ArticlePDF AvailableLiterature Review

DNA Methyltransferase Inhibitors and Their Emerging Role in Epigenetic Therapy of Cancer

Authors:

Abstract and Figures

The DNA methyltransferase (DNMT) inhibitors azacytidine and decitabine are the most successful epigenetic drugs to date and are still the most widely used as epigenetic modulators, even though their application for oncological diseases is restricted by their relative toxicity and poor chemical stability. Zebularine (1-(β-D-ribofuranosyl)-1,2-dihydropyrimidin-2-one), a more stable and less toxic cytidine analog, is another inhibitor of DNMT with concomitant inhibitory activity towards cytidine deaminase. Unfortunately, there is no new information related to the possible clinical applications of zebularine. Although many new inhibitors of DNMT have been identified, none of them can so far replace azacytidine, decitabine and, to a lesser degree, zebularine. This review summarizes the current data and knowledge about azacytidine, decitabine and zebularine, and their role in present and possible future epigenetic cancer therapy. We also discuss the molecular modes of action of these agents with consideration of their different toxicities and demethylation profiles, reflecting their complex and partially overlapping biological effects.
Content may be subject to copyright.
Abstract. The DNA methyltransferase (DNMT) inhibitors
azacytidine and decitabine are the most successful epigenetic
drugs to date and are still the most widely used as epigenetic
modulators, even though their application for oncological
diseases is restricted by their relative toxicity and poor
chemical stability. Zebularine (1-(β-D-ribofuranosyl)-1,2-
dihydropyrimidin-2-one), a more stable and less toxic
cytidine analog, is another inhibitor of DNMT with
concomitant inhibitory activity towards cytidine deaminase.
Unfortunately, there is no new information related to the
possible clinical applications of zebularine. Although many
new inhibitors of DNMT have been identified, none of them
can so far replace azacytidine, decitabine and, to a lesser
degree, zebularine. This review summarizes the current data
and knowledge about azacytidine, decitabine and zebularine,
and their role in present and possible future epigenetic
cancer therapy. We also discuss the molecular modes of
action of these agents with consideration of their different
toxicities and demethylation profiles, reflecting their complex
and partially overlapping biological effects.
The term ‘epigenetic’ was coined by C. H. Waddington in
1942 and from that time, the definition of ‘epigenetic’ has
evolved (1). Finally, in 2008 a consensus definition of
epigenetics’ was established as “stably-heritable phenotype
resulting from changes in a chromosome without alterations
in the DNA sequence”, which may lead to disease e.g.
Beckwith-Wiedemann and Prader Willi/Angelman syndromes,
as well as cellular aging and cancer (2). Although for many
years scientists have been reporting that epigenetic changes
may influence cancer development, the methodology to prove
this had some limitations. Nowadays, it is known that
epigenetic alterations are as equally responsible for
carcinogenesis as are genetic mutations. In cancer, gene
silencing through methylation occurs at least as frequently as
mutations or deletions, and leads to aberrant silencing of
normal tumor-suppressor function (3). It has also been proven
that epigenetic changes can be detected in carcinogenesis
earlier than the well-known genetic origins of cancer (4).
Epigenetic modifications rely on re-building of chromatin
structure resulting in an open or closed configuration and
thereby expressing or repressing genes which control such
basic cellular processes such as differentiation, proliferation
and apoptosis, and as a consequence, cell functions. Two
covalent modifications are responsible for these processes:
DNA methylation and nucleosomal histone tail acetylation,
which influence the epigenetic regulation of the gene
expression pattern.
DNA methylation is the most characterized epigenetic
phenomenon described as a stable epigenetic marker (5). This
process involves enzymes belonging to the DNA
methyltransferase family (DNMTs). In humans, DNMTs bind
a methyl group (-CH
3
) at the carbon 5-position of the
pyrimidine ring of cytosine in CpG dinucleotides (Figure 1A).
In somatic cells, most CpG dinucleotides are methylated,
except those located in CpG islands (6-8). These islands are
mainly located near or in the promoter regions (nearly 60% of
mammalian gene promoters) in repeated sequences as long
interspersed nuclear elements (LINEs) and short interspersed
nuclear elements (SINEs) (9), as well as in CpG island shores,
where the methylation status depends on the tissue origin (10).
The regulation of histone acetylation is controlled by two
enzyme families: histone acetyltransferases (HATs) and
histone deacetylases (HDACs). The latter promotes a higher-
order chromatin structure, which is equated with
transcriptional gene repression.
Histone proteins are not the only target of HATs and
HDACs. It is important to note that HDACs might also
2989
*These Authors contributed equally to this work.
Correspondence to: Sylwia Flis, Ph.D., Department of
Pharmacology, National Medicines Institute, Chelmska Street 30/34,
00-725 Warsaw, Poland. E-mail: sylwia.flis@yahoo.pl
Key Words: Epigenetics, DNA methylation, DNMT inhibitors,
cancer therapy, review.
ANTICANCER RESEARCH 33: 2989-2996 (2013)
Review
DNA Methyltransferase Inhibitors and Their Emerging
Role in Epigenetic Therapy of Cancer
AGNIESZKA GNYSZKA*, ZENON JASTRZĘBSKI and SYLWIA FLIS*
Department of Pharmacology, National Medicines Institute, Warsaw, Poland
0250-7005/2013 $2.00+.40
directly modulate acetylation of the non-histone proteins such
as p53, signal transducer and activator of transcription (STAT),
transcription factor E2F and others. Hence, HDACs do not
only act in an epigenetic manner. Nevertheless, DNA
methylation and histone modifications are closely related.
Methylated CpG sites in gene promoter regions are easily
recognized by specific methyl CpG binding proteins (MBPs)
which act as adapters between methylated DNA and chromatin
modifying factors. MBPs can recruit co-repressors such as
HDAC, methyltransferase and chromatin remodeling factors,
creating the protein complex which regulates gene expression
(11). If the promoter region is methylated, the corresponding
gene is repressed due to its poor recognition by transcription
factors (12). Indeed, DNMTs affect protein DNA interactions
by chromatin remodeling, determine the accessibility of DNA
to transcription factors, and are associated with under- or
overexpression of certain proteins, ultimately leading to
diverse pathologies, among which cancer (13).
Distinct changes of DNA methylation are termed
epimutations’ and appear to play an important role in
carcinogenesis. Consequences of epimutation are similar to
those of classic genetic mutations because the affected genes
are silenced and functional gene products cannot be
generated (14). In many types of tumors, the genomic DNA
methylation pattern is changed either through
hypermethylation (increased methylation) or
hypomethylation (decreased methylation). In cancer cells,
hypomethylation of CpG dinucleotides, especially in the
pericentromeric regions of the chromosome, may lead to
genomic instability. On the other hand, dense methylation of
CpG islands, particularly in the promoter region of tumor
suppressor genes, is associated with aberrant silencing of
transcription (15). An elevated level of methylation in cancer
cells probably results from increased activity of DNMTs,
often as a consequence of overexpression. De-regulation of
the DNMTs has been shown in many types of cancer
including of the lung, breast, stomach and colon, and as well
as in leukemia (16). Fortunately, epigenetic alterations are
potentially reversible, unlike genetic mutations. Therefore,
such alterations have become an attractive target for cancer
therapy. Since hypermethylation of tumor suppressor genes
and overexpression of DNMTs have been established as the
major key players in carcinogenesis, demethylating agents
seem to be especially promising as anticancer drugs. Re-
expression of aberrantly silenced genes and restoration of
their normal function can be achieved through the use of
DNMT inhibitors which are incorporated into the growing
DNA strand and covalently bind DNMTs.
