Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us
Publications > Journals > Gene Expression > Article Full Text
OPEN ACCESS

Potential Epigenetic Modifiers Targeting the Alteration of Methylation in Colorectal Cancer

  • Abdeslam Jaafari1,2,* 

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Jaafari A. Potential Epigenetic Modifiers Targeting the Alteration of Methylation in Colorectal Cancer. Gene Expr. 2024;23(2):139-152. doi: 10.14218/GE.2023.00039S.

Abstract

Colorectal cancer (CRC) is one of the most frequent causes of cancer-related death worldwide. Chemotherapeutic agents used in CRC treatment include oxaliplatin, irinotecan, leucovorin, Tegafur-Uracil, capecitabine, 5-fluorouracil, and monoclonal antibodies. The development of other effective drugs is urgently needed for CRC patients. As the epigenetics of CRC is increasingly understood, epigenetic modifiers (or epidrugs) targeting epigenetic mechanisms could play an important role in this process. During the past two decades, many studies have demonstrated that many specific genes are silenced by hypermethylation of their promoters in CRC, which means that the expression of these genes could be restored since epigenetic alterations are reversible. In fact, some molecules have been studied for their ability to inhibit DNA methyltransferases, and the results showed that silenced genes were reactivated. These molecules could be natural, such as curcumin, tea polyphenols, quercetin, and nanaomycycin A, or synthetic, such as 5-azacytidine, decitabine, procainamide, and zebularine. On the other hand, we hypothesized in this article that ten-eleven translocation inhibitors could be another class of epigenetic modifiers since they could prevent chromosomal instability through decreasing the global hypomethylation of genomic DNA. Some studies have reported that some ten-eleven translocation inhibitors exhibit anticancer effects, which supports our hypothesis. Additionally, we have proposed combinations of these epigenetic modifiers according to different parameters.

Keywords

CRC, Colorectal cancer, Epigenetics, Methylation, Epidrugs, DNA methyltransferase inhibitors, DNMTi, TETi

Introduction

Colorectal cancer (CRC) is considered one of the most frequent causes of cancer-related death worldwide. In 2020, 1.9 million people were diagnosed with colorectal cancer (≈10% of all new cancer cases worldwide). This number will increase to 3.2 million new cases per year. The CRC incidence increases with age (≈80% of all new cases in individuals aged more than 55). Despite recent progress in CRC treatment, the relative five-year survival rate has decreased to ≈70%.1 About 10% of cancer deaths are directly attributed to CRC, which is the second leading cause of cancer mortality. In 2020, the number of deaths due to CRC was estimated at 935,000 worldwide (50% occurring between ages 50 and 74) and will reach 1.6 million deaths per year by 2040 according to the WHO. Furthermore, it has been reported that CRC causes important treatment costs over time depending on the stage of disease, cancer subtype, country, and individual. On the other hand, CRC has been reported to have high mortality rates of 45%, 35% and 47.8% in Europe, the USA and worldwide, respectively, which makes it the third most frequent cancer worldwide in both sexes.1,2

Regarding CRC treatment, Chemotherapy can be used before surgery for some patients to shrink the tumor before it is eradicated. In the late stages, it is used to increase the life expectancy of patients. Chemotherapeutic agents may include oxaliplatin, irinotecan, leucovorin, Tegafur-Uracil capecitabine, or 5-fluorouracil. Additionally, cetuximab, panitumumab, or bevacizumab could be used as monoclonal antibodies in CRC immunotherapy.3 The development of other effective drugs is urgently needed for CRC patients. As the epigenetics of CRC are increasingly understood, epigenetic modifiers (or epidrugs) targeting epigenetic mechanisms constitute a way to explore this process.

In this review, we will first elucidate epigenetic modifications involved in CRC with a focus on DNA methylation (hypermethylation and hypomethylation) and then provide a list of potential molecules (natural and synthetic) that could be used as epigenetic modifiers to target alterations in gene methylation, one of the most involved epigenetic alterations in CRC. Finally, we propose combinations of these epigenetic modifiers according to different parameters, such as their source, mechanism of action, target and chemical structure. Information given in this paper could help researchers in the development of new molecules and combinations of molecules to test as potential epigenetic modifiers in the treatment of methylation alterations in CRC and other cancers.

Colorectal cancer epigenetics

Epigenetics is a heritable alteration of gene expression that does not affect the coding sequence of a gene. It has been confirmed that epigenetic alterations play a central role in cancer pathogenesis, especially in colorectal cancer.4 During the past two decades, many studies have reported a correlation between certain CRC-specific gene expression patterns and the absence of gene mutations. For example, microsatellite instability, one of the hallmarks of the CRC molecular subgroup, is the result of a deficiency in the DNA mismatch repair system. This instability, in addition to genetic mutations in one of the mismatch repair genes, can also be the consequence of epigenetic silencing of the MLH1 gene by hypermethylation of its promoter.5 On the other hand, it has been demonstrated that global hypomethylation of DNA could also lead to CRC through chromosomal instability (CIN).6

DNA methylation in CRC

DNA hypermethylation

Compared with those in normal cells, there are hundreds of hypermethylated genes in CRC tumors, as revealed by CRC epigenome sequencing.7 In the same study, researchers also showed that epigenetic alterations in tumors are significantly more prevalent than genetic alterations among patients.7 Another study reported that some groups of CRC patients have greater hypermethylation of promoter regions, known as the CpG island methylator phenotype.8 In a large study, genome methylation profiles were compared between CRC cells and normal cells using the MBD-capture protocol.9 A total of 322,551 genomic regions (249.5 Mb of the human genome including 7 million CpG sites) were analyzed. According to the results of this study, most of the differentially methylated regions (DMRs) between CRC and normal cells were hypermethylated.9 Furthermore, hypermethylated DMRs were more frequent in intragenic, gene-regulatory, or CpG shelf-shore island segments, as shown in Table 1.10–32

Table 1

Genes silenced by hypermethylation in CRC classified according to their function

Gene (Protein)FunctionTargetEffect of gene silencingReference
APC (Adenomatous polyposis coli)Wnt signaling pathway inhibitionWnt signaling pathway inhibitionIncreased Wnt/β-catenin signaling10
SFRP2 (Secreted frizzled-related protein 2)Wnt antagonistIncreased Wnt/β-catenin signaling11
SFRP1 (Secreted frizzled-related protein 1)Wnt antagonistIncreased Wnt/β-catenin signaling11
MGMT (O-6-methylguanine-DNA methyltransferase)Repair of alkylation DNA damageDNA repairIncreased G→A mutation frequency12
MLH1 (MutL homolog 1)DNA Mismatch repairMicrosatellite instability13
HLTF (Helicase-like transcription factor)dsDNA translocase, fork remodeling activity, ubiquitin ligaseImpaired DNA repair14
RUNX3 (Runt-related transcription factor 3)Transcription factorDNA TranscriptionDecreased TGF-β/BMP signaling15
ID4 (Inhibitor of DNA binding 4)Transcription factor?16
IRF8 (Interferon regulatory factor 8)Transcription factorInterferon signaling17
ESR1 (Estrogen receptor 1)Ligand-activated transcription factorLoss of estrogen receptor signaling18
CDH1 (E-Cadherin)Calcium dependent cell-cell adhesion glycoproteinAdhesionLoss of cell adhesion, possible increased Wnt/β-catenin signaling19
CDH13 (Cadherin 13)Selective cell recognition and adhesion, anti-apoptoticIncreased PI3K/Akt/mTOR signaling, MAPK signaling20
THBS1/TSP1 (Thrombospondin 1)Cell-to-cell and cell-to-matrix adhesive glycoproteinDecreased TGF-β1 signaling21
CRABP1 (Retinol-binding protein 1)Carrier protein for transport of retinol, promotes apoptosisTransport?22
SLC5A8 (Sodium solute symporter family 5 member 8)Sodium and short-chain fatty acid transporter, suppress colony formation?23
DAPK (Death associated protein kinase)Induction of cell deathApoptosisInterferon gamma signaling, TNF alpha signaling, Fas/APO1 signaling24
RASSF1A (Ras association domain family 1 (isoform A))Negative RAS effector, pro-apoptotic, microtubule stabilizationIncreased RAS/RAF/MAP kinase signaling, death receptor-dependent apoptosis25
CDKN2A (P14, ARF) (Protein 14)Inhibits E3 ubiquitin ligaseCell cycle regulationDecreased p53 stabilization and activation26
CDKN2A/p16 (Cyclin-dependent kinase inhibitor 2A)Regulates cell cycle G1 progressionIncreased cell proliferation27
SEPT9 (Septin 9)GTPase, formation of filamentsImpaired Cytokinesis and loss of cell cycle control28
SEPT9 (Septin 9)GTPase, formation of filamentsInvasion and MigrationImpaired Cytokinesis and loss of cell cycle control29
CXCL12 (Chemokine)(C-X-C motif) ligand 12 Alpha chemokineIncreased tumor cell metastases30
VIM (Vimentin)Stabilizing cytoskeletonIncreased tumor cell metastases31
TIMP3 (Tissue inhibitor of metalloproteinase 3)Inhibition of MMPs and ADAMsAngiogenesisIncreased EGF receptor signaling, TNF alpha signaling32

