Next Article in Journal
Exploring Chalcone Derivatives as a Multifunctional Therapeutic Agent: Investigating Antioxidant Potential, Acetylcholinesterase Inhibition and Computational Insights
Previous Article in Journal
Current Therapies and Potential Strategies for Uveal Melanoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetic Alterations in Cancer: The Therapeutic Potential of Epigenetic Drugs in Cancer Therapy

Mouse Cancer Genetics Program, National Cancer Institute, NIH, Frederick, MD 21702, USA
Drugs Drug Candidates 2025, 4(2), 15; https://doi.org/10.3390/ddc4020015
Submission received: 2 March 2025 / Revised: 21 March 2025 / Accepted: 28 March 2025 / Published: 5 April 2025

Abstract

:
To date, numerous studies have emerged that indicate the possible role of epigenetic modulation in the development and progression of several diseases, including cancer. Epigenetic alterations participate in various steps of carcinogenesis. They play important regulatory roles in processes like cell division, proliferation, angiogenesis, and metastasis. Thus, epigenetic modifications such as DNA methylation, histone modifications, and non-coding RNAs serve as attractive and promising targets for cancer prevention and anti-cancer therapy. Epigenetic drugs or epi-drugs possess the ability to reverse many such epigenetic alterations and thus can help manage the clinical manifestations of cancer. Epigenetic drugs broadly target epigenetic modifications, including DNA methylation and histone post-translational modifications, to manifest their effects. Several naturally occurring as well as chemically synthesized compounds have been recognized as epigenetic drugs. Some of them are clinically approved, while many are in their preclinical and clinical trials. In this review, we aim to present a broad overview of the epigenetic modifications implicated in carcinogenesis. The review also compiles various epigenetic drugs that are approved for clinical practice, as well as those that are in the preclinical and clinical stages of investigation for anti-cancer therapy.

Graphical Abstract

1. Introduction

It has been well established that both genetic and epigenetic factors play a significant role in the development of disease. While genetic factors modify gene expression by making alterations in the DNA sequence, epigenetic factors contribute to phenotypic changes independent of genetic alterations. More recently, the term epigenetics has referred to the cellular events that control gene expression through structural modifications in chromatin. The correct regulation of such chromatin-based events is crucial for appropriate cell growth and development, and any de-regulation of epigenetic patterns leads to aberrant gene expression, resulting in cancer or other epigenetic diseases. Epigenetic processes are controlled by three distinct mechanisms, viz., DNA methylation, histone tail modifications and small noncoding RNAs or microRNAs [1]. These mechanisms function in a coordinated manner to fine-tune the chromatin structure and thus maintain precise control over gene expression and silencing. In particular, DNA methylation and histone tail modifications regulate the accessibility of transcription factors to the target genes via the modulation of heterochromatin to euchromatin transition and vice-versa [2,3].
Aberrant epigenetic modifications, along with genetic alterations, are key players in the development and progression of several diseases, including cancer [4]. Unlike irreversible genetic alterations or mutations, epigenetic modifications are more dynamic and thus can be reprogrammed to achieve a normal phenotype [5]. This unique and valuable feature makes scientists particularly interested in understanding the molecular features of epigenetic mechanisms. Better molecular insight into epigenetic factors would certainly be valuable in the development of drugs that fix the altered epigenetic regulation of gene expression. Moreover, targeting reversible epigenetic modifications is a more attractive and promising strategy compared to targeting stable genetic mutations, from a therapeutic standpoint [6].
Over the past decade, expanding research into chromatin biology has identified various new drug targets that largely belong to the chromatin regulatory factors. These include enzymes that covalently modify DNA and histones [6], chromatin remodeling complexes [7], and transcription activators or repressors [8]. Since these chromatin regulatory enzymes play crucial roles in epigenetic pathways of cancer and other human pathologies, targeting them with high-efficacy drugs is currently a research area with huge therapeutic relevance. In general, the term epigenetic drugs or epi-drugs is used to define a class of molecules that are used to reprogram the altered epigenome in a diseased state, leading to beneficial clinical manifestations. Currently, various epi-drugs are in clinical use, and many more are undergoing preclinical as well as clinical stages of drug development [9]. In this review, we discuss the current understanding of epigenetic alterations in carcinogenesis. A comprehensive list of the various epi-drugs that are currently in clinical practice for anti-cancer therapy is provided. The list also summarizes recent advances in epi-drugs at various stages of drug development in clinical settings.

2. Epigenetic Modifications

Epigenetics refers to the inherited genetic traits that arise due to alterations in gene expression rather than changes in the DNA sequence itself. An important regulatory mechanism for gene expression involves the temporal and spatial accessibility of DNA to various DNA-binding proteins, which in turn is regulated by the high-order chromatin structure, comprising DNA and histone proteins. The fundamental unit of chromatin is the nucleosome, which is made up of 147 base pairs of DNA wrapped around a histone octamer, comprising two units each of four canonical histones, i.e., H2A, H2B, H3, and H4. To facilitate a genome organization that fits into the nucleus, nucleosomes are arranged on DNA like “beads on a string”, which is further condensed to chromatin fibers and a high-order chromatin structure via inter-nucleosomal interactions (Figure 1) [10]. DNA accessibility is regulated by the degree of condensation of this high-order chromatin structure. Heterochromatin refers to the highly condensed chromatin state, which primarily comprises repetitive DNA sequences and silenced genes and is mostly inaccessible to DNA-binding machinery. In contrast, euchromatin represents de-condensed chromatin regions with actively transcribed genomic regions [11].
The fundamental epigenetic modifications that regulate chromatin accessibility include DNA methylation and post-translational modification of histone tails such as acetylation, methylation, citrullination, and phosphorylation [12]. Additionally, non-coding RNAs control gene expression at the level of protein translation by degrading mRNA or inhibiting its translation (Figure 1) [13]. The continuous interest in the growing field of epigenetics had led to the uncovering of the important role of epigenetic modifications in a multitude of diseases including cancer, neurodevelopmental disorders, neurological diseases, autoimmune diseases, endocrine disorders, and so on.

3. Epigenetic Modifications in Cancer

In cancers of various organs, genetic aberrations work in conjunction with epigenetic alterations—particularly of histone modifications—creating chaos in transcriptional control. Several biological processes are affected by epigenetic dysregulation, including cell proliferation, growth, differentiation, apoptosis, and DNA repair [4]. Unlike genetic perturbations, epigenetic mechanisms control transcription via changes in gene expression levels without compromising DNA sequence integrity [14]. Thus, it is wise to target dysregulated epigenetic functions rather than targeting stable and irreversible genetic mutations from a therapeutic standpoint. Altered histone modifications have been linked to the mis-regulated expression of multiple genes with crucial roles in tumorigenesis [15]. In fact, certain tumors exhibit specific altered histone modifications and DNA methylation patterns even at the individual cellular level. Such cell- and tumor-specific altered profiles of histone modifications act as markers for cancer prognosis as well as predicting the chances of relapse [16]. Moreover, DNA methylation is often observed to be dysregulated in various cancer types. Notably, different subtypes of breast cancer show different profiles of DNA methylation [17].
Because of the well-established link between epigenetic dysregulation and cancer development, various therapeutic strategies have been envisaged that target DNA methyltransferases (DNMT) and histone modifying enzymes, thereby reverting the pathogenic consequences of epigenetic alterations. Histone modifying enzymes are majorly categorized as histone methyltransferases (HMT), histone demethylases (HDM), histone acetyl transferases (HAT), and histone deacetylases (HDAC) [3]. The drugs that target DNA methyltransferase and various histone modifying enzymes are termed as epigenetic drugs or epi-drugs [18].

3.1. DNA Methylation

DNA methylation is a well-defined epigenetic marker that involves a family of enzymes known as DNA methyltransferases (DNMTs). DNMTs catalyze the transfer of methyl groups from S-adenosyl methionine (SAM) to the C-5 position of the pyrimidine ring of cytosine in CpG di-nucleotides in the genome [19]. Four different active isoforms of DNMTs have been identified in mammals, viz., DNMT1, TDRMT1 (or DNMT2), and DNMT3. DNMT1 and DNMT3 are localized in the nucleus, while TDRMT1 functions in the cytoplasm. DNMT1 is the most abundant form that maintains methylation patterns post-replication by predominantly methylating hemimethylated CpG di-nucleotides, whereas DNMT3 methylates both hemimethylated and unmethylated DNA with the same efficiency. Interestingly, TDRMT1 does not methylate DNA, it methylates aspartic acid transfer RNA (tRNAAsp), despite sharing strong structural similarity with classical DNMTs. DNA methylation also occurs at sites other than CpG sites [20]. DNA methylation in non-CpG sites is enriched at gene bodies and depleted at enhancers and protein binding sites [21].
DNA methylation is an important mechanism for gene silencing, especially of tumor suppressing genes and genes involved in DNA repair processes. It is often observed to be dysregulated in various cancers [22,23,24]. Aberrant DNA methylation, including both site-specific hypermethylation (e.g., CpG islands in gene regulatory elements) and genome-wide hypomethylation (e.g., repetitive sequences and intergenic regions), are common characteristics of cancer cells (Figure 2). Differential DNA methylation patterns may exist in different cancer types of the same tissue. For example, different breast cancer subtypes show different DNA methylation patterns. Aberrant hypermethylation is more prominently observed in estrogen receptor alpha (Erα)-positive breast cancer as compared to Erα-negative subtypes [25]. Abnormal DNA methylation in the promoters of cancer-linked genes often lead to their altered expression, as well as to the inactivation of tumor suppressor genes. One example is gastric cancer, where abnormal methylation occurs in genes associated with transcriptional regulation (like Helicase-like transcription factor), DNA repair (like MGMT and hMLH1), cell growth and differentiation (such as HoxD10), and apoptosis (like BNIP3) [26]. Notably, the hypermethylation of the promoter region of MutL homolog 1 (MLH1), a DNA mismatch repair gene, is observed in sporadic cases of colorectal cancers [27]. Likewise, the hypermethylation of promoters of epithelium-specific genes and the hypomethylation of epithelial to mesenchymal transition (EMT) markers are observed in prostate cancer, resulting in EMT and stemness [28].

3.2. Histone Modifications

The nucleosome, the fundamental unit of chromatin, is composed of 147 base pairs of DNA wrapped around a histone octamer. Each histone octamer has two each of the canonical histone proteins, viz., H2A, H2B, H3, and H4. The N-terminal tails of histones that protrude out from the nucleosome are accessible on its surface for various post-translational modifications (PTMs) such as methylation, acetylation, phosphorylation, ubiquitination, ADP ribosylation, and sumoylation. The enzymes that regulate various histone PTMs include histone methyl transferase (HMT), histone demethylase (HDM), histone acetyltransferase (HAT), histone deacetylase (HDAC), kinases, and E3-ubiquitin [16]. These histone modifications can affect the interaction of histones with DNA strands as well as the interaction of one nucleosome with another nucleosome. They also act as sites for the recruitment of transcription factors or other histone binding proteins such as PHD-, chromo-, and bromo-domain-containing proteins [3,29]. In this way, histone tail modifications can influence the chromatin state, and thus, gene expression. The chromatin organization or architecture depends upon histone configuration. Euchromatin, having a relaxed chromatin structure, facilitates the transcription process and usually contains high-expression genes. In contrast, heterochromatin has a denser configuration and is primarily associated with gene silencing. Notably, both euchromatin and heterochromatin are recognized by specific epigenetic markers. Euchromatin is rich in histone acetylation, whereas heterochromatin shows high methylation levels (Figure 3) [30].
Altered patterns of histone PTMs, both at specific gene loci and at the global genome level, have been implicated in various human cancers [31]. There is a proven role of altered histone modifications in the over-activation of oncogenes or suppression of tumor suppressors (Figure 3). Both transcriptionally active histone markers such as H3K4me3 and H3K36me3 as well as repressive markers including H3K9me2/3, H3K27me3, and H4K20me3 are involved in cancer progression [29]. MLL2, a H3K4 methyltransferase, is involved in breast cancer development; it interacts with estrogen receptor alpha, which regulates key genes involved in breast cancer proliferation [32]. One study also suggests that a H4K4 methyltransferase (MLL4) and a H3K27 demethylase (UTX) coordinate with each other to regulate the transcriptional programs for cell invasiveness in breast cancer MDA-MD-231 cells [33]. In the same line, it has been reported that the inactivation of KMT2C/MLL3, another H3K4 methyltransferase, promotes the development of colorectal cancer through transcriptional dysregulation in various pathways with established cancer relevance [34]. Deregulation of HMTs or HDMs is known to be associated with prostate cancer development and progression. SUV39H2 and SETDB1, which catalyze H3K9me1/2 and H3K9me3, respectively, are upregulated in prostate cancer [35,36]. Similarly, SMYD3, a H3K4 methyltransferase, is overexpressed in prostate, colorectal, hepatocellular, and breast cancer [37,38,39]. Reduced levels of H4K20me3 are found to be associated with HER-2-independent breast cancer. In fact, cancer cell invasiveness is overcome by the ectopic expression of the H3K20 methyltransferases SUV420H1 and SUV420H2 [40].
Functional dysregulation of histone acetyltransferases (HATs), just like HMTs and HDMs, is strongly correlated with carcinogenesis [41]. Mutations in HATs have been reported in various cancers, with involvement at various stages of cancer progression [42,43,44]. Mutations in CREBBP and EP300, two histone acetyltransferases (HATs), are linked to many lymphoid malignancies including acute lymphoblastic leukemia, B-cell lymphoma, and follicular lymphoma [43,45,46]. It has also been observed that the interaction of CBP/p300 with c-Myb activates its transforming potential and induces acute myeloid leukemia [47]. HAT1 is upregulated in pancreatic ductal adenocarcinoma (PDAC) and is associated with poor prognosis. Interestingly, the knockdown of HAT1 does in fact decrease cancer cell proliferation in PDAC both in vitro and in vivo [48]. A large body of growing evidence suggests that overexpression of HDAC occurs in cancers of various organs such as lung cancer, breast cancer, colon cancer, gastrointestinal cancer, neuroblastoma, and many more [49,50,51,52,53]. HDACs regulate various aspects of tumorigenesis ranging from cell growth/differentiation, autophagy, apoptosis, and angiogenesis to prognosis and chemo-resistance [54,55,56].
Other common post-translational modifications of histone proteins include ubiquitination and SUMOylation. Histone ubiquitination involves the addition of ubiquitin protein molecules onto target histone tails. Histones are frequently modified with one ubiquitin molecule (mono-ubiquitination), but can also acquire ubiquitin chains (polyubiquitination). The addition of ubiquitin to the histones is a reversible modification carried out by E3 ubiquitin ligases and ubiquitin-specific peptidases (deubiquitinating enzymes (DUBs)). It plays important roles in DNA replication, gene transcription, and DNA damage repair. It also affects the chromatin state as well as regulating the recruitment of repair proteins at DSB sites [57]. Moreover, histone ubiquitination regulates other histone PTMs including methylation and acetylation to preserve genome integrity. Dysregulation of histone ubiquitination is often observed in cancer and can promote cancer development by altering the expression of oncogenes and tumor suppressors. One example is the elevated expression of H2AK119ub1 in pancreatic ductal adenocarcinoma as compared to normal tissues. Interestingly, the tumors show reduced levels of H3K27Me3, indicating the importance of the intricate cross-talk between histone ubiquitination and methylation in carcinogenesis [58]. In gastric cancer, triple-negative breast cancer, and colorectal cancer, a loss of H2BK120ub has been observed [59,60,61]. Notably, the genes encoding E3 ubiquitin ligases are frequently mutated in cancer. E3 ligases regulate major growth-promoting pathways such as the MAPK or PI3K–AKT–mTOR pathways. Thus, dysregulated E3 ligases drive uncontrolled proliferation and genomic instability, which promote malignant transformation and tumor progression [62].
Another histone modification, i.e., SUMOylation, refers to the reversible addition of a small ubiquitin-like modifier (SUMO) moiety to a lysine residue on the histone protein. It is carried out by three principle enzymes over three main steps: activation via SUMO E1, conjugation via SUMO E2, and ligation via SUMO E3 [63]. SUMO-specific proteases (SENPs) can reverse this modification. There are seven SENPs identified in humans, of which, SENP5 is observed to be upregulated in oral cancer compared to normal epithelial cells [64]. Furthermore, Katayama et al. has shown that SUMO-1 is highly overexpressed in human oral squamous carcinoma and contributes to tumor cell proliferation [65]. In another study, Shiio et al. demonstrated that SUMOylation promotes transcriptional repression via recruitment of heterochromatin protein 1 and histone deacetylases [66]. SUMOylation regulates the functional activity of many of the transcription factors governing transitions between epithelial and mesenchymal states, which points towards the important role of this modification in tumor growth and metastasis [67,68]. Given the important role of histone modifying enzymes in cancer development and progression, they serve as key targets of epigenetic drugs.