ANTICANCER RESEARCH 33: 2989-2996 (2013)
2990
Figure 1. Scheme of DNA methylation (A) and DNA methyltansferase inhibition (B). A: DNA methylation at the 5-position of the pyrimidine ring of
cytosine is catalyzed by DNA methyltransferase. The methyl group (-CH
3
) is transferred from the cofactor S-adenosyl-L methionine (SAM), resulting
in creation of 5-methylcytosine, then the enzyme is released by β elimination. B: Trapping reaction relies on prevention of β elimination due to the
presence of the nitrogen atom at the 5-position of azacytidine, resulting in a covalent irreversible complex (6).
Currently, two DNMT inhibitors (azanucleosides) have
been approved by the US Food and Drug Administration
(FDA): azacytidine (Vidaza; Celgene) and decitabine (5 aza
2’ deoxycytidine) (Dacogen; SuperGen). These two types of
drugs are the first molecules that have been characterized as
the archetypal DNMT inhibitors and the only epidrugs that
have been approved for the treatment of patients with acute
myeloid leukemia (AML) and myelodyplastic syndrome
(MDS). Azacytidine has also been approved by the FDA and
the European Medicines Agency (EMA) for use against
chronic myelomonocytic leukemia (CMML) (10).
Therapeutic use of DNMT inhibitors can provide new and
effective solutions for patients not only with hematological
malignancies but also with other tumor types (especially
since azacytidine and decitabine are currently in phase I
clinical trials in patients with solid tumors) (17). The third
novel member of the nucleoside DNMT inhibitor family is
zebularine, a cytidine analog, which has been characterized
as a potent and promising agent because of good results
achieved in in vitro experiments, encouraging zebularine use
for future clinical trials.
In the present article, we discuss the action mechanism of
DNMT inhibitors (azacytidine, decitabine and zebularine)
with consideration of their different activities towards cancer
cells which may impact future clinical trials.
Azacytidine and decitabine are cytidine analogs modified
in position 5 of the pyrimidine ring (Figure 2). Both
compounds were synthesized by Sorm and co-workers in
1964 (18). The compounds were initially used as
antimetabolite agents in leukemia chemotherapy until their
hypomethylating properties were discovered. These cytidine
analogs are transported into cells by human concentrative
nucleoside transporter-1 and converted to active triphosphate
forms, i.e. azacytidine by uridine cytidine kinase to 5-
azacytidine 5’-triphosphate and decitabine by deoxycytidine
kinase to 5-aza-2’-deoxycytidine-5’-triphosphate and then
degraded by cytidine deaminase (CDA). Azacytidine, being a
ribonucleoside, is incorporated into RNA and, to a lesser
extent, into DNA, whereas decitabine, as a deoxyribose
analog, is incorporated only into DNA strands (Figure 3A).
To be active, these compounds need to be integrated into the
genome of rapidly proliferating cells during the S phase of
the cell cycle (19). The incorporated 5-azanucleoside disrupts
the interaction between DNA and DNMTs through nitrogen
instead of carbon, in the 5-position of the modified
pyrimidine, which precludes the resolution of the complex
and finally promotes its proteosomal degradation (Figure
1B). Thus, the enzyme remains covalently bound to DNA
and its DNMT and the function is blocked. In addition, the
covalent protein adduction also compromises the
functionality of DNA and triggers DNA damage signaling,
resulting in the degradation of the trapped DNMTs (20, 21).
Therefore, further methylation of cytosine residues is
inhibited, causing the passive loss of cytosine methylation in
the daughter cells after replication.
Another demethylating agent in the family of nucleoside
analogs is zebularine, which is characterized by chemical
stability, apparent bioavailability and low cytotoxicity (22).
Such properties distinguish zebularine among nucleoside
inhibitors (Figure 2). Zebularine was originally synthesized
and evaluated as an inhibitor of CDA to prevent de-amination
of nucleoside analogs. It acts, however, primarily as a DNMT
inhibitor by trapping the DNMT protein and forming tight
covalent complexes between the DNMT protein and
zebularine-substituted DNA (23). Zebularine is also activated
after incorporation into DNA and metabolized presumably in
a similar way to azacytidine. The initial phosphorylation is
most likely mediated by uridine-cytidine kinase, followed by
conversion to the 2’-deoxyzebularine-5’-diphosphate by
ribonucleotide reductase. Furthermore, 2’ deoxyzebularine-
5’-diphosphate is converted to 2’-deoxyzebularine-5’
triphosphate, which appears to be crucial for its incorporation
into DNA, but this step of metabolic activation still needs to
be clarified. In brief, inactivation of the DNMT by zebularine
may be related to the absence of the 4-amino group in the
6-position of its pyrimidine ring (Figure 3B). In this way,
zebularine does not allow the activation of cytosine C5
Gnyszka et al: DNMT Inhibitors in Cancer Therapy (Review)
2991
Figure 2. Nucleoside analog inhibitors of DNA methyltranferase.
position and methyl group transfer. The lack of methylation,
even if it is reversible, can clarify the stabilization of the
zebularine containing DNA binding complex and slowing its
dissociation (24).
Even though azacytidine and decitabine are used in the
clinic, they are characterized by poor chemical stability,
which depends on temperature and pH. In alkaline solutions
both agents undergo irreversible decomposition. In vitro
studies have shown that at 37˚C, in neutral aqueous
solutions, the half-lives were 7 hours for azacytidine and 21
hours for decitabine, whereas the corresponding times in vivo
are 41 minutes for azacytidine and from 15 to 25 minutes for
decitabine. At 4˚C, both agents have considerable chemical
stability, but with elevation of the temperature, they undergo
more rapid degradation (21, 25, 26).
In humans, azacytidine and decitabine are metabolized by
CDA, the enzyme which renders these drugs inactive by
converting them into 5-azauridine compounds. The high level
of CDA in the human liver and spleen is largely responsible
for shorter half-lives of both agents in vivo than in vitro.
Therefore, an increase in CDA activity may reduce efficacy
by lowering drug levels and shortening the half-life times.
Unlike azacytidine and decitabine, zebularine is highly
stable at acid and neutral pH. Zebularine has a half-life of
~44 hours at 37˚C in phosphate buffered saline (PBS) at pH
1.0 and ~508 hours at pH 7.0; such properties allow for its
oral administration (27). Moreover, taking into consideration
the half-life times of zebularine after intravenous and oral
dosing in mice, rats and rhesus monkeys, it is likely that
relatively frequent dosing or continuous intravenous infusion
of zebularine will be necessary to maintain prolonged
DNMT inhibition, which seems to be possible because of the
low toxicity of zebularine (28).
