez, A. , Chavez-Blanco, A. , Trejo-Becerril, C. , Perez-Cardenas, E. , Taja-Chayeb, L. , Bargallo, E. , 2006. A proof-of-principle study of epigenetic therapy added to neoadjuvant doxorubicin cyclophosphamide for locally advanced breast cancer. PLoS One. 1, e98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Archin, N.M. , Cheema, M. , Parker, D. , Wiegand, A. , Bosch, R.J. , Coffin, J.M. , Eron, J. , Cohen, M. , Margolis, D.M. , 2010. Antiretroviral intensification and valproic acid lack sustained effect on residual HIV-1 viremia or resting CD4+ cell infection. PLoS One. 5, e9390 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arts, J. , King, P. , Marien, A. , Floren, W. , Belien, A. , Janssen, L. , Pilatte, I. , Roux, B. , Decrane, L. , Gilissen, R. , 2009. JNJ-26481585, a novel “second-generation” oral histone deacetylase inhibitor, shows broad-spectrum preclinical antitumoral activity. Clinical Cancer Research. 15, 6841–6851. [DOI] [PubMed] [Google Scholar]
- Ashraf, N. , Zino, S. , Macintyre, A. , Kingsmore, D. , Payne, A.P. , George, W.D. , Shiels, P.G. , 2006. Altered sirtuin expression is associated with node-positive breast cancer. British Journal of Cancer. 95, 1056–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Badros, A. , Burger, A.M. , Philip, S. , Niesvizky, R. , Kolla, S.S. , Goloubeva, O. , Harris, C. , Zwiebel, J. , Wright, J.J. , Espinoza-Delgado, I. , 2009. Phase I study of vorinostat in combination with bortezomib for relapsed and refractory multiple myeloma. Clinical Cancer Research. 15, 5250–5257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balasubramanyam, K. , Swaminathan, V. , Ranganathan, A. , Kundu, T.K. , 2003. Small molecule modulators of histone acetyltransferase p300. The Journal of Biological Chemistry. 278, 19134–19140. [DOI] [PubMed] [Google Scholar]
- Balasubramanyam, K. , Altaf, M. , Varier, R.A. , Swaminathan, V. , Ravindran, A. , Sadhale, P.P. , Kundu, T.K. , 2004. Polyisoprenylated benzophenone, garcinol, a natural histone acetyltransferase inhibitor, represses chromatin transcription and alters global gene expression. The Journal of Biological Chemistry. 279, 33716–33726. [DOI] [PubMed] [Google Scholar]
- Balasubramanyam, K. , Varier, R.A. , Altaf, M. , Swaminathan, V. , Siddappa, N.B. , Ranga, U. , Kundu, T.K. , 2004. Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription. The Journal of Biological Chemistry. 279, 51163–51171. [DOI] [PubMed] [Google Scholar]
- Baliga, M.S. , Joseph, N. , Venkataranganna, M.V. , Saxena, A. , Ponemone, V. , Fayad, R. , 2012. Curcumin, an active component of turmeric in the prevention and treatment of ulcerative colitis: preclinical and clinical observations. Food & Function. 3, http://dx.doi.org/10.1039/C2FO30097D [DOI] [PubMed] [Google Scholar]
- Banerji, U. , van Doorn, L. , Papadatos-Pastos, D. , Kristeleit, R. , Debnam, P. , Tall, M. , Stewart, A. , Raynaud, F. , Garrett, M.D. , Toal, M. , 2012. A phase I pharmacokinetic and pharmacodynamic study of CHR-3996, an oral class I selective histone deacetylase inhibitor in refractory solid tumors. Clinical Cancer Research. 18, 2687–2694. [DOI] [PubMed] [Google Scholar]
- Bannister, A.J. , Kouzarides, T. , 2011. Regulation of chromatin by histone modifications. Cell Research. 21, 381–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartel, D.P. , 2004. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 116, 281–297. [DOI] [PubMed] [Google Scholar]
- Baud, M.G. , Leiser, T. , Haus, P. , Samlal, S. , Wong, A.C. , Wood, R.J. , Petrucci, V. , Gunaratnam, M. , Hughes, S.M. , Buluwela, L. , 2012. Defining the mechanism of action and enzymatic selectivity of psammaplin A against its epigenetic targets. Journal of Medicinal Chemistry. 55, 1731–1750. [DOI] [PubMed] [Google Scholar]
- Bernstein, B.E. , Meissner, A. , Lander, E.S. , 2007. The mammalian epigenome. Cell. 128, 669–681. [DOI] [PubMed] [Google Scholar]
- Binion, D.G. , Otterson, M.F. , Rafiee, P. , 2008. Curcumin inhibits VEGF-mediated angiogenesis in human intestinal microvascular endothelial cells through COX-2 and MAPK inhibition. Gut. 57, 1509–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blackwell, L. , Norris, J. , Suto, C.M. , Janzen, W.P. , 2008. The use of diversity profiling to characterize chemical modulators of the histone deacetylases. Life Sciences. 82, 1050–1058. [DOI] [PubMed] [Google Scholar]
- Blum, K.A. , Advani, A. , Fernandez, L. , Van Der Jagt, R. , Brandwein, J. , Kambhampati, S. , Kassis, J. , Davis, M. , Bonfils, C. , Dubay, M. , 2009. Phase II study of the histone deacetylase inhibitor MGCD0103 in patients with previously treated chronic lymphocytic leukaemia. British Journal of Haematology. 147, 507–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bolden, J.E. , Peart, M.J. , Johnstone, R.W. , 2006. Anticancer activities of histone deacetylase inhibitors. Nature Reviews Drug Discovery. 5, 769–784. [DOI] [PubMed] [Google Scholar]
- Borbone, E. , Berlingieri, M.T. , De Bellis, F. , Nebbioso, A. , Chiappetta, G. , Mai, A. , Altucci, L. , Fusco, A. , 2010. Histone deacetylase inhibitors induce thyroid cancer-specific apoptosis through proteasome-dependent inhibition of TRAIL degradation. Oncogene. 29, 105–116. [DOI] [PubMed] [Google Scholar]
- Borra, M.T. , Smith, B.C. , Denu, J.M. , 2005. Mechanism of human SIRT1 activation by resveratrol. The Journal of Biological Chemistry. 280, 17187–17195. [DOI] [PubMed] [Google Scholar]
- Bosch-Presegue, L. , Vaquero, A. , 2011. The dual role of sirtuins in cancer. Genes & Cancer. 2, 648–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bouzar, A.B. , Boxus, M. , Defoiche, J. , Berchem, G. , Macallan, D. , Pettengell, R. , Willis, F. , Burny, A. , Lagneaux, L. , Bron, D. , 2009. Valproate synergizes with purine nucleoside analogues to induce apoptosis of B-chronic lymphocytic leukaemia cells. British Journal of Haematology. 144, 41–52. [DOI] [PubMed] [Google Scholar]
- Bracker, T.U. , Sommer, A. , Fichtner, I. , Faus, H. , Haendler, B. , Hess-Stumpp, H. , 2009. Efficacy of MS-275, a selective inhibitor of class I histone deacetylases, in human colon cancer models. International Journal of Oncology. 35, 909–920. [DOI] [PubMed] [Google Scholar]
- Bratland, A. , Dueland, S. , Hollywood, D. , Flatmark, K. , Ree, A.H. , 2011. Gastrointestinal toxicity of vorinostat: reanalysis of phase 1 study results with emphasis on dose-volume effects of pelvic radiotherapy. Radiation Oncology. 6, 33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braun, J. , Boittiaux, I. , Tilborg, A. , Lambert, D. , Wouters, J. , 2010. The dicyclo-hexyl-amine salt of RG108 (N-phthalyl-l-tryptophan), a potential epigenetic modulator. Acta Crystallographica Section E, Structure Reports Online. 66, o3175–3176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brooks, C.L. , Gu, W. , 2009. How does SIRT1 affect metabolism, senescence and cancer?. Nature Reviews Cancer. 9, 123–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brueckner, B. , Garcia Boy, R. , Siedlecki, P. , Musch, T. , Kliem, H.C. , Zielenkiewicz, P. , Suhai, S. , Wiessler, M. , Lyko, F. , 2005. Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases. Cancer Research. 65, 6305–6311. [DOI] [PubMed] [Google Scholar]
- Buglio, D. , Mamidipudi, V. , Khaskhely, N.M. , Brady, H. , Heise, C. , Besterman, J. , Martell, R.E. , MacBeth, K. , Younes, A. , 2010. The class-I HDAC inhibitor MGCD0103 induces apoptosis in Hodgkin lymphoma cell lines and synergizes with proteasome inhibitors by an HDAC6-independent mechanism. British Journal of Haematology. 151, 387–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burgess, A. , Ruefli, A. , Beamish, H. , Warrener, R. , Saunders, N. , Johnstone, R. , Gabrielli, B. , 2004. Histone deacetylase inhibitors specifically kill nonproliferating tumour cells. Oncogene. 23, 6693–6701. [DOI] [PubMed] [Google Scholar]
- Butler, K.V. , Kozikowski, A.P. , 2008. Chemical origins of isoform selectivity in histone deacetylase inhibitors. Current Pharmaceutical Design. 14, 505–528. [DOI] [PubMed] [Google Scholar]
- Butler, L.M. , Webb, Y. , Agus, D.B. , Higgins, B. , Tolentino, T.R. , Kutko, M.C. , LaQuaglia, M.P. , Drobnjak, M. , Cordon-Cardo, C. , Scher, H.I. , 2001. Inhibition of transformed cell growth and induction of cellular differentiation by pyroxamide, an inhibitor of histone deacetylase. Clinical Cancer Research. 7, 962–970. [PubMed] [Google Scholar]
- Candelaria, M. , Cetina, L. , Perez-Cardenas, E. , de la Cruz-Hernandez, E. , Gonzalez-Fierro, A. , Trejo-Becerril, C. , Taja-Chayeb, L. , Chanona, J. , Arias, D. , Duenas-Gonzalez, A. , 2010. Epigenetic therapy and cisplatin chemoradiation in FIGO stage IIIB cervical cancer. European Journal of Gynaecological Oncology. 31, 386–391. [PubMed] [Google Scholar]
- Candelaria, M. , Gallardo-Rincon, D. , Arce, C. , Cetina, L. , Aguilar-Ponce, J.L. , Arrieta, O. , Gonzalez-Fierro, A. , Chavez-Blanco, A. , de la Cruz-Hernandez, E. , Camargo, M.F. , 2007. A phase II study of epigenetic therapy with hydralazine and magnesium valproate to overcome chemotherapy resistance in refractory solid tumors. Annals of Oncology. 18, 1529–1538. [DOI] [PubMed] [Google Scholar]
- Candelaria, M. , Herrera, A. , Labardini, J. , Gonzalez-Fierro, A. , Trejo-Becerril, C. , Taja-Chayeb, L. , Perez-Cardenas, E. , de la Cruz-Hernandez, E. , Arias-Bofill, D. , Vidal, S. , 2011. Hydralazine and magnesium valproate as epigenetic treatment for myelodysplastic syndrome. Preliminary results of a phase-II trial. Annals of Hematology. 90, 379–387. [DOI] [PubMed] [Google Scholar]
- Carafa, V. , Nebbioso, A. , Altucci, L. , 2012. Sirtuins and disease: the road ahead. Frontiers in Pharmacology. 3, 4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carew, J.S. , Giles, F.J. , Nawrocki, S.T. , 2008. Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Letters. 269, 7–17. [DOI] [PubMed] [Google Scholar]