3.3. Non-Coding RNAs

Recently, several studies have emerged that suggest that processes other than the deregulation of DNA methylation and histone modifications contribute to tumorigenesis and its progression. One such well-defined process is the altered expression of non-coding RNAs (ncRNAs). They are divided into two main classes based on the length of the nucleotides, viz., short ncRNAs (<200 nucleotides) and long ncRNAs (>200 nucleotides). Short ncRNAs include microRNA (miRNA), germline-restricted PIWI-interacting RNA (pi-RNA), and small-interfering RNA (siRNA). Of all these, miRNAs are the best studied and serve as key players in epigenetics, playing important roles in cell proliferation and differentiation [69].

3.3.1. MicroRNAs

MicroRNAs are ~22 nucleotides long and complementary to messenger RNA, which is obtained via the processing of short RNA hairpins. miRNA does not translate into proteins; instead, it downregulates or inhibits the translation of mRNA that bears complementary sequences [70]. About 60% of protein coding genes in the human genome are targeted by miRNAs [71]. miRNA targets the 3′-UTR region of the target mRNA and inhibits its translation via mRNA cleavage and degradation.
Various recent studies have suggested that miRNAs play a key role in various regulatory processes such as cell division, proliferation, and apoptosis. Abnormal expression of miRNAs is observed in several human diseases such as cardiovascular diseases, various inflammatory diseases, autoimmune diseases, rare genetic disorders, and cancers of various organs [72,73,74]. OncomiRs are miRNAs that are associated with carcinogenesis and metastasis. In normal cells, some oncomiR genes function as oncogenes in the sense that they lead to the development of cancer upon overexpression. One example is miR-155, which is upregulated in breast cancer [75]. Strikingly, some other oncomiR genes function as tumor suppressors, and their downregulation promotes cancerous growth (Figure 4). One example is the downregulation of miR-15a and miR-16-1, which negatively regulate the anti-apoptotic gene BCL2 in chronic lymphocytic leukemia patients [76]. Also, miRNAs show differential expression patterns in cancerous tissues as well as in the blood of cancer patients [77]. Some of the miRNAs that are overexpressed and contribute to tumor growth include miR-10b, miR-16, miR-21, miR-34a, miR-155, and miR-221/222 [75,78,79,80,81,82]. miRNAs can also indicate the presence and clinical stage of the tumor. Hence, they serve as key diagnostic biomarkers for cancer and offer new paths for the development of cancer therapies.

3.3.2. Long Non-Coding RNA

Another type of ncRNA, i.e., long non-coding RNA (lncRNA), is a very heterogeneous class and includes RNAs with both housekeeping and gene regulatory functions. Just like miRNAs, lncRNAs interact with the DNA via sequence complementarity and form non-canonical, triple-stranded structures such as RNA:DNA triplexes, R-loops, and G-quadruplexes (Figure 5) [84]. RNA:DNA triplexes bind directly to the major groove of the DNA double helix via Hoogsteen hydrogen bonding following base-pairing like Watson–Crick interactions. Thus, RNA:DNA triplex-forming lncRNAs act as sequence-specific labels that target their protein partners to specific genomic locations [85]. In fact, various studies have suggested that lncRNAs form stable ‘domains’ for protein binding and chromatin localization, which enables the sequence-specific localization of chromatin modifiers or transcription factors. This in turn alters the epigenetic signatures and thus regulates gene expression at distinct genomic sites [86]. Another non-canonical three-stranded nucleic acid structures are R-loops, which consist of a DNA–RNA hybrid and a displaced single-stranded DNA [84]. They are formed during transcription when the newly synthesized RNA transcript remains bound to its DNA template [87]. These DNA:RNA hybrids nucleate from G-rich clusters in the DNA template and extend across GC-rich spans of transcribed genes during elongation. R-loops are very stable structures and thus need to be resolved enzymatically to restore the native double helix DNA. A balance between the constant formation and removal of R-loops creates a homeostasis at nucleation sites throughout the genome. An alteration in R-loop homeostasis leads to their accumulation in cells. This promotes genomic instability significantly, which is often associated with neurodegenerative disorders and cancer (Figure 5) [88].
LncRNAs affect various cellular properties such as differentiation, proliferation, cell cycle, apoptosis, invasion, and migration, suggesting that they play an important regulatory role in cancer development [89]. For example, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is one of the best-characterized druggable lncRNAs found to be dysregulated in multiple human malignancies [90]. The interaction of MALAT1 with zeste homolog 2 (EZH2) downregulates E-cadherin expression and promotes cell metastasis in osteosarcoma [91]. Likewise, MALAT1 plays an important role in the development of malignant esophageal squamous cell carcinoma (ESCC) via the targeting of β-catenin and Lin28 via Ezh2 [92]. The upregulation of MALAT1 is also observed in various hematological malignancies, including multiple myeloma and lymphoma, and is often associated with disease progression and reduced overall survival of patients [90]. Lung cancer-related transcript 1 (LUCAT1) is another lncRNA involved in various malignant tumor types such as liver cancer, esophageal cancer, lung cancer, thyroid cancer, ovarian cancer, breast cancer, gastric cancer, and renal cell carcinoma. LUCAT1 is overexpressed in esophageal squamous cell carcinoma (ESCC), which downregulates the tumor suppressor genes GADD45G and SFRP2 by stabilizing DNMT1, thereby stimulating the progression of ESCC [93]. Recently, a lncRNA growth arrest-specific 5 (GAS5) has gained attention as it functions as a tumor suppressor with significantly low expression levels in several malignancies, including breast cancer, gastric cancer, cervical cancer, non-small cell lung cancer (NSCLC), ovarian cancer, renal cancer, colorectal cancer, and so on [94]. GAS5 regulates multiple cellular functions such as cell proliferation, migration, EMT, apoptosis, and migration through multiple mechanisms. These include transcriptional regulation of GAS5 through microRNA sponges, binding to target mRNAs, and riborepression of steroid (glucocorticoid) hormones, thereby regulating the expression of both coding and non-coding genes at the transcriptional and translational levels [95].
R-loops have been found to be associated with various malignancies. They strikingly affect cancer progression and therapeutic efficiency by regulating crucial genes such as BRCA1/2, TOP1, PARP, DHX9, and sex hormone receptors [96]. The progressing R-loop collides with the replication fork, leading to fork stalling or reversal, leading to DNA single or double- stranded breaks and threatening genomic stability [97]. BRCA1 binds to R-loops that are formed over a subset of termination regions (TRs) of actively transcribed genes. It then mediates the recruitment of the R-loop resolving protein Senataxin (SETX). Various insertion/deletion mutations of these genes around TRs are observed in BRCA1-mutated breast tumors. This suggests that BRCA1/SETX functions as a DNA repair mechanism that resolves R-loop-mediated DNA breaks at transcription termination/pause sites, thereby halting tumorigenesis [95,98].
RNA G-quadruplexes (rG4s) are another structural motif in lncRNAs that have gained immense attention recently due to their varying roles in cell behavior and disease pathogenesis. The four guanines in G-rich RNA connect via Hoogsteen hydrogen bonds to form a square planar G-quartet structure. Several G-quartets stack on top of one another to create non-canonical structures called G-quadruplexes (G4s) [99,100]. In the nucleus, rG4s play an important role in the splicing of mRNA and ncRNAs, as well as in transcriptional regulation. Sequential G-tracts within the R-loops can also promote the formation of rG4s and inhibit transcription [101]. rG4s regulate mRNA translation, stability, and RNA biogenesis in the cytoplasm. The formation of rG4 in the 3′-UTR prevents the miRNA binding, thereby regulating RNA interference and promoting RNA stability [102]. In the genome, G4 structures are enriched at transcription factor binding sites, promoters, and telomeres, and play regulatory roles in protein expression, genome stability, and telomerase activity. In around 90% of all cancer cells, telomerase activity is upregulated, which enables the cell to replicate without the shortening of telomeres [103]. The formation of G4 structures at telomeres prevents telomerase binding and blocks its activity [104]. This strategy can be applied in the therapeutic intervention of cancer, wherein G4 formation blocks the telomerase activity in tumor cells at the telomeric region, thereby preventing uncontrolled DNA replication. Since somatic cells lack telomerase, they do not get affected. G4s are also described to be formed in the promoter regions of several oncogenes such as c-Myc and K-Ras, thereby playing a potential role in regulating gene expression and contributing to cancer development. One therapeutic approach is to stabilize G4s using ligands or oligonucleotide aptamers. The stabilization of these structures hinders the replication and transcription processes, as polymerases face steric clashes while moving through these stabilized structures [105,106]. Several chemical agents are known to stabilize G4 structures in the telomeres and promoters of oncogenes, and thus, they can be used as promising therapeutics; the list includes pyridostatin, TMPyP4, MM41 (naphthalenediimide derivative), and GQC-05 (ellipticine analog). These molecules bind to G4 structures through π-π stacking interactions or ionic coordination and stabilize their unique topology [107].
G4 ligands that tend to stabilize G4 structures can be used as potential anti-cancer drugs; however, no G4 binder has yet been approved in clinical trials [108]. G4 ligands bind to G4 motifs and affect cancer cells in multiple ways: These include (1) inhibition of telomerase function; (2) regulation of oncogene expression via stabilization of G4 motifs at their promoters; (3) gene regulation via targeting of the mRNA 5′-untranslated region; or (4) regulation by blocking the helicase unwinding function [108,109,110,111].
There are three fundamental strategies to targeting lncRNA for therapeutic intervention: These include the use of (a) antisense oligonucleotides (ASOs) that bind to lncRNAs, thereby inducing their degradation by RNaseH1; (b) small interfering RNAs (siRNAs) (20–25 nucleotides long) that bind target lncRNAs based on sequence complementarity and promote its degradation via Argonaute endonucleases; and (c) small molecules that are targeted to a specific lncRNA and inhibit it binding to its molecular partner or interfere with correct structure formation [112].

4. Epigenetic Drugs

Unlike mutations in DNA, epigenetic modifications are reversible. They also play important regulatory roles in processes like cell division, proliferation, angiogenesis, and metastasis. Altogether, this makes these epigenetic modifications, i.e., DNA methylation, histone modifications, and miRNA, attractive and promising targets for cancer prevention and therapy (Figure 6). Several naturally occurring as well as chemically synthesized compounds have been recognized as epigenetic modifiers or epi-drugs. They target enzymes that catalyze various epigenetic modifications such as DNMT, HDAC, HDM, HAT, HMT, Ubiquitin ligases, and non-coding RNAs (Figure 6). Mutations in these enzymes result in the altered expression of important regulatory genes involved in carcinogenesis, including the re-expression of tumor-suppressor genes. Various emerging studies have shown that epigenetic alterations regulate the tumor microenvironment, which can influence the response to chemo/immunotherapy. Table 1, Table 2 and Table 3 lists various epi-drugs as cancer therapeutics with their clinical status. Some of them are clinically approved, while many are in preclinical and clinical trials. Table 4 shows a list of lncRNAs targeted via these three strategies, viz., ASOs, siRNAs, or small molecules. Interestingly, it has been observed that epigenetic drugs that are either approved or undergoing clinical trials modulate the tumor immune microenvironment, thereby inducing a strong antitumor immune response [113,114,115,116,117].
Many therapies with epi-drugs have demonstrated encouraging results against hematological malignancies, but have also showed limited efficacy and poor clinical response against solid tumors [228]. This is partly attributed to the fact that solid tumors tend to originate from more differentiated or even terminally differentiated cells with a reduced capacity for epigenetic reprogramming [229]. Additionally, the first-generation epi-drugs were developed using a ‘one size fits all’ approach without considering biomarkers for patient selection. This probably hindered the development of first-generation and second-generation epi-drugs, which are almost exclusively DNMT or HDAC inhibitors. However, some success has now been achieved with third-generation epi-drugs, which have been developed following a precision-medicine approach [229].
Another major drawback with epi-drugs is their non-selectivity, which leads to severe toxic side effects [230]. Many of the first-generation DNMT and HDAC inhibitors are not biomarker-driven and target broadly all their isoforms. This lack of specificity causes severe toxicity in subjects/patients. Consequently, many clinical trials that have aimed to evaluate the efficacy of epi-drugs on solid tumors have either failed or terminated early. Early attempts have also been made to develop isoform-specific inhibitor molecules, but these were also largely disappointing, particularly under monotherapy settings [228]. This further necessitates the rigorous work needed to be done in the development of more selective inhibitors that specifically target single isoforms or proteins in a family. The therapeutic impact of the drug can be immensely enhanced by focusing on selective inhibition with fewer unwanted interactions.
Epi-drugs have also been tested in combination with other anticancer agents (chemotherapy, immunotherapy, radiotherapy, targeted therapy), but this has also not improved clinical outcomes significantly. Advancements in the field of precision medicine might unlock the potential of epi-drugs by mitigating their non-selective or off-target side effects [229]. The development of sophisticated drug delivery systems based on lipid-based nanoparticles or targeted delivery vehicles could improve the specificity and efficacy of epi-drug-based therapies by directing them to precise intracellular sites. Lipid nanoparticles elicit a range of benefits, including biocompatibility, biodegradability, and entrapment effectiveness. The use of liposomes, dendrimers, and small-molecule drug conjugates can also be explored as delivery systems to unleash their full potential for targeted drug delivery. The critical investigation of lower doses, sequential scheduling, and targeted delivery may certainly improve the therapeutic potential of epi-drugs.