DNA hypomethylation

The development and progression of CRC involves a series of events. These different steps, starting with the transformation of normal colonic epithelium to an adenomatous intermediate and then to an adenocarcinoma, require multiple genetic mutations. Among these genetic events, genomic instability is now described as the main molecular driving force in CRC. In fact, CIN has been reported in 65%–70% of sporadic CRC patients.33 There are at least three different pathways associated with genomic instability: CIN, microsatellite instability, and the CpG island methylator phenotype. The majority of CRC cases are caused by the CIN pathway. CIN is characterized by aneuploidy (imbalances in chromosome number) and loss of heterozygosity (LOH), and it can result from defects in chromosomal segregation, telomere stability, and DNA damage repair.34

Hypomethylation is recognized as an early molecular event leading to CIN that can cause deletion, translocation, inversion or duplication in the entire or part of the chromosome.35 As a consequence of global DNA hypomethylation, some genes can be released from inhibition induced by the methylation of their promoter and become overexpressed, which leads to the activation of oncogenic pathways that are crucial for CRC pathogenesis. Additionally, the exploration of global DNA methylation could have potential clinical applications, such as early cancer formation by cell-free DNA (cfDNA) fraction analysis. In fact, there is a good correlation between global DNA methylation and long interspersed nuclear element (LINE-1) methylation levels since LINE-1 (mobile genetic elements) retrotransposon composes approximately 17% of the human genome.33

Histone modifications

Histone modifications also play a key role in carcinogenesis. In the context of CRC, histone acetylation and methylation status have been widely studied, and it has been shown that they are associated with various clinicopathological features of CRC. In fact, it has been shown that H3K9me (methylation of lysine 9 on histone 3) is more prevalent in CRC and adenomas compared to normal colonic mucosa, and H3K27ac (acetylation of lysine 27 on H3) and H4K12ac (acetylation of lysine 12 on H4) are more prevalent in CRC in comparison with normal mucosa.36 Additionally, ChIP (chromatin immunoprecipitation technique) of circulating nucleosomes revealed decreased levels of H3K9me3 (trimethylation of lysine 9 on H3) and H4K20me3 (trimethylation of lysine 20 on H4) in individuals with CRC compared with healthy individuals.37

Noncoding RNAs (ncRNAs)

ncRNAs (lncRNAs (long ncRNAs) and miRNAs (microRNAs)), which are considered to act via another epigenetic mechanism, can inhibit protein expression and consequently influence many cancer-related pathways, especially at the post-transcriptional level. MicroRNAs are involved in all CRC stages (initiation, progression and metastasis).38 For example, miR-143 prevents cell proliferation through the inhibition of the KRAS mRNA transcript. This mRNA was found to be frequently downregulated in CRC.39

LncRNAs have attracted increasing interest as markers of CRC over the past few years. Many lncRNAs related specifically to colon cancer have been identified in different databases (NONCODE database, PubMed, etc.). Most of these lncRNAs are upregulated, and it seems that they function as miRNA sponges. HOX transcript antisense intergenic RNA (HOTAIR), a lncRNA that has been analyzed in both serum and tissue, was shown to be upregulated in the early stages of CRC development.40 Colon cancer-associated transcript 1 (CCAT1) is another CRC-related lncRNA that is reported to be upregulated in both cancer cells and blood.41 In addition, it has been demonstrated that the expression of the lncRNA growth arrest specific 5 (GAS5) is downregulated in human colon cancer cells compared to normal tissues.42

Signaling pathways

In CRC, as well as in all other cancers, alterations in several signaling pathways, such as the mitogen-activated protein kinase (MAPK) pathway, PI3K pathway, and Wnt/β-catenin pathway, can lead to the onset and development of cancer.43 Crosstalk between these pathways can also promote colon cancer invasion and increase resistance to drugs. Other pathways were reported to be involved in CRC, such as the Janus-activated kinase/signal transducers and activators of transcription pathway, 67 kDa laminin receptor pathway, nuclear factor-kappa B pathway, nuclear factor-erythroid 2-related factor pathway, and Hippo pathway. The Hippo pathway was shown to be responsible for cell proliferation, differentiation, apoptosis, and tumorigenesis,44 and its interaction with the Wnt/β-catenin pathway is crucial for colorectal cancer development (Fig. 1).45

Some epigenetic alterations detected in CRC.
Fig. 1  Some epigenetic alterations detected in CRC.

Epigenetic alterations leading to CRC include DNA methylation (hypermethylation of TSG promotors and global hypomethylation that leads to CIN), histone modifications, and ncRNA (miRNA and lncRNA). ↗ (increased), + (presence), → (leads to). CRC, coloractal cancer; TSG, tumor suppressor gene, CIN, chromosomal instability; miRNA, microRNA; ncRNA, non coding RNA; lncRNA, long non coding RNA.

Inhibitors targeting molecular signal transduction have modest efficacy in nonhematologic malignancies because of the complexity of the genome in solid tumors. Thus, molecules designed to target abnormal DNA methylation, particularly in CRC, might be a more efficacious anticancer treatment strategy. As epigenetic modifications are reversible, the molecules that can target the enzymes involved in these mechanisms may be good drug candidates to fix these alterations and subsequently restore normal gene expression. These molecules are referred to as epidrugs or epigenetic modifiers. In fact, many recent studies have reported that certain molecules are able to inhibit enzymes like DNMT (DNA methyltransferase) and HDAC (histone deacetylase), which are responsible for DNA methylation and histone deacetylation, respectively.46

Epigenetic modifiers as potential epidrugs for CRC treatment

As we have mentioned before, many studies have confirmed that epigenetic mechanisms, such as DNA methylation, histone modifications and ncRNAs, are strongly involved in many processes of cellular physiology and development, and the alteration of these mechanisms could, under certain conditions, lead to carcinogenesis through alteration of the expression of oncogenes or tumor suppressor genes. Thus, since epigenetic modifications are reversible, molecules able to restore normal gene expression by fixing epigenetic alterations could be good candidates for use as agents in the treatment of cancer, either alone or in combination with conventional treatment. These molecules, called epigenetic modifiers or epidrugs, can target DNA methylation, histone modifications or ncRNAs. In this article, we will focus on molecules that target DNA methylation in CRC, namely, DNMT inhibitors (as hypomethylating or demethylating agents) and ten-eleven translocation (TET) inhibitors (as potential hypomethylating agents).

Demethylating (hypomethylating) molecules: DNA methyltransferase inhibitors (DNMTis)

DNA methyltransferases

DNA methylation is the most studied epigenetic mechanism, and it could be considered the most important. In fact, DNA methylation is involved in many physiological mechanisms, such as cellular differentiation, parental imprinting, and X-chromosome inactivation. DNA methylation is catalyzed by enzymes called DNA methyltransferases (DNMTs). These enzymes lead to the formation of 5-methylcytosine (5mC) by transferring a methyl group from S-adenosyl-L-methionine to cytosine at CpG sites in the gene promoter. In humans, de novo DNA methylation is ensured by DNMT3, while DNA methylation is maintained by DNMT1 (a multidomain protein consisting of 1,616 amino acids).

Currently, there is no doubt that deregulation of DNA methylation is associated with diseases, especially cancers. In fact, this epigenetic modification, when it occurs on oncogenes or tumor suppressor genes, could lead to carcinogenesis.47 Additionally, because of alterations in DNA methylation, cancer cells can escape apoptosis and resist chemotherapy.48

Recently, some studies reported that certain drugs approved for other indications could be good candidates for use as DNA methylation inhibitors. Among these drugs, hydralazine, procaine,49 procainamide,50 and certain antibiotics, such as mithramycin A.51 Other natural and synthetic molecules could also be good candidates for DNMT inhibitors.52 Recently, researchers have been working on the rational development of small-molecule non-nucleoside inhibitors.53 This family of molecules is steadily growing and comprises a large variety of different chemical scaffolds like polyphenolic compounds such as epigallocatechin-3-gallate (EGCG) or compounds with acidic functions such as caffeic acid or methylenedisalicylic acid.54–56

Synthetic molecules

5-Azacytidine

5-Azacytidine, shown in the Figure 2, is a cytidine analog modified at position 5 of the pyrimidine ring (with nitrogen instead of carbon). Its incorporation into DNA disrupts the interaction between DNA and DNMTs (1 and 3). Consequently, the enzyme remains covalently bound to DNA, and its function is inactivated.57 Furthermore, because of the inhibition of cytosine methylation, there is a passive loss of methylation in daughter cells after replication. Azacitidine, known to reactivate the expression of some tumor suppressor genes (TSGs), is approved by the US FDA for the treatment of myelodysplastic syndrome.58 This molecule restores the normal growth and differentiation of cells via the demethylation of TSG.59

Synthetic demethylating (hypomethylating) molecules.
Fig. 2  Synthetic demethylating (hypomethylating) molecules.