- Cashen, A. , Juckett, M. , Jumonville, A. , Litzow, M. , Flynn, P.J. , Eckardt, J. , LaPlant, B. , Laumann, K. , Erlichman, C. , DiPersio, J. , 2012. Phase II study of the histone deacetylase inhibitor belinostat (PXD101) for the treatment of myelodysplastic syndrome (MDS). Annals of Hematology. 91, 33–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cebrat, M. , Kim, C.M. , Thompson, P.R. , Daugherty, M. , Cole, P.A. , 2003. Synthesis and analysis of potential prodrugs of coenzyme A analogues for the inhibition of the histone acetyltransferase p300. Bioorganic & Medicinal Chemistry. 11, 3307–3313. [DOI] [PubMed] [Google Scholar]
- Chaib, H. , Prebet, T. , Vey, N. , Collette, Y. , 2011. Histone methyltransferases: a new class of therapeutic targets in cancer treatment?. Medecine Sciences. 27, 725–732. [DOI] [PubMed] [Google Scholar]
- Chen, W.F. , Huang, F. , Zha, J. , 2012. The comparison of short-term efficacies between decitabine and HA regimen for MDS/AML patients. Zhonghua Xue Ye Xue Za Zhi. 33, 143–144. [PubMed] [Google Scholar]
- Chestnut, B.A. , Chang, Q. , Price, A. , Lesuisse, C. , Wong, M. , Martin, L.J. , 2011. Epigenetic regulation of motor neuron cell death through DNA methylation. The Journal of Neuroscience. 31, 16619–16636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chin, S.P. , Dickinson, J.L. , Holloway, A.F. , 2011. Epigenetic regulation of prostate cancer. Clinical Epigenetics. 2, 151–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Choo, K.B. , 2011. Epigenetics in disease and cancer. The Malaysian Journal of Pathology. 33, 61–70. [PubMed] [Google Scholar]
- Coiffier, B. , Pro, B. , Prince, H.M. , Foss, F. , Sokol, L. , Greenwood, M. , Caballero, D. , Borchmann, P. , Morschhauser, F. , Wilhelm, M. , 2012. Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. Journal of Clinical Oncology. 30, 631–636. [DOI] [PubMed] [Google Scholar]
- Copeland, R.A. , Solomon, M.E. , Richon, V.M. , 2009. Protein methyltransferases as a target class for drug discovery. Nature Reviews Drug Discovery. 8, 724–732. [DOI] [PubMed] [Google Scholar]
- Crisanti, M.C. , Wallace, A.F. , Kapoor, V. , Vandermeers, F. , Dowling, M.L. , Pereira, L.P. , Coleman, K. , Campling, B.G. , Fridlender, Z.G. , Kao, G.D. , Albelda, S.M. , 2009. The HDAC inhibitor panobinostat (LBH589) inhibits mesothelioma and lung cancer cells in vitro and in vivo with particular efficacy for small cell lung cancer. Molecular Cancer Therapeutics. 8, 2221–2231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cudkowicz, M.E. , Andres, P.L. , Macdonald, S.A. , Bedlack, R.S. , Choudry, R. , Brown, R.H. , Zhang, H. , Schoenfeld, D.A. , Shefner, J. , Matson, S. , 2009. Phase 2 study of sodium phenylbutyrate in ALS. Amyotrophic Lateral Sclerosis. 10, 99–106. [DOI] [PubMed] [Google Scholar]
- Dandawate, P.R. , Vyas, A. , Ahmad, A. , Banerjee, S. , Deshpande, J. , Swamy, K.V. , Jamadar, A. , Dumhe-Klaire, A.C. , Padhye, S. , Sarkar, F.H. , 2012. Inclusion complex of novel curcumin analogue CDF and beta-cyclodextrin (1:2) and its enhanced in vivo anticancer activity against pancreatic cancer. Pharmaceutical Research. 29, 1775–1786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darkin-Rattray, S.J. , Gurnett, A.M. , Myers, R.W. , Dulski, P.M. , Crumley, T.M. , Allocco, J.J. , Cannova, C. , Meinke, P.T. , Colletti, S.L. , Bednarek, M.A. , 1996. Apicidin: a novel antiprotozoal agent that inhibits parasite histone deacetylase. Proceedings of the National Academy of Sciences of the United States of America. 93, 13143–13147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darvesh, A.S. , Carroll, R.T. , Bishayee, A. , Novotny, N.A. , Geldenhuys, W.J. , Van der Schyf, C.J. , 2012. Curcumin and neurodegenerative diseases: a perspective. Expert Opinion on Investigational Drugs. 21, 1123–1140. [DOI] [PubMed] [Google Scholar]
- Das, P.M. , Singal, R. , 2004. DNA methylation and cancer. Journal of Clinical Oncology. 22, 4632–4642. [DOI] [PubMed] [Google Scholar]
- Datta, J. , Ghoshal, K. , Denny, W.A. , Gamage, S.A. , Brooke, D.G. , Phiasivongsa, P. , Redkar, S. , Jacob, S.T. , 2009. A new class of quinoline-based DNA hypomethylating agents reactivates tumor suppressor genes by blocking DNA methyltransferase 1 activity and inducing its degradation. Cancer Research. 69, 4277–4285. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- Daud, A.I. , Dawson, J. , DeConti, R.C. , Bicaku, E. , Marchion, D. , Bastien, S. , Hausheer, F.A. , Lush, R. , Neuger, A. , Sullivan, D.M. , Munster, P.N. , 2009. Potentiation of a topoisomerase I inhibitor, karenitecin, by the histone deacetylase inhibitor valproic acid in melanoma: translational and phase I/II clinical trial. Clinical Cancer Research. 15, 2479–2487. [DOI] [PubMed] [Google Scholar]
- Davis, A.J. , Gelmon, K.A. , Siu, L.L. , Moore, M.J. , Britten, C.D. , Mistry, N. , Klamut, H. , D'Aloisio, S. , MacLean, M. , Wainman, N. , 2003. Phase I and pharmacologic study of the human DNA methyltransferase antisense oligodeoxynucleotide MG98 given as a 21-day continuous infusion every 4 weeks. Investigational New Drugs. 21, 85–97. [DOI] [PubMed] [Google Scholar]
- Deng, C.X. , 2009. SIRT1, is it a tumor promoter or tumor suppressor?. International Journal of Biological Sciences. 5, 147–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Croce, L. , Raker, V.A. , Corsaro, M. , Fazi, F. , Fanelli, M. , Faretta, M. , Fuks, F. , Lo Coco, F. , Kouzarides, T. , Nervi, C. , 2002. Methyltransferase recruitment and DNA hypermethylation of target promoters by an oncogenic transcription factor. Science. 295, 1079–1082. [DOI] [PubMed] [Google Scholar]
- Dokmanovic, M. , Clarke, C. , Marks, P.A. , 2007. Histone deacetylase inhibitors: overview and perspectives. Molecular Cancer Research. 5, 981–989. [DOI] [PubMed] [Google Scholar]
- Dong, S.W. , Cui, Y.T. , Zhong, R.R. , Liang, D.C. , Liu, Y.M. , Wang, Y.G. , He, Z. , Wang, S. , Liang, S.J. , Zhang, P. , 2012. Decreased expression of retinoblastoma protein-interacting zinc-finger gene 1 in human esophageal squamous cell cancer by DNA methylation. Clinical Laboratory. 58, 41–51. [PubMed] [Google Scholar]
- Dowdell, K.C. , Pesnicak, L. , Hoffmann, V. , Steadman, K. , Remaley, A.T. , Cohen, J.I. , Straus, S.E. , Rao, V.K. , 2009. Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, diminishes lymphoproliferation in the Fas -deficient MRL/lpr(-/-) murine model of autoimmune lymphoproliferative syndrome (ALPS). Experimental Hematology. 37, 487–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dummer, R. , Beyer, M. , Hymes, K. , Epping, M.T. , Bernards, R. , Steinhoff, M. , Sterry, W. , Kerl, H. , Heath, K. , Ahern, J.D. , 2012. Vorinostat combined with bexarotene for treatment of cutaneous T-cell lymphoma: in vitro and phase I clinical evidence supporting augmentation of retinoic acid receptor/retinoid X receptor activation by histone deacetylase inhibition. Leukemia & Lymphoma. 53, 1501–1508. [DOI] [PubMed] [Google Scholar]
- Duvic, M. , Vu, J. , 2007. Vorinostat: a new oral histone deacetylase inhibitor approved for cutaneous T-cell lymphoma. Expert Opinion on Investigational Drugs. 16, 1111–1120. [DOI] [PubMed] [Google Scholar]
- Egger, G. , Liang, G. , Aparicio, A. , Jones, P.A. , 2004. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 429, 457–463. [DOI] [PubMed] [Google Scholar]
- Eglen, R.M. , Reisine, T. , 2011. Screening for compounds that modulate epigenetic regulation of the transcriptome: an overview. Journal of Biomolecular Screening. 16, 1137–1152. [DOI] [PubMed] [Google Scholar]
- El-Khoury, V. , Moussay, E. , Janji, B. , Palissot, V. , Aouali, N. , Brons, N.H. , Van Moer, K. , Pierson, S. , Van Dyck, E. , Berchem, G. , 2010. The histone deacetylase inhibitor MGCD0103 induces apoptosis in B-cell chronic lymphocytic leukemia cells through a mitochondria-mediated caspase activation cascade. Molecular Cancer Therapeutics. 9, 1349–1360. [DOI] [PubMed] [Google Scholar]
- Ellis, L. , Pan, Y. , Smyth, G.K. , George, D.J. , McCormack, C. , Williams-Truax, R. , Mita, M. , Beck, J. , Burris, H. , Ryan, G. , 2008. Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma. Clinical Cancer Research. 14, 4500–4510. [DOI] [PubMed] [Google Scholar]
- Estiu, G. , Greenberg, E. , Harrison, C.B. , Kwiatkowski, N.P. , Mazitschek, R. , Bradner, J.E. , Wiest, O. , 2008. Structural origin of selectivity in class II-selective histone deacetylase inhibitors. Journal of Medicinal Chemistry. 51, 2898–2906. [DOI] [PubMed] [Google Scholar]
- Fakih, M.G. , Groman, A. , McMahon, J. , Wilding, G. , Muindi, J.R. , 2012. A randomized phase II study of two doses of vorinostat in combination with 5-FU/LV in patients with refractory colorectal cancer. Cancer Chemotherapy and Pharmacology. 69, 743–751. [DOI] [PubMed] [Google Scholar]
- Fandy, T.E. , 2009. Development of DNA methyltransferase inhibitors for the treatment of neoplastic diseases. Current Medicinal Chemistry. 16, 2075–2085. [DOI] [PubMed] [Google Scholar]
- Fandy, T.E. , Herman, J.G. , Kerns, P. , Jiemjit, A. , Sugar, E.A. , Choi, S.H. , Yang, A.S. , Aucott, T. , Dauses, T. , Odchimar-Reissig, R. , 2009. Early epigenetic changes and DNA damage do not predict clinical response in an overlapping schedule of 5-azacytidine and entinostat in patients with myeloid malignancies. Blood. 114, 2764–2773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fang, M. , Chen, D. , Yang, C.S. , 2007. Dietary polyphenols may affect DNA methylation. The Journal of Nutrition. 137, 223S–228S. [DOI] [PubMed] [Google Scholar]
- Feinberg, A.P. , Ohlsson, R. , Henikoff, S. , 2006. The epigenetic progenitor origin of human cancer. Nature Reviews Genetics. 7, 21–33. [DOI] [PubMed] [Google Scholar]