5. Summary and Future Perspectives

Cancer is a complex disease that involves several factors. Various genetic alterations like gene fusion, deletion, duplication, and mutations in tumor suppressors and oncogenes are closely related to cancer development. Additionally, epigenetic mechanisms, as discussed in the review, also play an important role in carcinogenesis and metastasis. Genetic aberrations are irreversible and are difficult to manage or treat. Given that targeting dynamically reversible epigenetic alterations appears to be a more attractive and promising strategy to improve cancer therapy, some epigenetic drugs have been approved by the FDA and are currently in clinical use for the treatment of various hematological malignancies. Furthermore, many naturally occurring as well as chemically synthesized molecules are under investigation in preclinical and clinical studies. Although epi-drugs have shown better clinical outcomes in combination with other anticancer therapies including chemotherapy, immunotherapy, and radiotherapy, several issues must be addressed. Advancements in the field of precision medicine might unlock the potential of epi-drugs by mitigating their non-selective or off-target side effects. Improvements in drug engineering, drug targeting, delivery methods, and the designing of clinical trials to evaluate the safety and efficacy of epi-drugs would certainly be fruitful in the development of anticancer therapies for different cancer types and patients in need.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Szyf, M. Epigenetics, DNA methylation, and chromatin modifying drugs. Annu. Rev. Pharmacol. Toxicol. 2009, 49, 243–263. [Google Scholar] [CrossRef]
  2. Grewal, S.I.S. The molecular basis of heterochromatin assembly and epigenetic inheritance. Mol. Cell 2023, 83, 1767–1785. [Google Scholar] [CrossRef] [PubMed]
  3. Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef] [PubMed]
  4. Darwiche, N. Epigenetic mechanisms and the hallmarks of cancer: An intimate affair. Am. J. Cancer Res. 2020, 10, 1954–1978. [Google Scholar]
  5. Sharma, S.; Kelly, T.K.; Jones, P.A. Epigenetics in cancer. Carcinogenesis 2010, 31, 27–36. [Google Scholar] [CrossRef] [PubMed]
  6. Mohammad, H.P.; Barbash, O.; Creasy, C.L. Targeting epigenetic modifications in cancer therapy: Erasing the roadmap to cancer. Nat. Med. 2019, 25, 403–418. [Google Scholar] [CrossRef]
  7. Zhang, F.L.; Li, D.Q. Targeting Chromatin-Remodeling Factors in Cancer Cells: Promising Molecules in Cancer Therapy. Int. J. Mol. Sci. 2022, 23, 2815. [Google Scholar] [CrossRef]
  8. Bushweller, J.H. Targeting transcription factors in cancer—From undruggable to reality. Nat. Rev. Cancer 2019, 19, 611–624. [Google Scholar] [CrossRef]
  9. Miranda Furtado, C.L.; Dos Santos Luciano, M.C.; Silva Santos, R.D.; Furtado, G.P.; Moraes, M.O.; Pessoa, C. Epidrugs: Targeting epigenetic marks in cancer treatment. Epigenetics 2019, 14, 1164–1176. [Google Scholar] [CrossRef]
  10. Li, G.; Reinberg, D. Chromatin higher-order structures and gene regulation. Curr. Opin. Genet. Dev. 2011, 21, 175–186. [Google Scholar] [CrossRef]
  11. Klemm, S.L.; Shipony, Z.; Greenleaf, W.J. Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 2019, 20, 207–220. [Google Scholar] [CrossRef] [PubMed]
  12. Gagnidze, K.; Pfaff, D.W. Epigenetic Mechanisms: DNA Methylation and Histone Protein Modification. In Neuroscience in the 21st Century: From Basic to Clinical; Pfaff, D.W., Volkow, N.D., Rubenstein, J.L., Eds.; Springer International Publishing: Cham, Switzerland, 2022; pp. 2677–2716. [Google Scholar]
  13. Kaikkonen, M.U.; Lam, M.T.; Glass, C.K. Non-coding RNAs as regulators of gene expression and epigenetics. Cardiovasc. Res. 2011, 90, 430–440. [Google Scholar] [CrossRef]
  14. Al Aboud, N.M.; Tupper, C.; Jialal, I. Genetics, Epigenetic Mechanism. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2025. [Google Scholar]
  15. Farooq, Z.; Shah, A.; Tauseef, M.; Rather, R.A.; Anwar, M. Evolution of Epigenome as the Blueprint for Carcinogenesis. In Epigenetics to Optogenetics-A New Paradigm in the Study of Biology; IntechOpen: Rijeka, Croatia, 2021. [Google Scholar]
  16. Neganova, M.E.; Klochkov, S.G.; Aleksandrova, Y.R.; Aliev, G. Histone modifications in epigenetic regulation of cancer: Perspectives and achieved progress. Semin. Cancer Biol. 2022, 83, 452–471. [Google Scholar] [CrossRef] [PubMed]
  17. Szyf, M. DNA methylation signatures for breast cancer classification and prognosis. Genome Med. 2012, 4, 26. [Google Scholar] [CrossRef]
  18. Dai, W.; Qiao, X.; Fang, Y.; Guo, R.; Bai, P.; Liu, S.; Li, T.; Jiang, Y.; Wei, S.; Na, Z.; et al. Epigenetics-targeted drugs: Current paradigms and future challenges. Signal Transduct. Target. Ther. 2024, 9, 332. [Google Scholar] [CrossRef] [PubMed]
  19. Wang, S.; Wu, W. Chapter 5—DNA Methylation Alterations in Human Cancers. In Epigenetics in Human Disease, 2nd ed.; Tollefsbol, T.O., Ed.; Academic Press: Cambridge, MA, USA, 2018; Volume 6, pp. 109–139. [Google Scholar]
  20. Ramsahoye, B.H.; Biniszkiewicz, D.; Lyko, F.; Clark, V.; Bird, A.P.; Jaenisch, R. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc. Natl. Acad. Sci. USA 2000, 97, 5237–5242. [Google Scholar] [CrossRef]
  21. Lister, R.; Pelizzola, M.; Dowen, R.H.; Hawkins, R.D.; Hon, G.; Tonti-Filippini, J.; Nery, J.R.; Lee, L.; Ye, Z.; Ngo, Q.M.; et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009, 462, 315–322. [Google Scholar] [CrossRef]
  22. Batra, R.N.; Lifshitz, A.; Vidakovic, A.T.; Chin, S.-F.; Sati-Batra, A.; Sammut, S.-J.; Provenzano, E.; Ali, H.R.; Dariush, A.; Bruna, A.; et al. DNA methylation landscapes of 1538 breast cancers reveal a replication-linked clock, epigenomic instability and cis-regulation. Nat. Commun. 2021, 12, 5406. [Google Scholar] [CrossRef]
  23. Xiao, M.; Liang, X.; Yan, Z.; Chen, J.; Zhu, Y.; Xie, Y.; Li, Y.; Li, X.; Gao, Q.; Feng, F.; et al. A DNA-Methylation-Driven Genes Based Prognostic Signature Reveals Immune Microenvironment in Pancreatic Cancer. Front. Immunol. 2022, 13, 803962. [Google Scholar]
  24. Hoang PH, L.M. DNA Methylation in Lung Cancer: Mechanisms and Associations with Histological Subtypes, Molecular Alterations, and Major Epidemiological Factors. Cancers 2022, 14, 961. [Google Scholar] [CrossRef]
  25. Shi, J.-F.; Li, X.-J.; Si, X.-X.; Li, A.-D.; Ding, H.-J.; Han, X.; Sun, Y.-J. ERα positively regulated DNMT1 expression by binding to the gene promoter region in human breast cancer MCF-7 cells. Biochem. Biophys. Res. Commun. 2012, 427, 47–53. [Google Scholar] [PubMed]
  26. Qu, Y.; Dang, S.; Hou, P. Gene methylation in gastric cancer. Clin. Chim. Acta 2013, 424, 53–65. [Google Scholar] [PubMed]
  27. Kientz, C.; Prieur, F.; Clemenson, A.; Joly, M.-O.; Stachowicz, M.-L.; Auclair, J.; Attignon, V.; Schiappa, R.; Wang, Q. MLH1 promoter hypermethylation: Are you absolutely sure about the absence of MLH1 germline mutation? About a new case. Fam. Cancer 2020, 19, 11–14. [Google Scholar] [CrossRef]
  28. Pistore, C.; Giannoni, E.; Colangelo, T.; Rizzo, F.; Magnani, E.; Muccillo, L.; Giurato, G.; Mancini, M.; Rizzo, S.; Riccardi, M.; et al. DNA methylation variations are required for epithelial-to-mesenchymal transition induced by cancer-associated fibroblasts in prostate cancer cells. Oncogene 2017, 36, 5551–5566. [Google Scholar] [CrossRef]
  29. Zhao, Z.; Shilatifard, A. Epigenetic modifications of histones in cancer. Genome Biol. 2019, 20, 245. [Google Scholar] [CrossRef]
  30. Morrison, O.; Thakur, J. Molecular Complexes at Euchromatin, Heterochromatin and Centromeric Chromatin. Int. J. Mol. Sci. 2021, 22, 6922. [Google Scholar] [CrossRef]
  31. Audia, J.E.; Campbell, R.M. Histone modifications and cancer. Cold Spring Harb. Perspect. Biol. 2016, 8, a019521. [Google Scholar] [CrossRef] [PubMed]
  32. Mo, R.; Rao, S.M.; Zhu, Y.-J. Identification of the MLL2 complex as a coactivator for estrogen receptor α. J. Biol. Chem. 2006, 281, 15714–15720. [Google Scholar] [CrossRef]
  33. Kim, J.-H.; Sharma, A.; Dhar, S.S.; Lee, S.-H.; Gu, B.; Chan, C.-H.; Lin, H.-K.; Lee, M.G. UTX and MLL4 coordinately regulate transcriptional programs for cell proliferation and invasiveness in breast cancer cells. Cancer Res. 2014, 74, 1705–1717. [Google Scholar]
  34. Larsson, C.; Cordeddu, L.; Siggens, L.; Pandzic, T.; Kundu, S.; He, L.; Ali, M.A.; Pristovšek, N.; Hartman, K.; Ekwall, K. Restoration of KMT2C/MLL3 in human colorectal cancer cells reinforces genome-wide H3K4me1 profiles and influences cell growth and gene expression. Clin. Epigenetics 2020, 12, 74. [Google Scholar] [CrossRef]
  35. Xu, C.; Zhao, S.; Cai, L. Epigenetic (De)regulation in Prostate Cancer. In Epigenetics in Oncology; Chen, J., Wang, G.G., Lu, J., Eds.; Springer International Publishing: Cham, Switzerland, 2023; pp. 321–360. [Google Scholar]
  36. Strepkos, D.; Markouli, M.; Klonou, A.; Papavassiliou, A.G.; Piperi, C. Histone methyltransferase SETDB1: A common denominator of tumorigenesis with therapeutic potential. Cancer Res. 2021, 81, 525–534. [Google Scholar]
  37. Vieira, F.Q.; Costa-Pinheiro, P.; Ramalho-Carvalho, J.; Pereira, A.; Menezes, F.D.; Antunes, L.; Carneiro, I.; Oliveira, J.; Henrique, R.; Jeronimo, C. Deregulated expression of selected histone methylases and demethylases in prostate carcinoma. Endocr.-Relat. Cancer 2014, 21, 51–61. [Google Scholar]
  38. Sanese, P.; Fasano, C.; Lepore Signorile, M.; De Marco, K.; Forte, G.; Disciglio, V.; Grossi, V.; Simone, C. Methyltransferases in cancer drug resistance: Unlocking the potential of targeting SMYD3 to sensitize cancer cells. Biochim. Et Biophys. Acta (BBA)-Rev. Cancer 2024, 1879, 189203. [Google Scholar] [CrossRef]
  39. Sun, J.; Li, Y.; Shi, M.; Tian, H.; Li, J.; Zhu, K.; Guo, Y.; Mu, Y.; Geng, J.; Li, Z. A Positive Feedback Loop of lncRNA HOXD-AS2 and SMYD3 Facilitates Hepatocellular Carcinoma Progression via the MEK/ERK Pathway. J. Hepatocell. Carcinoma 2023, 10, 1237–1256. [Google Scholar] [CrossRef]
  40. Yokoyama, Y.; Matsumoto, A.; Hieda, M.; Shinchi, Y.; Ogihara, E.; Hamada, M.; Nishioka, Y.; Kimura, H.; Yoshidome, K.; Tsujimoto, M.; et al. Loss of histone H4K20 trimethylation predicts poor prognosis in breast cancer and is associated with invasive activity. Breast Cancer Res. 2014, 16, R66. [Google Scholar] [CrossRef] [PubMed]
  41. Di Martile, M.; Del Bufalo, D.; Trisciuoglio, D. The multifaceted role of lysine acetylation in cancer: Prognostic biomarker and therapeutic target. Oncotarget 2016, 7, 55789. [Google Scholar] [PubMed]
  42. Iyer, A.; Fairlie, D.P.; Brown, L. Lysine acetylation in obesity, diabetes and metabolic disease. Immunol. Cell Biol. 2012, 90, 39–46. [Google Scholar]
  43. Pasqualucci, L.; Dominguez-Sola, D.; Chiarenza, A.; Fabbri, G.; Grunn, A.; Trifonov, V.; Kasper, L.H.; Lerach, S.; Tang, H.; Ma, J. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 2011, 471, 189–195. [Google Scholar]
  44. Trisciuoglio, D.; Di Martile, M.; Del Bufalo, D. Emerging Role of Histone Acetyltransferase in Stem Cells and Cancer. Stem Cells Int. 2018, 2018, 8908751. [Google Scholar] [CrossRef]
  45. Pastore, A.; Jurinovic, V.; Kridel, R.; Hoster, E.; Staiger, A.M.; Szczepanowski, M.; Pott, C.; Kopp, N.; Murakami, M.; Horn, H. Integration of gene mutations in risk prognostication for patients receiving first-line immunochemotherapy for follicular lymphoma: A retrospective analysis of a prospective clinical trial and validation in a population-based registry. Lancet Oncol. 2015, 16, 1111–1122. [Google Scholar]
  46. Qian, M.; Zhang, H.; Kham, S.K.-Y.; Liu, S.; Jiang, C.; Zhao, X.; Lu, Y.; Goodings, C.; Lin, T.-N.; Zhang, R. Whole-transcriptome sequencing identifies a distinct subtype of acute lymphoblastic leukemia with predominant genomic abnormalities of EP300 and CREBBP. Genome Res. 2017, 27, 185–195. [Google Scholar] [PubMed]
  47. Sun, X.-J.; Man, N.; Tan, Y.; Nimer, S.D.; Wang, L. The role of histone acetyltransferases in normal and malignant hematopoiesis. Front. Oncol. 2015, 5, 108. [Google Scholar] [CrossRef] [PubMed]
  48. Fan, P.; Zhao, J.; Meng, Z.; Wu, H.; Wang, B.; Wu, H.; Jin, X. Overexpressed histone acetyltransferase 1 regulates cancer immunity by increasing programmed death-ligand 1 expression in pancreatic cancer. J. Exp. Clin. Cancer Res. 2019, 38, 47. [Google Scholar]
  49. Wang, W.; Zhao, M.; Cui, L.; Ren, Y.; Zhang, J.; Chen, J.; Jia, L.; Zhang, J.; Yang, J.; Chen, G.; et al. Characterization of a novel HDAC/RXR/HtrA1 signaling axis as a novel target to overcome cisplatin resistance in human non-small cell lung cancer. Mol. Cancer 2020, 19, 134. [Google Scholar] [CrossRef] [PubMed]
  50. Maccallini, C.; Ammazzalorso, A.; De Filippis, B.; Fantacuzzi, M.; Giampietro, L.; Amoroso, R. HDAC Inhibitors for the Therapy of Triple Negative Breast Cancer. Pharmaceuticals 2022, 15, 667. [Google Scholar] [CrossRef]
  51. Zhang, S.-L.; Zhu, H.-Y.; Zhou, B.-Y.; Chu, Y.; Huo, J.-R.; Tan, Y.-Y.; Liu, D.-L. Histone deacetylase 6 is overexpressed and promotes tumor growth of colon cancer through regulation of the MAPK/ERK signal pathway. OncoTargets Ther. 2019, 12, 2409–2419. [Google Scholar] [CrossRef]
  52. Cao, L.L.; Yue, Z.; Liu, L.; Pei, L.; Yin, Y.; Qin, L.; Zhao, J.; Liu, H.; Wang, H.; Jia, M. The expression of histone deacetylase HDAC1 correlates with the progression and prognosis of gastrointestinal malignancy. Oncotarget 2017, 8, 39241–39253. [Google Scholar] [CrossRef]
  53. Phimmachanh, M.; Han, J.Z.; O’Donnell, Y.E.; Latham, S.L.; Croucher, D.R. Histone deacetylases and histone deacetylase inhibitors in neuroblastoma. Front. Cell Dev. Biol. 2020, 8, 578770. [Google Scholar]