Azanucleosides in Cancer Therapy
Mechanism of Action
The importance of DNA hypermethylation of CpG islands as
a key player during cancer development and progress is not
questioned. Especially since this phenomenon occurs
infrequently in normal cells, the abnormal transcriptional
silencing of tumor suppressor genes by hypermethylation of
CpG islands has become an attractive and selective tumor
specific therapeutic cancer target (3, 27). During gene
methylation, the DNA sequence, as well as protein product,
remains unaltered. Therefore, pharmacological intervention
in the form of chemical inhibitors seems to be one of the
possible ways for de-repression of inappropriately silenced
genes and restoration of their normal functions.
The ability of azacytidine and decitabine to deplete the
DNA methylating activity of DNMT can be achieved at low
doses of both agents. Stresemann et al. have shown that
azacytidine and decitabine are able to induce demethylation
of epigenetically silenced genes at least at concentrations that
ANTICANCER RESEARCH 33: 2989-2996 (2013)
2992
Figure 3. Metabolic activation of 5 azanucleosides (A) and zabularine (B). After cellular uptake 5-azanucleosides and zebularine are modified by
different metabolic pathways as described in the text. CDA: Cytidine deaminase; UCK: uridine cytidine kinase; dCK: deoxycytidine kinase; CMP
kinase: cytidylate kinases; dCMP: deoxycytidylate kinase; 5-Aza-U: 5-aza-uridine; 5-Aza-CTP: 5 azacytidine 5-triphosphate; 5-Aza-dCTP: 5-aza
2-deoxycytidine-5-triphosphate; ZTP: zebularine-5-triphosphate; dZTP: 2-deoxyzebularine-5-triphosphate (19, 22).
exceed 20% inhibition of cell growth (IC
20
). In vitro
experiments on whole-genome methylation, as well as local
methylation at a defined genomic locus revealed that
azacytidine and decitabine at 5-fold the IC
20
concentration
strongly reduces the genomic DNA methylation level in
lymphoid cancer cells as compared to controls, i.e. by 60%
and 40%, respectively. A similarly strong concentration-
dependent demethylation was observed after incubation of
colon carcinoma cells with both agents at concentrations
centered on the respective drug specific IC
20
concentration
(29). Independent studies have shown that these two epidrugs
may significantly reactivate silenced genes [tissue inhibitor
of metalloproteinase 3 (TIMP3), p15, p16, cyclin-dependent
kinase inhibitor 1C (CDKN1C), RAS-association domain
family 1 (RASSF1)] responsible for basic cellular mechanisms
such as apoptosis, cell cycle, and DNA repair (29), and that
the concentrations required of both agents to achieve this
effect are not high. Azacytidine and decitabine should be used
at low inhibitory concentrations because at higher
concentrations these agents exert strong cytotoxicity, interfere
with DNA synthesis and cause DNA damage (30, 31). Apart
from the use of low concentrations, for their demethylation
function, the S phase of the cell cycle is needed. The DNA
replication phase of the cell cycle enables selective and
effective incorporation of these substances into the DNA of
rapidly dividing cancer cells, reducing, thereby,
hypomethylation in normal cycling cells (32). However, the
hypomethylation activity of these two agents is not equal. It
has been shown that azacytidine has only about 10% the
potency of decitabine at inhibiting DNA methylation (33).
Decitabine as a deoxyribonucleotide is directly incorporated
into the DNA after phosphorylation to inhibit DNA
methylation, whereas azacytidine is additionally incorporated
into RNA. The overall incorporation of azacitydine into RNA
can account for about 80-90% (34). Decrease of tRNA
acceptor activity, polyribosome breakdown and incorporation
into mRNA causing the subsequent inhibition of protein
synthesis and enzyme induction are the functional
consequences of RNA synthesis inhibition by azacytidine
(35). All these in effect may influence both cancer and normal
cells, resulting in greater in vitro and in vivo side-effects.
It has been proven that the mechanism of action of both
these agents is dose-dependent. Therefore, the concept of a
dual mechanism of action of these two agents has arisen. For
decitabine, the ‘dual mechanism’ refers to the inhibition of
cell proliferation at high doses and to the DNA
hypomethylation-mediated effect on gene re-expression at
low doses affecting the processes of cell differentiation and
tumor suppression, whereas for azacytidine the ‘dual
mechanism’ refers to the cytotoxicity at high doses, via
incorporation into RNA and DNA, and to the DNA
hypomethylation effect at lower doses (34). Their mechanism
of action at the highest doses is related to the formation of
covalent DNMT-DNA adducts in aza-containing DNA,
leading to DNA damage and cytotoxicity. Experiments
conducted on human tumor cell lines have shown that
treatment with these agents causes growth inhibition by cell
cycle arrest (specific to G
2
/M phase for decitabine and cell
cycle non-specific for azacytidine) and reduction in
clonogenic survival. Moreover, decitabine and azacytidine
can induce apoptosis in p53-dependent or p53-independent
manners, respectively (36-38).
Unfortunately, these agents also have some limitations. In
spite of their clinical efficacy, azacytidine and decitabine are
characterized by poor bioavailability, instability in
physiological media and high toxicity, restricting their use.
For this reason, one of the most promising molecules appears
to be zebularine. It has been characterized as a potential
antitumor agent, based on its stability (half-life of >500
hours at pH 7.4) and minimal toxicity both in vitro and in
vivo (39-41). Once incorporated into DNA, zebularine is
involved in the reactivation of the silenced genes, such as
cyclin-dependent kinase inhibitors (CKI) p15 in AML cells,
p16 in bladder, colon and pancreatic cells, and p57 in
myeloid leukemia cells (42, 43). Zebularine also leads to the
re-expression of the cell cycle and apoptosis modulator Ras
association domain family 1 isoform A (RASSF1A) as a
result of induced demethylation of its promoter region in
ovarian cancer cells. Moreover, microarray analysis
demonstrated that decitabine and zebularine may
demethylate 78 genes, with 32 exclusive to decitabine and
only eight specific for zebularine (44). This might suggest
that both agents exert their demethylating effects by different
mechanisms. Cheng et al. observed that zebularine caused
complete depletion of DNMT1 and partial depletion of
DNMT3A and DNMT3B2/3 in cancer cells (45). This
suggests zebularine preference for DNMT1 over DNMT3A
and DNMT3B. Moreover, DNMT1 may be an important
indicator of the demethylating ability of zebularine. It has
also been reported that DNMT3A/DNMT3B double-null
embryonic stem cells are more resistant to decitabine than
are DNMT1 null cells, suggesting that decitabine may be
more effective for selected types of cancer cells, in which
DNMT3 expression is up-regulated (46). Furthermore,
DNMTs have a higher affinity for zebularine-containing
DNA than for the unmodified DNA (24). Such results
indicate that the differential specificity and affinity of these
drugs for DNMT isoforms could lead to divergent cellular
responses. It has been demonstrated that higher doses of
zebularine are required to obtain demethylation and gene re-
expression levels comparable to those that are induced by
azacytidine and decitabine (29). This might be a result of
lower binding affinity of uridine-cytidine kinase for
zebularine and its slow conversion to 2’-deoxyzebularine-5’-
diphosphate (40). However, owing to its minimal toxicity
profile, zebularine, unlike azacytidine and decitabine, can be
Gnyszka et al: DNMT Inhibitors in Cancer Therapy (Review)
2993
used at high micromolar concentrations in prolonged
treatment periods (41, 47). The lower toxicity of zebularine,
as a ribonucleoside, may result from its different
incorporation into RNA and DNA in normal and
neoplastically-transformed cells, due to the overexpression
of uridine-cytidine kinase in cancer cells, facilitating
zebularine insertion into nucleic acids (44). Moreover,
Champion et al. have shown that the absence of the 4-amino
group in zebularine does not allow for the activation of the
cytosine C5 position after covalent intermediate formation
and methyl group transfer (24). Such lack of zebularine
methylation seems to prevent dissociation of the DNA-
enzyme complex, indicating the stabilization of the
zebularine-containing DNA-binding complex.