- Feinberg, A.P. , Tycko, B. , 2004. The history of cancer epigenetics. Nature Reviews Cancer. 4, 143–153. [DOI] [PubMed] [Google Scholar]
- Feng, R. , Oton, A. , Mapara, M.Y. , Anderson, G. , Belani, C. , Lentzsch, S. , 2007. The histone deacetylase inhibitor, PXD101, potentiates bortezomib-induced anti-multiple myeloma effect by induction of oxidative stress and DNA damage. British Journal of Haematology. 139, 385–397. [DOI] [PubMed] [Google Scholar]
- Fiskus, W. , Wang, Y. , Sreekumar, A. , Buckley, K.M. , Shi, H. , Jillella, A. , Ustun, C. , Rao, R. , Fernandez, P. , Chen, J. , 2009. Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells. Blood. 114, 2733–2743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fonseca, A.L. , Kugelberg, J. , Starker, L.F. , Scholl, U. , Choi, M. , Hellman, P. , Akerstrom, G. , Westin, G. , Lifton, R.P. , Bjorklund, P. , Carling, T. , 2012. Comprehensive DNA methylation analysis of benign and malignant adrenocortical tumors. Genes, Chromosomes & Cancer. 51, (10) 949–960. [DOI] [PubMed] [Google Scholar]
- Force, J. , Rajan, A. , Dombi, E. , Steinberg, S.M. , Giaccone, G. , 2011. Assessment of objective responses using volumetric evaluation in advanced thymic malignancies and metastatic non-small cell lung cancer. Journal of Thoracic Oncology. 6, 1267–1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Forneris, F. , Binda, C. , Vanoni, M.A. , Battaglioli, E. , Mattevi, A. , 2005. Human histone demethylase LSD1 reads the histone code. The Journal of Biological Chemistry. 280, 41360–41365. [DOI] [PubMed] [Google Scholar]
- Foulks, J.M. , Parnell, K.M. , Nix, R.N. , Chau, S. , Swierczek, K. , Saunders, M. , Wright, K. , Hendrickson, T.F. , Ho, K.K. , McCullar, M.V. , Kanner, S.B. , 2012. Epigenetic drug discovery: targeting DNA methyltransferases. Journal of Biomolecular Screening. 17, 2–17. [DOI] [PubMed] [Google Scholar]
- Fournel, M. , Bonfils, C. , Hou, Y. , Yan, P.T. , Trachy-Bourget, M.C. , Kalita, A. , Liu, J. , Lu, A.H. , Zhou, N.Z. , Robert, M.F. , 2008. MGCD0103, a novel isotype-selective histone deacetylase inhibitor, has broad spectrum antitumor activity in vitro and in vivo. Molecular Cancer Therapeutics. 7, 759–768. [DOI] [PubMed] [Google Scholar]
- Friday, B.B. , Anderson, S.K. , Buckner, J. , Yu, C. , Giannini, C. , Geoffroy, F. , Schwerkoske, J. , Mazurczak, M. , Gross, H. , Pajon, E. , 2012. Phase II trial of vorinostat in combination with bortezomib in recurrent glioblastoma: a north central cancer treatment group study. Neuro-oncology. 14, 215–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fukutomi, A. , Hatake, K. , Matsui, K. , Sakajiri, S. , Hirashima, T. , Tanii, H. , Kobayashi, K. , Yamamoto, N. , 2012. A phase I study of oral panobinostat (LBH589) in Japanese patients with advanced solid tumors. Investigational New Drugs. 30, 1096–1106. [DOI] [PubMed] [Google Scholar]
- Fulda, S. , Debatin, K.M. , 2004. Sensitization for anticancer drug-induced apoptosis by the chemopreventive agent resveratrol. Oncogene. 23, 6702–6711. [DOI] [PubMed] [Google Scholar]
- Galli, M. , Salmoiraghi, S. , Golay, J. , Gozzini, A. , Crippa, C. , Pescosta, N. , Rambaldi, A. , 2010. A phase II multiple dose clinical trial of histone deacetylase inhibitor ITF2357 in patients with relapsed or progressive multiple myeloma. Annals of Hematology. 89, 185–190. [DOI] [PubMed] [Google Scholar]
- Galmarini, C.M. , Mackey, J.R. , Dumontet, C. , 2001. Nucleoside analogues: mechanisms of drug resistance and reversal strategies. Leukemia. 15, 875–890. [DOI] [PubMed] [Google Scholar]
- Garcia-Manero, G. , 2012. Myelodysplastic syndromes: 2012 update on diagnosis, risk-stratification, and management. American Journal of Hematology. 87, 692–701. [DOI] [PubMed] [Google Scholar]
- Garcia-Manero, G. , Assouline, S. , Cortes, J. , Estrov, Z. , Kantarjian, H. , Yang, H. , Newsome, W.M. , Miller, W.H. , Rousseau, C. , Kalita, A. , 2008. Phase 1 study of the oral isotype specific histone deacetylase inhibitor MGCD0103 in leukemia. Blood. 112, 981–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia-Manero, G. , Yang, H. , Bueso-Ramos, C. , Ferrajoli, A. , Cortes, J. , Wierda, W.G. , Faderl, S. , Koller, C. , Morris, G. , Rosner, G. , 2008. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood. 111, 1060–1066. [DOI] [PubMed] [Google Scholar]
- Gardian, G. , Browne, S.E. , Choi, D.K. , Klivenyi, P. , Gregorio, J. , Kubilus, J.K. , Ryu, H. , Langley, B. , Ratan, R.R. , Ferrante, R.J. , Beal, M.F. , 2005. Neuroprotective effects of phenylbutyrate in the N171-82Q transgenic mouse model of Huntington's disease. The Journal of Biological Chemistry. 280, 556–563. [DOI] [PubMed] [Google Scholar]
- George, P. , Bali, P. , Annavarapu, S. , Scuto, A. , Fiskus, W. , Guo, F. , Sigua, C. , Sondarva, G. , Moscinski, L. , Atadja, P. , Bhalla, K. , 2005. Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3. Blood. 105, 1768–1776. [DOI] [PubMed] [Google Scholar]
- Ghoneum, M. , Gollapudi, S. , 2011. Synergistic apoptotic effect of arabinoxylan rice bran (MGN-3/Biobran) and curcumin (turmeric) on human multiple myeloma cell line U266 in vitro. Neoplasma. 58, 118–123. [DOI] [PubMed] [Google Scholar]
- Gillenwater, A.M. , Zhong, M. , Lotan, R. , 2007. Histone deacetylase inhibitor suberoylanilide hydroxamic acid induces apoptosis through both mitochondrial and Fas (Cd95) signaling in head and neck squamous carcinoma cells. Molecular Cancer Therapeutics. 6, 2967–2975. [DOI] [PubMed] [Google Scholar]
- Gimsing, P. , Hansen, M. , Knudsen, L.M. , Knoblauch, P. , Christensen, I.J. , Ooi, C.E. , Buhl-Jensen, P. , 2008. A phase I clinical trial of the histone deacetylase inhibitor belinostat in patients with advanced hematological neoplasia. European Journal of Haematology. 81, 170–176. [DOI] [PubMed] [Google Scholar]
- Glozak, M.A. , Seto, E. , 2007. Histone deacetylases and cancer. Oncogene. 26, 5420–5432. [DOI] [PubMed] [Google Scholar]
- Goffin, J. , Eisenhauer, E. , 2002. DNA methyltransferase inhibitors-state of the art. Annals of Oncology. 13, 1699–1716. [DOI] [PubMed] [Google Scholar]
- Gojo, I. , Jiemjit, A. , Trepel, J.B. , Sparreboom, A. , Figg, W.D. , Rollins, S. , Tidwell, M.L. , Greer, J. , Chung, E.J. , Lee, M.J. , 2007. Phase 1 and pharmacologic study of MS-275, a histone deacetylase inhibitor, in adults with refractory and relapsed acute leukemias. Blood. 109, 2781–2790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goll, M.G. , Bestor, T.H. , 2005. Eukaryotic cytosine methyltransferases. Annual Review of Biochemistry. 74, 481–514. [DOI] [PubMed] [Google Scholar]
- Gore, L. , Rothenberg, M.L. , O'Bryant, C.L. , Schultz, M.K. , Sandler, A.B. , Coffin, D. , McCoy, C. , Schott, A. , Scholz, C. , Eckhardt, S.G. , 2008. A phase I and pharmacokinetic study of the oral histone deacetylase inhibitor, MS-275, in patients with refractory solid tumors and lymphomas. Clinical Cancer Research. 14, 4517–4525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gravina, G.L. , Marampon, F. , Piccolella, M. , Motta, M. , Ventura, L. , Pomante, R. , Popov, V.M. , Zani, B.M. , Pestell, R.G. , Tombolini, V. , 2011. Hormonal therapy promotes hormone-resistant phenotype by increasing DNMT activity and expression in prostate cancer models. Endocrinology. 152, 4550–4561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gray, J. , Cubitt, C.L. , Zhang, S. , Chiappori, A. , 2012. Combination of HDAC and topoisomerase inhibitors in small cell lung cancer. Cancer Biology & Therapy. 13, 614–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greer, E.L. , Shi, Y. , 2012. Histone methylation: a dynamic mark in health, disease and inheritance. Nature Reviews Genetics. 13, 343–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greiner, D. , Bonaldi, T. , Eskeland, R. , Roemer, E. , Imhof, A. , 2005. Identification of a specific inhibitor of the histone methyltransferase SU(VAR)3-9. Nature Chemical Biology. 1, 143–145. [DOI] [PubMed] [Google Scholar]
- Gu, B. , Ding, Q. , Xia, G. , Fang, Z. , 2009. EGCG inhibits growth and induces apoptosis in renal cell carcinoma through TFPI-2 overexpression. Oncology Reports. 21, 635–640. [PubMed] [Google Scholar]
- Guo, L.D. , Chen, X.J. , Hu, Y.H. , Yu, Z.J. , Wang, D. , Liu, J.Z. , 2012. Curcumin inhibits proliferation and induces apoptosis of human colorectal cancer cells by activating the mitochondria apoptotic pathway. Phytotherapy Research. 26, http://dx.doi.org/10.1002/ptr.4731 [DOI] [PubMed] [Google Scholar]
- Gupta, S.C. , Kannappan, R. , Reuter, S. , Kim, J.H. , Aggarwal, B.B. , 2011. Chemosensitization of tumors by resveratrol. Annals of the New York Academy of Sciences. 1215, 150–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guz, J. , Foksinski, M. , Olinski, R. , 2010. Global DNA hipomethylation – the meaning in carcinogenesis. Postepy Biochemii. 56, 16–21. [PubMed] [Google Scholar]
- Hagemann, S. , Heil, O. , Lyko, F. , Brueckner, B. , 2011. Azacytidine and decitabine induce gene-specific and non-random DNA demethylation in human cancer cell lines. PLoS One. 6, e17388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haggarty, S.J. , Koeller, K.M. , Wong, J.C. , Grozinger, C.M. , Schreiber, S.L. , 2003. Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation. Proceedings of the National Academy of Sciences of the United States of America. 100, 4389–4394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hahn, W.C. , Weinberg, R.A. , 2002. Rules for making human tumor cells. The New England Journal of Medicine. 347, 1593–1603. [DOI] [PubMed] [Google Scholar]
- Harfe, B.D. , 2005. MicroRNAs in vertebrate development. Current Opinion in Genetics & Development. 15, 410–415. [DOI] [PubMed] [Google Scholar]