  54. Liang, T.; Wang, F.; Elhassan, R.M.; Cheng, Y.; Tang, X.; Chen, W.; Fang, H.; Hou, X. Targeting histone deacetylases for cancer therapy: Trends and challenges. Acta Pharm. Sin. B 2023, 13, 2425–2463. [Google Scholar] [CrossRef]
  55. Zhang, J.; Zhong, Q. Histone deacetylase inhibitors and cell death. Cell. Mol. Life Sci. 2014, 71, 3885–3901. [Google Scholar]
  56. Wang, Z.; Hu, P.; Tang, F.; Lian, H.; Chen, X.; Zhang, Y.; He, X.; Liu, W.; Xie, C. HDAC6 promotes cell proliferation and confers resistance to temozolomide in glioblastoma. Cancer Lett. 2016, 379, 134–142. [Google Scholar] [CrossRef] [PubMed]
  57. Ghate, N.B.; Nadkarni, K.S.; Barik, G.K.; Tat, S.S.; Sahay, O.; Santra, M.K. Histone ubiquitination: Role in genome integrity and chromatin organization. Biochim. Et Biophys. Acta (BBA)-Gene Regul. Mech. 2024, 1867, 195044. [Google Scholar] [CrossRef] [PubMed]
  58. Chen, S.; Chen, J.; Zhan, Q.; Zhu, Y.; Chen, H.; Deng, X.; Hou, Z.; Shen, B.; Chen, Y.; Peng, C. H2AK119Ub1 and H3K27Me3 in molecular staging for survival prediction of patients with pancreatic ductal adenocarcinoma. Oncotarget 2014, 5, 10421–10433. [Google Scholar] [CrossRef]
  59. Wang, Z.; Zhu, L.; Guo, T.; Wang, Y.; Yang, J. Decreased H2B monoubiquitination and overexpression of ubiquitin-specific protease enzyme 22 in malignant colon carcinoma. Hum. Pathol. 2015, 46, 1006–1014. [Google Scholar] [CrossRef] [PubMed]
  60. Prenzel, T.; Begus-Nahrmann, Y.; Kramer, F.; Hennion, M.; Hsu, C.; Gorsler, T.; Hintermair, C.; Eick, D.; Kremmer, E.; Simons, M. Estrogen-dependent gene transcription in human breast cancer cells relies upon proteasome-dependent monoubiquitination of histone H2B. Cancer Res. 2011, 71, 5739–5753. [Google Scholar] [CrossRef]
  61. Wang, Z.-J.; Yang, J.-L.; Wang, Y.-P.; Lou, J.-Y.; Chen, J.; Liu, C.; Guo, L.-D. Decreased histone H2B monoubiquitination in malignant gastric carcinoma. World J. Gastroenterol. WJG 2013, 19, 8099. [Google Scholar]
  62. Senft, D.; Qi, J.; Ronai, Z.e.A. Ubiquitin ligases in oncogenic transformation and cancer therapy. Nat. Rev. Cancer 2018, 18, 69–88. [Google Scholar] [CrossRef]
  63. Vertegaal, A.C.O. Signalling mechanisms and cellular functions of SUMO. Nat. Rev. Mol. Cell Biol. 2022, 23, 715–731. [Google Scholar] [CrossRef]
  64. Ding, X.; Sun, J.; Wang, L.; Li, G.; Shen, Y.; Zhou, X.; Chen, W. Overexpression of SENP5 in oral squamous cell carcinoma and its association with differentiation. Oncol. Rep. 2008, 20, 1041–1045. [Google Scholar]
  65. Katayama, A.; Ogino, T.; Bandoh, N.; Takahara, M.; Kishibe, K.; Nonaka, S.; Harabuchi, Y. Overexpression of small ubiquitin-related modifier-1 and sumoylated Mdm2 in oral squamous cell carcinoma: Possible involvement in tumor proliferation and prognosis. Int. J. Oncol. 2007, 31, 517–524. [Google Scholar] [CrossRef]
  66. Shiio, Y.; Eisenman, R.N. Histone sumoylation is associated with transcriptional repression. Proc. Natl. Acad. Sci. USA 2003, 100, 13225–13230. [Google Scholar] [CrossRef] [PubMed]
  67. Bogachek, M.V.; De Andrade, J.P.; Weigel, R.J. Regulation of Epithelial–Mesenchymal Transition through SUMOylation of Transcription Factors. Cancer Res. 2015, 75, 11–15. [Google Scholar] [CrossRef]
  68. Wang, Y.; Chen, Y.; Zhao, M. N6-methyladenosine modification and post-translational modification of epithelial-mesenchymal transition in colorectal cancer. Discov. Oncol. 2024, 15, 209. [Google Scholar] [CrossRef] [PubMed]
  69. Sandberg, K.; Samson, W.K.; Ji, H. Decoding noncoding RNA: Da Vinci redux? Circ. Res. 2013, 113, 240–241. [Google Scholar] [CrossRef] [PubMed]
  70. Chen, L.-L.; Kim, V.N. Small and long non-coding RNAs: Past, present, and future. Cell 2024, 187, 6451–6485. [Google Scholar]
  71. Friedman, R.C.; Farh, K.K.-H.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef]
  72. Laggerbauer, B.; Engelhardt, S. MicroRNAs as therapeutic targets in cardiovascular disease. J. Clin. Investig. 2022, 132, 11. [Google Scholar] [CrossRef]
  73. Abdallah, H.Y.; Faisal, S.; Tawfik, N.Z.; Soliman, N.H.; Kishk, R.M.; Ellawindy, A. Expression signature of immune-related MicroRNAs in autoimmune skin disease: Psoriasis and vitiligo insights. Mol. Diagn. Ther. 2023, 27, 405–423. [Google Scholar]
  74. Renaudineau, Y.; Berindan-Neagoe, I.; Stanciu, L.A. role of macrophage MicroRNAs in inflammatory diseases and cancer. Front. Immunol. 2021, 12, 764525. [Google Scholar]
  75. Mattiske, S.; Suetani, R.J.; Neilsen, P.M.; Callen, D.F. The Oncogenic Role of miR-155 in Breast Cancer. Cancer Epidemiol. Biomark. Prev. 2012, 21, 1236–1243. [Google Scholar] [CrossRef]
  76. Braga, T.V.; Evangelista, F.C.G.; Gomes, L.C.; Araújo, S.S.d.S.; Carvalho, M.d.G.; Sabino, A.d.P. Evaluation of MiR-15a and MiR-16-1 as prognostic biomarkers in chronic lymphocytic leukemia. Biomed. Pharmacother. 2017, 92, 864–869. [Google Scholar] [CrossRef] [PubMed]
  77. Xiong, Q.; Zhang, Y.; Li, J.; Zhu, Q. Small Non-Coding RNAs in Human Cancer. Genes 2022, 13, 2072. [Google Scholar] [CrossRef]
  78. Wang, H.; Tan, Z.; Hu, H.; Liu, H.; Wu, T.; Zheng, C.; Wang, X.; Luo, Z.; Wang, J.; Liu, S. microRNA-21 promotes breast cancer proliferation and metastasis by targeting LZTFL1. BMC Cancer 2019, 19, 738. [Google Scholar] [CrossRef] [PubMed]
  79. Zong, Y.; Zhang, Y.; Sun, X.; Xu, T.; Cheng, X.; Qin, Y. miR-221/222 promote tumor growth and suppress apoptosis by targeting lncRNA GAS5 in breast cancer. Biosci. Rep. 2019, 39, BSR20181859. [Google Scholar]
  80. Sheedy, P.; Medarova, Z. The fundamental role of miR-10b in metastatic cancer. Am. J. Cancer Res. 2018, 8, 1674. [Google Scholar]
  81. Ghafouri-Fard, S.; Khoshbakht, T.; Hussen, B.M.; Abdullah, S.T.; Taheri, M.; Samadian, M. A review on the role of mir-16-5p in the carcinogenesis. Cancer Cell Int. 2022, 22, 342. [Google Scholar] [CrossRef]
  82. Li, X.J.; Ren, Z.J.; Tang, J.H. MicroRNA-34a: A potential therapeutic target in human cancer. Cell Death Dis. 2014, 5, e1327. [Google Scholar] [CrossRef]
  83. Lee, S.W.L.; Paoletti, C.; Campisi, M.; Osaki, T.; Adriani, G.; Kamm, R.D.; Mattu, C.; Chiono, V. MicroRNA delivery through nanoparticles. J. Control. Release 2019, 313, 80–95. [Google Scholar] [CrossRef]
  84. Greifenstein, A.A.; Jo, S.; Bierhoff, H. RNA:DNA triple helices: From peculiar structures to pervasive chromatin regulators. Essays Biochem. 2021, 65, 731–740. [Google Scholar] [CrossRef]
  85. Forrest, M.E.; Khalil, A.M. Review: Regulation of the cancer epigenome by long non-coding RNAs. Cancer Lett. 2017, 407, 106–112. [Google Scholar] [CrossRef]
  86. Mattick, J.S.; Amaral, P.P.; Carninci, P.; Carpenter, S.; Chang, H.Y.; Chen, L.-L.; Chen, R.; Dean, C.; Dinger, M.E.; Fitzgerald, K.A. Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat. Rev. Mol. Cell Biol. 2023, 24, 430–447. [Google Scholar] [CrossRef] [PubMed]
  87. Al-Hadid, Q.; Yang, Y. R-loop: An emerging regulator of chromatin dynamics. Acta Biochim. Et Biophys. Sin. 2016, 48, 623–631. [Google Scholar] [CrossRef]
  88. Allison, D.F.; Wang, G.G. R-loops: Formation, function, and relevance to cell stress. Cell Stress 2019, 3, 38–46. [Google Scholar] [CrossRef]
  89. Li, Y.; Jiang, T.; Zhou, W.; Li, J.; Li, X.; Wang, Q.; Jin, X.; Yin, J.; Chen, L.; Zhang, Y. Pan-cancer characterization of immune-related lncRNAs identifies potential oncogenic biomarkers. Nat. Commun. 2020, 11, 1000. [Google Scholar]
  90. Amodio, N.; Raimondi, L.; Juli, G.; Stamato, M.A.; Caracciolo, D.; Tagliaferri, P.; Tassone, P. MALAT1: A druggable long non-coding RNA for targeted anti-cancer approaches. J. Hematol. Oncol. 2018, 11, 63. [Google Scholar] [CrossRef]
  91. Huo, Y.; Li, Q.; Wang, X.; Jiao, X.; Zheng, J.; Li, Z.; Pan, X. MALAT1 predicts poor survival in osteosarcoma patients and promotes cell metastasis through associating with EZH2. Oncotarget 2017, 8, 46993. [Google Scholar] [PubMed]
  92. Wang, W.; Zhu, Y.; Li, S.; Chen, X.; Jiang, G.; Shen, Z.; Qiao, Y.; Wang, L.; Zheng, P.; Zhang, Y. Long noncoding RNA MALAT1 promotes malignant development of esophageal squamous cell carcinoma by targeting β-catenin via Ezh2. Oncotarget 2016, 7, 25668–25682. [Google Scholar] [CrossRef]
  93. Xing, C.; Sun, S.-g.; Yue, Z.-Q.; Bai, F. Role of lncRNA LUCAT1 in cancer. Biomed. Pharmacother. 2021, 134, 111158. [Google Scholar] [CrossRef]
  94. Ma, C.; Shi, X.; Zhu, Q.; Li, Q.; Liu, Y.; Yao, Y.; Song, Y. The growth arrest-specific transcript 5 (GAS5): A pivotal tumor suppressor long noncoding RNA in human cancers. Tumor Biol. 2016, 37, 1437–1444. [Google Scholar] [CrossRef]
  95. Elsakrmy, N.; Cui, H. R-Loops and R-Loop-Binding Proteins in Cancer Progression and Drug Resistance. Int. J. Mol. Sci. 2023, 24, 64. [Google Scholar] [CrossRef]
  96. Li, D.; Shao, F.; Li, X.; Yu, Q.; Wu, R.; Wang, J.; Wang, Z.; Wusiman, D.; Ye, L.; Guo, Y.; et al. Advancements and challenges of R-loops in cancers: Biological insights and future directions. Cancer Lett. 2025, 610, 217359. [Google Scholar] [CrossRef]
  97. Kim, S.; Shin, W.H.; Kang, Y.; Kim, H.; Lee, J.Y. Direct visualization of replication and R-loop collision using single-molecule imaging. Nucleic Acids Res. 2024, 52, 259–273. [Google Scholar] [CrossRef] [PubMed]
  98. Hatchi, E.; Skourti-Stathaki, K.; Ventz, S.; Pinello, L.; Yen, A.; Kamieniarz-Gdula, K.; Dimitrov, S.; Pathania, S.; McKinney, K.M.; Eaton, M.L.; et al. BRCA1 Recruitment to Transcriptional Pause Sites Is Required for R-Loop-Driven DNA Damage Repair. Mol. Cell 2015, 57, 636–647. [Google Scholar] [CrossRef] [PubMed]
  99. Dumas, L.; Herviou, P.; Dassi, E.; Cammas, A.; Millevoi, S. G-Quadruplexes in RNA biology: Recent advances and future directions. Trends Biochem. Sci. 2021, 46, 270–283. [Google Scholar]
  100. Zenkov, R.G.; Kirsanov, K.I.; Ogloblina, A.M.; Vlasova, O.A.; Naberezhnov, D.S.; Karpechenko, N.Y.; Fetisov, T.I.; Lesovaya, E.A.; Belitsky, G.A.; Dolinnaya, N.G.; et al. Effects of G-Quadruplex-Binding Plant Secondary Metabolites on c-MYC Expression. Int. J. Mol. Sci. 2022, 23, 9209. [Google Scholar] [CrossRef]
  101. Wanrooij, P.H.; Uhler, J.P.; Simonsson, T.; Falkenberg, M.; Gustafsson, C.M. G-quadruplex structures in RNA stimulate mitochondrial transcription termination and primer formation. Proc. Natl. Acad. Sci. USA 2010, 107, 16072–16077. [Google Scholar]
  102. Varshney, D.; Spiegel, J.; Zyner, K.; Tannahill, D.; Balasubramanian, S. The regulation and functions of DNA and RNA G-quadruplexes. Nat. Rev. Mol. Cell Biol. 2020, 21, 459–474. [Google Scholar] [PubMed]
  103. Kim, N.W.; Piatyszek, M.A.; Prowse, K.R.; Harley, C.B.; West, M.D.; Ho, P.L.; Coviello, G.M.; Wright, W.E.; Weinrich, S.L.; Shay, J.W. Specific association of human telomerase activity with immortal cells and cancer. Science 1994, 266, 2011–2015. [Google Scholar] [CrossRef]
  104. Moye, A.L.; Porter, K.C.; Cohen, S.B.; Phan, T.; Zyner, K.G.; Sasaki, N.; Lovrecz, G.O.; Beck, J.L.; Bryan, T.M. Telomeric G-quadruplexes are a substrate and site of localization for human telomerase. Nat. Commun. 2015, 6, 7643. [Google Scholar]
  105. Bahls, B.; Aljnadi, I.M.; Emídio, R.; Mendes, E.; Paulo, A. G-Quadruplexes in c-MYC Promoter as Targets for Cancer Therapy. Biomedicines 2023, 11, 969. [Google Scholar] [CrossRef]
  106. Xu, J.; Huang, H.; Zhou, X. G-Quadruplexes in Neurobiology and Virology: Functional Roles and Potential Therapeutic Approaches. JACS Au 2021, 1, 2146–2161. [Google Scholar] [CrossRef] [PubMed]
  107. Awadasseid, A.; Ma, X.; Wu, Y.; Zhang, W. G-quadruplex stabilization via small-molecules as a potential anti-cancer strategy. Biomed. Pharmacother. 2021, 139, 111550. [Google Scholar] [CrossRef]
  108. Figueiredo, J.; Mergny, J.-L.; Cruz, C. G-quadruplex ligands in cancer therapy: Progress, challenges, and clinical perspectives. Life Sci. 2024, 340, 122481. [Google Scholar] [CrossRef]
  109. Huppert, J.L.; Balasubramanian, S. G-quadruplexes in promoters throughout the human genome. Nucleic Acids Res. 2006, 35, 406–413. [Google Scholar] [CrossRef]
  110. Gomez, D.; Guédin, A.; Mergny, J.-L.; Salles, B.; Riou, J.-F.; Teulade-Fichou, M.-P.; Calsou, P. A G-quadruplex structure within the 5′-UTR of TRF2 mRNA represses translation in human cells. Nucleic Acids Res. 2010, 38, 7187–7198. [Google Scholar] [CrossRef] [PubMed]
  111. Gray, L.T.; Vallur, A.C.; Eddy, J.; Maizels, N. G quadruplexes are genomewide targets of transcriptional helicases XPB and XPD. Nat. Chem. Biol. 2014, 10, 313–318. [Google Scholar] [CrossRef]
  112. Coan, M.; Haefliger, S.; Ounzain, S.; Johnson, R. Targeting and engineering long non-coding RNAs for cancer therapy. Nat. Rev. Genet. 2024, 25, 578–595. [Google Scholar] [CrossRef]
  113. Cao, J.; Yan, Q. Cancer Epigenetics, Tumor Immunity, and Immunotherapy. Trends Cancer 2020, 6, 580–592. [Google Scholar] [CrossRef]
  114. Dawson, M.A. The cancer epigenome: Concepts, challenges, and therapeutic opportunities. Science 2017, 355, 1147–1152. [Google Scholar]
  115. Roulois, D.; Yau, H.L.; Singhania, R.; Wang, Y.; Danesh, A.; Shen, S.Y.; Han, H.; Liang, G.; Jones, P.A.; Pugh, T.J. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell 2015, 162, 961–973. [Google Scholar]
  116. Chiappinelli, K.B.; Strissel, P.L.; Desrichard, A.; Li, H.; Henke, C.; Akman, B.; Hein, A.; Rote, N.S.; Cope, L.M.; Snyder, A. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 2015, 162, 974–986. [Google Scholar] [CrossRef]
  117. Sheng, W.; LaFleur, M.W.; Nguyen, T.H.; Chen, S.; Chakravarthy, A.; Conway, J.R.; Li, Y.; Chen, H.; Yang, H.; Hsu, P.-H. LSD1 ablation stimulates anti-tumor immunity and enables checkpoint blockade. Cell 2018, 174, 549–563. e519. [Google Scholar] [CrossRef] [PubMed]