Future medical use of zebularine could involve
combinations of this drug with other therapeutic modalities,
such as chemotherapy, immunotherapy or radiotherapy.
Clinical trials with azacytidine or decitabine administered as
single agents or in co-administration with other
chemotherapeutics resulted in significant toxicity (48, 49).
Therefore, zebularine and other demethylation agents with
similar properties could be less detrimental and more
promising drug candidates.
Conclusion and Future Perspectives
Azacytidine, decitabine and zebularine have differential
activity, complex and partially overlapping mechanisms of
action. Studies have indicated that the methylation patterns
of tumor suppressor genes might differ depending on the
drug being used. Azacytidine and decitabine induce a strong
demethylating effect, leading at lower doses to re expression
of aberrantly silenced genes associated with reduced
proliferation, cell differentiation, apoptosis, and senescence,
while at higher doses, their main effect consists of DNA
damage after incorporation into genomic DNA. The impact
of zebularine on the DNA methylation level is more
moderate. However, this cytidine analog is less toxic and
can, therefore, be given continuously at high doses.
Additionally, zebularine seems to target tumor cells
preferentially.
The use of demethylating agents azacytidine and decitabine
in the treatment of myelodysplastic syndromes is well-
documented, in spite of their high toxicity. But the status of
knowledge for using demethylating agents for solid tumors is
still insufficient and needs to be further evaluated. It appears
to be reasonable to use demethylating agents in combination
with chemotherapeutic agents. In vitro experiments indicate
that decitabine can potentiate the cytotoxic effect of classical
chemotherapeutics, such as doxorubicin, 5-fluorouracil and
oxaliplatin, and induce highly synergistic effects (50, 51).
Interestingly, encouraging results were obtained with
combination of decitabine and carboplatin in patients with
solid tumors (52). The authors concluded that decitabine
combines safely with carboplatin and that the specific
regimen causes epigenetic changes. In another phase I study,
a combination of cisplatin with decitabine resulted in one
partial response in a patient with cervical cancer, and two
minor responses: one in a patient with non-small cell lung
cancer and another in a patient with cervical cancer (17).
Generally, combinations of azacytidine or decitabine with
standard chemotherapy clearly have clinical activity, but it is
difficult to distinguish the effect of the epigenetic therapy
from the cytotoxic therapy (17). On the other hand, the
present demethylating agents used in clinics are cytotoxic,
mutagenic and exhibit lack of specificity towards genes,
which may limit their clinical application. Thus, the next
generation of DNMT inhibitors with lower toxicity might be
more fruitful for future research. Zebularine is an alternative
derivative of cytidine and promises to be a better drug than
azacytidine and decitabine for epigenetic cancer therapy.
Based on its stability, it was the first DNMT inhibitor
showing in vivo antitumor activity against T-cell lymphoma
after oral administration (53). Attention has also been
attributed on the identification of small non-nucleoside
DNMT inhibitors, such as epigallocatechin-3-gallate,
hydralazine, procainamide, procaine and RG108, which bind
directly to the catalytic region of DNMTs without
incorporation into DNA. However, the results of in vitro
studies showed that non-nucleoside compounds induce
limited epigenetic changes in living cells (54). Among the
novel agents of demethylation, the most intensively studied
are DNMT1 antisense and siRNA. Down-regulation of
DNMT1 by antisense or siRNA is sufficient to restore the
expression of aberrantly hypermethylated genes (55, 56), but
such therapy is still controversial mainly because of the
problems associated with the administration to humans,
which include instability, toxicity, risk of non-specific effects,
and complexity in developing a suitable delivery system.
Even though many DNMT inhibitors other then cytidine
analogues have been developed, their effects are not
satisfactory and they do not seem to be able to replace
azacytidine and decitabine, as yet. Therefore, azacytidine and
decitabine, in spite of their limitations, are still used for
combination epigenetic therapy. However, attempts to
discover and to develop novel compounds targeting DNMTs
should be continued. It is important to find more selective and
less toxic DNMT inhibitors which will be effective especially
in patients with solid tumors. It would be a challenge to
design inhibitors whose mechanism of action will rely only
on reactivation of abnormally silenced suppressor genes.
Acknowledgements
This work was supported by National Science Centre, Poland (grant
N405 139139).
ANTICANCER RESEARCH 33: 2989-2996 (2013)
2994
References
1 Waddington CH: The epigenotype. Endeavour 1: 18-20, 1942.
2 Berger SL, Kouzarides T, Shiekhattar R and Shilatifard A: An
operational definition of epigenetics. Genes Dev 23: 781-783,
2009.
3 Baylin SB: DNA methylation and gene silencing in cancer. Nat
Clin Pract Oncol 2: 4-11, 2005.
4 Sharma S, Kelly TK and Jones PA: Epigenetics in cancer.
Carcinogenesis 31: 27-36, 2010.
5 Reik W: Stability and flexibility of epigenetic gene regulation in
mammalian development. Nature 447: 425-432, 2007.
6 Schermelleh L, Spada F, Easwaran HP, Zolghadr K, Margot JB,
Cardoso MC, Leonhardt H: Trapped in action: Direct
visualization of DNA methyltransferase activity in living cells.
Nat Methods 2: 751-756, 2005.
7 Suzuki MM and Bird A: DNA methylation landscapes: Provocative
insights from epigenomics. Nat Rev Genet 9: 465-476, 2008.
8 Patai AV, Molnár B, Kalmár A, Schöller A, Tóth K and Tulassay
Z: Role of DNA methylation in colorectal carcinogenesis. Dig
Dis 30: 310-315, 2012.
9 Bestor TH: The DNA methyltransferases of mammals. Hum Mol
Genet 9: 2395-2402, 2000.
10 Gros C, Fahy J, Halby L, Dufau I, Erdmann A, Gregoire JM,
Ausseil F, Vispé S and Arimondo PB: DNA methylation
inhibitors in cancer: Recent and future approaches. Biochimie
94: 2280-2296, 2012.
11 Sasai N and Defossez PA: Many paths to one goal? The proteins
that recognize methylated DNA in eukaryotes. Int J Dev Biol 53:
323-334, 2009.