- Hatziapostolou, M. , Iliopoulos, D. , 2011. Epigenetic aberrations during oncogenesis. Cellular and Molecular Life Sciences. 68, 1681–1702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hogarth, P. , Lovrecic, L. , Krainc, D. , 2007. Sodium phenylbutyrate in Huntington's disease: a dose-finding study. Movement Disorders. 22, 1962–1964. [DOI] [PubMed] [Google Scholar]
- Huang, H.C. , Chang, P. , Dai, X.L. , Jiang, Z.F. , 2012. Protective effects of curcumin on amyloid-beta-induced neuronal oxidative damage. Neurochemical Research. 37, 1584–1597. [DOI] [PubMed] [Google Scholar]
- Ikehata, M. , Ueda, K. , Iwakawa, S. , 2012. Different involvement of DNA methylation and histone deacetylation in the expression of solute-carrier transporters in 4 colon cancer cell lines. Biological & Pharmaceutical Bulletin. 35, 301–307. [DOI] [PubMed] [Google Scholar]
- Isham, C.R. , Tibodeau, J.D. , Jin, W. , Xu, R. , Timm, M.M. , Bible, K.C. , 2007. Chaetocin: a promising new antimyeloma agent with in vitro and in vivo activity mediated via imposition of oxidative stress. Blood. 109, 2579–2588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iwamoto, F.M. , Lamborn, K.R. , Kuhn, J.G. , Wen, P.Y. , Yung, W.K. , Gilbert, M.R. , Chang, S.M. , Lieberman, F.S. , Prados, M.D. , Fine, H.A. , 2011. A phase I/II trial of the histone deacetylase inhibitor romidepsin for adults with recurrent malignant glioma: North American Brain Tumor Consortium Study 03-03. Neuro-oncology. 13, 509–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Iyer, L.M. , Abhiman, S. , Aravind, L. , 2011. Natural history of eukaryotic DNA methylation systems. Progress in Molecular Biology and Translational Science. 101, 25–104. [DOI] [PubMed] [Google Scholar]
- Jenuwein, T. , Allis, C.D. , 2001. Translating the histone code. Science. 293, 1074–1080. [DOI] [PubMed] [Google Scholar]
- Jones, P. , Bottomley, M.J. , Carfi, A. , Cecchetti, O. , Ferrigno, F. , Lo Surdo, P. , Ontoria, J.M. , Rowley, M. , Scarpelli, R. , Schultz-Fademrecht, C. , Steinkuhler, C. , 2008. 2-Trifluoroacetylthiophenes, a novel series of potent and selective class II histone deacetylase inhibitors. Bioorganic & Medicinal Chemistry Letters. 18, 3456–3461. [DOI] [PubMed] [Google Scholar]
- Jones, P.A. , Baylin, S.B. , 2002. The fundamental role of epigenetic events in cancer. Nature Reviews Genetics. 3, 415–428. [DOI] [PubMed] [Google Scholar]
- Jones, P.A. , Baylin, S.B. , 2007. The epigenomics of cancer. Cell. 128, 683–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jurkowska, R.Z. , Jurkowski, T.P. , Jeltsch, A. , 2011. Structure and function of mammalian DNA methyltransferases. Chembiochem. 12, 206–222. [DOI] [PubMed] [Google Scholar]
- Jurkowski, T.P. , Jeltsch, A. , 2011. On the evolutionary origin of eukaryotic DNA methyltransferases and Dnmt2. PLoS One. 6, e28104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kadia, T.M. , Jabbour, E. , Kantarjian, H. , 2011. Failure of hypomethylating agent-based therapy in myelodysplastic syndromes. Seminars in Oncology. 38, 682–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaminskyy, V.O. , Surova, O.V. , Vaculova, A. , Zhivotovsky, B. , 2011. Combined inhibition of DNA methyltransferase and histone deacetylase restores caspase-8 expression and sensitizes SCLC cells to TRAIL. Carcinogenesis. 32, 1450–1458. [DOI] [PubMed] [Google Scholar]
- Karam, J.A. , Fan, J. , Stanfield, J. , Richer, E. , Benaim, E.A. , Frenkel, E. , Antich, P. , Sagalowsky, A.I. , Mason, R.P. , Hsieh, J.T. , 2007. The use of histone deacetylase inhibitor FK228 and DNA hypomethylation agent 5-azacytidine in human bladder cancer therapy. International Journal of Cancer. 120, 1795–1802. [DOI] [PubMed] [Google Scholar]
- Keating, G.M. , 2012. Azacitidine: a review of its use in the management of myelodysplastic syndromes/acute myeloid leukaemia. Drugs. 72, 1111–1136. [DOI] [PubMed] [Google Scholar]
- Khan, N. , Jeffers, M. , Kumar, S. , Hackett, C. , Boldog, F. , Khramtsov, N. , Qian, X. , Mills, E. , Berghs, S.C. , Carey, N. , 2008. Determination of the class and isoform selectivity of small-molecule histone deacetylase inhibitors. The Biochemical Journal. 409, 581–589. [DOI] [PubMed] [Google Scholar]
- Khan, S.B. , Maududi, T. , Barton, K. , Ayers, J. , Alkan, S. , 2004. Analysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myeloma. British Journal of Haematology. 125, 156–161. [DOI] [PubMed] [Google Scholar]
- Kijima, M. , Yoshida, M. , Sugita, K. , Horinouchi, S. , Beppu, T. , 1993. Trapoxin, an antitumor cyclic tetrapeptide, is an irreversible inhibitor of mammalian histone deacetylase. The Journal of Biological Chemistry. 268, 22429–22435. [PubMed] [Google Scholar]
- Kikuchi, J. , Wada, T. , Shimizu, R. , Izumi, T. , Akutsu, M. , Mitsunaga, K. , Noborio-Hatano, K. , Nobuyoshi, M. , Ozawa, K. , Kano, Y. , Furukawa, Y. , 2010. Histone deacetylases are critical targets of bortezomib-induced cytotoxicity in multiple myeloma. Blood. 116, 406–417. [DOI] [PubMed] [Google Scholar]
- Kim, H.S. , Patel, K. , Muldoon-Jacobs, K. , Bisht, K.S. , Aykin-Burns, N. , Pennington, J.D. , van der Meer, R. , Nguyen, P. , Savage, J. , Owens, K.M. , 2010. SIRT3 is a mitochondria-localized tumor suppressor required for maintenance of mitochondrial integrity and metabolism during stress. Cancer Cell. 17, 41–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kimura, S. , Kuramoto, K. , Homan, J. , Naruoka, H. , Ego, T. , Nogawa, M. , Sugahara, S. , Naito, H. , 2012. Antiproliferative and antitumor effects of azacitidine against the human myelodysplastic syndrome cell line SKM-1. Anticancer Research. 32, 795–798. [PubMed] [Google Scholar]
- Kirschbaum, M. , Frankel, P. , Popplewell, L. , Zain, J. , Delioukina, M. , Pullarkat, V. , Matsuoka, D. , Pulone, B. , Rotter, A.J. , Espinoza-Delgado, I. , 2011. Phase II study of vorinostat for treatment of relapsed or refractory indolent non-Hodgkin's lymphoma and mantle cell lymphoma. Journal of Clinical Oncology. 29, 1198–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitazono, M. , Robey, R. , Zhan, Z. , Sarlis, N.J. , Skarulis, M.C. , Aikou, T. , Bates, S. , Fojo, T. , 2001. Low concentrations of the histone deacetylase inhibitor, depsipeptide (FR901228), increase expression of the Na(+)/I(-) symporter and iodine accumulation in poorly differentiated thyroid carcinoma cells. The Journal of Clinical Endocrinology and Metabolism. 86, 3430–3435. [DOI] [PubMed] [Google Scholar]
- Klimek, V.M. , Fircanis, S. , Maslak, P. , Guernah, I. , Baum, M. , Wu, N. , Panageas, K. , Wright, J.J. , Pandolfi, P.P. , Nimer, S.D. , 2008. Tolerability, pharmacodynamics, and pharmacokinetics studies of depsipeptide (romidepsin) in patients with acute myelogenous leukemia or advanced myelodysplastic syndromes. Clinical Cancer Research. 14, 826–832. [DOI] [PubMed] [Google Scholar]
- Klisovic, R.B. , Stock, W. , Cataland, S. , Klisovic, M.I. , Liu, S. , Blum, W. , Green, M. , Odenike, O. , Godley, L. , Burgt, J.V. , 2008. A phase I biological study of MG98, an oligodeoxynucleotide antisense to DNA methyltransferase 1, in patients with high-risk myelodysplasia and acute myeloid leukemia. Clinical Cancer Research. 14, 2444–2449. [DOI] [PubMed] [Google Scholar]
- Knipstein, J. , Gore, L. , 2011. Entinostat for treatment of solid tumors and hematologic malignancies. Expert Opinion on Investigational Drugs. 20, 1455–1467. [DOI] [PubMed] [Google Scholar]
- Kobayashi, T. , Hashimoto, S. , Horie, T. , 1997. Curcumin inhibition of Dermatophagoides farinea-induced interleukin-5 (IL-5) and granulocyte macrophage-colony stimulating factor (GM-CSF) production by lymphocytes from bronchial asthmatics. Biochemical Pharmacology. 54, 819–824. [DOI] [PubMed] [Google Scholar]
- Kozikowski, A.P. , Chen, Y. , Gaysin, A. , Chen, B. , D'Annibale, M.A. , Suto, C.M. , Langley, B.C. , 2007. Functional differences in epigenetic modulators-superiority of mercaptoacetamide-based histone deacetylase inhibitors relative to hydroxamates in cortical neuron neuroprotection studies. Journal of Medicinal Chemistry. 50, 3054–3061. [DOI] [PubMed] [Google Scholar]
- Kramer, O.H. , Gottlicher, M. , Heinzel, T. , 2001. Histone deacetylase as a therapeutic target. Trends in Endocrinology and Metabolism. 12, 294–300. [DOI] [PubMed] [Google Scholar]
- Kubicek, S. , O'Sullivan, R.J. , August, E.M. , Hickey, E.R. , Zhang, Q. , Teodoro, M.L. , Rea, S. , Mechtler, K. , Kowalski, J.A. , Homon, C.A. , 2007. Reversal of H3K9me2 by a small-molecule inhibitor for the G9a histone methyltransferase. Molecular Cell. 25, 473–481. [DOI] [PubMed] [Google Scholar]
- Kuendgen, A. , Bug, G. , Ottmann, O.G. , Haase, D. , Schanz, J. , Hildebrandt, B. , Nachtkamp, K. , Neukirchen, J. , Dienst, A. , Haas, R. , 2011. Treatment of poor-risk myelodysplastic syndromes and acute myeloid leukemia with a combination of 5-azacytidine and valproic acid. Clinical Epigenetics. 2, 389–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kulis, M. , Esteller, M. , 2010. DNA methylation and cancer. Advances in Genetics. 70, 27–56. [DOI] [PubMed] [Google Scholar]
- Kurd, S.K. , Smith, N. , VanVoorhees, A. , Troxel, A.B. , Badmaev, V. , Seykora, J.T. , Gelfand, J.M. , 2008. Oral curcumin in the treatment of moderate to severe psoriasis vulgaris: a prospective clinical trial. Journal of the American Academy of Dermatology. 58, 625–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwon, H.J. , Owa, T. , Hassig, C.A. , Shimada, J. , Schreiber, S.L. , 1998. Depudecin induces morphological reversion of transformed fibroblasts via the inhibition of histone deacetylase. Proceedings of the National Academy of Sciences of the United States of America. 95, 3356–3361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lagneaux, L. , Gillet, N. , Stamatopoulos, B. , Delforge, A. , Dejeneffe, M. , Massy, M. , Meuleman, N. , Kentos, A. , Martiat, P. , Willems, L. , Bron, D. , 2007. Valproic acid induces apoptosis in chronic lymphocytic leukemia cells through activation of the death receptor pathway and potentiates TRAIL response. Experimental Hematology. 35, 1527–1537. [DOI] [PubMed] [Google Scholar]