  118. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/212576s000lbl.pdf (accessed on 28 December 2024).
  119. Garcia-Manero, G.; Griffiths, E.A.; Steensma, D.P.; Roboz, G.J.; Wells, R.; McCloskey, J.; Odenike, O.; DeZern, A.E.; Yee, K.; Busque, L.; et al. Oral cedazuridine/decitabine for MDS and CMML: A phase 2 pharmacokinetic/pharmacodynamic randomized crossover study. Blood 2020, 136, 674–683. [Google Scholar] [CrossRef] [PubMed]
  120. Garcia-Manero, G.; McCloskey, J.; Griffiths, E.A.; Yee, K.W.L.; Zeidan, A.M.; Al-Kali, A.; Deeg, H.J.; Patel, P.A.; Sabloff, M.; Keating, M.M.; et al. Oral decitabine-cedazuridine versus intravenous decitabine for myelodysplastic syndromes and chronic myelomonocytic leukaemia (ASCERTAIN): A registrational, randomised, crossover, pharmacokinetics, phase 3 study. Lancet Haematol. 2024, 11, e15–e26. [Google Scholar] [CrossRef] [PubMed]
  121. Bataller, A.; Montalban-Bravo, G.; Bazinet, A.; Alvarado, Y.; Chien, K.; Venugopal, S.; Ishizawa, J.; Hammond, D.; Swaminathan, M.; Sasaki, K.; et al. Oral decitabine plus cedazuridine and venetoclax in patients with higher-risk myelodysplastic syndromes or chronic myelomonocytic leukaemia: A single-centre, phase 1/2 study. Lancet Haematol. 2024, 11, e186–e195. [Google Scholar] [CrossRef]
  122. Kantarjian, H.; Oki, Y.; Garcia-Manero, G.; Huang, X.; O’Brien, S.; Cortes, J.; Faderl, S.; Bueso-Ramos, C.; Ravandi, F.; Estrov, Z.; et al. Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood 2006, 109, 52–57. [Google Scholar] [CrossRef]
  123. Zhao, Q.; Fan, J.; Hong, W.; Li, L.; Wu, M. Inhibition of cancer cell proliferation by 5-fluoro-2′-deoxycytidine, a DNA methylation inhibitor, through activation of DNA damage response pathway. SpringerPlus 2012, 1, 65. [Google Scholar] [CrossRef]
  124. O’Connell, C.L.; Baer, M.R.; Ørskov, A.D.; Saini, S.K.; Duong, V.H.; Kropf, P.; Hansen, J.W.; Tsao-Wei, D.; Jang, H.S.; Emadi, A. Safety, outcomes, and T-cell characteristics in patients with relapsed or refractory MDS or CMML treated with atezolizumab in combination with guadecitabine. Clin. Cancer Res. 2022, 28, 5306–5316. [Google Scholar] [CrossRef]
  125. Garcia-Manero, G.; Roboz, G.; Walsh, K.; Kantarjian, H.; Ritchie, E.; Kropf, P.; O’Connell, C.; Tibes, R.; Lunin, S.; Rosenblat, T. Guadecitabine (SGI-110) in patients with intermediate or high-risk myelodysplastic syndromes: Phase 2 results from a multicentre, open-label, randomised, phase 1/2 trial. Lancet Haematol. 2019, 6, e317–e327. [Google Scholar] [CrossRef]
  126. Chung, W.; Kelly, A.D.; Kropf, P.; Fung, H.; Jelinek, J.; Su, X.Y.; Roboz, G.J.; Kantarjian, H.M.; Azab, M.; Issa, J.-P.J. Genomic and epigenomic predictors of response to guadecitabine in relapsed/refractory acute myelogenous leukemia. Clin. Epigenetics 2019, 11, 106. [Google Scholar] [CrossRef]
  127. Kantarjian, H.M.; Roboz, G.J.; Kropf, P.L.; Yee, K.W.; O’Connell, C.L.; Tibes, R.; Walsh, K.J.; Podoltsev, N.A.; Griffiths, E.A.; Jabbour, E. Guadecitabine (SGI-110) in treatment-naive patients with acute myeloid leukaemia: Phase 2 results from a multicentre, randomised, phase 1/2 trial. Lancet Oncol. 2017, 18, 1317–1326. [Google Scholar] [PubMed]
  128. Noviello, T.M.R.; Di Giacomo, A.M.; Caruso, F.P.; Covre, A.; Mortarini, R.; Scala, G.; Costa, M.C.; Coral, S.; Fridman, W.H.; Sautès-Fridman, C.; et al. Guadecitabine plus ipilimumab in unresectable melanoma: Five-year follow-up and integrated multi-omic analysis in the phase 1b NIBIT-M4 trial. Nat. Commun. 2023, 14, 5914. [Google Scholar] [CrossRef] [PubMed]
  129. Albany, C.; Fazal, Z.; Singh, R.; Bikorimana, E.; Adra, N.; Hanna, N.H.; Einhorn, L.H.; Perkins, S.M.; Sandusky, G.E.; Christensen, B.C.; et al. A phase 1 study of combined guadecitabine and cisplatin in platinum refractory germ cell cancer. Cancer Med. 2021, 10, 156–163. [Google Scholar] [CrossRef] [PubMed]
  130. Prebet, T.; Goldberg, A.D.; Jurcic, J.G.; Khaled, S.; Dail, M.; Feng, Y.; Green, C.; Li, C.; Ma, C.; Medeiros, B.C. A phase 1b study of atezolizumab in combination with guadecitabine for the treatment of acute myeloid leukemia. Leuk. Lymphoma 2022, 63, 2180–2188. [Google Scholar]
  131. Papadatos-Pastos, D.; Yuan, W.; Pal, A.; Crespo, M.; Ferreira, A.; Gurel, B.; Prout, T.; Ameratunga, M.; Chénard-Poirier, M.; Curcean, A.; et al. Phase 1, dose-escalation study of guadecitabine (SGI-110) in combination with pembrolizumab in patients with solid tumors. J. Immunother. Cancer 2022, 10, e004495. [Google Scholar] [CrossRef]
  132. Bever, K.M.; Thomas, D.L., 2nd; Zhang, J.; Diaz Rivera, E.A.; Rosner, G.L.; Zhu, Q.; Nauroth, J.M.; Christmas, B.; Thompson, E.D.; Anders, R.A.; et al. A feasibility study of combined epigenetic and vaccine therapy in advanced colorectal cancer with pharmacodynamic endpoint. Clin. Epigenetics 2021, 13, 25. [Google Scholar] [CrossRef]
  133. Di Giacomo, A.M.; Covre, A.; Finotello, F.; Rieder, D.; Danielli, R.; Sigalotti, L.; Giannarelli, D.; Petitprez, F.; Lacroix, L.; Valente, M. Guadecitabine plus ipilimumab in unresectable melanoma: The NIBIT-M4 clinical trial. Clin. Cancer Res. 2019, 25, 7351–7362. [Google Scholar] [CrossRef]
  134. Wei, C.X.; Mamdani, H.; Gentzler, R.; Kalra, M.; Perkins, S.; Althouse, S.; Jalal, S.I. A brief report of a phase II trial evaluating efficacy and safety of hypomethylating agent guadecitabine in combination with carboplatin in extensive stage small cell lung cancer. Clin. Lung Cancer 2023, 24, 347–352. [Google Scholar] [CrossRef]
  135. Jang, H.J.; Hostetter, G.; Macfarlane, A.W.; Madaj, Z.; Ross, E.A.; Hinoue, T.; Kulchycki, J.R.; Burgos, R.S.; Tafseer, M.; Alpaugh, R.K. A phase II trial of guadecitabine plus atezolizumab in metastatic urothelial carcinoma progressing after initial immune checkpoint inhibitor therapy. Clin. Cancer Res. 2023, 29, 2052–2065. [Google Scholar]
  136. Chen, S.; Xie, P.; Cowan, M.; Huang, H.; Cardenas, H.; Keathley, R.; Tanner, E.J.; Fleming, G.F.; Moroney, J.W.; Pant, A.; et al. Epigenetic priming enhances antitumor immunity in platinum-resistant ovarian cancer. J Clin Investig 2022, 132, 14. [Google Scholar] [CrossRef]
  137. Sheikh, T.N.; Chen, X.; Xu, X.; McGuire, J.T.; Ingham, M.; Lu, C.; Schwartz, G.K. Growth inhibition and induction of innate immune signaling of chondrosarcomas with epigenetic inhibitors. Mol. Cancer Ther. 2021, 20, 2362–2371. [Google Scholar] [PubMed]
  138. Kaminskas, E.; Farrell, A.T.; Wang, Y.-C.; Sridhara, R.; Pazdur, R. FDA drug approval summary: Azacitidine (5-azacytidine, Vidaza™) for injectable suspension. Oncologist 2005, 10, 176–182. [Google Scholar]
  139. Winquist, E.; Knox, J.; Ayoub, J.-P.; Wood, L.; Wainman, N.; Reid, G.K.; Pearce, L.; Shah, A.; Eisenhauer, E. Phase II trial of DNA methyltransferase 1 inhibition with the antisense oligonucleotide MG98 in patients with metastatic renal carcinoma: A National Cancer Institute of Canada Clinical Trials Group investigational new drug study. Investig. New Drugs 2006, 24, 159–167. [Google Scholar]
  140. Stewart, D.; Donehower, R.; Eisenhauer, E.; Wainman, N.; Shah, A.; Bonfils, C.; MacLeod, A.; Besterman, J.; Reid, G. A phase I pharmacokinetic and pharmacodynamic study of the DNA methyltransferase 1 inhibitor MG98 administered twice weekly. Ann. Oncol. 2003, 14, 766–774. [Google Scholar]
  141. Candelaria, M.; Gallardo-Rincón, D.; Arce, C.; Cetina, L.; Aguilar-Ponce, J.L.; Arrieta, O.; Gonzalez-Fierro, A.; Chavez-Blanco, A.; de La Cruz-Hernandez, E.; Camargo, M. A phase II study of epigenetic therapy with hydralazine and magnesium valproate to overcome chemotherapy resistance in refractory solid tumors. Ann. Oncol. 2007, 18, 1529–1538. [Google Scholar] [PubMed]
  142. Liu, Y.C.; Su, C.W.; Ko, P.S.; Lee, R.C.; Liu, C.J.; Huang, Y.H.; Gau, J.P.; Liu, J.H. A clinical trial with valproic acid and hydralazine in combination with gemcitabine and cisplatin followed by doxorubicin and dacarbazine for advanced hepatocellular carcinoma. Asia Pac. J. Clin. Oncol. 2022, 18, 19–27. [Google Scholar] [CrossRef] [PubMed]
  143. Arce, C.; Pérez-Plasencia, C.; González-Fierro, A.; de la Cruz-Hernández, E.; Revilla-Vázquez, A.; Chávez-Blanco, A.; Trejo-Becerril, C.; Pérez-Cárdenas, E.; Taja-Chayeb, L.; Bargallo, E. A proof-of-principle study of epigenetic therapy added to neoadjuvant doxorubicin cyclophosphamide for locally advanced breast cancer. PLoS ONE 2006, 1, e98. [Google Scholar]
  144. Fang, M.Z.; Wang, Y.; Ai, N.; Hou, Z.; Sun, Y.; Lu, H.; Welsh, W.; Yang, C.S. Tea polyphenol (−)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res. 2003, 63, 7563–7570. [Google Scholar]
  145. Aldawsari, F.S.; Aguayo-Ortiz, R.; Kapilashrami, K.; Yoo, J.; Luo, M.; Medina-Franco, J.L.; Velázquez-Martínez, C.A. Resveratrol-salicylate derivatives as selective DNMT3 inhibitors and anticancer agents. J. Enzym. Inhib. Med. Chem. 2016, 31, 695–703. [Google Scholar] [CrossRef]
  146. Sun, N.; Zhang, J.; Zhang, C.; Zhao, B.; Jiao, A. DNMTs inhibitor SGI-1027 induces apoptosis in Huh7 human hepatocellular carcinoma cells. Oncol. Lett. 2018, 16, 5799–5806. [Google Scholar] [CrossRef]
  147. Fagan, R.L.; Cryderman, D.E.; Kopelovich, L.; Wallrath, L.L.; Brenner, C. Laccaic Acid A Is a Direct, DNA-competitive Inhibitor of DNA Methyltransferase 1*. J. Biol. Chem. 2013, 288, 23858–23867. [Google Scholar] [CrossRef] [PubMed]
  148. Brueckner, B.; Garcia Boy, R.; Siedlecki, P.; Musch, T.; Kliem, H.C.; Zielenkiewicz, P.; Suhai, S.; Wiessler, M.; Lyko, F. Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases. Cancer Res. 2005, 65, 6305–6311. [Google Scholar]
  149. Kuck, D.; Caulfield, T.; Lyko, F.; Medina-Franco, J.L. Nanaomycin A selectively inhibits DNMT3B and reactivates silenced tumor suppressor genes in human cancer cells. Mol. Cancer Ther. 2010, 9, 3015–3023. [Google Scholar]
  150. Mann, B.S.; Johnson, J.R.; Cohen, M.H.; Justice, R.; Pazdur, R. FDA Approval Summary: Vorinostat for Treatment of Advanced Primary Cutaneous T-Cell Lymphoma. Oncologist 2007, 12, 1247–1252. [Google Scholar] [CrossRef]
  151. Lee, H.Z.; Kwitkowski, V.E.; Del Valle, P.L.; Ricci, M.S.; Saber, H.; Habtemariam, B.A.; Bullock, J.; Bloomquist, E.; Li Shen, Y.; Chen, X.H.; et al. FDA Approval: Belinostat for the Treatment of Patients with Relapsed or Refractory Peripheral T-cell Lymphoma. Clin. Cancer Res. 2015, 21, 2666–2670. [Google Scholar] [CrossRef] [PubMed]
  152. Petrich, A.; Nabhan, C. Use of class I histone deacetylase inhibitor romidepsin in combination regimens. Leuk. Lymphoma 2016, 57, 1755–1765. [Google Scholar]
  153. Abaza, Y.M.; Kadia, T.M.; Jabbour, E.J.; Konopleva, M.Y.; Borthakur, G.; Ferrajoli, A.; Estrov, Z.; Wierda, W.G.; Alfonso, A.; Chong, T.H. Phase 1 dose escalation multicenter trial of pracinostat alone and in combination with azacitidine in patients with advanced hematologic malignancies. Cancer 2017, 123, 4851–4859. [Google Scholar] [PubMed]
  154. Yong, W.; Goh, B.; Soo, R.; Toh, H.; Ethirajulu, K.; Wood, J.; Novotny-Diermayr, V.; Lee, S.; Yeo, W.; Chan, D. Phase I and pharmacodynamic study of an orally administered novel inhibitor of histone deacetylases, SB939, in patients with refractory solid malignancies. Ann. Oncol. 2011, 22, 2516–2522. [Google Scholar]
  155. Razak, A.; Hotte, S.; Siu, L.; Chen, E.; Hirte, H.; Powers, J.; Walsh, W.; Stayner, L.; Laughlin, A.; Novotny-Diermayr, V. Phase I clinical, pharmacokinetic and pharmacodynamic study of SB939, an oral histone deacetylase (HDAC) inhibitor, in patients with advanced solid tumours. Br. J. Cancer 2011, 104, 756–762. [Google Scholar]
  156. Yalniz, F.F.; Berdeja, J.G.; Maris, M.B.; Lyons, R.M.; Reeves Jr, J.A.; Essell, J.H.; Patel, P.; Sekeres, M.; Hughes, A.; Mappa, S. A phase II study of addition of pracinostat to a hypomethylating agent in patients with myelodysplastic syndromes who have not responded to previous hypomethylating agent therapy. Br. J. Haematol. 2020, 188, 404–412. [Google Scholar]
  157. Garcia-Manero, G.; Montalban-Bravo, G.; Berdeja, J.G.; Abaza, Y.; Jabbour, E.; Essell, J.; Lyons, R.M.; Ravandi, F.; Maris, M.; Heller, B. Phase 2, randomized, double-blind study of pracinostat in combination with azacitidine in patients with untreated, higher-risk myelodysplastic syndromes. Cancer 2017, 123, 994–1002. [Google Scholar] [PubMed]
  158. Quintás-Cardama, A.; Kantarjian, H.; Estrov, Z.; Borthakur, G.; Cortes, J.; Verstovsek, S. Therapy with the histone deacetylase inhibitor pracinostat for patients with myelofibrosis. Leuk. Res. 2012, 36, 1124–1127. [Google Scholar]
  159. Chu, Q.-C.; Nielsen, T.; Alcindor, T.; Gupta, A.; Endo, M.; Goytain, A.; Xu, H.; Verma, S.; Tozer, R.; Knowling, M. A phase II study of SB939, a novel pan-histone deacetylase inhibitor, in patients with translocation-associated recurrent/metastatic sarcomas—NCIC-CTG IND 200. Ann. Oncol. 2015, 26, 973–981. [Google Scholar] [PubMed]
  160. Garcia-Manero, G.; Kazmierczak, M.; Wierzbowska, A.; Fong, C.Y.; Keng, M.K.; Ballinari, G.; Scarci, F.; Adès, L. Pracinostat combined with azacitidine in newly diagnosed adult acute myeloid leukemia (AML) patients unfit for standard induction chemotherapy: PRIMULA phase III study. Leuk. Res. 2024, 140, 107480. [Google Scholar] [CrossRef]
  161. Singh, A.; Patel, V.K.; Jain, D.K.; Patel, P.; Rajak, H. Panobinostat as Pan-deacetylase Inhibitor for the Treatment of Pancreatic Cancer: Recent Progress and Future Prospects. Oncol. Ther. 2016, 4, 73–89. [Google Scholar] [CrossRef] [PubMed]