A clinical study reported that treatment with 5-azacytidine in combination with entinostat (an HDAC inhibitor) restored TSG and inhibited the growth of CRC cell lines. The same study showed that the reversal of hypermethylation was observed in a subset of patients and correlated with improved performance status.60

Decitabine (and its derivative S-110)

Decitabineis a nucleoside analog of cytidine obtained by substitution of a carbon by a nitrogen at position 5 of the pyrimidine ring of deoxyribose (Fig. 2). Like 5-azacytidine, decitabine acts by incorporating into RNA and/or DNA during the S phase of the cell cycle. Decitabine is more specific and less toxic than azacitidine, but both are good DNMT inhibitors even at low concentrations.61 In addition, decitabine alone or in combination with other chemotherapeutic agents, such as oxaliplatin, inhibited CRC cell proliferation.62

Procainamide

Procainamide, shown in the Figure 2, is a drug used to treat a variety of atrial and ventricular dysrhythmias. Investigations have shown that this molecule is a specific inhibitor of DNMT1. It is a competitive non-nucleoside inhibitor that interacts with the binding pocket of the enzyme, which increases the affinity of DNMT1 for hemimethylated DNA and S-adenosyl-L-methionine. This inhibition of methylation leads to a reduction in the global 5mC content in the cell and decreases gene-specific hypermethylation at promoter CpG islands, which could lead to the reactivation of TSG inhibited by hypermethylation in cancer cells.63

Shih et al. showed that the IL27RA gene was downregulated in a group of rats with lung injury induced by LPS and upregulated in the LPS+procainamide group. Thus, the demethylation of the IL27RA promoter by procainamide restored the activation of this gene, which is known to have an anti-inflammatory effect in several models of inflammation.64 Another study reported that excessive methylation of antioxidant gene promoters increases oxidative stress in many diseases. The involvement of DNMT inhibitors in decreasing reactive oxygen species production has been observed in many diseases, such as osteoarthritis chondrocytes and lung adenocarcinoma cells.65 Moreover, Chih-Chin has shown that hypermethylation induced by microbial infection is associated with increased inflammation and oxidative stress.66 These results showed that procainamide may be a good candidate for use as a therapeutic agent in diseases caused by oxidative stress, like colorectal cancer (often associated with an alteration of the microbiota), because of its involvement in DNA demethylation, suppression of superoxide production and neutrophil infiltration.66 Gao et al. reported that Procainamide inhibits the Wnt (Wingless type) pathway, which is involved in carcinogenesis. Aberrant promoter methylation of Wnt inhibitory factor-1 (WIF-1) is a crucial mechanism of epigenetic silencing in human cancers, especially in colorectal cancer, where this hypermethylation targets the APC gene (adenomatous polyposis coli), which functions as an inhibitor of the Wnt signaling pathway.67

Procaine

Procaine, shown in the Figure 2, a local anesthetic drug, is a non-nucleoside inhibitor. Its DNMT inhibitor effect was first reported in breast cancer cells, where it induced global DNA demethylation and restored the activation of certain TSGs.68 Moreover, it has been demonstrated that procaine, at high concentrations, decreases the proliferation of different cancer cells.69 Procaine acts by specifically binding to sequences rich in CpG islands, which inhibits the DNMT-DNA interaction.70

Procaine inhibits the proliferation and migration of CRC cells. Chang et al. reported that, when tested on HCT116 cells, procaine significantly inhibited cell viability, increased apoptosis, and decreased the expression level of Ras homolog family member (RhoA) in a dose-dependent manner (p < 0.05). In fact, this drug increased the proportion of HCT116 cells in the G1 phase and downregulated cyclin D1 and cyclin E expression. In this study, it was demonstrated that procaine inhibits the proliferation and migration of CRC cells through inactivation of the ERK/MAPK/FAK pathways via the regulation of RhoA.71

Zebularine

Zebularine, shown in the Figure 2, is a nucleoside analog. Its DNMT inhibitor effect is specific since it forms a close covalent complex between DNMT and zebularine-substituted DNA, which prevents methyl group transfer and subsequently inhibits DNA methylation.72 In human squamous carcinoma cell lines, zebularine has been shown to reduce viability and DNA synthesis through cell cycle arrest at the G2/M phase and through apoptosis.73

The effect of zebularine on colorectal cancer was investigated by Yang et al., who reported that zebularine has cytotoxic effects on cancer cell cultures, tumor xenografts and a mouse model of colitis-associated CRC. This effect is achieved through the stabilization of p53 via the ribosomal protein S7 (RPS7)/MDM2 pathway and DNA damage.74

N-Phthalyl-L-tryptophan (RG-108)

N-Phthalyl-L-tryptophan is a non-nucleoside DNA methyltransferases inhibitor (IC50 = 115 nM) that blocks the active site of DNMTs (Fig. 2). It has been shown that N-phthalyl-L-tryptophan induces the demethylation and reactivation of TSG. In fact, incubation of NALM6 and HCT116 (human colon carcinoma) cell lines with low concentrations of this molecule results in significant DNA demethylation without any detectable toxicity.75

SGI-1027

SGI-1027, shown in the Figure 2, is a quinoline-based compound. It is a nonnucleoside inhibitor of DNMT3B, DNMT3A, and DNMT1 with IC50 values of 7.5 µM, 8 µM, and 12.5 µM, respectively (with poly-dI-dC as the substrate). Jharna Datta et al. demonstrated that SGI-1027 inhibits these DNMTs by competing with S-adenosylmethionine in the methylation reaction. Different cancer cells were treated with SGI-1027, and the results showed that there was selective degradation of DNMT1 with minimal or no effect on DNMT3A or DNMT3B. Moreover, prolonged treatment of RKO (colorectal cancer cells) with SGI-1027 led to demethylation and reactivation of the silenced TSG P16, MLH1, and TIMP3. Additionally, the same study reported the involvement of the proteasomal pathway in the mechanism of action of this molecule. In addition, no significant toxicity has been detected in a rat hepatoma (H4IIE) cell line.76

Laccaic acid A

Laccaic acids or laccainic acids, shown in the Figure 2, are a group of five anthraquinone derivatives. Laccaic acid A (LCA) is a tetrahydroxyanthraquinone (3,5,6,8-tetrahydroxy-9,10-anthraquinone) substituted by two carboxy groups at positions 1 and 2. It has been reported that LCA is a DNMT1 inhibitor with Ki = 310 nM and IC50 = 650 nM. In a study aiming to evaluate the anti-colorectal cancer activity of the combination (LCA+ 5-FU), the results demonstrated that LCA is a highly DNA-competitive inhibitor of DNMT1. According to in vitro methylation assays, LCA competes with DNA substrates and alters the expression of methylated genes in the MCF-7 cell line.77 In another study, it was shown that LCA combined with phenethyl isothiocyanate has a strong synergistic effect on CRC. Additionally, LCA inhibited human colon carcinoma HT29 cell growth with an IC50 value of 6.08 µM after 72 h of treatment and induced cell apoptosis and cell cycle arrest at the sub-G1 phase.78

MG-98

MG-98 is referred to as the second-generation DNMT inhibitor. It is a 20-mer antisense compound with a phosphorothioate backbone. MG-98 is a highly specific inhibitor of DNMT1 mRNA translation in humans. It acts by binding to the 3′ untranslated region of DNMT1 mRNA. In vitro studies have reported that this oligonucleotide restores the expression of the cyclin-dependent kinase inhibitor p16 (α-CDKN2A) through suppression of DNMT1 expression.79 In preclinical studies and clinical phase I/II trials, it has been demonstrated that MG-98 can lead to the reactivation of silenced tumor suppressor genes safely and effectively. Thirty-two patients with gastric, colonic, ovarian, breast, renal, lung cancer, and melanoma were treated with MG-98 administered as a 7-day continuous infusion every 14 days, and DNMT1 activity in PBMCs was monitored during two cycles of therapy. DNMT1 inhibition was reported in 26 of 32 patients, and MG-98 was well tolerated, with early evidence of a clinical effect.80

Natural molecules

Curcumin

Curcumin, shown in the Figure 3, a plant-derived polyphenol, has been shown to inhibit DNMT activity in various cancer cell lines, including colorectal cancer cells, through the regulation of multiple signaling pathways. These pathways include cell proliferation pathway (cyclin D1, c-myc), cell survival pathway (Bcl-2, Bcl-xL, cFLIP, XIAP, c-IAP1), caspase activation pathway (caspase-8, 3, 9), tumor suppressor pathway (p53, p21), death receptor pathway (DR4, DR5), mitochondrial pathways, and protein kinase pathway (JNK, Akt, and AMPK).81,82 Additionally, curcumin treatment has been reported to decrease global DNA methylation in a model of leukemia cells.83 In another study aiming to assess the antitumor effect of curcumin on CC531 colorectal cancer cells both in vitro and in vivo, the results showed that this natural product reduced cell proliferation by more than 30% after 48 h and 50% after 72 h. The same study demonstrated, using a wound healing test, that curcumin inhibited migration. Finally, in vivo, curcumin reduced the tumor volume of liver implants of CRC cells by 5.6-fold.84

Natural demethylating (hypomethylating) molecules.
Fig. 3  Natural demethylating (hypomethylating) molecules.