- Lai, M.T. , Yang, C.C. , Lin, T.Y. , Tsai, F.J. , Chen, W.C. , 2008. Depsipeptide (FK228) inhibits growth of human prostate cancer cells. Urologic Oncology. 26, 182–189. [DOI] [PubMed] [Google Scholar]
- Lau, O.D. , Kundu, T.K. , Soccio, R.E. , Ait-Si-Ali, S. , Khalil, E.M. , Vassilev, A. , Wolffe, A.P. , Nakatani, Y. , Roeder, R.G. , Cole, P.A. , 2000. HATs off: selective synthetic inhibitors of the histone acetyltransferases p300 and PCAF. Molecular Cell. 5, 589–595. [DOI] [PubMed] [Google Scholar]
- Lee, M.G. , Wynder, C. , Schmidt, D.M. , McCafferty, D.G. , Shiekhattar, R. , 2006. Histone H3 lysine 4 demethylation is a target of nonselective antidepressive medications. Chemistry & Biology. 13, 563–567. [DOI] [PubMed] [Google Scholar]
- Lee, W.J. , Shim, J.Y. , Zhu, B.T. , 2005. Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids. Molecular Pharmacology. 68, 1018–1030. [DOI] [PubMed] [Google Scholar]
- Leone, G. , Teofili, L. , Voso, M.T. , Lubbert, M. , 2002. DNA methylation and demethylating drugs in myelodysplastic syndromes and secondary leukemias. Haematologica. 87, 1324–1341. [PubMed] [Google Scholar]
- Lin, L. , Liu, Y. , Li, H. , Li, P.K. , Fuchs, J. , Shibata, H. , Iwabuchi, Y. , Lin, J. , 2011. Targeting colon cancer stem cells using a new curcumin analogue, GO-Y030. British Journal of Cancer. 105, 212–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu, F. , Chen, X. , Allali-Hassani, A. , Quinn, A.M. , Wasney, G.A. , Dong, A. , Barsyte, D. , Kozieradzki, I. , Senisterra, G. , Chau, I. , 2009. Discovery of a 2,4-diamino-7-aminoalkoxyquinazoline as a potent and selective inhibitor of histone lysine methyltransferase G9a. Journal of Medicinal Chemistry. 52, 7950–7953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu, Q.Y. , Chen, D.W. , Xie, L.P. , Zhang, R.Q. , Wang, H.Z. , 2011. Decitabine, independent of apoptosis, exerts its cytotoxic effects on cell growth in melanoma cells. Environmental Toxicology and Pharmacology. 32, 423–429. [DOI] [PubMed] [Google Scholar]
- Liu, Z. , Zhang, L. , Ding, F. , Li, J. , Guo, M. , Li, W. , Wang, Y. , Yu, Z. , Zhan, Q. , Wu, M. , Liu, Z. , 2005. 5-Aza-2'-deoxycytidine induces retinoic acid receptor-beta(2) demethylation and growth inhibition in esophageal squamous carcinoma cells. Cancer Letters. 230, 271–283. [DOI] [PubMed] [Google Scholar]
- Lokk, K. , Vooder, T. , Kolde, R. , Valk, K. , Vosa, U. , Roosipuu, R. , Milani, L. , Fischer, K. , Koltsina, M. , Urgard, E. , 2012. Methylation markers of early-stage non-small cell lung cancer. PLoS One. 7, e39813 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lu, Q. , Wu, A. , Richardson, B.C. , 2005. Demethylation of the same promoter sequence increases CD70 expression in lupus T cells and T cells treated with lupus-inducing drugs. Journal of Immunology. 174, 6212–6219. [DOI] [PubMed] [Google Scholar]
- Lucas, D.M. , Davis, M.E. , Parthun, M.R. , Mone, A.P. , Kitada, S. , Cunningham, K.D. , Flax, E.L. , Wickham, J. , Reed, J.C. , Byrd, J.C. , Grever, M.R. , 2004. The histone deacetylase inhibitor MS-275 induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia cells. Leukemia. 18, 1207–1214. [DOI] [PubMed] [Google Scholar]
- Luchenko, V.L. , Salcido, C.D. , Zhang, Y. , Agama, K. , Komlodi-Pasztor, E. , Murphy, R.F. , Giaccone, G. , Pommier, Y. , Bates, S.E. , Varticovski, L. , 2011. Schedule-dependent synergy of histone deacetylase inhibitors with DNA damaging agents in small cell lung cancer. Cell Cycle. 10, 3119–3128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mabaera, R. , Greene, M.R. , Richardson, C.A. , Conine, S.J. , Kozul, C.D. , Lowrey, C.H. , 2008. Neither DNA hypomethylation nor changes in the kinetics of erythroid differentiation explain 5-azacytidine's ability to induce human fetal hemoglobin. Blood. 111, 411–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mai, A. , Altucci, L. , 2009. Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. The International Journal of Biochemistry & Cell Biology. 41, 199–213. [DOI] [PubMed] [Google Scholar]
- Mai, A. , Massa, S. , Pezzi, R. , Valente, S. , Loidl, P. , Brosch, G. , 2005. Synthesis and biological evaluation of 2-, 3-, and 4-acylaminocinnamyl-N-hydroxyamides as novel synthetic HDAC inhibitors. Medicinal Chemistry. 1, 245–254. [DOI] [PubMed] [Google Scholar]
- Maiso, P. , Carvajal-Vergara, X. , Ocio, E.M. , Lopez-Perez, R. , Mateo, G. , Gutierrez, N. , Atadja, P. , Pandiella, A. , San Miguel, J.F. , 2006. The histone deacetylase inhibitor LBH589 is a potent antimyeloma agent that overcomes drug resistance. Cancer Research. 66, 5781–5789. [DOI] [PubMed] [Google Scholar]
- Marks, P.A. , Breslow, R. , 2007. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nature Biotechnology. 25, 84–90. [DOI] [PubMed] [Google Scholar]
- Marks, P.W. , 2012. Decitabine for acute myeloid leukemia. Expert Review of Anticancer Therapy. 12, 299–305. [DOI] [PubMed] [Google Scholar]
- Mazumder, A. , Vesole, D.H. , Jagannath, S. , 2010. Vorinostat plus bortezomib for the treatment of relapsed/refractory multiple myeloma: a case series illustrating utility in clinical practice. Clinical Lymphoma, Myeloma & Leukemia. 10, 149–151. [DOI] [PubMed] [Google Scholar]
- McGovern, A.P. , Powell, B.E. , Chevassut, T.J. , 2012. A dynamic multi-compartmental model of DNA methylation with demonstrable predictive value in hematological malignancies. Journal of Theoretical Biology. 310C, 14–20. [DOI] [PubMed] [Google Scholar]
- McGuinness, D. , McGuinness, D.H. , McCaul, J.A. , Shiels, P.G. , 2011. Sirtuins, bioageing, and cancer. Journal of Aging Research. 235754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKinsey, T.A. , Olson, E.N. , 2004. Cardiac histone acetylation – therapeutic opportunities abound. Trends in Genetics. 20, 206–213. [DOI] [PubMed] [Google Scholar]
- Meng, X.Y. , Zhu, S.T. , Zhou, Q.Z. , Li, P. , Wang, Y.J. , Zhang, S.T. , 2012. Promoter methylation regulates cigarette smoke-stimulated cyclooxygenase-2 expression in esophageal squamous cell carcinoma. Journal of Digestive Diseases. 13, 208–213. [DOI] [PubMed] [Google Scholar]
- Minucci, S. , Pelicci, P.G. , 2006. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nature Reviews Cancer. 6, 38–51. [DOI] [PubMed] [Google Scholar]
- Mirza, S. , Sharma, G. , Pandya, P. , Ralhan, R. , 2010. Demethylating agent 5-aza-2-deoxycytidine enhances susceptibility of breast cancer cells to anticancer agents. Molecular and Cellular Biochemistry. 342, 101–109. [DOI] [PubMed] [Google Scholar]
- Modesitt, S.C. , Sill, M. , Hoffman, J.S. , Bender, D.P. , 2008. A phase II study of vorinostat in the treatment of persistent or recurrent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. Gynecologic Oncology. 109, 182–186. [DOI] [PubMed] [Google Scholar]
- Moffat, D. , Patel, S. , Day, F. , Belfield, A. , Donald, A. , Rowlands, M. , Wibawa, J. , Brotherton, D. , Stimson, L. , Clark, V. , 2010. Discovery of 2-(6-{[(6-fluoroquinolin-2-yl)methyl]amino}bicyclo[3.1.0]hex-3-yl)-N-hydroxypyrim idine-5-carboxamide (CHR-3996), a class I selective orally active histone deacetylase inhibitor. Journal of Medicinal Chemistry. 53, 8663–8678. [DOI] [PubMed] [Google Scholar]
- Moradei, O. , Vaisburg, A. , Martell, R.E. , 2008. Histone deacetylase inhibitors in cancer therapy: new compounds and clinical update of benzamide-type agents. Current Topics in Medicinal Chemistry. 8, 841–858. [DOI] [PubMed] [Google Scholar]
- Mouzaoui, S. , Rahim, I. , Djerdjouri, B. , 2012. Aminoguanidine and curcumin attenuated tumor necrosis factor (TNF)-alpha-induced oxidative stress, colitis and hepatotoxicity in mice. International Immunopharmacology. 12, 302–311. [DOI] [PubMed] [Google Scholar]
- Munster, P.N. , Thurn, K.T. , Thomas, S. , Raha, P. , Lacevic, M. , Miller, A. , Melisko, M. , Ismail-Khan, R. , Rugo, H. , Moasser, M. , Minton, S.E. , 2011. A phase II study of the histone deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of patients with hormone therapy-resistant breast cancer. British Journal of Cancer. 104, 1828–1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nagaraju, G.P. , Aliya, S. , Zafar, S.F. , Basha, R. , Diaz, R. , El-Rayes, B.F. , 2012. The impact of curcumin on breast cancer. Integrative Biology. 4, (9) 996–1007. [DOI] [PubMed] [Google Scholar]
- Nakajima, H. , Kim, Y.B. , Terano, H. , Yoshida, M. , Horinouchi, S. , 1998. FR901228, a potent antitumor antibiotic, is a novel histone deacetylase inhibitor. Experimental Cell Research. 241, 126–133. [DOI] [PubMed] [Google Scholar]
- Nandakumar, V. , Vaid, M. , Katiyar, S.K. , 2011. (-)-Epigallocatechin-3-gallate reactivates silenced tumor suppressor genes, Cip1/p21 and p16INK4a, by reducing DNA methylation and increasing histones acetylation in human skin cancer cells. Carcinogenesis. 32, 537–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Newmark, H.L. , Lupton, J.R. , Young, C.W. , 1994. Butyrate as a differentiating agent: pharmacokinetics, analogues and current status. Cancer Letters. 78, 1–5. [DOI] [PubMed] [Google Scholar]
- Niesvizky, R. , Ely, S. , Mark, T. , Aggarwal, S. , Gabrilove, J.L. , Wright, J.J. , Chen-Kiang, S. , Sparano, J.A. , 2011. Phase 2 trial of the histone deacetylase inhibitor romidepsin for the treatment of refractory multiple myeloma. Cancer. 117, 336–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nightingale, K.P. , O'Neill, L.P. , Turner, B.M. , 2006. Histone modifications: signalling receptors and potential elements of a heritable epigenetic code. Current Opinion in Genetics & Development. 16, 125–136. [DOI] [PubMed] [Google Scholar]