12 Prendergast GC and Ziff EB: Methylation-sensitive sequence-
specific DNA binding by the c-Myc basic region. Science 251:
186-189, 1991.
13 Esteller M: Epigenetics in Cancer. N Engl J Med 358: 1148-
1159, 2008.
14 Brueckner B and Lyko F: DNA methyltransferase inhibitors: Old
and new drugs for an epigenetic cancer therapy. Trends
Pharmacol Sci 25: 551-554, 2004.
15 Herman JG and Baylin SB: Gene silencing in cancer in
association with promoter hypermethylation. N Engl J Med 349:
2042-2054, 2003.
16 Chik F, Szyf M and Rabbani SA: Role of epigenetics in cancer
initiation and progression. Adv Exp Med Biol 720: 91-104,
2011.
17 Cowan LA, Talwar S and Yang AS: Will DNA methylation
inhibitors work in solid tumors? A review of the clinical
experience with azacitidine and decitabine in solid tumors.
Epigenomics 2: 71-86, 2010.
18 Leone G, Voso MT, Teofili L and Lübbert M: Inhibitors of DNA
methylation in the treatment of hematological malignancies and
MDS. Clin Immunol 109: 89-102, 2003.
19 Yang X, Lay F, Han H and Jones PA: Targeting DNA
methylation for epigenetic therapy. Trends Pharmacol Sci 31:
536-546, 2010.
20 Santi DV, Norment A and Garrett CE: Covalent bond formation
between a DNA-cytosine methyltransferase and DNA containing
5-azacytosine. Proc Natl Acad Sci USA 81: 6993-6997, 1984.
21 Stresemann C and Lyko F: Modes of action of the DNA
methyltransferase inhibitors azacytidine and decitabine. Int J
Cancer 123: 8-13, 2008.
22 Ben-Kasus T, Ben-Zvi Z, Marquez VE, Kelley JA and Agbaria
R: Metabolic activation of zebularine, a novel DNA methylation
inhibitor, in human bladder carcinoma cells. Biochem Pharmacol
70: 121-133, 2005.
23 Billam M, Sobolewski MD and Davidson NE: Effects of a novel
DNA methyltransferase inhibitor zebularine on human breast
cancer cells. Breast Cancer Res Treat 120: 581-592, 2010.
24 Champion Ch, Guianvarc’h D, Sénamaud-Beaufort C, Jurkowska
RZ, Jeltsch A, Konger L, Arimondo PB and Guieysse-Peugeot
AL: Mechanistic insights on the inhibition of C5 DNA
methyltransferases by zebularine. PLoS One 5: e12388, 2010.
25 Kaminskas E, Farrell A, Abraham S, Baird A, Hsieh LS, Lee SL,
Leighton JK, Patel H, Rahman A, Sridhara R, Wang YC and
Pazdur R: Approval summary: Azacitidine for treatment of
myelodysplastic syndrome subtypes. Clin Cancer Res 15: 3604-
3608, 2005.
26 Momparler RL: Pharmacology of 5-aza-2’-deoxycytidine
(decitabine). Semin Hematol 42: 9-16, 2005.
27 Yoo CB, Cheng JC and Jones PA: Zebularine: A new drug for
epigenetic therapy. Biochem Soc Trans 32: 910-912, 2004.
28 Holleran JL, Parise RA, Joseph E, Eiseman JL, Covey JM, Glaze
ER, Lyubimov AV, Chen YF, D’Argenio DZ and Egorin MJ:
Plasma pharmacokinetics, oral bioavailability, and interspecies
scaling of the DNA methyltransferase inhibitor, zebularine. Clin
Cancer Res 15: 3862-3868, 2005.
29 Stresemann C, Brueckner B, Musch T, Stopper H and Lyko F:
Functional diversity of DNA methyltransferase inhibitors in
human cancer cell lines. Cancer Res 66: 2794-2800, 2006.
30 Qin T, Jelinek J, Si J, Shu J and Issa JP: Mechanisms of
resistance to 5-aza-2’-deoxycytidine in human cancer cell lines.
Blood 113: 659-667, 2009.
31 Yang X, Lay F, Han H and Jones PA: Targeting DNA
methylation for epigenetic therapy. Trends Pharmacol Sci 31:
536-546, 2010.
32 Cortez CC and Jones PA: Chromatin, cancer and drug therapies.
Mutat Res 647: 44-51, 2008.
33 Jones PA and Taylor SM: Cellular differentiation, cytidine
analogs and DNA methylation. Cell 20: 85-93, 1980.
34 Hollenbach PW, Nguyen AN, Brady H, Williams M, Ning Y,
Richard N, Krushel L, Aukerman SL, Heise C and MacBeth KJ:
A comparison of azacitidine and decitabine activities in acute
myeloid leukemia cell lines. PLoS One 5: e9001, 2010.
35 Li LH, Olin EJ, Buskirk HH and Reineke LM: Cytotoxicity and
mode of action of 5-azacytidine on L1210 leukemia. Cancer Res
30: 2760-2769, 1970.
36 Nieto M, Samper E, Fraga MF, González de Buitrago G, Esteller
M and Serrano M: The absence of p53 is critical for the
induction of apoptosis by 5-aza-2’-deoxycytidine. Oncogene 23:
735-743, 2004.
37 Schneider-Stock R, Diab-Assef M, Rohrbeck A, Foltzer-
Jourdainne C, Boltze C, Hartig R, Schönfeld P, Roessner A and
Gali-Muhtasib H: 5-Aza-cytidine is a potent inhibitor of DNA
methyltransferase 3A and induces apoptosis in HCT-116 colon
cancer cells via GADD45- and p53-dependent mechanisms. J
Pharmacol Exp Ther 312: 525-536, 2005.
38 Palii SS, Van Emburgh BO, Sankpal UT, Brown KD and
Robertson KD: DNA methylation inhibitor 5-aza-2’-
deoxycytidine induces reversible genome-wide DNA damage
that is distinctly influenced by DNA methyltransferases 1 and
3B. Mol Cell Biol 28: 752-771, 2008.
Gnyszka et al: DNMT Inhibitors in Cancer Therapy (Review)
2995
39 Zhou L, Cheng X, Connolly BA, Dickman MJ, Hurd PJ and
Hornby DP: Zebularine: A novel DNA methylation inhibitor that
forms a covalent complex with DNA methyltransferases. J Mol
Biol 321: 591-599, 2002.
40 Marquez VE, Eritja R, Kelley JA, Vanbemmel D and Christman
JK: Potent inhibition of HhaI DNA methylase by the aglycon of
2-(1H)-pyrimidinone riboside (zebularine) at the GCGC
recognition domain. Ann NY Acad Sci 1002: 154-164, 2003.
41 Cheng JC, Matsen CB, Gonzales FA, Ye W, Greer S, Marquez
VE, Jones PA and Selker EU: Inhibition of DNA methylation
and reactivation of silenced genes by zebularine. J Natl Cancer
Inst 95: 399-409, 2003.