Tea polyphenols: EGCG, catechin and epicatechin

Several tea catechins and bioflavonoids have been studied for their ability to modulate DNA methylation catalyzed by prokaryotic SssI DNA methyltransferase and human DNMT1 (Fig. 3). According to the results of these studies, catechin and epicatechin inhibited DNMT1 in a dose-dependent manner with IC50 values ranging from 1.0 to 8.4 µM. The IC50 of EGCG, the most potent inhibitor, ranged from 0.21 to 0.47 µM.85 Since EGCG is the most effective polyphenol, its mechanism of action was studied in silico. In silico studies demonstrated that the gallic acid moiety is responsible, in large part, for its high-affinity and direct inhibitory interaction with the DNMT1 catalytic site, and its interaction with the enzyme is stabilized by Mg2+.85

Other in vivo and epidemiological studies have reported that tea polyphenols can inhibit the growth and metastasis of CRC through anti-inflammatory, anti-oxidative, and pro-apoptotic effects. Some studies have shown that these natural molecules can influence several signaling pathways in tumor cells, such as mitogen-activated protein kinase pathway, phosphatidylinositol-3 kinase/Akt pathway, Wnt/β-catenin pathway, and 67 kDa laminin receptor pathway, leading to the inhibition of cell proliferation and apoptosis. Additionally, other studies have suggested that tea polyphenols can prevent the growth and metastasis of colorectal cancer by improving the immune response and decreasing inflammatory responses through the modulation of the gut microbiota composition.86

Quercitin

According to the results of a study aiming to evaluate the potential of quercetin, shown in the Figure 3, as an epigenetic modifier in cancer, this phytochemical decreases the activity of DNMTs, HDACs, and HMTs (histone methyltransferases) in a dose-dependent manner. The same study reported that quercetin decreased global DNA methylation levels in a dose- and time-dependent manner and restored TSG expression by demethylating their promoters. Additionally, an in silico study (molecular docking) showed that quercetin could act as a competitive inhibitor by interacting with residues in the catalytic site of several DNMTs and HDACs.87

The protective effect of quercetin on colon cancer was investigated in 45 rats using azotoxin methane (15 mg/kg s.c.) as a carcinogen. The results showed that quercetin reduces cytological changes in colon cancer cells, decreases beta-catenin and Bcl-2 (anti-apoptotic gene) expression, and increases caspase 3 (apoptotic gene) expression.88

Nanaomycin A

Nanaomycin A (see Figure 3 bellow) is an anthracycline antibiotic belonging to the quinone class. It was isolated from Streptomyces. Its mechanism of action depends on its reduction by respiratory chain-linked NADH or flavin dehydrogenase. The reduced form of this molecule produces singlet molecular oxygen (O2) after its quick auto-oxidation by molecular oxygen, which is responsible for its antimicrobial activity.89

Since it induces antiproliferative activity against cancer cell lines, the epigenetic effect of nanaomycin A was first studied by an in silico screening method for the inhibition of DNMTs. Kuck et al., in a biochemical study, reported that this molecule interacts with the catalytic site of DNMT3B at specific AAs (Glu697, Arg731 and Arg733) of the enzyme binding pocket. It has been shown that this drug decreases the expression of DNMT1, 3A and 3B.56 This molecule can also inhibit DNMT3B activity by reactivating the RASSF1A tumor suppressor gene, which reduces cell proliferation and viability.56,90 In fact, RASSF1A (Ras association domain family 1), a pro-apoptotic gene involved in microtubule stabilization, is among the genes that are commonly silenced by methylation in CRC. The loss of this gene leads to an increase in RAS/RAF/MAP kinase signaling and death receptor-dependent apoptosis.91

Resveratrol

Resveratrol is a natural polyphenol that has many biological activities. Recently, Resveratrol was shown to reactivate silenced tumor suppressor genes through decreasing DNMT expression. Additionally, synthesized derivatives of resveratrol (resveratrol-salicylate) could exhibit an important DNMT inhibitor effect. Some of these analogs selectively inhibit DNMT3. Additionally, the most active derivative showed an important cytotoxic effect (greater than that of resveratrol) against three human cancer cell lines: Hep-G2, SK-BR-3, and especially HT-29 (colorectal adenocarcinoma cells).92,93

Other natural molecules

Other natural molecules, such as fisetin,94 myricetin,95 theaflavin,96 thearubigin,96 trichostatin,97 kazinol,98 genistein, silibinin, luteolin, boswellic acid, mahanine and selenium, have been reported to exhibit a DNMT inhibitory effect,99 which makes them good candidates for use as hypomethylating agents for CRC treatment (Table 2).

Table 2

Some DNMT inhibitors used as potential epigenetic modifiers in CRC, classified according to their target, their origin, and their mechanism of action

DNMT iTargetNucleoside/non nucleosideMechanism of actionNatural/synthetic
AzacitidineGlobal
DNMT
Nucleoside inhibitorIncorporation into the RNA and/or genomic DNASynthetic
DecitabineGlobal
DNMT
Nucleoside inhibitorSynthetic
S110 (derivative of Decitabine)Global
DNMT
Nucleoside inhibitor,Synthetic
CurcuminGlobal
DNMT
Non-nucleoside inhibitor,Direct inhibitory interaction of the catalytic siteNatural
EGCGDNMT1Non-nucleoside inhibitor,Natural
CatechinDNMT1Non-nucleoside inhibitor,Natural
EpicatechinDNMT1Non-nucleoside inhibitor,Natural
QuercetinDNMT1Non-nucleoside inhibitor,Natural
FisetinDNMT1Non-nucleoside inhibitor,Natural
ResveratrolDNMTNon-nucleoside inhibitorNatural
GenisteinDNMT1Non-nucleoside inhibitorNatural
resveratrol-salicylate derivativesDNMT3BSynthetic
MyricetinDNMT1Non-nucleoside inhibitor,Natural
block the binding of DNMTs to DNA
ProcainamideDNMT1Non-nucleoside inhibitor,Synthetic
RG108 (found by virtual screening)DNMT1Non-nucleoside inhibitor,Synthetic
ProcaineGlobal
DNMT
Non-nucleoside inhibitor,Synthetic
SGI-1027 (lipophilic, quinoline-based compound)Global
DNMT
Non-nucleoside inhibitor,competitive inhibitor of SAMSynthetic
ZebularineGlobal
DNMT
Nucleoside inhibitor,form a covalent complex with DNMT and cytidine deaminase in DNASynthetic
Laccaic Acid ADNMT1DNA-competitive inhibitorSynthetic
MG-98 (20 bp anti-sense oligonucleotide)DNMT1Non-nucleoside inhibitor,antisense oligonucleotide binding DNMT1Synthetic
Theaflavin 3, 3′-digallate N6DNMT3ANon-nucleoside inhibitor,dietary polyphenols from black tea and coffeeNatural
ThearubiginDNMT3ANon-nucleoside inhibitor,Natural
Nanaomycin ADNMT3BNon-nucleoside inhibitor,quinone antibioticNatural
Trichostatin ADNMT3BNon-nucleoside inhibitorNatural
Kazinol CDNMTNon-nucleoside inhibitorPro-apoptotic (via AMPK activation)Natural

Hypermethylating molecules

TET Inhibitors (ten-eleven translocation inhibitors)

DNA methylation contributes widely to the dynamic chromatin states that impart precise epigenetic landscapes, which maintain cell type-specific transcriptional programs. Before the discovery of TET protein enzymes, it was believed that DNA methylation is an irreversible epigenetic event. These enzymes can modify methylcytosine and potentially erase DNA methylation.100 They also play a key role in the efficient transcription of target genes necessary for the proliferation and survival of tumor cells.101 TETs are a family of three proteins, TET1, TET2, and TET3, that are responsible for the catalysis of successive oxidation reactions of 5mC to 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine, respectively as seen in the Figure 4 bellow.102,103

Successive oxidation reactions of 5mC.
Fig. 4  Successive oxidation reactions of 5mC.

DNMT, DNA methyltransferase; SAM, s-adenosylmethionine; 5mC, 5-methylcytosine; 5hmC, 5-hydroxymethylcytosine; 5-fC, 5-formylcytosine; 5-caC, 5-carboxycytosine; TET, ten-elleven translocation, 5hmC, alpha ketoglutarate; TDG, thymine DNA glycosylase; BER, base excision repair.