- Nolan, L. , Johnson, P.W. , Ganesan, A. , Packham, G. , Crabb, S.J. , 2008. Will histone deacetylase inhibitors require combination with other agents to fulfil their therapeutic potential?. British Journal of Cancer. 99, 689–694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Novotny-Diermayr, V. , Hart, S. , Goh, K.C. , Cheong, A. , Ong, L.C. , Hentze, H. , Pasha, M.K. , Jayaraman, R. , Ethirajulu, K. , Wood, J.M. , 2012. The oral HDAC inhibitor pracinostat (SB939) is efficacious and synergistic with the JAK2 inhibitor pacritinib (SB1518) in preclinical models of AML. Blood Cancer Journal. 2, e69 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Novotny-Diermayr, V. , Sangthongpitag, K. , Hu, C.Y. , Wu, X. , Sausgruber, N. , Yeo, P. , Greicius, G. , Pettersson, S. , Liang, A.L. , Loh, Y.K. , 2010. SB939, a novel potent and orally active histone deacetylase inhibitor with high tumor exposure and efficacy in mouse models of colorectal cancer. Molecular Cancer Therapeutics. 9, 642–652. [DOI] [PubMed] [Google Scholar]
- Oki, Y. , Kondo, Y. , Yamamoto, K. , Ogura, M. , Kasai, M. , Kobayashi, Y. , Watanabe, T. , Uike, N. , Ohyashiki, K. , Okamoto, S.I. , 2012. A phase I/II study of decitabine in patients with myelodysplastic syndrome: a multi-center study in Japan. Cancer Science. 103, (10) 1839–18347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olindo, S. , Belrose, G. , Gillet, N. , Rodriguez, S. , Boxus, M. , Verlaeten, O. , Asquith, B. , Bangham, C. , Signate, A. , Smadja, D. , 2011. Safety of long-term treatment of HAM/TSP patients with valproic acid. Blood. 118, 6306–6309. [DOI] [PubMed] [Google Scholar]
- Olivieri, N.F. , Saunthararajah, Y. , Thayalasuthan, V. , Kwiatkowski, J. , Ware, R.E. , Kuypers, F.A. , Kim, H.Y. , Trachtenberg, F.L. , Vichinsky, E.P. , 2011. A pilot study of subcutaneous decitabine in beta-thalassemia intermedia. Blood. 118, 2708–2711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Otterson, G.A. , Hodgson, L. , Pang, H. , Vokes, E.E. , 2010. Phase II study of the histone deacetylase inhibitor romidepsin in relapsed small cell lung cancer (Cancer and Leukemia Group B 30304). Journal of Thoracic Oncology. 5, 1644–1648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patutina, O.A. , Mironova, N.L. , Vlassov, V.V. , Zenkova, M.A. , 2009. New approaches for cancer treatment: antitumor drugs based on gene-targeted nucleic acids. Acta Naturae. 1, 44–60. [PMC free article] [PubMed] [Google Scholar]
- Peedicayil, J. , 2012. The role of DNA methylation in the pathogenesis and treatment of cancer. Current Clinical Pharmacology. 7, (4) 333–340. [DOI] [PubMed] [Google Scholar]
- Peng, X. , Pentassuglia, L. , Sawyer, D.B. , 2010. Emerging anticancer therapeutic targets and the cardiovascular system: is there cause for concern?. Circulation Research. 106, 1022–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phiel, C.J. , Zhang, F. , Huang, E.Y. , Guenther, M.G. , Lazar, M.A. , Klein, P.S. , 2001. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. The Journal of Biological Chemistry. 276, 36734–36741. [DOI] [PubMed] [Google Scholar]
- Piekarz, R.L. , Frye, R. , Prince, H.M. , Kirschbaum, M.H. , Zain, J. , Allen, S.L. , Jaffe, E.S. , Ling, A. , Turner, M. , Peer, C.J. , 2011. Phase 2 trial of romidepsin in patients with peripheral T-cell lymphoma. Blood. 117, 5827–5834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Platzbecker, U. , Schetelig, J. , Finke, J. , Trenschel, R. , Scott, B.L. , Kobbe, G. , Schaefer-Eckart, K. , Bornhauser, M. , Itzykson, R. , Germing, U. , 2012. Allogeneic hematopoietic cell transplantation in patients age 60-70 years with de novo high-risk myelodysplastic syndrome or secondary acute myelogenous leukemia: comparison with patients lacking donors who received azacitidine. Biology of Blood and Marrow Transplantation. 18, (9) 1415–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plitta, B. , Giel-Pietraszuk, M. , Markiewicz, W.T. , Barciszewski, J. , 2011. Cytosine methylation in DNA and its role in cancer therapy. Postepy Biochemii. 57, 24–32. [PubMed] [Google Scholar]
- Plummer, R. , Vidal, L. , Griffin, M. , Lesley, M. , de Bono, J. , Coulthard, S. , Sludden, J. , Siu, L.L. , Chen, E.X. , Oza, A.M. , 2009. Phase I study of MG98, an oligonucleotide antisense inhibitor of human DNA methyltransferase 1, given as a 7-day infusion in patients with advanced solid tumors. Clinical Cancer Research. 15, 3177–3183. [DOI] [PubMed] [Google Scholar]
- Pontiki, E. , Hadjipavlou-Litina, D. , 2012. Histone deacetylase inhibitors (HDACIs). Structure–activity relationships: history and new QSAR perspectives. Medicinal Research Reviews. 32, 1–165. [DOI] [PubMed] [Google Scholar]
- Prebet, T. , Vey, N. , 2011. Vorinostat in acute myeloid leukemia and myelodysplastic syndromes. Expert Opinion on Investigational Drugs. 20, 287–295. [DOI] [PubMed] [Google Scholar]
- Prince, H.M. , Dickinson, M. , 2012. Romidepsin for cutaneous T-cell lymphoma. Clinical Cancer Research. 18, 3509–3515. [DOI] [PubMed] [Google Scholar]
- Rabizadeh, E. , Merkin, V. , Belyaeva, I. , Shaklai, M. , Zimra, Y. , 2007. Pivanex, a histone deacetylase inhibitor, induces changes in BCR-ABL expression and when combined with STI571, acts synergistically in a chronic myelocytic leukemia cell line. Leukemia Research. 31, 1115–1123. [DOI] [PubMed] [Google Scholar]
- Rajendran, G. , Shanmuganandam, K. , Bendre, A. , Muzumdar, D. , Goel, A. , Shiras, A. , 2011. Epigenetic regulation of DNA methyltransferases: DNMT1 and DNMT3B in gliomas. Journal of Neuro-oncology. 104, 483–494. [DOI] [PubMed] [Google Scholar]
- Rambaldi, A. , Dellacasa, C.M. , Finazzi, G. , Carobbio, A. , Ferrari, M.L. , Guglielmelli, P. , Gattoni, E. , Salmoiraghi, S. , Finazzi, M.C. , Di Tollo, S. , 2010. A pilot study of the Histone-Deacetylase inhibitor Givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms. British Journal of Haematology. 150, 446–455. [DOI] [PubMed] [Google Scholar]
- Rapin, J.R. , Wiernsperger, N. , 2010. Possible links between intestinal permeability and food processing: a potential therapeutic niche for glutamine. Clinics (Sao Paulo). 65, 635–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rasheed, W.K. , Johnstone, R.W. , Prince, H.M. , 2007. Histone deacetylase inhibitors in cancer therapy. Expert Opinion on Investigational Drugs. 16, 659–678. [DOI] [PubMed] [Google Scholar]
- Raver-Shapira, N. , Marciano, E. , Meiri, E. , Spector, Y. , Rosenfeld, N. , Moskovits, N. , Bentwich, Z. , Oren, M. , 2007. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Molecular Cell. 26, 731–743. [DOI] [PubMed] [Google Scholar]
- Razak, A.R. , Hotte, S.J. , Siu, L.L. , Chen, E.X. , Hirte, H.W. , Powers, J. , Walsh, W. , Stayner, L.A. , Laughlin, A. , Novotny-Diermayr, V. , 2011. Phase I clinical, pharmacokinetic and pharmacodynamic study of SB939, an oral histone deacetylase (HDAC) inhibitor, in patients with advanced solid tumours. British Journal of Cancer. 104, 756–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ree, A.H. , Dueland, S. , Folkvord, S. , Hole, K.H. , Seierstad, T. , Johansen, M. , Abrahamsen, T.W. , Flatmark, K. , 2010. Vorinostat, a histone deacetylase inhibitor, combined with pelvic palliative radiotherapy for gastrointestinal carcinoma: the Pelvic Radiation and Vorinostat (PRAVO) phase 1 study. The Lancet Oncology. 11, 459–464. [DOI] [PubMed] [Google Scholar]
- Reid, T. , Valone, F. , Lipera, W. , Irwin, D. , Paroly, W. , Natale, R. , Sreedharan, S. , Keer, H. , Lum, B. , Scappaticci, F. , Bhatnagar, A. , 2004. Phase II trial of the histone deacetylase inhibitor pivaloyloxymethyl butyrate (Pivanex, AN-9) in advanced non-small cell lung cancer. Lung Cancer. 45, 381–386. [DOI] [PubMed] [Google Scholar]
- Ren, J. , Singh, B.N. , Huang, Q. , Li, Z. , Gao, Y. , Mishra, P. , Hwa, Y.L. , Li, J. , Dowdy, S.C. , Jiang, S.W. , 2011. DNA hypermethylation as a chemotherapy target. Cellular Signalling. 23, 1082–1093. [DOI] [PubMed] [Google Scholar]
- Rennolds, J. , Malireddy, S. , Hassan, F. , Tridandapani, S. , Parinandi, N. , Boyaka, P.N. , Cormet-Boyaka, E. , 2012. Curcumin regulates airway epithelial cell cytokine responses to the pollutant cadmium. Biochemical and Biophysical Research Communications. 417, 256–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rice, J.C. , Allis, C.D. , 2001. Histone methylation versus histone acetylation: new insights into epigenetic regulation. Current Opinion in Cell Biology. 13, 263–273. [DOI] [PubMed] [Google Scholar]
- Richardson, P. , Mitsiades, C. , Colson, K. , Reilly, E. , McBride, L. , Chiao, J. , Sun, L. , Ricker, J. , Rizvi, S. , Oerth, C. , 2008. Phase I trial of oral vorinostat (suberoylanilide hydroxamic acid, SAHA) in patients with advanced multiple myeloma. Leukemia & Lymphoma. 49, 502–507. [DOI] [PubMed] [Google Scholar]
- Richon, V.M. , Emiliani, S. , Verdin, E. , Webb, Y. , Breslow, R. , Rifkind, R.A. , Marks, P.A. , 1998. A class of hybrid polar inducers of transformed cell differentiation inhibits histone deacetylases. Proceedings of the National Academy of Sciences of the United States of America. 95, 3003–3007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ritchie, E.K. , 2012. Safety and efficacy of azacitidine in the treatment of elderly patients with myelodysplastic syndrome. Clinical Interventions in Aging. 7, 165–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robertson, K.D. , 2001. DNA methylation, methyltransferases, and cancer. Oncogene. 20, 3139–3155. [DOI] [PubMed] [Google Scholar]
- Robertson, K.D. , Wolffe, A.P. , 2000. DNA methylation in health and disease. Nature Reviews Genetics. 1, 11–19. [DOI] [PubMed] [Google Scholar]