42 Lemaire M, Momparler LF, Bernstein ML, Marquez VE and
Momparler RL: Enhancement of antineoplastic action of 5-aza-
2’-deoxycytidine by zebularine on L1210 leukemia. Anti-cancer
Drug 16: 301-308, 2005.
43 Scott SA, Lakshimikuttysamma A, Sheridan DP, Sanche SE,
Geyer CR and DeCoteau GF: Zebularine inhibits human acute
myeloid leukaemia cell growth in vitro in association with
p15
INK4B
demethylation and reexpression. Exp Hematol 35: 263-
273, 2007.
44 Balch C, Yan P, Craft T, Young S, Skalnik DG, Huang TH and
Nephew KP: Antimitogenic and chemosensitizing effects of the
methylation inhibitor zebularine in ovarian cancer. Mol Cancer
Ther 4: 1505-1514, 2005.
45 Cheng JC, Weisenberger DJ, Gonzales FA, Liang G, Xu GL, Hu
YG, Marquez VE and Jones PA: Continuous zebularine
treatment effectively sustains demethylation in human bladder
cancer cells. Mol Cell Biol 24: 1270-1278, 2004.
46 Oka M, Meacham AM, Hamazaki T, Rodić N, Chang LJ and
Terada N: De novo DNA methyltransferases Dnmt3a and
Dnmt3b primarily mediate the cytotoxic effect of 5-aza-2’-
deoxycytidine. Oncogene 24: 3091-3099, 2005.
47 Hurd PJ, Whitmarsh AJ, Baldwin GS, Kelly SM, Waltho JP,
Connolly BA and Hornby DP: Mechanism-based inhibition of
C5-cytosine DNA methyltransferases by 2-H pyrimidinone. J
Mol Biol 286: 389-401, 1999.
48 Schwartsmann G, Schunemann H, Gorini CN, Filho AF, Garbino
C, Sabini G, Muse I, DiLeone L and Mans DR: A phase I trial of
cisplatin plus decitabine, a new DNA-hypomethylating agent, in
patients with advanced solid tumors and a follow-up early phase
II evaluation in patients with inoperable non-small cell lung
cancer. Invest New Drugs 18: 83-91, 2000.
49 Pohlmann P, DiLeone LP, Cancella AI, Caldas AP, Dal Lago L,
Campos O Jr., Monego E, Rivoire W and Schwartsmann G:
Phase II trial of cisplatin plus decitabine, a new DNA
hypomethylating agent, in patients with advanced squamous cell
carcinoma of the cervix. Am J Clin Oncol 25: 496-501, 2002.
50 Flis S, Gnyszka A, Misiewicz-Krzemińska I and Spławiński J:
Decytabine enhances cytotoxicity induced by oxaliplatin and 5-
fluorouracil in the colorectal cancer cell line Colo-205. Cancer
Cell Int 9: 1-10, 2009.
51 Vijayaraghavalu S, Dermawan JK, Cheriyath V and Labhasetwar
V: Highly synergistic effect of sequential treatment with
epigenetic and anticancer drugs to overcome drug resistance in
breast cancer cells is mediated via activation of p21 gene
expression leading to G
2
/M cycle arrest. Mol Pharm 10: 337-
352, 2013.
52 Appleton K, Mackay HJ, Judson I, Plumb JA, McCormick C,
Strathdee G, Lee C, Barrett S, Reade S, Jadayel D, Tang A,
Bellenger K, Mackay L, Setanoians A, Schätzlein A, Twelves C,
Kaye SB and Brown R: Phase I and pharmacodynamic trial of
the DNA methyltransferase inhibitor decitabine and carboplatin
in solid tumors. J Clin Oncol 25: 4603-4609, 2007.
53 Herranz M, Martín-Caballero J, Fraga MF, Ruiz-Cabello J,
Flores JM, Desco M, Marquez V and Esteller M: The novel
DNA methylation inhibitor zebularine is effective against the
development of murine T-cell lymphoma. Blood 107: 1174-1177,
2006.
54 Ren J, Singh BN, Huang Q, Li Z, Gao Y, Mishra P, Hwa YL, Li
J, Dowdy SC and Jiang SW: DNA hypermethylation as a
chemotherapy target. Cell Signal 23: 1082-1093, 2011.
55 Jung Y, Park J, Kim TY, Park JH, Jong HS, Im SA, Robertson
KD, Bang YJ and Kim TY: Potential advantages of DNA
methyltransferase 1 (DNMT1)-targeted inhibition for cancer
therapy. J Mol Med 85: 1137-1148, 2007.
56 Foulks JM, Parnell KM, Nix RN, Chau S, Swierczek K,
Saunders M, Wright K, Hendrickson TF, Ho KK, McCullar MV
and Kanner SB: Epigenetic drug discovery: Targeting DNA
methyltransferases. J Biomol Screen 17: 2-17, 2012.
Received May 10, 2013
Revised May 24, 2013
Accepted May 27, 2013
ANTICANCER RESEARCH 33: 2989-2996 (2013)
2996
... In fact, inhibition of DNA methyltransferase (DNMT) or DNMT gene deletion was shown to be protective against delayed ischemic brain injury in mice [37]. The use of decitabine or azacytidine is limited by their poor chemical stability and relative toxicity [38]. This has been partly overcome by the introduction of zebularine, a DNMT inhibitor with a more stable structure and low cytotoxicity [38]. ...
... The use of decitabine or azacytidine is limited by their poor chemical stability and relative toxicity [38]. This has been partly overcome by the introduction of zebularine, a DNMT inhibitor with a more stable structure and low cytotoxicity [38]. ...
Article
Full-text available
Recent studies suggest that differential DNA methylation could play a role in the mechanism of cerebral vasospasm (CVS) and delayed cerebral ischemia (DCI) after aneurysmal subarachnoid hemorrhage (aSAH). Considering the significance of this matter and a lack of effective prophylaxis against DCI, we aim to summarize the current state of knowledge regarding their associations with DNA methylation and identify the gaps for a future trial. PubMed MEDLINE, Scopus, and Web of Science were searched by two authors in three waves for relevant DNA methylation association studies in DCI after aSAH. PRISMA checklist was followed for a systematic structure. STROBE statement was used to assess the quality and risk of bias within studies. This research was funded by the National Science Centre, Poland (grant number 2021/41/N/NZ2/00844). Of 70 records, 7 peer-reviewed articles met the eligibility criteria. Five studies used a candidate gene approach, three were epigenome-wide association studies (EWAS), one utilized bioinformatics of the previous EWAS, with two studies using more than one approach. Methylation status of four cytosine-guanine dinucleotides (CpGs) related to four distinct genes (ITPR3, HAMP, INSR, CDHR5) have been found significantly or suggestively associated with DCI after aSAH. Analysis of epigenetic clocks yielded significant association of lower age acceleration with radiological CVS but not with DCI. Hub genes for hypermethylation (VHL, KIF3A, KIFAP3, RACGAP1, OPRM1) and hypomethylation (ALB, IL5) in DCI have been indicated through bioinformatics analysis. As none of the CpGs overlapped across the studies, meta-analysis was not applicable. The identified methylation sites might potentially serve as a biomarker for early diagnosis of DCI after aSAH in future. However, a lack of overlapping results prompts the need for large-scale multicenter studies. Challenges and prospects are discussed.