As with DNMTs, the activity of TET enzymes, which are involved in removing epigenetic marks, is also a part of the disruption of epigenetic landscapes characterizing malignant transformation. Moreover, since TETs function is to demethylate CpG islands of genomic DNA, this could be responsible for, or at least increase, global DNA hypomethylation, which could lead to chromosomal instability. Consequently, we can hypothesize that this family of enzymes could be an interesting target for epigenetic modifiers in the treatment of cancer. Indeed, in principle, inhibition of TETs could lead to a decrease in global DNA hypomethylation and therefore prevent chromosomal instability. The fact that some TET inhibitors exhibit anticancer activity supports our hypothesis.104,105

Cytosine-based TET enzyme inhibitors

Some cytosine derivatives, shown in the Figure 5 bellow, have been synthesized by substitution at the 5 position and evaluated for their capacity to inhibit TET1 and TET2. The results showed that the derivative obtained by chlorination at this position, Bobcat212, had the most inhibitory effect, at 57% and 43%, on TET1 and TET2, respectively. Another interesting derivative, Bobcat339, which is substituted at the R2 position with 3-biphenyl, significantly increased the inhibitory effect of TET1 and TET2 without inhibiting DNMT3a.106

Hypermethylating molecules.
Fig. 5  Hypermethylating molecules.

(a) Cytosine derivatives; (b) C35.

αKG-dependent dioxygenases

2-Hydroxyglutarate (2HG), N-oxalylglycine (NOG), and dimethyl fumarate (DMF) are known for their inhibitory effects on a variety of αKG-dependent dioxygenases.107 Several molecules were synthesized by substitution of the C4 position with either -keto, -olefin, -methyl, or -cyclopropyl functional groups, and the C2 position was single or double substituted with -chloro, -fluoro, -hydroxy, -methyl, or –trifluoromethyl groups. These derivatives were subsequently tested in vitro and in vivo for their ability to induce cancer cell death and TET dioxygenase inhibition. These results suggested that TET inhibitors need to be further investigated as a new class of targeted agents for cancer treatment.104

C35

Forty TET inhibitors were designed using a virtual ligand screening pipeline (Lvspipe), and tested in vitro for their inhibitory activity. The results demonstrated that C35, shown in the Figure 5 below, can inhibit the catalytic activities of both TET1 and TET3 with IC50 values of 3.48 µM and 2.31 µM, respectively. Using molecular modeling, the same study reported that there is an interaction between C35 and the TET2 catalytic domain.108

Proposed epidrug combinations

The epigenetic modifiers discussed in this article could be used alone or in combination. Combinations of molecules with different mechanisms of action are widely used to treat different diseases, especially cancer.109 Here, we propose some potential combinations of epidrugs that could be useful for the treatment of CRC. These combinations are designed according to different parameters, as shown in the Table 3.

Table 3

Some proposed combinations of epigenetic modifiers targeting alterations in DNA methylation in CRC based on their mechanism of action

ParameterCombinationExample of combination
Targeted enzyme (DNMT)Inhibitor of DNMT1
+
Inhibitor of DNMT3
Procainamide + nanaoycin A
Laccaic acid + Trichostatin
+ Thearubigin
Mechanism of actionIncorporation into the RNA and/or genomic DNA
+
Direct inhibitory interaction of the catalytic site
Decitabine + curcumin
block the binding of DNMTs to DNA
+
competitive inhibitor of SAM
Procaine + SGI-1027
SourceNatural
+
Synthetic
Decitabine + EGCG
Quercetin + resveratrol-salicylate
Zebularine + Epichatechin
Chemical structureNucleoside + Non-nucleosideS-110 + Procainamide
5-Azacytidine + RG-108

Conclusions

Currently, epigenetic alterations are being increasingly explored as targets in the treatment of cancer, especially CRC. This is because epigenetic modifications are reversible, which means that they can be fixed by molecules called epigenetic modifiers. Among these epigenetic alterations that could lead to CRC carcinogenesis, alterations in the methylation of gene promoters are the most studied, which prompted researchers to look for molecules targeting these alterations. As we have detailed in this article, many natural and synthetic molecules could be interesting candidates as epigenetic modifiers targeting the alteration of methylation in CRC. Two categories of these molecules have been described. The first group consists of DNMT inhibitors, which are hypomethylating (or demethylating) agents that can restore the expression of genes silenced by hypermethylation of their promoters. In the second group, we identified TET inhibitors that could prevent chromosomal instability through decreasing the global hypomethylation of genomic DNA. Finally, these epidrugs could be tested alone, in combination with each other, or with other conventional chemotherapeutic drugs. Although several DNMTis are used in the clinic, there are still some limitations of DNMTis, such as toxicity and lack of selectivity, that could be overcome by strategies like combination with other molecules and/or modification of their chemical structure to develop novel molecules with more efficacy and less limitations.

Abbreviations

ADAMs: 

a disintegrin and metalloproteinase

AMPK: 

AMP-activated protein kinase

CIN: 

chromosomal instability

CRC: 

colorectal cancer

DNA: 

deoxyribonucleic acid

DNMT: 

DNA methyltransferases

DR: 

death receptor

EGCG: 

epigallocatechin gallate

EGF: 

epidermal growth factor

HDAC: 

histone deacetylase

IC50 with lower case: 

half maximal inhibitory concentration

KG: 

alpha ketoglutarate

LCA: 

laccaic acid A

LINE-1: 

long interspersed nuclear element

lncRNA: 

long non coding RNA

microRNAs: 

miRNAs

MMPs: 

matrix metalloproteinases

MSI: 

microsatellite instability

RNA: 

ribonucleic acid

SAM: 

S-adenosyl-L-methionine

TET: 

ten-eleven translocation

TNF: 

tumor necrosis factor

TSG: 

tumor suppressor genes

5hmC: 

5-hydroxymethylcytosine

5mC: 

5-methylcytosine

Declarations

Acknowledgement

The author acknowledges Mohamed Amine Jaafri for his help in revising the English.

Funding

No funding was given to support this publication.

Conflict of interest

The author has no conflict of interest related to this publication.