- Rosato, R. , Hock, S. , Dent, P. , Dai, Y. , Grant, S. , 2012. LBH-589 (panobinostat) potentiates fludarabine anti-leukemic activity through a JNK- and XIAP-dependent mechanism. Leukemia Research. 36, 491–498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosato, R.R. , Maggio, S.C. , Almenara, J.A. , Payne, S.G. , Atadja, P. , Spiegel, S. , Dent, P. , Grant, S. , 2006. The histone deacetylase inhibitor LAQ824 induces human leukemia cell death through a process involving XIAP down-regulation, oxidative injury, and the acid sphingomyelinase-dependent generation of ceramide. Molecular Pharmacology. 69, 216–225. [DOI] [PubMed] [Google Scholar]
- Rose, C. , Fournier, M. , Nibourel, O. , Herbaux, C. , Charpentier, A. , Renneville, A. , Pissard, S. , Badens, C. , Preudhomme, C. , 2011. Acquired alpha thalassemia myelodyslastic/myeloproliferative syndrome (ATMDS): evolution on hypomethylating agent therapy. Leukemia Research. 35, e203–205. [DOI] [PubMed] [Google Scholar]
- Ruana, B.F. , Lua, X.Q. , Songa, J. , Zhu, H.L. , 2012. Derivatives of resveratrol: potential agents in prevention and treatment of cardiovascular disease. Current Medicinal Chemistry. 19, (24) 4175–4183. [DOI] [PubMed] [Google Scholar]
- Ryu, H. , Smith, K. , Camelo, S.I. , Carreras, I. , Lee, J. , Iglesias, A.H. , Dangond, F. , Cormier, K.A. , Cudkowicz, M.E. , Brown, R.H. , Ferrante, R.J. , 2005. Sodium phenylbutyrate prolongs survival and regulates expression of anti-apoptotic genes in transgenic amyotrophic lateral sclerosis mice. Journal of Neurochemistry. 93, 1087–1098. [DOI] [PubMed] [Google Scholar]
- Saito, A. , Yamashita, T. , Mariko, Y. , Nosaka, Y. , Tsuchiya, K. , Ando, T. , Suzuki, T. , Tsuruo, T. , Nakanishi, O. , 1999. A synthetic inhibitor of histone deacetylase, MS-27-275, with marked in vivo antitumor activity against human tumors. Proceedings of the National Academy of Sciences of the United States of America. 96, 4592–4597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sato, A. , Asano, T. , Horiguchi, A. , Ito, K. , Sumitomo, M. , Asano, T. , 2010. Combination of suberoylanilide hydroxamic acid and ritonavir is effective against renal cancer cells. Urology. 76, 764.e7–764.e13. [DOI] [PubMed] [Google Scholar]
- Sato, N. , Ohta, T. , Kitagawa, H. , Kayahara, M. , Ninomiya, I. , Fushida, S. , Fujimura, T. , Nishimura, G. , Shimizu, K. , Miwa, K. , 2004. FR901228, a novel histone deacetylase inhibitor, induces cell cycle arrest and subsequent apoptosis in refractory human pancreatic cancer cells. International Journal of Oncology. 24, 679–685. [PubMed] [Google Scholar]
- Saunders, L.R. , Verdin, E. , 2007. Sirtuins: critical regulators at the crossroads between cancer and aging. Oncogene. 26, 5489–5504. [DOI] [PubMed] [Google Scholar]
- Saunthararajah, Y. , Hillery, C.A. , Lavelle, D. , Molokie, R. , Dorn, L. , Bressler, L. , Gavazova, S. , Chen, Y.H. , Hoffman, R. , DeSimone, J. , 2003. Effects of 5-aza-2'-deoxycytidine on fetal hemoglobin levels, red cell adhesion, and hematopoietic differentiation in patients with sickle cell disease. Blood. 102, 3865–3870. [DOI] [PubMed] [Google Scholar]
- Schaefer, M. , Lyko, F. , 2010. Solving the Dnmt2 enigma. Chromosoma. 119, 35–40. [DOI] [PubMed] [Google Scholar]
- Schaefer, M. , Pollex, T. , Hanna, K. , Tuorto, F. , Meusburger, M. , Helm, M. , Lyko, F. , 2010. RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes & Development. 24, 1590–1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schirrmacher, E. , Beck, C. , Brueckner, B. , Schmitges, F. , Siedlecki, P. , Bartenstein, P. , Lyko, F. , Schirrmacher, R. , 2006. Synthesis and in vitro evaluation of biotinylated RG108: a high affinity compound for studying binding interactions with human DNA methyltransferases. Bioconjugate Chemistry. 17, 261–266. [DOI] [PubMed] [Google Scholar]
- Schmidt, D.M. , McCafferty, D.G. , 2007. trans-2-Phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1. Biochemistry. 46, 4408–4416. [DOI] [PubMed] [Google Scholar]
- Schneider, B.J. , Kalemkerian, G.P. , Bradley, D. , Smith, D.C. , Egorin, M.J. , Daignault, S. , Dunn, R. , Hussain, M. , 2012. Phase I study of vorinostat (suberoylanilide hydroxamic acid, NSC 701852) in combination with docetaxel in patients with advanced and relapsed solid malignancies. Investigational New Drugs. 30, 249–257. [DOI] [PubMed] [Google Scholar]
- Schrump, D.S. , Fischette, M.R. , Nguyen, D.M. , Zhao, M. , Li, X. , Kunst, T.F. , Hancox, A. , Hong, J.A. , Chen, G.A. , Kruchin, E. , 2008. Clinical and molecular responses in lung cancer patients receiving romidepsin. Clinical Cancer Research. 14, 188–198. [DOI] [PubMed] [Google Scholar]
- Schrump, D.S. , Fischette, M.R. , Nguyen, D.M. , Zhao, M. , Li, X. , Kunst, T.F. , Hancox, A. , Hong, J.A. , Chen, G.A. , Pishchik, V. , 2006. Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura. Clinical Cancer Research. 12, 5777–5785. [DOI] [PubMed] [Google Scholar]
- Scuto, A. , Kirschbaum, M. , Kowolik, C. , Kretzner, L. , Juhasz, A. , Atadja, P. , Pullarkat, V. , Bhatia, R. , Forman, S. , Yen, Y. , Jove, R. , 2008. The novel histone deacetylase inhibitor, LBH589, induces expression of DNA damage response genes and apoptosis in Ph- acute lymphoblastic leukemia cells. Blood. 111, 5093–5100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shabbeer, S. , Williams, S.A. , Simons, B.W. , Herman, J.G. , Carducci, M.A. , 2012. Progression of prostate carcinogenesis and dietary methyl donors: temporal dependence. Cancer Prevention Research (Philadelphia, PA). 5, 229–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shang, D. , Liu, Y. , Matsui, Y. , Ito, N. , Nishiyama, H. , Kamoto, T. , Ogawa, O. , 2008. Demethylating agent 5-aza-2'-deoxycytidine enhances susceptibility of bladder transitional cell carcinoma to Cisplatin. Urology. 71, 1220–1225. [DOI] [PubMed] [Google Scholar]
- Shankar, S. , Srivastava, R.K. , 2008. Histone deacetylase inhibitors: mechanisms and clinical significance in cancer: HDAC inhibitor-induced apoptosis. Advances in Experimental Medicine and Biology. 615, 261–298. [DOI] [PubMed] [Google Scholar]
- Shao, W. , Growney, J.D. , Feng, Y. , O'Connor, G. , Pu, M. , Zhu, W. , Yao, Y.M. , Kwon, P. , Fawell, S. , Atadja, P. , 2010. Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T-cell lymphoma models: defining molecular mechanisms of resistance. International Journal of Cancer. 127, 2199–2208. [DOI] [PubMed] [Google Scholar]
- Shi, Y. , Desponts, C. , Do, J.T. , Hahm, H.S. , Scholer, H.R. , Ding, S. , 2008. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell Stem Cell. 3, 568–574. [DOI] [PubMed] [Google Scholar]
- Shi, Y.K. , Li, Z.H. , Han, X.Q. , Yi, J.H. , Wang, Z.H. , Hou, J.L. , Feng, C.R. , Fang, Q.H. , Wang, H.H. , Zhang, P.F. , 2010. The histone deacetylase inhibitor suberoylanilide hydroxamic acid induces growth inhibition and enhances taxol-induced cell death in breast cancer. Cancer Chemotherapy and Pharmacology. 66, 1131–1140. [DOI] [PubMed] [Google Scholar]
- Shukla, V. , Vaissiere, T. , Herceg, Z. , 2008. Histone acetylation and chromatin signature in stem cell identity and cancer. Mutation Research. 637, 1–15. [DOI] [PubMed] [Google Scholar]
- Siegel, D. , Hussein, M. , Belani, C. , Robert, F. , Galanis, E. , Richon, V.M. , Garcia-Vargas, J. , Sanz-Rodriguez, C. , Rizvi, S. , 2009. Vorinostat in solid and hematologic malignancies. Journal of Hematology & Oncology. 2, 31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simon, J.A. , Lange, C.A. , 2008. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutation Research. 647, 21–29. [DOI] [PubMed] [Google Scholar]
- Singh, N. , Duenas-Gonzalez, A. , Lyko, F. , Medina-Franco, J.L. , 2009. Molecular modeling and molecular dynamics studies of hydralazine with human DNA methyltransferase 1. ChemMedChem. 4, 792–799. [DOI] [PubMed] [Google Scholar]
- Siu, L.L. , Pili, R. , Duran, I. , Messersmith, W.A. , Chen, E.X. , Sullivan, R. , MacLean, M. , King, S. , Brown, S. , Reid, G.K. , 2008. Phase I study of MGCD0103 given as a three-times-per-week oral dose in patients with advanced solid tumors. Journal of Clinical Oncology. 26, 1940–1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith, B.C. , Denu, J.M. , 2009. Chemical mechanisms of histone lysine and arginine modifications. Biochimica et Biophysica Acta. 1789, 45–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Somoza, J.R. , Skene, R.J. , Katz, B.A. , Mol, C. , Ho, J.D. , Jennings, A.J. , Luong, C. , Arvai, A. , Buggy, J.J. , Chi, E. , 2004. Structural snapshots of human HDAC8 provide insights into the class I histone deacetylases. Structure. 12, 1325–1334. [DOI] [PubMed] [Google Scholar]
- Son, D.S. , Wilson, A.J. , Parl, A.K. , Khabele, D. , 2010. The effects of the histone deacetylase inhibitor romidepsin (FK228) are enhanced by aspirin (ASA) in COX-1 positive ovarian cancer cells through augmentation of p21. Cancer Biology & Therapy. 9, 928–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song, S.H. , Han, S.W. , Bang, Y.J. , 2011. Epigenetic-based therapies in cancer: progress to date. Drugs. 71, 2391–2403. [DOI] [PubMed] [Google Scholar]
- Sorm, F. , Piskala, A. , Cihak, A. , Vesely, J. , 1964. 5-Azacytidine, a new, highly effective cancerostatic. Experientia. 20, 202–203. [DOI] [PubMed] [Google Scholar]
- Souto, J.A. , Benedetti, R. , Otto, K. , Miceli, M. , Alvarez, R. , Altucci, L. , de Lera, A.R. , 2010. New anacardic acid-inspired benzamides: histone lysine acetyltransferase activators. ChemMedChem. 5, 1530–1540. [DOI] [PubMed] [Google Scholar]
- Souto, J.A. , Conte, M. , Alvarez, R. , Nebbioso, A. , Carafa, V. , Altucci, L. , de Lera, A.R. , 2008. Synthesis of benzamides related to anacardic acid and their histone acetyltransferase (HAT) inhibitory activities. ChemMedChem. 3, 1435–1442. [DOI] [PubMed] [Google Scholar]
- Spannhoff, A. , Hauser, A.T. , Heinke, R. , Sippl, W. , Jung, M. , 2009. The emerging therapeutic potential of histone methyltransferase and demethylase inhibitors. ChemMedChem. 4, 1568–1582. [DOI] [PubMed] [Google Scholar]