... The existing DNMT1 inhibitors 5-azacytidine (Vidaza) and 5-aza-2′-deoxycytidine (decitabine) are nucleoside analogs given approval by the USFDA for the management of myelodysplastic syndrome and leukemia (Fig. 1). However, these nucleoside analogs are reportedly making covalent binding with DNA resulting in cytotoxicity and nonspecificity [22,23]. Therefore, the identification of specific small-molecule DNMT1 inhibitors with improved potency and less toxic effects has a significant interest in anticancer drug development. ...
Article
Full-text available
DNA methyl transferases (DNMTs) are one of the crucial epigenetic modulators associated with a wide variety of cancer conditions. Among the DNMT isoforms, DNMT1 is correlated with bladder, pancreatic, and breast cancer, as well as acute myeloid leukemia and esophagus squamous cell carcinoma. Therefore, the inhibition of DNMT1 could be an attractive target for combating cancers and other metabolic disorders. The disadvantages of the existing nucleoside and non-nucleoside DNMT1 inhibitors are the main motive for the discovery of novel promising inhibitors. Here, pharmacophore modeling, 3D-QSAR, and e-pharmacophore modeling of DNMT1 inhibitors were performed for the large fragment database screening. The resulting fragments with high dock scores were combined into molecules. The current study revealed several constitutional pharmacophoric features that can be essential for selective DNMT1 inhibition. The fragment docking and virtual screening identified 10 final hit molecules that exhibited good binding affinities in terms of docking score, binding free energies, and acceptable ADME properties. Also, the modified lead molecules (GL1b and GL2b) designed in this study showed effective binding with DNMT1 confirmed by their docking scores, binding free energies, 3D-QSAR predicted activities and acceptable drug-like properties. The MD simulation studies also suggested that leads (GL1b and GL2b) formed stable complexes with DNMT1. Therefore, the findings of this study can provide effective information for the development/identification of novel DNMT1 inhibitors as effective anticancer agents.
... DNMTs remain attached to the DNA even in the presence of DNMTi, but the proteasome pathway breaks them down later on (Santi et al. 1984). The ribonucleoside analogue 5-AZA is present in the RNA molecule as well as in the DNA molecule in limited quantity but the deoxyribose analogue decitabine is present only in DNA (Gnyszka et al. 2013). ...
Article
Full-text available
Scientists are making efforts to search for new metabolites as they are essential lead molecules for the drug discovery, much required due to the evolution of multi drug resistance and new diseases. Moreover, higher production of known drugs is required because of the ever growing population. Microorganisms offer a vast collection of chemically distinct compounds that exhibit various biological functions. They play a crucial role in safeguarding crops, agriculture, and combating several infectious ailments and cancer. Research on fungi have grabbed a lot of attention after the discovery of penicillin, most of the compounds produced by fungi under normal cultivation conditions are discovered and now rarely new compounds are discovered. Treatment of fungi with the epigenetic modifiers has been becoming very popular since the last few years to boost the discovery of new molecules and enhance the production of already known molecules. Epigenetic literally means above genetics that actually does not alter the genome but alter its expression by altering the state of chromatin from heterochromatin to euchromatin. Chromatin in heterochromatin state usually doesn’t express because it is closely packed by histones in this state. Epigenetic modifiers loosen the packing of chromatin by inhibiting DNA methylation and histone deacetylation and thus permit the expression of genes that usually remain dormant. This study delves into the possibility of utilizing epigenetic modifying agents to generate pharmacologically significant secondary metabolites from fungi.
... Topoisomerase inhibitors are well known for their ability to covalently link their respective enzyme targets to DNA 48 . DNA methyltransferases are also known to be trapped on DNA by their nucleotide analog inhibitors 49 . In this report, we present the first evidence of small-molecule inhibitor trapping of Polθ-pol on DNA as the underlying mechanism of action of an effective Polθi class. ...
Article
Full-text available
The DNA damage response (DDR) protein DNA Polymerase θ (Polθ) is synthetic lethal with homologous recombination (HR) factors and is therefore a promising drug target in BRCA1/2 mutant cancers. We discover an allosteric Polθ inhibitor (Polθi) class with 4–6 nM IC50 that selectively kills HR-deficient cells and acts synergistically with PARP inhibitors (PARPi) in multiple genetic backgrounds. X-ray crystallography and biochemistry reveal that Polθi selectively inhibits Polθ polymerase (Polθ-pol) in the closed conformation on B-form DNA/DNA via an induced fit mechanism. In contrast, Polθi fails to inhibit Polθ-pol catalytic activity on A-form DNA/RNA in which the enzyme binds in the open configuration. Remarkably, Polθi binding to the Polθ-pol:DNA/DNA closed complex traps the polymerase on DNA for more than forty minutes which elucidates the inhibitory mechanism of action. These data reveal a unique small-molecule DNA polymerase:DNA trapping mechanism that induces synthetic lethality in HR-deficient cells and potentiates the activity of PARPi.
Article
Full-text available
Epigenetic alterations my play a role in the aggressive behavior of Non-Small Cell Lung Cancer (NSCLC). Treatment with the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA, vorinostat) has been reported to interfere with the proliferative and invasive potential of NSCLC cells. In addition, the DNA methyltransferase inhibitor azacytidine (AZA, vidaza) can modulate the levels of the metastasis suppressor KiSS-1. Thus, since cisplatin is still clinically available for NSCLC therapy, the aim of this study was to evaluate drug combinations between cisplatin and SAHA as well as AZA using cisplatin-sensitive H460 and -resistant H460/Pt NSCLC cells in relation to KiSS-1 modulation. An analysis of drug interaction according to the Combination-Index values indicated a more marked synergistic effect when the exposure to SAHA or AZA preceded cisplatin treatment with respect to a simultaneous schedule. A modulation of proteins involved in apoptosis (p53, Bax) was found in both sensitive and resistant cells, and compared to the treatment with epigenetic agents alone, the combination of cisplatin and SAHA or AZA increased apoptosis induction. The epigenetic treatments, both as single agents and in combination, increased the release of KiSS-1. Finally, the exposure of cisplatin-sensitive and -resistant cells to the kisspeptin KP10 enhanced cisplatin induced cell death. The efficacy of the combination of SAHA and cisplatin was tested in vivo after subcutaneous inoculum of parental and resistant cells in immunodeficient mice. A significant tumor volume inhibition was found when mice bearing advanced tumors were treated with the combination of SAHA and cisplatin according to the best schedule identified in cellular studies. These results, together with the available literature, support that epigenetic drugs are amenable for the combination treatment of NSCLC, including patients bearing cisplatin-resistant tumors.