References

  1. Klimeck L, Heisser T, Hoffmeister M, Brenner H. Colorectal cancer: A health and economic problem. Best Pract Res Clin Gastroenterol 2023;66:101839 View Article PubMed/NCBI
  2. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68(6):394-424 View Article PubMed/NCBI
  3. Hoyle M, Crathorne L, Peters J, Jones-Hughes T, Cooper C, Napier M, et al. The clinical effectiveness and cost-effectiveness of cetuximab (mono- or combination chemotherapy), bevacizumab (combination with non-oxaliplatin chemotherapy) and panitumumab (monotherapy) for the treatment of metastatic colorectal cancer after first-line chemotherapy (review of technology appraisal No.150 and part review of technology appraisal No. 118): a systematic review and economic model. Health Technol Assess 2013;17(14):1-237 View Article PubMed/NCBI
  4. Goel A, Boland CR. Epigenetics of colorectal cancer. Gastroenterology 2012;143(6):1442-1460.e1 View Article PubMed/NCBI
  5. Joo JE, Mahmood K, Walker R, Georgeson P, Candiloro I, Clendenning M, et al. Identifying primary and secondary MLH1 epimutation carriers displaying low-level constitutional MLH1 methylation using droplet digital PCR and genome-wide DNA methylation profiling of colorectal cancers. Clin Epigenetics 2023;15(1):95 View Article PubMed/NCBI
  6. Suter CM, Martin DI, Ward RL. Hypomethylation of L1 retrotransposons in colorectal cancer and adjacent normal tissue. Int J Colorectal Dis 2004;19(2):95-101 View Article PubMed/NCBI
  7. Schuebel KE, Chen W, Cope L, Glöckner SC, Suzuki H, Yi JM, et al. Comparing the DNA hypermethylome with gene mutations in human colorectal cancer. PLoS Genet 2007;3(9):1709-1723 View Article PubMed/NCBI
  8. Weisenberger DJ, Levine AJ, Long TI, Buchanan DD, Walters R, Clendenning M, et al. Association of the colorectal CpG island methylator phenotype with molecular features, risk factors, and family history. Cancer Epidemiol Biomarkers Prev 2015;24(3):512-519 View Article PubMed/NCBI
  9. Orjuela S, Menigatti M, Schraml P, Kambakamba P, Robinson MD, Marra G. The DNA hypermethylation phenotype of colorectal cancer liver metastases resembles that of the primary colorectal cancers. BMC Cancer 2020;20(1):290 View Article PubMed/NCBI
  10. Liang TJ, Wang HX, Zheng YY, Cao YQ, Wu X, Zhou X, et al. APC hypermethylation for early diagnosis of colorectal cancer: a meta-analysis and literature review. Oncotarget 2017;8(28):46468-46479 View Article PubMed/NCBI
  11. Kong C, Fu T. Value of methylation markers in colorectal cancer (Review). Oncol Rep 2021;46(2):177 View Article PubMed/NCBI
  12. Randon G, Pagani F, Pietrantonio F. MGMT Promoter Methylation as a Target In Metastatic Colorectal Cancer: Rapid Turnover and Use of Folates Alter its Study-Response. Clin Cancer Res 2020;26(13):3495 View Article PubMed/NCBI
  13. Li X, Yao X, Wang Y, Hu F, Wang F, Jiang L, et al. MLH1 promoter methylation frequency in colorectal cancer patients and related clinicopathological and molecular features. PLoS One 2013;8(3):e59064 View Article PubMed/NCBI
  14. Moinova HR, Chen WD, Shen L, Smiraglia D, Olechnowicz J, Ravi L, et al. HLTF gene silencing in human colon cancer. Proc Natl Acad Sci U S A 2002;99(7):4562-4567 View Article PubMed/NCBI
  15. Nishio M, Sakakura C, Nagata T, Komiyama S, Myashita A, Hamada T, et al. RUNX3 Promoter Methylation in Colorectal Cancer: Its Relationship with Microsatellite Instability and its Suitability as a Novel Serum Tumor Marker. Anticancer Res 2010;30(7):2673-2682 View Article PubMed/NCBI
  16. Umetani N, Takeuchi H, Fujimoto A, Shinozaki M, Bilchik AJ, Hoon DS. Epigenetic inactivation of ID4 in colorectal carcinomas correlates with poor differentiation and unfavorable prognosis. Clin Cancer Res 2004;10(22):7475-7483 View Article PubMed/NCBI
  17. Gutierrez A, Demond H, Brebi P, Ili CG. Novel Methylation Biomarkers for Colorectal Cancer Prognosis. Biomolecules 2021;11(11):1722 View Article PubMed/NCBI
  18. Wu AH, Siegmund KD, Long TI, Cozen W, Wan P, Tseng CC, et al. Hormone therapy, DNA methylation and colon cancer. Carcinogenesis 2010;31(6):1060-1067 View Article PubMed/NCBI
  19. Wheeler JM, Kim HC, Efstathiou JA, Ilyas M, Mortensen NJ, Bodmer WF. Hypermethylation of the promoter region of the E-cadherin gene (CDH1) in sporadic and ulcerative colitis associated colorectal cancer. Gut 2001;48(3):367-371 View Article PubMed/NCBI
  20. Ye M, Huang T, Li J, Zhou C, Yang P, Ni C, et al. Role of CDH13 promoter methylation in the carcinogenesis, progression, and prognosis of colorectal cancer: A systematic meta-analysis under PRISMA guidelines. Medicine (Baltimore) 2017;96(4):e5956 View Article PubMed/NCBI
  21. Hu XY, Ling ZN, Hong LL, Yu QM, Li P, Ling ZQ. Circulating methylated THBS1 DNAs as a novel marker for predicting peritoneal dissemination in gastric cancer. J Clin Lab Anal 2021;35(9):e23936 View Article PubMed/NCBI
  22. Lind GE, Kleivi K, Meling GI, Teixeira MR, Thiis-Evensen E, Rognum TO, et al. ADAMTS1, CRABP1, and NR3C1 identified as epigenetically deregulated genes in colorectal tumorigenesis. Cell Oncol 2006;28(5-6):259-272 View Article PubMed/NCBI
  23. Li H, Myeroff L, Smiraglia D, Romero MF, Pretlow TP, Kasturi L, et al. SLC5A8, a sodium transporter, is a tumor suppressor gene silenced by methylation in human colon aberrant crypt foci and cancers. Proc Natl Acad Sci U S A 2003;100(14):8412-8417 View Article PubMed/NCBI
  24. Borinstein SC, Conerly M, Dzieciatkowski S, Biswas S, Washington MK, Trobridge P, et al. Aberrant DNA methylation occurs in colon neoplasms arising in the azoxymethane colon cancer model. Mol Carcinog 2010;49(1):94-103 View Article PubMed/NCBI
  25. Sun X, Yuan W, Hao F, Zhuang W. Promoter Methylation of RASSF1A indicates Prognosis for Patients with Stage II and III Colorectal Cancer Treated with Oxaliplatin-Based Chemotherapy. Med Sci Monit 2017;23:5389-5395 View Article PubMed/NCBI
  26. Duggan C, Yu M, Willbanks AR, Tapsoba JD, Wang CY, Grady WM, et al. Exercise effects on DNA methylation in EVL, CDKN2A (p14, ARF), and ESR1 in colon tissue from healthy men and women. Epigenetics 2022;17(10):1070-1079 View Article PubMed/NCBI
  27. Shima K, Nosho K, Baba Y, Cantor M, Meyerhardt JA, Giovannucci EL, et al. Prognostic significance of CDKN2A (p16) promoter methylation and loss of expression in 902 colorectal cancers: Cohort study and literature review. Int J Cancer 2011;128(5):1080-1094 View Article PubMed/NCBI
  28. Wasserkort R, Kalmar A, Valcz G, Spisak S, Krispin M, Toth K, et al. Aberrant septin 9 DNA methylation in colorectal cancer is restricted to a single CpG island. BMC Cancer 2013;13:398 View Article PubMed/NCBI
  29. Lu DC, Zhang QF, Li L, Luo XK, Liang B, Lu YH, et al. Methylated Septin9 has moderate diagnostic value in colorectal cancer detection in Chinese population: A multicenter study. BMC Gastroenterol 2022;22(1):232 View Article PubMed/NCBI
  30. Goïta AA, Guenot D. Colorectal Cancer: The Contribution of CXCL12 and Its Receptors CXCR4 and CXCR7. Cancers (Basel) 2022;14(7):1810 View Article PubMed/NCBI
  31. Mojtabanezhad Shariatpanahi A, Yassi M, Nouraie M, Sahebkar A, Varshoee Tabrizi F, Kerachian MA. The importance of stool DNA methylation in colorectal cancer diagnosis: A meta-analysis. PLoS One 2018;13(7):e0200735 View Article PubMed/NCBI
  32. Ashktorab H, Brim H. DNA Methylation and Colorectal Cancer. Curr Colorectal Cancer Rep 2014;10(4):425-430 View Article PubMed/NCBI
  33. Rowan A, Halford S, Gaasenbeek M, Kemp Z, Sieber O, Volikos E, et al. Refining molecular analysis in the pathways of colorectal carcinogenesis. Clin Gastroenterol Hepatol 2005;3(11):1115-1123 View Article PubMed/NCBI
  34. Pino MS, Chung DC. The chromosomal instability pathway in colon cancer. Gastroenterology 2010;138(6):2059-2072 View Article PubMed/NCBI
  35. Scott EC, Gardner EJ, Masood A, Chuang NT, Vertino PM, Devine SE. A hot L1 retrotransposon evades somatic repression and initiates human colorectal cancer. Genome Res 2016;26(6):745-755 View Article PubMed/NCBI
  36. Karczmarski J, Rubel T, Paziewska A, Mikula M, Bujko M, Kober P, et al. Histone H3 lysine 27 acetylation is altered in colon cancer. Clin Proteomics 2014;11(1):24 View Article PubMed/NCBI