  162. Eleutherakis-Papaiakovou, E.; Kanellias, N.; Kastritis, E.; Gavriatopoulou, M.; Terpos, E.; Dimopoulos, M.A. Efficacy of Panobinostat for the Treatment of Multiple Myeloma. J. Oncol. 2020, 2020, 7131802. [Google Scholar] [CrossRef]
  163. Ikeda, M.; Ohno, I.; Ueno, H.; Mitsunaga, S.; Hashimoto, Y.; Okusaka, T.; Kondo, S.; Sasaki, M.; Sakamoto, Y.; Takahashi, H. Phase I study of resminostat, an HDAC inhibitor, combined with S-1 in patients with pre-treated biliary tract or pancreatic cancer. Investig. New Drugs 2019, 37, 109–117. [Google Scholar] [CrossRef] [PubMed]
  164. Walewski, J.; Paszkiewicz-Kozik, E.; Borsaru, G.; Hellmann, A.; Janikova, A.; Warszewska, A.; Mais, A.; Ammendola, A.; Herz, T.; Krauss, B. Resminostat in patients with relapsed or refractory Hodgkin lymphoma: Results of the phase II SAPHIRE study. Leuk. Lymphoma 2019, 60, 675–684. [Google Scholar] [CrossRef]
  165. Bitzer, M.; Horger, M.; Giannini, E.G.; Ganten, T.M.; Wörns, M.A.; Siveke, J.T.; Dollinger, M.M.; Gerken, G.; Scheulen, M.E.; Wege, H. Resminostat plus sorafenib as second-line therapy of advanced hepatocellular carcinoma–the SHELTER study. J. Hepatol. 2016, 65, 280–288. [Google Scholar]
  166. Tambo, Y.; Hosomi, Y.; Sakai, H.; Nogami, N.; Atagi, S.; Sasaki, Y.; Kato, T.; Takahashi, T.; Seto, T.; Maemondo, M. Phase I/II study of docetaxel combined with resminostat, an oral hydroxamic acid HDAC inhibitor, for advanced non-small cell lung cancer in patients previously treated with platinum-based chemotherapy. Investig. New Drugs 2017, 35, 217–226. [Google Scholar]
  167. Streubel, G.; Schrepfer, S.; Kallus, H.; Parnitzke, U.; Wulff, T.; Hermann, F.; Borgmann, M.; Hamm, S. Histone deacetylase inhibitor resminostat in combination with sorafenib counteracts platelet-mediated pro-tumoral effects in hepatocellular carcinoma. Sci. Rep. 2021, 11, 9587. [Google Scholar] [CrossRef]
  168. Venugopal, B.; Baird, R.; Kristeleit, R.S.; Plummer, R.; Cowan, R.; Stewart, A.; Fourneau, N.; Hellemans, P.; Elsayed, Y.; Mcclue, S. A phase I study of quisinostat (JNJ-26481585), an oral hydroxamate histone deacetylase inhibitor with evidence of target modulation and antitumor activity, in patients with advanced solid tumors. Clin. Cancer Res. 2013, 19, 4262–4272. [Google Scholar] [PubMed]
  169. Moreau, P.; Facon, T.; Touzeau, C.; Benboubker, L.; Delain, M.; Badamo-Dotzis, J.; Phelps, C.; Doty, C.; Smit, H.; Fourneau, N. Quisinostat, bortezomib, and dexamethasone combination therapy for relapsed multiple myeloma. Leuk. Lymphoma 2016, 57, 1546–1559. [Google Scholar] [PubMed]
  170. Child, F.; Ortiz-Romero, P.; Alvarez, R.; Bagot, M.; Stadler, R.; Weichenthal, M.; Alves, R.; Quaglino, P.; Beylot-Barry, M.; Cowan, R. Phase II multicentre trial of oral quisinostat, a histone deacetylase inhibitor, in patients with previously treated stage IB–IVA mycosis fungoides/Sézary syndrome. Br. J. Dermatol. 2016, 175, 80–88. [Google Scholar]
  171. Booth, S.W.; Eyre, T.A.; Whittaker, J.; Campo, L.; Wang, L.M.; Soilleux, E.; Royston, D.; Rees, G.; Kesavan, M.; Hildyard, C. A Phase 2a cohort expansion study to assess the safety, tolerability, and preliminary efficacy of CXD101 in patients with advanced solid-organ cancer expressing HR23B or lymphoma. BMC Cancer 2021, 21, 851. [Google Scholar]
  172. Saunders, M.P.; Graham, J.; Cunningham, D.; Plummer, R.; Church, D.; Kerr, R.; Cook, S.; Zheng, S.; La Thangue, N.; Kerr, D. CXD101 and nivolumab in patients with metastatic microsatellite-stable colorectal cancer (CAROSELL): A multicentre, open-label, single-arm, phase II trial. ESMO Open 2022, 7, 100594. [Google Scholar]
  173. Aggarwal, R.; Thomas, S.; Pawlowska, N.; Bartelink, I.; Grabowsky, J.; Jahan, T.; Cripps, A.; Harb, A.; Leng, J.; Reinert, A. Inhibiting histone deacetylase as a means to reverse resistance to angiogenesis inhibitors: Phase I study of abexinostat plus pazopanib in advanced solid tumor malignancies. J. Clin. Oncol. 2017, 35, 1231–1239. [Google Scholar]
  174. Evens, A.M.; Balasubramanian, S.; Vose, J.M.; Harb, W.; Gordon, L.I.; Langdon, R.; Sprague, J.; Mani, C.; Yue, J.; Luan, Y. A phase I/II multicenter, open-label study of the oral histone deacetylase inhibitor abexinostat in relapsed/refractory lymphoma. Clin. Cancer Res. 2016, 22, 1059–1066. [Google Scholar]
  175. Choy, E.; Flamand, Y.; Balasubramanian, S.; Butrynski, J.E.; Harmon, D.C.; George, S.; Cote, G.M.; Wagner, A.J.; Morgan, J.A.; Mani, C. Phase 1 study of oral abexinostat, a histone deacetylase inhibitor, in combination with doxorubicin in patients with metastatic sarcoma. Cancer 2015, 121, 1223–1230. [Google Scholar]
  176. Morschhauser, F.; Terriou, L.; Coiffier, B.; Bachy, E.; Varga, A.; Kloos, I.; Lelièvre, H.; Sarry, A.-L.; Depil, S.; Ribrag, V. Phase 1 study of the oral histone deacetylase inhibitor abexinostat in patients with Hodgkin lymphoma, non-Hodgkin lymphoma, or chronic lymphocytic leukaemia. Investig. New Drugs 2015, 33, 423–431. [Google Scholar]
  177. Ribrag, V.; Kim, W.S.; Bouabdallah, R.; Lim, S.T.; Coiffier, B.; Illes, A.; Lemieux, B.; Dyer, M.J.; Offner, F.; Felloussi, Z. Safety and efficacy of abexinostat, a pan-histone deacetylase inhibitor, in non-Hodgkin lymphoma and chronic lymphocytic leukemia: Results of a phase II study. Haematologica 2017, 102, 903. [Google Scholar] [PubMed]
  178. Xu, Y.; Zhang, P.; Liu, Y. Chidamide tablets: HDAC inhibition to treat lymphoma. Drugs Today 2017, 53, 167–176. [Google Scholar] [CrossRef]
  179. Collier, K.A.; Valencia, H.; Newton, H.; Hade, E.M.; Sborov, D.W.; Cavaliere, R.; Poi, M.; Phelps, M.A.; Liva, S.G.; Coss, C.C. A phase 1 trial of the histone deacetylase inhibitor AR-42 in patients with neurofibromatosis type 2-associated tumors and advanced solid malignancies. Cancer Chemother. Pharmacol. 2021, 87, 599–611. [Google Scholar] [PubMed]
  180. Liva, S.G.; Coss, C.C.; Wang, J.; Blum, W.; Klisovic, R.; Bhatnagar, B.; Walsh, K.; Geyer, S.; Zhao, Q.; Garzon, R. Phase I study of AR-42 and decitabine in acute myeloid leukemia. Leuk. Lymphoma 2020, 61, 1484–1492. [Google Scholar] [CrossRef]
  181. Lin, J.; Elkon, J.; Ricart, B.; Palmer, E.; Zevallos-Delgado, C.; Noonepalle, S.; Burgess, B.; Siegel, R.; Ma, Y.; Villagra, A. Phase I study of entinostat in combination with enzalutamide for treatment of patients with metastatic castration-resistant prostate cancer. Oncologist 2021, 26, e2136–e2142. [Google Scholar]
  182. Gojo, I.; Jiemjit, A.; Trepel, J.B.; Sparreboom, A.; Figg, W.D.; Rollins, S.; Tidwell, M.L.; Greer, J.; Chung, E.J.; Lee, M.-J. Phase 1 and pharmacologic study of MS-275, a histone deacetylase inhibitor, in adults with refractory and relapsed acute leukemias. Blood 2007, 109, 2781–2790. [Google Scholar]
  183. Kummar, S.; Gutierrez, M.; Gardner, E.R.; Donovan, E.; Hwang, K.; Chung, E.J.; Lee, M.-J.; Maynard, K.; Kalnitskiy, M.; Chen, A. Phase I trial of MS-275, a histone deacetylase inhibitor, administered weekly in refractory solid tumors and lymphoid malignancies. Clin. Cancer Res. 2007, 13, 5411–5417. [Google Scholar]
  184. Iwata, H.; Nakamura, R.; Masuda, N.; Yamashita, T.; Yamamoto, Y.; Kobayashi, K.; Tsurutani, J.; Iwasa, T.; Yonemori, K.; Tamura, K. Efficacy and exploratory biomarker analysis of entinostat plus exemestane in advanced or recurrent breast cancer: Phase II randomized controlled trial. Jpn. J. Clin. Oncol. 2023, 53, 4–15. [Google Scholar]
  185. Hellmann, M.D.; Jänne, P.A.; Opyrchal, M.; Hafez, N.; Raez, L.E.; Gabrilovich, D.I.; Wang, F.; Trepel, J.B.; Lee, M.-J.; Yuno, A. Entinostat plus pembrolizumab in patients with metastatic NSCLC previously treated with anti–PD-(L) 1 therapy. Clin. Cancer Res. 2021, 27, 1019–1028. [Google Scholar]
  186. Xu, B.; Zhang, Q.; Hu, X.; Li, Q.; Sun, T.; Li, W.; Ouyang, Q.; Wang, J.; Tong, Z.; Yan, M. Entinostat, a class I selective histone deacetylase inhibitor, plus exemestane for Chinese patients with hormone receptor-positive advanced breast cancer: A multicenter, randomized, double-blind, placebo-controlled, phase 3 trial. Acta Pharm. Sin. B 2023, 13, 2250–2258. [Google Scholar]
  187. Bauer, T.M.; Besse, B.; Martinez-Marti, A.; Trigo, J.M.; Moreno, V.; Garrido, P.; Ferron-Brady, G.; Wu, Y.; Park, J.; Collingwood, T.; et al. Phase I, Open-Label, Dose-Escalation Study of the Safety, Pharmacokinetics, Pharmacodynamics, and Efficacy of GSK2879552 in Relapsed/Refractory SCLC. J. Thorac. Oncol. 2019, 14, 1828–1838. [Google Scholar] [CrossRef] [PubMed]
  188. Wang, N.; Ma, T.; Yu, B. Targeting epigenetic regulators to overcome drug resistance in cancers. Signal Transduct Target Ther. 2023, 8, 69. [Google Scholar] [CrossRef] [PubMed]
  189. Wass, M.; Göllner, S.; Besenbeck, B.; Schlenk, R.F.; Mundmann, P.; Göthert, J.R.; Noppeney, R.; Schliemann, C.; Mikesch, J.-H.; Lenz, G. A proof of concept phase I/II pilot trial of LSD1 inhibition by tranylcypromine combined with ATRA in refractory/relapsed AML patients not eligible for intensive therapy. Leukemia 2021, 35, 701–711. [Google Scholar] [PubMed]
  190. Ribrag, V.; Iglesias, L.; De Braud, F.; Ma, B.; Yokota, T.; Zander, T.; Spreafico, A.; Subbiah, V.; Illert, A.L.; Tan, D.; et al. A first-in-human phase 1/2 dose-escalation study of MAK683 (EED inhibitor) in patients with advanced malignancies. Eur. J. Cancer 2025, 216, 115122. [Google Scholar] [CrossRef]
  191. Waters, N.J.; Daigle, S.R.; Rehlaender, B.N.; Basavapathruni, A.; Campbell, C.T.; Jensen, T.B.; Truitt, B.F.; Olhava, E.J.; Pollock, R.M.; Stickland, K.A.; et al. Exploring drug delivery for the DOT1L inhibitor pinometostat (EPZ-5676): Subcutaneous administration as an alternative to continuous IV infusion, in the pursuit of an epigenetic target. J. Control. Release 2015, 220, 758–765. [Google Scholar] [CrossRef]
  192. Watts, J.; Minden, M.D.; Bachiashvili, K.; Brunner, A.M.; Abedin, S.; Crossman, T.; Zajac, M.; Moroz, V.; Egger, J.L.; Tarkar, A.; et al. Phase I/II study of the clinical activity and safety of GSK3326595 in patients with myeloid neoplasms. Ther. Adv. Hematol. 2024, 15, 20406207241275376. [Google Scholar] [CrossRef]
  193. Gajer, J.M.; Furdas, S.D.; Gründer, A.; Gothwal, M.; Heinicke, U.; Keller, K.; Colland, F.; Fulda, S.; Pahl, H.L.; Fichtner, I.; et al. Histone acetyltransferase inhibitors block neuroblastoma cell growth in vivo. Oncogenesis 2015, 4, e137. [Google Scholar] [CrossRef]
  194. Michaelides, M.R.; Kluge, A.; Patane, M.; Van Drie, J.H.; Wang, C.; Hansen, T.M.; Risi, R.M.; Mantei, R.; Hertel, C.; Karukurichi, K.; et al. Discovery of Spiro Oxazolidinediones as Selective, Orally Bioavailable Inhibitors of p300/CBP Histone Acetyltransferases. ACS Med. Chem. Lett. 2018, 9, 28–33. [Google Scholar] [CrossRef]
  195. Lu, W.; Xiong, H.; Chen, Y.; Wang, C.; Zhang, H.; Xu, P.; Han, J.; Xiao, S.; Ding, H.; Chen, Z.; et al. Discovery and biological evaluation of thiobarbituric derivatives as potent p300/CBP inhibitors. Bioorganic Med. Chem. 2018, 26, 5397–5407. [Google Scholar] [CrossRef]
  196. Oike, T.; Komachi, M.; Ogiwara, H.; Amornwichet, N.; Saitoh, Y.; Torikai, K.; Kubo, N.; Nakano, T.; Kohno, T. C646, a selective small molecule inhibitor of histone acetyltransferase p300, radiosensitizes lung cancer cells by enhancing mitotic catastrophe. Radiother. Oncol. 2014, 111, 222–227. [Google Scholar] [CrossRef]
  197. Milite, C.; Feoli, A.; Sasaki, K.; La Pietra, V.; Balzano, A.L.; Marinelli, L.; Mai, A.; Novellino, E.; Castellano, S.; Tosco, A. A novel cell-permeable, selective, and noncompetitive inhibitor of KAT3 histone acetyltransferases from a combined molecular pruning/classical isosterism approach. J. Med. Chem. 2015, 58, 2779–2798. [Google Scholar] [PubMed]
  198. Lasko, L.M.; Jakob, C.G.; Edalji, R.P.; Qiu, W.; Montgomery, D.; Digiammarino, E.L.; Hansen, T.M.; Risi, R.M.; Frey, R.; Manaves, V. Discovery of a selective catalytic p300/CBP inhibitor that targets lineage-specific tumours. Nature 2017, 550, 128–132. [Google Scholar] [PubMed]
  199. Zeng, X.; Sigoillot, F.; Gaur, S.; Choi, S.; Pfaff, K.L.; Oh, D.-C.; Hathaway, N.; Dimova, N.; Cuny, G.D.; King, R.W. Pharmacologic Inhibition of the Anaphase-Promoting Complex Induces A Spindle Checkpoint-Dependent Mitotic Arrest in the Absence of Spindle Damage. Cancer Cell 2010, 18, 382–395. [Google Scholar] [CrossRef]
  200. Vu, B.; Wovkulich, P.; Pizzolato, G.; Lovey, A.; Ding, Q.; Jiang, N.; Liu, J.-J.; Zhao, C.; Glenn, K.; Wen, Y. Discovery of RG7112: A small-molecule MDM2 inhibitor in clinical development. ACS Med. Chem. Lett. 2013, 4, 466–469. [Google Scholar] [PubMed]
  201. Montesinos, P.; Beckermann, B.M.; Catalani, O.; Esteve, J.; Gamel, K.; Konopleva, M.Y.; Martinelli, G.; Monnet, A.; Papayannidis, C.; Park, A.; et al. MIRROS: A randomized, placebo-controlled, Phase III trial of cytarabine ± idasanutlin in relapsed or refractory acute myeloid leukemia. Future Oncol. 2020, 16, 807–815. [Google Scholar] [CrossRef]
  202. Sun, D.; Li, Z.; Rew, Y.; Gribble, M.; Bartberger, M.D.; Beck, H.P.; Canon, J.; Chen, A.; Chen, X.; Chow, D.; et al. Discovery of AMG 232, a potent, selective, and orally bioavailable MDM2-p53 inhibitor in clinical development. J. Med. Chem. 2014, 57, 1454–1472. [Google Scholar] [CrossRef]
  203. van Zandwijk, N.; Pavlakis, N.; Kao, S.; Clarke, S.; Lee, A.; Brahmbhatt, H.; Macdiarmid, J.; Pattison, S.; Leslie, F.; Huynh, Y.; et al. P1.02—MesomiR 1: A Phase I study of TargomiRs in patients with refractory malignant pleural mesothelioma (MPM) and lung cancer (NSCLC). Ann. Oncol. 2015, 26, ii16. [Google Scholar] [CrossRef]
  204. Beg, M.S.; Brenner, A.J.; Sachdev, J.; Borad, M.; Kang, Y.-K.; Stoudemire, J.; Smith, S.; Bader, A.G.; Kim, S.; Hong, D.S. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Investig. New Drugs 2017, 35, 180–188. [Google Scholar]