Article
Full-text available
Over the past thirty years, epigenetic regulation of gene expression has gained increasing interest as it was shown to be implicated in illnesses ranging from cancers to parasitic diseases. In the malaria parasite, epigenetics was shown to be involved in several key steps of the complex life cycle of Plasmodium, among which asexual development and sexual commitment, but also in major biological processes like immune evasion, response to environmental changes or DNA repair. Because epigenetics plays such paramount roles in the Plasmodium parasite, enzymes involved in these regulating pathways represent a reservoir of potential therapeutic targets. This review focuses on epigenetic regulatory processes and their effectors in the malaria parasite, as well as the inhibitors of epigenetic pathways and their potential as new anti-malarial drugs. Such types of drugs could be formidable tools that may contribute to malaria eradication in a context of widespread resistance to conventional anti-malarials.
Article
Full-text available
Colorectal cancer is the most common malignancy of the gastrointestinal tract and a leading cause of cancer-related deaths worldwide. In order to detect early precursor lesions, colonoscopy is widely used. Unfortunately, patient adherence to colonoscopy is poor, which is partially due to the modest performance of currently used prescreening tests. Recently, epigenetics added an additional layer to the understanding of colorectal carcinogenesis. DNA methylation as part of the epigenetic gene-silencing complex is a universally occurring change in colorectal cancer and arises prior to the onset of recognizable preneoplastic changes, which may have huge preventive implications. Herein we discuss the major developments in the field of colorectal carcinogenesis and DNA methylation, including alterations in non-neoplastic conditions such as aging and ulcerative colitis. We try to demonstrate how this epigenetic modification can be harnessed to address some of the key issues impeding the successful clinical management of colorectal cancer.
Article
Full-text available
Mechanism-based inhibitors of enzymes, which mimic reactive intermediates in the reaction pathway, have been deployed extensively in the analysis of metabolic pathways and as candidate drugs. The inhibition of cytosine-[C5]-specific DNA methyltransferases (C5 MTases) by oligodeoxynucleotides containing 5-azadeoxycytidine (AzadC) and 5-fluorodeoxycytidine (FdC) provides a well-documented example of mechanism-based inhibition of enzymes central to nucleic acid metabolism. Here, we describe the interaction between the C5 MTase from Haemophilus haemolyticus (M.HhaI) and an oligodeoxynucleotide duplex containing 2-H pyrimidinone, an analogue often referred to as zebularine and known to give rise to high-affinity complexes with MTases. X-ray crystallography has demonstrated the formation of a covalent bond between M.HhaI and the 2-H pyrimidinone-containing oligodeoxynucleotide. This observation enables a comparison between the mechanisms of action of 2-H pyrimidinone with other mechanism-based inhibitors such as FdC. This novel complex provides a molecular explanation for the mechanism of action of the anti-cancer drug zebularine.
Article
Full-text available
Epigenetic modification of DNA leads to changes in gene expression. DNA methyltransferases (DNMTs) comprise a family of nuclear enzymes that catalyze the methylation of CpG dinucleotides, resulting in an epigenetic methylome distinguished between normal cells and those in disease states such as cancer. Disrupting gene expression patterns through promoter methylation has been implicated in many malignancies and supports DNMTs as attractive therapeutic targets. This review focuses on the rationale of targeting DNMTs in cancer, the historical approach to DNMT inhibition, and current marketed hypomethylating therapeutics azacytidine and decitabine. In addition, we address novel DNMT inhibitory agents emerging in development, including CP-4200 and SGI-110, analogs of azacytidine and decitabine, respectively; the oligonucleotides MG98 and miR29a; and a number of reversible inhibitors, some of which appear to be selective against particular DNMT isoforms. Finally, we discuss future opportunities and challenges for next-generation therapeutics.
Article
Epigenetic alterations such as aberrant DNA methylation and histone modifications contribute substantially to both the cause and maintenance of drug resistance. These epigenetic changes lead to silencing of tumor suppressor genes involved in key DNA damage-response pathways, making drug-resistant cancer cells nonresponsive to conventional anticancer drug therapies. Our hypothesis is that treating drug-resistant cells with epigenetic drugs could restore the sensitivity to anticancer drugs by reactivating previously silenced genes. To test our hypothesis, we used drug-resistant breast cancer cells (MCF-7/ADR) and two epigenetic drugs that act via different mechanisms-5-aza-2' deoxycytidine (Decitabine, DAC), a demethylating agent and suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor-in combination with doxorubicin. We show that the sequential treatment of resistant cells, first with an epigenetic drug (DAC), and then with doxorubicin, induces a highly synergistic effect, thus reducing the IC50 of doxorubicin by several thousand folds. The sequential treatment caused over 90% resistant cells to undergo G2/M cell cycle arrest, determined to be due to upregulation of p21WAF1/CIP1 expression, which is responsible for cell-cycle regulation. The induction of p21WAF1/CIP1 correlated well with the depletion of DNA methyltransferase1 (DNMT1), an enzyme that promotes methylation of DNA, suggesting that the p21WAF1/CIP1 gene may have been methylated and hence is inactive in MCF-7/ADR cells. Microarray analysis shows expression of several tumor suppressor genes and downregulation of tumor promoter genes, particularly in sequentially treated resistant cells. Sequential treatment was found to be significantly more effective than simultaneous treatment, and DAC was more effective than SAHA in overcoming doxorubicin resistance. Synergistic effect with sequential treatment was also seen in drug-sensitive breast cancer cells, but the effect was significantly more pronounced in resistant cells. In conclusion, the sequential treatment of an epigenetic drug in combination with doxorubicin induces a highly synergistic effect that overcomes doxorubicin resistance in breast cancer cells.
Article
This review presents the different human DNA methyltransferases (DNMTs), their biological roles, their mechanisms of action and their role in cancer. The description of assays for detecting DNMT inhibitors (DNMTi) follows. The different known DNMTi are reported along with their advantages, drawbacks and clinical trials. A discussion on the features of the future DNMT inhibitors will conclude this review.
Article
The recent approval of azacitidine (Vidaza®), decitabine (Dacogen®) and vorinostat (Zolinza™) for myelodysplastic syndrome and cutaneous T-cell lymphoma has led to a wave of interest in epigenetic therapy. These DNA methylation inhibitors and the histone deacetylase inhibitor clearly have demonstrated activity in hematologic malignancies, but the future role of epigenetic therapy in solid tumors is still unknown. What is not commonly known is that azacitidine and decitabine were originally developed as cytotoxic nucleoside analogs and clinical trials were previously conducted in a variety of cancer types prior to the knowledge of their ability to inhibit DNA methylation. We review the experience of azacitidine and decitabine in early clinical trials and demonstrate the activity of epigenetic therapy in solid tumors.
Article
The epigenome which comprises DNA methylation, histone modifications, chromatin structures and non-coding RNAs controls gene expression patterns. In cancer cells, there are aberrant changes in the epigenome. The question in cancer epigenetics is that whether these changes are the cause of cell transformation, or rather the consequence of it. We will discuss the epigenetic phenomenon in cancer, as well as the recent interests in the epigenetic reprogramming events, and their implications in the cancer stem cell theory. We will also look at the progression of cancers as they become more aggressive, with focus on the role of epigenetics in tumor metastases exemplified with the urokinase plasminogen activator (uPA) system. Last but not least, with therapeutics intervention in mind, we will highlight the importance of balance in the design of epigenetic based anti-cancer therapeutic strategies.