  37. Gezer U, Ustek D, Yörüker EE, Cakiris A, Abaci N, Leszinski G, et al. Characterization of H3K9me3- and H4K20me3-associated circulating nucleosomal DNA by high-throughput sequencing in colorectal cancer. Tumour Biol 2013;34(1):329-336 View Article PubMed/NCBI
  38. Strubberg AM, Madison BB. MicroRNAs in the etiology of colorectal cancer: pathways and clinical implications. Dis Model Mech 2017;10(3):197-214 View Article PubMed/NCBI
  39. Wang D, Liu Q, Ren Y, Zhang Y, Wang X, Liu B. Association analysis of miRNA-related genetic polymorphisms in miR-143/145 and KRAS with colorectal cancer susceptibility and survival. Biosci Rep 2021;41(4):BSR20204136 View Article PubMed/NCBI
  40. Gharib E, Nazemalhosseini-Mojarad E, Baghdar K, Nayeri Z, Sadeghi H, Rezasoltani S, et al. Identification of a stool long non-coding RNAs panel as a potential biomarker for early detection of colorectal cancer. J Clin Lab Anal 2021;35(2):e23601 View Article PubMed/NCBI
  41. Kalmár A, Nagy ZB, Galamb O, Csabai I, Bodor A, Wichmann B, et al. Genome-wide expression profiling in colorectal cancer focusing on lncRNAs in the adenoma-carcinoma transition. BMC Cancer 2019;19(1):1059 View Article PubMed/NCBI
  42. Ni W, Yao S, Zhou Y, Liu Y, Huang P, Zhou A, et al. Long noncoding RNA GAS5 inhibits progression of colorectal cancer by interacting with and triggering YAP phosphorylation and degradation and is negatively regulated by the m(6)A reader YTHDF3. Mol Cancer 2019;18(1):143 View Article PubMed/NCBI
  43. Pennel KAF, Park JH, McMillan DC, Roseweir AK, Edwards J. Signal interaction between the tumour and inflammatory cells in patients with gastrointestinal cancer: Implications for treatment. Cell Signal 2019;54:81-90 View Article PubMed/NCBI
  44. Zhou S, Xu H, Tang Q, Xia H, Bi F. Dipyridamole Enhances the Cytotoxicities of Trametinib against Colon Cancer Cells through Combined Targeting of HMGCS1 and MEK Pathway. Mol Cancer Ther 2020;19(1):135-146 View Article PubMed/NCBI
  45. Hu D, Meng RY, Nguyen TV, Chai OH, Park BH, Lee JS, et al. Inhibition of colorectal cancer tumorigenesis by ursolic acid and doxorubicin is mediated by targeting the Akt signaling pathway and activating the Hippo signaling pathway. Mol Med Rep 2023;27(1):11 View Article PubMed/NCBI
  46. Jin Y, Liu T, Luo H, Liu Y, Liu D. Targeting Epigenetic Regulatory Enzymes for Cancer Therapeutics: Novel Small-Molecule Epidrug Development. Front Oncol 2022;12:848221 View Article PubMed/NCBI
  47. Grady WM, Yu M, Markowitz SD. Epigenetic Alterations in the Gastrointestinal Tract: Current and Emerging Use for Biomarkers of Cancer. Gastroenterology 2021;160(3):690-709 View Article PubMed/NCBI
  48. Zeng Y, Rong H, Xu J, Cao R, Li S, Gao Y, et al. DNA Methylation: An Important Biomarker and Therapeutic Target for Gastric Cancer. Front Genet 2022;13:823905 View Article PubMed/NCBI
  49. Segura-Pacheco B, Perez-Cardenas E, Taja-Chayeb L, Chavez-Blanco A, Revilla-Vazquez A, Benitez-Bribiesca L, et al. Global DNA hypermethylation-associated cancer chemotherapy resistance and its reversion with the demethylating agent hydralazine. J Transl Med 2006;4:32 View Article PubMed/NCBI
  50. Lee BH, Yegnasubramanian S, Lin X, Nelson WG. Procainamide is a specific inhibitor of DNA methyltransferase 1. J Biol Chem 2005;280(49):40749-40756 View Article PubMed/NCBI
  51. Lin RK, Hsu CH, Wang YC. Mithramycin A inhibits DNA methyltransferase and metastasis potential of lung cancer cells. Anticancer Drugs 2007;18(10):1157-1164 View Article PubMed/NCBI
  52. Yu N, Wang M. Anticancer drug discovery targeting DNA hypermethylation. Curr Med Chem 2008;15(14):1350-1375 View Article PubMed/NCBI
  53. Stresemann C, Brueckner B, Musch T, Stopper H, Lyko F. Functional diversity of DNA methyltransferase inhibitors in human cancer cell lines. Cancer Res 2006;66(5):2794-2800 View Article PubMed/NCBI
  54. Gao Z, Xu Z, Hung MS, Lin YC, Wang T, Gong M, et al. Promoter demethylation of WIF-1 by epigallocatechin-3-gallate in lung cancer cells. Anticancer Res 2009;29(6):2025-2030 View Article PubMed/NCBI
  55. Hernandes LC, Machado ART, Tuttis K, Ribeiro DL, Aissa AF, Dévoz PP, et al. Caffeic acid and chlorogenic acid cytotoxicity, genotoxicity and impact on global DNA methylation in human leukemic cell lines. Genet Mol Biol 2020;43(3):e20190347 View Article PubMed/NCBI
  56. Kuck D, Singh N, Lyko F, Medina-Franco JL. Novel and selective DNA methyltransferase inhibitors: Docking-based virtual screening and experimental evaluation. Bioorg Med Chem 2010;18(2):822-829 View Article PubMed/NCBI
  57. Issa JP, Kantarjian HM. Targeting DNA methylation. Clin Cancer Res 2009;15(12):3938-3946 View Article PubMed/NCBI
  58. Xie M, Jiang Q, Xie Y. Comparison between decitabine and azacitidine for the treatment of myelodysplastic syndrome: a meta-analysis with 1,392 participants. Clin Lymphoma Myeloma Leuk 2015;15(1):22-28 View Article PubMed/NCBI
  59. Christman JK. 5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene 2002;21(35):5483-5495 View Article PubMed/NCBI
  60. Azad NS, El-Khoueiry A, Yin J, Oberg AL, Flynn P, Adkins D, et al. Combination epigenetic therapy in metastatic colorectal cancer (mCRC) with subcutaneous 5-azacitidine and entinostat: a phase 2 consortium/stand up 2 cancer study. Oncotarget 2017;8(21):35326-35338 View Article PubMed/NCBI
  61. Qin T, Jelinek J, Si J, Shu J, Issa JP. Mechanisms of resistance to 5-aza-2′-deoxycytidine in human cancer cell lines. Blood 2009;113(3):659-667 View Article PubMed/NCBI
  62. Hosokawa M, Tanaka S, Ueda K, Iwakawa S, Ogawara KI. Decitabine exerted synergistic effects with oxaliplatin in colorectal cancer cells with intrinsic resistance to decitabine. Biochem Biophys Res Commun 2019;509(1):249-254 View Article PubMed/NCBI
  63. Lötsch J, Schneider G, Reker D, Parnham MJ, Schneider P, Geisslinger G, et al. Common non-epigenetic drugs as epigenetic modulators. Trends Mol Med 2013;19(12):742-753 View Article PubMed/NCBI
  64. Shih CC, Liao MH, Hsiao TS, Hii HP, Shen CH, Chen SJ, et al. Procainamide inhibits DNA methylation and alleviates multiple organ dysfunction in rats with endotoxic shock. PLoS One 2016;11(9):e0163690 View Article PubMed/NCBI
  65. Teoh-Fitzgerald ML, Fitzgerald MP, Jensen TJ, Futscher BW, Domann FE. Genetic and epigenetic inactivation of extracellular superoxide dismutase promotes an invasive phenotype in human lung cancer by disrupting ECM homeostasis. Mol Cancer Res 2012;10(1):40-51 View Article PubMed/NCBI
  66. Shih CC, Hii HP, Tsao CM, Chen SJ, Ka SM, Liao MH, et al. Therapeutic Effects of Procainamide on Endotoxin-Induced Rhabdomyolysis in Rats. PLoS One 2016;11(2):e0150319 View Article PubMed/NCBI
  67. Gao Z, Xu Z, Hung MS, Lin YC, Wang T, Gong M, et al. Procaine and procainamide inhibit the Wnt canonical pathway by promoter demethylation of WIF-1 in lung cancer cells. Oncol Rep 2009;22(6):1479-1484 View Article PubMed/NCBI
  68. Villar-Garea A, Fraga MF, Espada J, Esteller M. Procaine is a DNA-demethylating agent with growth-inhibitory effects in human cancer cells. Cancer Res 2003;63(16):4984-4989 View Article PubMed/NCBI
  69. Li YC, Wang Y, Li DD, Zhang Y, Zhao TC, Li CF. Procaine is a specific DNA methylation inhibitor with anti-tumor effect for human gastric cancer. J Cell Biochem 2018;119(2):2440-2449 View Article PubMed/NCBI
  70. Lin X, Asgari K, Putzi MJ, Gage WR, Yu X, Cornblatt BS, et al. Reversal of GSTP1 CpG island hypermethylation and reactivation of pi-class glutathione S-transferase (GSTP1) expression in human prostate cancer cells by treatment with procainamide. Cancer Res 2001;61(24):8611-8616 View Article PubMed/NCBI
  71. Li C, Gao S, Li X, Li C, Ma L. Procaine Inhibits the Proliferation and Migration of Colon Cancer Cells Through Inactivation of the ERK/MAPK/FAK Pathways by Regulation of RhoA. Oncol Res 2018;26(2):209-217 View Article PubMed/NCBI
  72. Champion C, Guianvarc’h D, Sénamaud-Beaufort C, Jurkowska RZ, Jeltsch A, Ponger L, et al. Mechanistic insights on the inhibition of c5 DNA methyltransferases by zebularine. PLoS One 2010;5(8):e12388 View Article PubMed/NCBI