- Spannhoff, A. , Sippl, W. , Jung, M. , 2009. Cancer treatment of the future: inhibitors of histone methyltransferases. The International Journal of Biochemistry & Cell Biology. 41, 4–11. [DOI] [PubMed] [Google Scholar]
- Stadler, W.M. , Margolin, K. , Ferber, S. , McCulloch, W. , Thompson, J.A. , 2006. A phase II study of depsipeptide in refractory metastatic renal cell cancer. Clinical Genitourinary Cancer. 5, 57–60. [DOI] [PubMed] [Google Scholar]
- Stamatopoulos, B. , Meuleman, N. , De Bruyn, C. , Mineur, P. , Martiat, P. , Bron, D. , Lagneaux, L. , 2009. Antileukemic activity of valproic acid in chronic lymphocytic leukemia B cells defined by microarray analysis. Leukemia. 23, 2281–2289. [DOI] [PubMed] [Google Scholar]
- Stathis, A. , Hotte, S.J. , Chen, E.X. , Hirte, H.W. , Oza, A.M. , Moretto, P. , Webster, S. , Laughlin, A. , Stayner, L.A. , McGill, S. , 2011. Phase I study of decitabine in combination with vorinostat in patients with advanced solid tumors and non-Hodgkin's lymphomas. Clinical Cancer Research. 17, 1582–1590. [DOI] [PubMed] [Google Scholar]
- Sterner, D.E. , Berger, S.L. , 2000. Acetylation of histones and transcription-related factors. Microbiology and Molecular Biology Reviews. 64, 435–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stewart, D.J. , Donehower, R.C. , Eisenhauer, E.A. , Wainman, N. , Shah, A.K. , Bonfils, C. , MacLeod, A.R. , Besterman, J.M. , Reid, G.K. , 2003. A phase I pharmacokinetic and pharmacodynamic study of the DNA methyltransferase 1 inhibitor MG98 administered twice weekly. Annals of Oncology. 14, 766–774. [DOI] [PubMed] [Google Scholar]
- Strahl, B.D. , Allis, C.D. , 2000. The language of covalent histone modifications. Nature. 403, 41–45. [DOI] [PubMed] [Google Scholar]
- Stuhmer, T. , Arts, J. , Chatterjee, M. , Borawski, J. , Wolff, A. , King, P. , Einsele, H. , Leo, E. , Bargou, R.C. , 2010. Preclinical anti-myeloma activity of the novel HDAC-inhibitor JNJ-26481585. British Journal of Haematology. 149, 529–536. [DOI] [PubMed] [Google Scholar]
- Stunkel, W. , Campbell, R.M. , 2011. Sirtuin 1 (SIRT1): the misunderstood HDAC. Journal of Biomolecular Screening. 16, 1153–1169. [DOI] [PubMed] [Google Scholar]
- Suzuki, T. , Tanaka, R. , Hamada, S. , Nakagawa, H. , Miyata, N. , 2010. Design, synthesis, inhibitory activity, and binding mode study of novel DNA methyltransferase 1 inhibitors. Bioorganic & Medicinal Chemistry Letters. 20, 1124–1127. [DOI] [PubMed] [Google Scholar]
- Szyf, M. , 2009. Epigenetics, DNA methylation, and chromatin modifying drugs. Annual Review of Pharmacology and Toxicology. 49, 243–263. [DOI] [PubMed] [Google Scholar]
- Talero, E. , Avila-Rom, J. , Motilva, V. , 2012. Chemoprevention with phytonutrients and microalgae products in chronic inflammation and colon cancer. Current Pharmaceutical Design. 18, (26) 3939–3965. [DOI] [PubMed] [Google Scholar]
- Tarasenko, N. , Nudelman, A. , Tarasenko, I. , Entin-Meer, M. , Hass-Kogan, D. , Inbal, A. , Rephaeli, A. , 2008. Histone deacetylase inhibitors: the anticancer, antimetastatic and antiangiogenic activities of AN-7 are superior to those of the clinically tested AN-9 (Pivanex). Clinical & Experimental Metastasis. 25, 703–716. [DOI] [PubMed] [Google Scholar]
- Tate, C.R. , Rhodes, L.V. , Segar, H.C. , Driver, J.L. , Pounder, F.N. , Burow, M.E. , Collins-Burow, B.M. , 2012. Targeting triple-negative breast cancer cells with the histone deacetylase inhibitor panobinostat. Breast Cancer Research. 14, R79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tibodeau, J.D. , Benson, L.M. , Isham, C.R. , Owen, W.G. , Bible, K.C. , 2009. The anticancer agent chaetocin is a competitive substrate and inhibitor of thioredoxin reductase. Antioxidants & Redox Signaling. 11, 1097–1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tinari, N. , De Tursi, M. , Grassadonia, A. , Zilli, M. , Stuppia, L. , Iacobelli, S. , Natoli, C. , 2012. An epigenetic approach to pancreatic cancer treatment: the prospective role of histone deacetylase inhibitors. Current Cancer Drug Targets. 12, 439–452. [DOI] [PubMed] [Google Scholar]
- Tong, W.G. , Wei, Y. , Stevenson, W. , Kuang, S.Q. , Fang, Z. , Zhang, M. , Arts, J. , Garcia-Manero, G. , 2010. Preclinical antileukemia activity of JNJ-26481585, a potent second-generation histone deacetylase inhibitor. Leukemia Research. 34, 221–228. [DOI] [PubMed] [Google Scholar]
- Tseng, C.K. , Marquez, V.E. , Fuller, R.W. , Goldstein, B.M. , Haines, D.R. , McPherson, H. , Parsons, J.L. , Shannon, W.M. , Arnett, G. , Hollingshead, M. , 1989. Synthesis of 3-deazaneplanocin A, a powerful inhibitor of S-adenosyl homocysteine hydrolase with potent and selective in vitro and in vivo antiviral activities. Journal of Medicinal Chemistry. 32, 1442–1446. [DOI] [PubMed] [Google Scholar]
- Vaquero, A. , 2009. The conserved role of sirtuins in chromatin regulation. The International Journal of Developmental Biology. 53, 303–322. [DOI] [PubMed] [Google Scholar]
- Veeck, J. , Esteller, M. , 2010. Breast cancer epigenetics: from DNA methylation to microRNAs. Journal of Mammary Gland Biology and Neoplasia. 15, 5–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Veeraraghavan, J. , Natarajan, M. , Lagisetty, P. , Awasthi, V. , Herman, T.S. , Aravindan, N. , 2011. Impact of curcumin, raspberry extract, and neem leaf extract on rel protein-regulated cell death/radiosensitization in pancreatic cancer cells. Pancreas. 40, 1107–1119. [DOI] [PubMed] [Google Scholar]
- Villa, R. , De Santis, F. , Gutierrez, A. , Minucci, S. , Pelicci, P.G. , Di Croce, L. , 2004. Epigenetic gene silencing in acute promyelocytic leukemia. Biochemical Pharmacology. 68, 1247–1254. [DOI] [PubMed] [Google Scholar]
- Warrell, R.P. , He, L.Z. , Richon, V. , Calleja, E. , Pandolfi, P.P. , 1998. Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. Journal of the National Cancer Institute. 90, 1621–1625. [DOI] [PubMed] [Google Scholar]
- Watanabe, Y. , Maekawa, M. , 2010. Methylation of DNA in cancer. Advances in Clinical Chemistry. 52, 145–167. [DOI] [PubMed] [Google Scholar]
- Westphal, C.H. , Dipp, M.A. , Guarente, L. , 2007. A therapeutic role for sirtuins in diseases of aging?. Trends in Biochemical Sciences. 32, 555–560. [DOI] [PubMed] [Google Scholar]
- Whitehead, R.P. , Rankin, C. , Hoff, P.M. , Gold, P.J. , Billingsley, K.G. , Chapman, R.A. , Wong, L. , Ward, J.H. , Abbruzzese, J.L. , Blanke, C.D. , 2009. Phase II trial of romidepsin (NSC-630176) in previously treated colorectal cancer patients with advanced disease: a Southwest Oncology Group study (S0336). Investigational New Drugs. 27, 469–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wild, L. , Flanagan, J.M. , 2010. Genome-wide hypomethylation in cancer may be a passive consequence of transformation. Biochimica et Biophysica Acta. 1806, 50–57. [DOI] [PubMed] [Google Scholar]
- Winquist, E. , Knox, J. , Ayoub, J.P. , Wood, L. , Wainman, N. , Reid, G.K. , Pearce, L. , Shah, A. , Eisenhauer, E. , 2006. Phase II trial of DNA methyltransferase 1 inhibition with the antisense oligonucleotide MG98 in patients with metastatic renal carcinoma: a National Cancer Institute of Canada Clinical Trials Group investigational new drug study. Investigational New Drugs. 24, 159–167. [DOI] [PubMed] [Google Scholar]
- Wu, F. , Hu, C.H. , 2011. Reversal effect of 5-Aza-dc on cisplatin-resistance in human NSCLC cells in vitro. Zhonghua Zhong Liu Za Zhi [Chinese Journal of Oncology]. 33, 349–353. [PubMed] [Google Scholar]
- Xu, F. , Mao, C. , Ding, Y. , Rui, C. , Wu, L. , Shi, A. , Zhang, H. , Zhang, L. , Xu, Z. , 2010. Molecular and enzymatic profiles of mammalian DNA methyltransferases: structures and targets for drugs. Current Medicinal Chemistry. 17, 4052–4071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu, W.S. , Parmigiani, R.B. , Marks, P.A. , 2007. Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene. 26, 5541–5552. [DOI] [PubMed] [Google Scholar]
- Yamamoto, H. , Schoonjans, K. , Auwerx, J. , 2007. Sirtuin functions in health and disease. Molecular Endocrinology. 21, 1745–1755. [DOI] [PubMed] [Google Scholar]
- Yan, L. , Nass, S.J. , Smith, D. , Nelson, W.G. , Herman, J.G. , Davidson, N.E. , 2003. Specific inhibition of DNMT1 by antisense oligonucleotides induces re-expression of estrogen receptor-alpha (ER) in ER-negative human breast cancer cell lines. Cancer Biology & Therapy. 2, 552–556. [DOI] [PubMed] [Google Scholar]
- Yang, D. , Torres, C.M. , Bardhan, K. , Zimmerman, M. , McGaha, T.L. , Liu, K. , 2012. Decitabine and vorinostat cooperate to sensitize colon carcinoma cells to Fas ligand-induced apoptosis in vitro and tumor suppression in vivo. Journal of Immunology. 188, 4441–4449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang, X.J. , 2004. The diverse superfamily of lysine acetyltransferases and their roles in leukemia and other diseases. Nucleic Acids Research. 32, 959–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshida, M. , Kijima, M. , Akita, M. , Beppu, T. , 1990. Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A. The Journal of Biological Chemistry. 265, 17174–17179. [PubMed] [Google Scholar]
- Younes, A. , Oki, Y. , Bociek, R.G. , Kuruvilla, J. , Fanale, M. , Neelapu, S. , Copeland, A. , Buglio, D. , Galal, A. , Besterman, J. , 2011. Mocetinostat for relapsed classical Hodgkin's lymphoma: an open-label, single-arm, phase 2 trial. The Lancet Oncology. 12, 1222–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou, Q. , Atadja, P. , Davidson, N.E. , 2007. Histone deacetylase inhibitor LBH589 reactivates silenced estrogen receptor alpha (ER) gene expression without loss of DNA hypermethylation. Cancer Biology & Therapy. 6, 64–69. [DOI] [PubMed] [Google Scholar]