  205. Kotecki, N.; Opdam, F.; Robbrecht, D.; Strijbos, M.; Kroon, K.; Janicot, M.; Yahyanejad, S.; Telford, B.; van den Bosch, M.; Alemdehy, F. Phase I/Ib study with INT-1B3, a novel LNP-formulated micro-RNA (miR-193a-3p mimic) therapeutic for patients with advanced solid cancer. J. Clin. Oncol. 2021, 39, TPS2666. [Google Scholar]
  206. Abplanalp, W.T.; Fischer, A.; John, D.; Zeiher, A.M.; Gosgnach, W.; Darville, H.; Montgomery, R.; Pestano, L.; Allée, G.; Paty, I. Efficiency and target derepression of anti-miR-92a: Results of a first in human study. Nucleic Acid Ther. 2020, 30, 335–345. [Google Scholar]
  207. Romano, G.; Acunzo, M.; Nana-Sinkam, P. microRNAs as Novel Therapeutics in Cancer. Cancers 2021, 13, 1526. [Google Scholar] [CrossRef] [PubMed]
  208. Lei, Y.; Chen, L.; Zhang, G.; Shan, A.; Ye, C.; Liang, B.; Sun, J.; Liao, X.; Zhu, C.; Chen, Y.; et al. MicroRNAs target the Wnt/β-catenin signaling pathway to regulate epithelial-mesenchymal transition in cancer (Review). Oncol. Rep. 2020, 44, 1299–1313. [Google Scholar] [CrossRef] [PubMed]
  209. Varkaris, A.; Medarova, Z. 383P Clinical experience with TTX-MC138: A first-in-class therapy against metastatic cancer. Ann. Oncol. 2024, 35, S379. [Google Scholar] [CrossRef]
  210. Gong, N.; Teng, X.; Li, J.; Liang, X.-J. Antisense oligonucleotide-conjugated nanostructure-targeting lncRNA MALAT1 inhibits cancer metastasis. ACS Appl. Mater. Interfaces 2018, 11, 37–42. [Google Scholar]
  211. Amodio, N.; Stamato, M.A.; Juli, G.; Morelli, E.; Fulciniti, M.; Manzoni, M.; Taiana, E.; Agnelli, L.; Cantafio, M.E.G.; Romeo, E. Drugging the lncRNA MALAT1 via LNA gapmeR ASO inhibits gene expression of proteasome subunits and triggers anti-multiple myeloma activity. Leukemia 2018, 32, 1948–1957. [Google Scholar]
  212. Liang, H.; Peng, J. LncRNA HOTAIR promotes proliferation, invasion and migration in NSCLC cells via the CCL22 signaling pathway. PLoS ONE 2022, 17, e0263997. [Google Scholar]
  213. Suzuki, M.M.; Iijima, K.; Ogami, K.; Shinjo, K.; Murofushi, Y.; Xie, J.; Wang, X.; Kitano, Y.; Mamiya, A.; Kibe, Y. TUG1-mediated R-loop resolution at microsatellite loci as a prerequisite for cancer cell proliferation. Nat. Commun. 2023, 14, 4521. [Google Scholar]
  214. Tasaki, Y.; Suzuki, M.; Katsushima, K.; Shinjo, K.; Iijima, K.; Murofushi, Y.; Naiki-Ito, A.; Hayashi, K.; Qiu, C.; Takahashi, A. Cancer-specific targeting of taurine-upregulated gene 1 enhances the effects of chemotherapy in pancreatic cancer. Cancer Res. 2021, 81, 1654–1666. [Google Scholar]
  215. Li, M.; Ding, X.; Zhang, Y.; Li, X.; Zhou, H.; Yang, L.; Li, Y.; Yang, P.; Zhang, X.; Hu, J. Antisense oligonucleotides targeting lncRNA AC104041. 1 induces antitumor activity through Wnt2B/β-catenin pathway in head and neck squamous cell carcinomas. Cell Death Dis. 2020, 11, 672. [Google Scholar]
  216. Taiana, E.; Favasuli, V.; Ronchetti, D.; Todoerti, K.; Pelizzoni, F.; Manzoni, M.; Barbieri, M.; Fabris, S.; Silvestris, I.; Gallo Cantafio, M.E. Long non-coding RNA NEAT1 targeting impairs the DNA repair machinery and triggers anti-tumor activity in multiple myeloma. Leukemia 2020, 34, 234–244. [Google Scholar]
  217. Zhu, Z.; Du, S.; Yin, K.; Ai, S.; Yu, M.; Liu, Y.; Shen, Y.; Liu, M.; Jiao, R.; Chen, X. Knockdown long noncoding RNA nuclear paraspeckle assembly transcript 1 suppresses colorectal cancer through modulating miR-193a-3p/KRAS. Cancer Med. 2019, 8, 261–275. [Google Scholar] [CrossRef]
  218. Esposito, R.; Polidori, T.; Meise, D.F.; Pulido-Quetglas, C.; Chouvardas, P.; Forster, S.; Schaerer, P.; Kobel, A.; Schlatter, J.; Kerkhof, E. Multi-hallmark long noncoding RNA maps reveal non-small cell lung cancer vulnerabilities. Cell Genom. 2022, 2, 100171. [Google Scholar] [CrossRef] [PubMed]
  219. Tian, L.; Huang, Y.; Zhang, B.; Song, Y.; Yang, L.; Chen, Q.; Wang, Z.; Wang, Y.; He, Q.; Yang, W. Targeting LncRNA LLNLR-299G3. 1 with antisense oligonucleotide inhibits malignancy of esophageal squamous cell carcinoma cells in vitro and in vivo. Oncol. Res. 2023, 31, 463. [Google Scholar] [CrossRef] [PubMed]
  220. Gupta, R.A.; Shah, N.; Wang, K.C.; Kim, J.; Horlings, H.M.; Wong, D.J.; Tsai, M.-C.; Hung, T.; Argani, P.; Rinn, J.L. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010, 464, 1071–1076. [Google Scholar] [CrossRef]
  221. Zhao, H.; Wang, Y.; Hou, W.; Ding, X.; Wang, W. Long non-coding RNA MALAT1 promotes cell proliferation, migration and invasion by targeting miR-590-3p in osteosarcoma. Exp. Ther. Med. 2022, 24, 672. [Google Scholar] [CrossRef] [PubMed]
  222. Ma, X.; Zhang, W.; Zhao, M.; Li, S.; Jin, W.; Wang, K. Oncogenic role of lncRNA CRNDE in acute promyelocytic leukemia and NPM1-mutant acute myeloid leukemia. Cell Death Discov. 2020, 6, 121. [Google Scholar] [CrossRef]
  223. Zhang, C.; Wang, W.; Lin, J.; Xiao, J.; Tian, Y. lncRNA CCAT1 promotes bladder cancer cell proliferation, migration and invasion. Int. Braz. J. Urol. 2019, 45, 549–559. [Google Scholar] [CrossRef]
  224. Fang, H.; Liu, H.-M.; Wu, W.-H.; Liu, H.; Pan, Y.; Li, W.-J. Upregulation of long noncoding RNA CCAT1-L promotes epithelial–mesenchymal transition in gastric adenocarcinoma. OncoTargets Ther. 2018, 11, 5647–5655. [Google Scholar] [CrossRef]
  225. Chen, F.; Li, Y.; Feng, Y.; He, X.; Wang, L. Evaluation of antimetastatic effect of lncRNA-ATB siRNA delivered using ultrasound-targeted microbubble destruction. DNA Cell Biol. 2016, 35, 393–397. [Google Scholar] [CrossRef]
  226. Aguilar, R.; Spencer, K.B.; Kesner, B.; Rizvi, N.F.; Badmalia, M.D.; Mrozowich, T.; Mortison, J.D.; Rivera, C.; Smith, G.F.; Burchard, J. Targeting Xist with compounds that disrupt RNA structure and X inactivation. Nature 2022, 604, 160–166. [Google Scholar] [CrossRef]
  227. Abulwerdi, F.A.; Xu, W.; Ageeli, A.A.; Yonkunas, M.J.; Arun, G.; Nam, H.; Schneekloth Jr, J.S.; Dayie, T.K.; Spector, D.; Baird, N. Selective small-molecule targeting of a triple helix encoded by the long noncoding RNA, MALAT1. ACS Chem. Biol. 2019, 14, 223–235. [Google Scholar] [PubMed]
  228. Rossi, A.; Zacchi, F.; Reni, A.; Rota, M.; Palmerio, S.; Menis, J.; Zivi, A.; Milleri, S.; Milella, M. Progresses and Pitfalls of Epigenetics in Solid Tumors Clinical Trials. Int. J. Mol. Sci. 2024, 25, 11740. [Google Scholar] [CrossRef] [PubMed]
  229. Morel, D.; Jeffery, D.; Aspeslagh, S.; Almouzni, G.; Postel-Vinay, S. Combining epigenetic drugs with other therapies for solid tumours—Past lessons and future promise. Nat. Rev. Clin. Oncol. 2020, 17, 91–107. [Google Scholar] [CrossRef] [PubMed]
  230. Feehley, T.; O’Donnell, C.W.; Mendlein, J.; Karande, M.; McCauley, T. Drugging the epigenome in the age of precision medicine. Clin. Epigenetics 2023, 15, 6. [Google Scholar] [CrossRef]
Figure 1. Illustration of chromatin organization and various epigenetic mechanisms used by cells to control gene expression. These include DNA methylation, histone modifications, and noncoding RNAs (microRNAs).
Figure 1. Illustration of chromatin organization and various epigenetic mechanisms used by cells to control gene expression. These include DNA methylation, histone modifications, and noncoding RNAs (microRNAs).
Ddc 04 00015 g001
Figure 2. Outline of DNA methylation in normal and cancer cells. Epigenetic changes during carcinogenesis include (1) gain of DNA methylation (red triangles) at CpG islands, which is often located in gene promoter regions (CG hypermethylation) and is associated with epigenetic silencing of gene transcription; (2) global loss of DNA methylation (DNA hypomethylation, open circles) at intergenic regions and repetitive sequences. Both epigenetic alterations contribute to genomic instability, leading to tumorigenesis.
Figure 2. Outline of DNA methylation in normal and cancer cells. Epigenetic changes during carcinogenesis include (1) gain of DNA methylation (red triangles) at CpG islands, which is often located in gene promoter regions (CG hypermethylation) and is associated with epigenetic silencing of gene transcription; (2) global loss of DNA methylation (DNA hypomethylation, open circles) at intergenic regions and repetitive sequences. Both epigenetic alterations contribute to genomic instability, leading to tumorigenesis.
Ddc 04 00015 g002
Figure 3. Histone modifications, viz., methylation and acetylation function as master control switches that regulate the overall chromatin state (euchromatin/active state or heterochromatin/repressed), and thus, the expression of genes. Altered histone modification causes over-activation of oncogenes or suppression of tumor suppressor genes, thereby playing a crucial role in cancer development and progression.
Figure 3. Histone modifications, viz., methylation and acetylation function as master control switches that regulate the overall chromatin state (euchromatin/active state or heterochromatin/repressed), and thus, the expression of genes. Altered histone modification causes over-activation of oncogenes or suppression of tumor suppressor genes, thereby playing a crucial role in cancer development and progression.
Ddc 04 00015 g003
Figure 4. Role of microRNAs in cancer. microRNAs suppress the expression of the target gene. Depending on whether the target gene is a tumor suppressor gene or an oncogene, miRNAs either promote or block the tumorigenesis process. Image modified from Lee, S.W.L. et al., Journal of Controlled Release 313 (2019) 80-95 [83].
Figure 4. Role of microRNAs in cancer. microRNAs suppress the expression of the target gene. Depending on whether the target gene is a tumor suppressor gene or an oncogene, miRNAs either promote or block the tumorigenesis process. Image modified from Lee, S.W.L. et al., Journal of Controlled Release 313 (2019) 80-95 [83].
Ddc 04 00015 g004
Figure 5. Role of non-canonical DNA structures in inducing genomic instability and carcinogenesis.
Figure 5. Role of non-canonical DNA structures in inducing genomic instability and carcinogenesis.
Ddc 04 00015 g005
Figure 6. Inhibition of epigenetic modifications in cancer therapy. This involves targeting DNMT, histone modifying enzymes, and microRNAs with their specific inhibitors.
Figure 6. Inhibition of epigenetic modifications in cancer therapy. This involves targeting DNMT, histone modifying enzymes, and microRNAs with their specific inhibitors.
Ddc 04 00015 g006
Table 1. List of epi-drugs targeting DNMTs in cancer therapy.
Table 1. List of epi-drugs targeting DNMTs in cancer therapy.
Modification Epi-DrugTargetCancerClinical PhaseOther Drug(s)/Intervention(s)Outcome(s)/MechanismBrand Name/Trial ID/Reference(s)
DNMTDecitabine
(5-aza-2′-deoxycytidine)
DNMT1MDS, CMLFDA approved, 2020In combination with cedazuridineProlonged overall survivalINQOVI, Astex Pharmaceuticals, Inc. [118] NCT02103478 [119] NCT03306264 [120]
Decitabine DNMT1High risk MDS, CMLPhase 1/2 studyIn combination with cedazuridine and venetoclaxHigher response rate in short period of time; displays tolerable toxicity and satisfactory activityNCT04655755 [121]
Decitabine DNMT1MDSPhase 3-Higher overall response rate, prolonged survival; well-tolerated dosage, with a manageable toxicity profile[122]
5-fluoro-2′-deoxycytidine DNMT1Breast cancer and other solid tumorsPhase 2-Arrest cell cycle at G2/M phase; activates p53 signaling and DNA damage response pathway; upregulates tumor suppressor genes[123]
Guadecitabine (SGI-110)DNMT1AML, MDS, CMMLPhase 1/2-Well-tolerated with clinical and biological activityNCT01261312 [124,125,126,127,128]
Guadecitabine DNMT1AML, MDS, CMMLPhase 1/2In combination with AtezolizumabAcceptable tolerance and clinically activeNCT02935361 [124]
Guadecitabine DNMT1Platinum refractory germ cell cancerPhase 1 In combination with CisplatinExhibits good tolerance and demonstrates clinical activityNCT02429466 [129]
GuadecitabineDNMT1AMLPhase 1bIn combination with AtezolizumabLimited clinical activity and an overall unfavorable benefit–risk profile at tested dosesNCT02892318 [130]
GuadecitabineDNMT1Solid tumors (NSCLS)Phase 1In combination with PembrolizumabTolerable, with biological and anticancer activityNCT02998567 [131]
GuadecitabineDNMT1Colorectal cancerPhase 1In combination with Cy/GVAX (cyclophosphamide with GM-CSF secreting colon vaccine)Tolerable, but no significant immunologic activity NCT01966289 [132]
GuadecitabineDNMT1MelanomaPhase 1bIn combination with ipilimumab (anti-CTLA-4 antibody) Safe and tolerable, with initial signs of clinical and immunologic activityNCT0260843 [128,133]
GuadecitabineDNMT1SCLCPhase 2In combination with carboplatinExhibits good efficacy along with possible adverse eventsNCT03913455 [134]
GuadecitabineDNMT1Urothelial carcinomaPhase 2In combination with atezolizumabPossible prolonged patient survivalNCT03179943 [135]
GuadecitabineDNMT1Ovarian, Primary Peritoneal, or Fallopian Tube CancerPhase 2In combination with PembrolizumabExhibits clinical activity and possibly activates antitumor immunityNCT02901899 [136]
GuadecitabineDNMT1ChondrosarcomasPhase 2In combination with Belinostat or ASTX727Active trial; not recruitingNCT04340843 [137]
5-AzacitidineDNMT1MDS, AML, CMMLFDA approved, 2004; EMEA approved 2009-Satisfactory safety profile and provides clinical benefitVidaza®; Celgene Corp., Summit, NJ, USA [138]
MG98DNMT1Renal carcinomaPhase 2 (terminated)-Exhibits no anti-cancer activity[139]
MG98DNMT1Solid tumorsPhase 1-Shows early evidence of clinical activity with good toleranceNCT00003890 [140]
HydralazineDNMT1/3A/3BRefractory solid tumorsPhase 2In combination with Magnesium valproateShows clinical benefits with potential to overcome resistance to chemotherapyNCT00404508 [141]