  73. Napso T, Fares F. Zebularine induces prolonged apoptosis effects via the caspase-3/PARP pathway in head and neck cancer cells. Int J Oncol 2014;44(6):1971-1979 View Article PubMed/NCBI
  74. Yang PM, Lin YT, Shun CT, Lin SH, Wei TT, Chuang SH, et al. Zebularine inhibits tumorigenesis and stemness of colorectal cancer via p53-dependent endoplasmic reticulum stress. Sci Rep 2013;3:3219 View Article PubMed/NCBI
  75. Brueckner B, Garcia Boy R, Siedlecki P, Musch T, Kliem HC, Zielenkiewicz P, et al. Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases. Cancer Res 2005;65(14):6305-6311 View Article PubMed/NCBI
  76. Datta J, Ghoshal K, Denny WA, Gamage SA, Brooke DG, Phiasivongsa P, et al. A new class of quinoline-based DNA hypomethylating agents reactivates tumor suppressor genes by blocking DNA methyltransferase 1 activity and inducing its degradation. Cancer Res 2009;69(10):4277-4285 View Article PubMed/NCBI
  77. Fagan RL, Cryderman DE, Kopelovich L, Wallrath LL, Brenner C. Laccaic acid A is a direct, DNA-competitive inhibitor of DNA methyltransferase 1. J Biol Chem 2013;288(33):23858-23867 View Article PubMed/NCBI
  78. Bhatt L, Gupta R. Potent anticolorectal cancer activity of 5-fluorouracil and laccaic acid combination via modulation of epigenetic regulation. Annals Oncol 2019;30:28 View Article PubMed/NCBI
  79. Reu FJ, Mark W. Chawla-Sarkar FM, Beaulieu N, Douglas W. Leaman A. et al. Genes involved in sensitization of renal cancer cells to interferon-induced apoptosis after selective depletion of DNA methyltransferase-1 by antisense oligonucleotide (MG98). Cancer Res 2004;64(7):370 View Article PubMed/NCBI
  80. Plummer R, Vidal L, Griffin M, Lesley M, de Bono J, Coulthard S, et al. 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. Clin Cancer Res 2009;15(9):3177-3183 View Article PubMed/NCBI
  81. Giordano A, Tommonaro G. Curcumin and Cancer. Nutrients 2019;11(10):E2376 View Article PubMed/NCBI
  82. Ravindran J, Prasad S, Aggarwal BB. Curcumin and cancer cells: how many ways can curry kill tumor cells selectively?. AAPS J 2009;11(3):495-510 View Article PubMed/NCBI
  83. Liu Z, Xie Z, Jones W, Pavlovicz RE, Liu S, Yu J, et al. Curcumin is a potent DNA hypomethylation agent. Bioorg Med Chem Lett 2009;19(3):706-709 View Article PubMed/NCBI
  84. Herrero de la Parte B, Rodeño-Casado M, Iturrizaga Correcher S, Mar Medina C, García-Alonso I. Curcumin Reduces Colorectal Cancer Cell Proliferation and Migration and Slows In Vivo Growth of Liver Metastases in Rats. Biomedicines 2021;9(9):1183 View Article PubMed/NCBI
  85. Lee WJ, Shim JY, Zhu BT. Mechanisms for the inhibition of DNA methyltransferases by tea catechins and bioflavonoids. Mol Pharmacol 2005;68(4):1018-1030 View Article PubMed/NCBI
  86. Wang ST, Cui WQ, Pan D, Jiang M, Chang B, Sang LX. Tea polyphenols and their chemopreventive and therapeutic effects on colorectal cancer. World J Gastroenterol 2020;26(6):562-597 View Article PubMed/NCBI
  87. Kedhari Sundaram M, Hussain A, Haque S, Raina R, Afroze N. Quercetin modifies 5′CpG promoter methylation and reactivates various tumor suppressor genes by modulating epigenetic marks in human cervical cancer cells. J Cell Biochem 2019;120(10):18357-18369 View Article PubMed/NCBI
  88. Tezerji S, Fallah A, Talaei B. The effect of resveratrol and quercetin intervention on azoxymethane-induced colon cancer in Rats model. Clin Nutrition Open Sci 2022;45:91-102 View Article PubMed/NCBI
  89. Hayashi M, Unemoto T, Minami-Kakinuma S, Tanaka H, Omura S. The mode of action of nanaomycins D and A on a gram-negative marine bacterium Vibrio alginolyticus. J Antibiot (Tokyo) 1982;35(8):1078-1085 View Article PubMed/NCBI
  90. Izumi K, Aoki H, Kakita H, Takeshita S, Ueda H, Inoue Y, Aoyama M. The DNMT3B Inhibitor Nanaomycin A as a Neuroblastoma Therapeutic Agent. Current Cancer Drug Targets 2023;23(11):837-842 View Article PubMed/NCBI
  91. Jung G, Hernández-Illán E, Moreira L, Balaguer F, Goel A. Epigenetics of colorectal cancer: biomarker and therapeutic potential. Nat Rev Gastroenterol Hepatol 2020;17(2):111-130 View Article PubMed/NCBI
  92. Maugeri A, Barchitta M, Mazzone MG, Giuliano F, Basile G, Agodi A. Resveratrol Modulates SIRT1 and DNMT Functions and Restores LINE-1 Methylation Levels in ARPE-19 Cells under Oxidative Stress and Inflammation. Int J Mol Sci 2018;19(7):2118 View Article PubMed/NCBI
  93. Aldawsari FS, Aguayo-Ortiz R, Kapilashrami K, Yoo J, Luo M, Medina-Franco JL, et al. Resveratrol-salicylate derivatives as selective DNMT3 inhibitors and anticancer agents. J Enzyme Inhib Med Chem 2016;31(5):695-703 View Article PubMed/NCBI
  94. Suh Y, Afaq F, Johnson JJ, Mukhtar H. A plant flavonoid fisetin induces apoptosis in colon cancer cells by inhibition of COX2 and Wnt/EGFR/NF-kappaB-signaling pathways. Carcinogenesis 2009;30(2):300-307 View Article PubMed/NCBI
  95. Kim ME, Ha TK, Yoon JH, Lee JS. Myricetin induces cell death of human colon cancer cells via BAX/BCL2-dependent pathway. Anticancer Res 2014;34(2):701-706 View Article PubMed/NCBI
  96. Juárez-Mercado KE, Prieto-Martínez FD, Sánchez-Cruz N, Peña-Castillo A, Prada-Gracia D, Medina-Franco JL. Expanding the Structural Diversity of DNA Methyltransferase Inhibitors. Pharmaceuticals (Basel) 2020;14(1):17 View Article PubMed/NCBI
  97. Sanaei M, Kavoosi F. Effect of 5-Aza-2′-Deoxycytidine in Comparison to Valproic Acid and Trichostatin A on Histone Deacetylase 1, DNA Methyltransferase 1, and CIP/KIP Family (p21, p27, and p57) Genes Expression, Cell Growth Inhibition, and Apoptosis Induction in Colon Cancer SW480 Cell Line. Adv Biomed Res 2019;8:52 View Article PubMed/NCBI
  98. Lee Y, Kwon J, Jeong JH, Ryu JH, Kim KI. Kazinol C from Broussonetia kazinoki stimulates autophagy via endoplasmic reticulum stress-mediated signaling. Anim Cells Syst (Seoul) 2022;26(1):28-36 View Article PubMed/NCBI
  99. Zwergel C, Valente S, Mai A. DNA Methyltransferases Inhibitors from Natural Sources. Curr Top Med Chem 2016;16(7):680-696 View Article PubMed/NCBI
  100. Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, Brudno Y, et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009;324(5929):930-935 View Article PubMed/NCBI
  101. Chen LL, Lin HP, Zhou WJ, He CX, Zhang ZY, Cheng ZL, et al. SNIP1 Recruits TET2 to Regulate c-MYC Target Genes and Cellular DNA Damage Response. Cell Rep 2018;25(6):1485-1500.e4 View Article PubMed/NCBI
  102. Storebjerg TM, Strand SH, Høyer S, Lynnerup AS, Borre M, Ørntoft TF, et al. Dysregulation and prognostic potential of 5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) levels in prostate cancer. Clin Epigenetics 2018;10(1):105 View Article PubMed/NCBI
  103. Slyvka A, Mierzejewska K, Bochtler M. Nei-like 1 (NEIL1) excises 5-carboxylcytosine directly and stimulates TDG-mediated 5-formyl and 5-carboxylcytosine excision. Sci Rep 2017;7(1):9001 View Article PubMed/NCBI
  104. Guan Y, Tiwari AD, Phillips JG, Hasipek M, Grabowski DR, Pagliuca S, et al. A Therapeutic Strategy for Preferential Targeting of TET2 Mutant and TET-dioxygenase Deficient Cells in Myeloid Neoplasms. Blood Cancer Discov 2021;2(2):146-161 View Article PubMed/NCBI
  105. Chia N, Wang L, Rouden D. TET inhibitors as potential new cancer drugs - An enzyme that Converts 5-methylcytosine to 5-hydroxymethylcytosine. Int Drug Discovery 2012;1:21-23 View Article PubMed/NCBI
  106. Chua GNL, Wassarman KL, Sun H, Alp JA, Jarczyk EI, Kuzio NJ, et al. Cytosine-Based TET Enzyme Inhibitors. ACS Med Chem Lett 2019;10(2):180-185 View Article PubMed/NCBI
  107. Das AT, Zhou X, Metz SW, Vink MA, Berkhout B. Selecting the optimal Tet-On system for doxycycline-inducible gene expression in transiently transfected and stably transduced mammalian cells. Biotechnol J 2016;11:71-9 View Article PubMed/NCBI
  108. Singh AK, Zhao B, Liu X, Wang X, Li H, Qin H, et al. Selective targeting of TET catalytic domain promotes somatic cell reprogramming. Proc Natl Acad Sci USA 2020;117(7):3621-3626 View Article PubMed/NCBI
  109. Pothongsrisit S, Pongrakhananon V, Iksen. Targeting the PI3K/AKT/mTOR Signaling Pathway in Lung Cancer: An Update Regarding Potential Drugs and Natural Products. Molecules 2021;26(13):4100 View Article PubMed/NCBI