HydralazineDNMT1/3A/3BHepatocellular carcinomaPhase 2In combination with valproic acid Exhibits manageable toxicity and clinical efficacyTPVGH97-07-07 [142]
HydralazineDNMT1/3A/3BBreast cancerPhase 2 (terminated)In combination with Magnesium valproateWell-tolerated and appears to increase the efficacy of chemotherapyNCT00395655 [143]
HydralazineDNMT1/3A/3BCervical cancerPhase 3In combination with Magnesium valproate; placebo-controlledUnknownNCT00532818 (Unpublished)
HydralazineDNMT1/3A/3BOvarian cancerPhase 3In combination with Magnesium valproate; placebo-controlledUnknownNCT00533299 (Unpublished)
Epigallocatechin-3-gallate (EGCG)DNMTColon cancer (HT-29 cells), esophageal cancer (KYSE 150 cells), and prostate cancer (PC3 cells)Preclinical phase-Reactivation of some methylation-silenced genes[144]
Resveratrol-salicylate derivativesDNMT3Colon cancer (HT-29), liver cancer (HepG2) and breast cancer (SK-BR-3)Preclinical phase-Unknown[145]
SGI-1027 (4-anilinoquinoline)DNMT1/3A/3BHepatocellular carcinoma (Huh7)Preclinical phase-Reactivates silenced tumor suppressor genes and induces apoptosis in cancer cells[146]
Laccaic acid A (LCA)DNMT1Breast cancer (MCF-7)Preclinical phase-Reactivates the expression of silenced tumor suppressor genes [147]
RG-108DNMT1Colorectal cancer (HCT116) and acute lymphoblastic leukemia (NALM-6)Preclinical phase-Reactivates key tumor suppressor genes [148]
Nanaomycin ADNMT3BLung cancer (A549), leukemia (HL60), and colorectal cancer (HCT116)Preclinical phase-Reactivates silenced tumor suppressor genes [149]
Table 2. List of epi-drugs targeting histone modifying enzymes in cancer therapy.
Table 2. List of epi-drugs targeting histone modifying enzymes in cancer therapy.
ModificationEpi-drugTargetCancerClinical PhaseOther drug(s)/Intervention(s)Outcome(s)/MechanismBrand Name/Trial ID/Reference(s)
HDACVorinostat (suberoylanilide hydroxamic acid)Pan HDACCutaneous T-cell lymphoma (CTCL)FDA approved 2006-Shows clinical benefitZolinza®; Merck & Co., Inc., Whitehouse Station, NJ [150]
BelinostatPan HDACPeripheral T cell lymphoma (PTCL)FDA approved 2006-Shows clinical benefitBeleodaq; Spectrum Pharmaceuticals, Inc. [151]
RomidepsinHDAC IT-cell lymphoma (both CTCL and PTCL)FDA approved-Shows clinical benefitIstodax, Celgene Corporation [152]
Pracinostat (SB939)HDAC I/II/IVLeukemia, solid tumorsPhase 1-UnknownNCT01184274 (Unpublished)
PracinostatHDAC I/II/IVSolid tumors, MDS, hematologic malignanciesPhase 1Alone or in combination with azacitidineSafe, with modest single-agent activityNCT00741234 [153]
PracinostatHDAC I/II/IVSolid malignanciesPhase 1-Well-tolerated and show inhibitory effectsSCS-PN0022 [154]
PracinostatHDAC I/II/IVSolid tumorsPhase 1-Shows good tolerability[155]
PracinostatHDAC I/II/IVMDSPhase 2In combination with azacitidine and decitabineExhibits improved efficacy and tolerance at reduced dosesNCT01993641 [156]
PracinostatHDAC I/II/IVMDSPhase 2In combination with azacitidineFails to improve outcomes with high rate of treatment discontinuationNCT01873703 [157]
PracinostatHDAC I/II/IVMyelofibrosisPhase 2-Exhibits modest tolerability and clinical activityNCT01200498 [158]
PracinostatHDAC I/II/IVTranslocation-associated sarcoma (TAS)Phase 2-Stopped prematurely due to prolonged unavailability of pracinostatNCT01112384 [159]
PracinostatHDAC I/II/IVAMLPhase 3In combination with azacitidineTerminated due to lack of clinical responseNCT03151408 [160]
Panobinostat (LBH-589)HDAC I/II/III/IVMultiple myelomaFDA and EMA approvedIn combination with bortezomib and dexamethasone Potential inhibitory activityFarydac® (Novartis) [161,162]
Resminostat HDAC I/IIb/IVBiliary tract or pancreatic cancerPhase 1In combination with chemotherapy (S1)Well-toleratedJapicCTI-152864 [163]
Resminostat HDAC I/IIb/IVCTCL, MFPhase 1-UnknownNCT04955340 (Unpublished)
Resminostat HDAC I/IIb/IVHodgkin lymphomaPhase 2-Exhibits acceptable tolerance and clinical activityNCT01037478 [164]
Resminostat HDAC I/IIb/IVHepatocellular carcinomaPhase 1/2In combination with sorafenibDisplay early signs of efficacy and well-toleratedNCT00943449 [165]
Resminostat HDAC I/IIb/IVNSCLCPhase 1/2In combination with docetaxelFails to improve progression-free survival and increases toxicityJapicCTI-132123 [166]
ResminostatHDAC I/IIb/IVHepatocellular carcinoma cellsPreclinical phaseIn combination with sorafenibBlocked platelet-induced hepatocellular carcinoma cell invasion [167]
QuisinostatHDAC I/IIb/IVSolid Malignancies and LymphomaPhase 1-Intermittent schedules show better tolerance than continuous schedulesNCT00677105 [168]
QuisinostatHDAC I/IIb/IVMultiple myelomaPhase 1In combination with dexamethasone and bortezonibShows clinical efficacy and safe toleranceNCT01464112 [169]
QuisinostatHDAC I/IIb/IVCTCLPhase 2-Shows an acceptable safety profileNCT01486277 [170]
CDX101HDAC ILymphoma or advanced solid organ cancers Phase 2a-Shows acceptable tolerance with clinical efficacy NCT01977638 [171]
CDX101HDAC IColorectal carcinomaPhase 2In combination with nivolumabWell tolerated and efficaciousNCT03993626 [172]
AbexinostatPan HDACNSCLC, melanoma, urothelial carcinoma, squamous cell carcinoma of head and neckPhase 1In combination with pembrolizumabUnknownNCT03590054 (Unpublished)
AbexinostatPan HDACMultiple myeloma, Hodgkin, and non-Hodgkin lymphomaPhase 1-UnknownNCT01149668 (Unpublished)
AbexinostatPan HDACSolid tumorsPhase 1 In combination with pazopanibShows good tolerability and anticancer effects NCT01543763 [173]
AbexinostatPan HDACFollicular lymphomaPhase 2-UnknownNCT03934567 NCT03600441
(Unpublished)
AbexinostatPan HDACNon-Hodgkin lymphomaPhase 1/2-UnknownNCT04024696 (Unpublished)
AbexinostatPan HDACFollicular lymphoma or mantle cell lymphoma.Phase 1/2-Well-tolerated and exhibits significant clinical activityNCT00724984 [174]
AbexinostatPan HDACSarcomaPhase 1/2In combination with doxorubicin and GCSFShows manageable toxicity and clinical responseNCT01027910 [175]
AbexinostatPan HDACCML, Hodgkin and non-Hodgkin lymphomaPhase 1/2-Exhibits manageable toxicity and partial clinical responseEudraCT 2009-013691-47 [176,177]
Chidamide (Epidaza)HDAC IPeripheral T-cell lymphomaApproved in China-Exhibits clinical benefit[178]
AR42Pan HDACNeurofibromatosis type 2-associated tumors and advanced solid malignanciesPhase 1-Safe and well-toleratedNCT01129193 [179]
AR42Pan HDACAMLPhase 1In combination with decitabineExhibits multi-organ failure as severe adverse effectNCT01798901 [180]
AR42Pan HDACMultiple myelomaPhase 1In combination with pomalidomideUnknownNCT02569320 (Unpublished)
AR42Pan HDACNeurofibromatosis type 2Phase 2/3 (recruiting)Placebo-controlledUnknownNCT05130866 (Unpublished)
EntinostatHDAC ICastration-resistant prostate cancerPhase 1In combination with enzalutamidShows acceptable safety profileNCT03829930 [181]
EntinostatHDAC IAcute leukemiasPhase 1-Effective inhibition of HDAC in vivoNCT00015925 [182]
EntinostatHDAC ISolid tumors and lymphoid malignanciesPhase 1-Shows good tolerability at the tested dosesNCT00020579 [183]
EntinostatHDAC IBreast cancerPhase 2In combination with exemestane; placebo-controlledShows acceptable safetyNCT03291886 [184]
EntinostatHDAC INSCLCPhase 1/2In combination with pembrolizumabExhibits clinical benefitNCT02437136 [185]
EntinostatHDAC IHR-positive breast cancerPhase 3In combination with exemestane; placebo-controlledUnknownNCT03538171 [186]
HDMGSK2879552LSD 1AMLPhase 1 (terminated)Alone or in combination with All-Trans Retinoic Acid (ATRA)Exhibits toxicity and adverse effectN CT02177812 (Unpublished)
GSK2879552LSD 1SCLCPhase 1 (terminated)-Shows many adverse effects with poor disease controlNCT02034123 [187]
INCB059872LSD1Solid Tumors and Hematologic MalignancyPhase 1/2 (terminated)Alone or in combination with ATRA, azacitidine, and nivolumabUnknown NCT02712905 [188]
TranylcypromineLSD1AMLPhase 1/2In combination with ATRAExhibits clinical response with acceptable toxicityNCT02261779 [189]
HMTTazemetostat EZH2Epithelioid sarcoma (ES), follicular lymphomaFDA approved 2020-Exhibits clinical benefitTAZVERIK, Epizyme, Inc.
MAK683EED/PRC2Diffuse large B-cell lymphoma (DLBCL) and epithelioid sarcoma (ES)Phase 1/2-Well-tolerated with observed clinical activity NCT02900651 [190]
Pinometostat (EPZ-5676)DOT1LHematologic malignancies, leukemia Phase 1/2-Shows inhibition of tumor growthNCT02141828, NCT01684150 [191]
GSK3326595PRMT5MDS, CMML, AMLPhase 1/2-Exhibits limited clinical activity NCT03614728 [192]
HATPU139 and PU141Pan HATNeuroblastomaPreclinical phase-PU139 blocks the HATs Gcn5, p300/CBP-associated factor (PCAF), CREB (cAMP response element-binding) protein (CBP) and p300, whereas PU141 is selective toward CBP and p300; Blocks growth of SK-N-SH neuroblastoma xenografts in mice[193]
Spiro Oxazolidinediones derivativesEP300/CBP histone acetyltransferase Lung squamous cell carcinoma cell line LK2-xenografted mouse modelPreclinical phase-Inhibits acetylation of H3K27 in the human lung cancer cell line LK2 [194]
DCH36_06 (thiobarbituric acid derivative)p300/CBPLeukemia cell linesPreclinical phase-Shows anti-tumor activity in leukemia xenograft[195]
C646 and its derivativesp300NSCLC (A549, H460 and H157 cells)Preclinical phase-Radio sensitization of NSCLC cells by enhancing mitotic catastrophe through the abrogation of G2 checkpoint maintenance[196]
EML425KAT3 (p300/CBP)Leukemia (U937 cells)Preclinical phase-Cell cycle arrest in G0/G1 phase [197]
A-485 p300/CBPHematological malignancies, androgen receptor-positive prostate cancerPreclinical phase-Shows potent anti-tumor activity[198]
Ubiquitin ligaseTosyl-L-arginine methyl ester (TAME)Anaphase-promoting complex (APC)-Preclinical stage-Induces mitotic arrest and cell death (xenopus cell extract)[199]
Nutlin, RG7112 (RO5045337)MDM2Advanced Solid Tumors and hematologic neoplasmsPhase 1-Inhibits p53-MDM2 interaction; induces cell cycle arrest, apoptosis, and inhibition or regression of human tumor xenograftsNCT00559533, NCT00623870 [200]
Idasanutlin, RG7388 (RO5503781)MDM2AMLPhase 3In combination with cytarabine; placebo-controlledExhibits good clinical response with acceptable toxicityNCT02545283 [201]
AMG 232MDM2Breast carcinoma, malignant solid tumor, multiple myelomaPhase 1-Exhibit adverse events, dose limiting toxicities, and clinically significant changes in vital signsNCT01723020 [202]
SAR405838MDM2Solid tumorsPhase 1In combination with pimasertibUnknownNCT01985191 (Unpublished)
Table 3. List of epi-drugs targeting small non-coding RNAs (microRNAs) in cancer therapy.
Table 3. List of epi-drugs targeting small non-coding RNAs (microRNAs) in cancer therapy.
Modification Epi-DrugTargetCancerClinical PhaseOther Drug(s)/Intervention(s)Outcome(s)/MechanismBrand Name/Trial ID/Reference(s)
miRNAMRG-106 (Cobomarsen)miR-155 CTCL, mycosis fungoides (MF) subtypePhase 2 (terminated)VorinostatUnknownNCT03713320 (Unpublished)
MRG-106miR-155Mycosis Fungoides MF, CLL, DLBCL or ATLLPhase 1-UnknownNCT02580552 (Unpublished)
MRG-106miR-155 CTCL, MF subtypePhase 1 (Terminated)-UnknownNCT03837457 (Unpublished)
TargomiRsmiR-16 Malignant pleural mesothelioma (MPM) and NSCLCPhase 1-Well-tolerated with early signs of clinical activityNCT02369198 [203]
MRX34miR-34aLiver cancer, SCLC, lymphoma, melanoma, MM, RCC, NSLCLPhase 1 (Terminated)-Exhibits serious adverse effectsNCT01829971 [204]
INT-1B3miR-193a-3pAdvanced solid tumorsPhase 1-Potential clinical benefitNCT04675996 [205]
MRG-110miR-92a-3p Healthy adultsPhase 1Placebo-controlledDe-repression miR-92a targetsNCT03494712 [206]
MRG-201miR-29bKeloidPhase 2Placebo-controlledExhibits clinical response and manageable adverse effectsNCT03601052 (Unpublished)
RGLS5579miR-10bGlioblastoma multiformePreclinical phase-Improved survival in animal model[207,208]
TTX-MC138miR-10bMetastatic breast cancerPreclinical phase-Shows early signs of clinical activity[209]
Table 4. List of epi-drugs targeting long non-coding RNAs in cancer therapy.
Table 4. List of epi-drugs targeting long non-coding RNAs in cancer therapy.
Drug ModuleTargetCancerReference
Antisense oligonucleotides (ASOs)MALAT1Lung cancer, breast cancer, multiple myeloma[210,211]
HOTAIRLung cancer[212]
TUG1Glioma, glioblastoma, pancreatic cancer[213,214]
AC104041.1Head and neck cancer[215]
NEAT1Neuroblastoma, multiple myeloma, colorectal cancer[216,217]
GCAWKR, CHiLL1Lung cancer[218]
LLNLR-299G3.1Esophageal cancer[219]
siRNAsHOTAIRBreast cancer[220]
MALAT1Lung, cervical, esophageal, and colorectal cancer, osteosarcoma, glioblastoma and lymphoma[112,221]
CRNDEAcute promyelocytic leukemia[222]
CCAT1Gastric, bladder and colorectal cancer[223,224]
lncRNA-ATBLiver cancer[225]
Small moleculesXISTBreast cancer[226]
MALAT1Breast cancer[227]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gupta, P. Epigenetic Alterations in Cancer: The Therapeutic Potential of Epigenetic Drugs in Cancer Therapy. Drugs Drug Candidates 2025, 4, 15. https://doi.org/10.3390/ddc4020015

AMA Style

Gupta P. Epigenetic Alterations in Cancer: The Therapeutic Potential of Epigenetic Drugs in Cancer Therapy. Drugs and Drug Candidates. 2025; 4(2):15. https://doi.org/10.3390/ddc4020015

Chicago/Turabian Style

Gupta, Preeti. 2025. "Epigenetic Alterations in Cancer: The Therapeutic Potential of Epigenetic Drugs in Cancer Therapy" Drugs and Drug Candidates 4, no. 2: 15. https://doi.org/10.3390/ddc4020015

APA Style

Gupta, P. (2025). Epigenetic Alterations in Cancer: The Therapeutic Potential of Epigenetic Drugs in Cancer Therapy. Drugs and Drug Candidates, 4(2), 15. https://doi.org/10.3390/ddc4020015

Article Metrics

Back to TopTop