Skip to main content
Journal of Oncology logoLink to Journal of Oncology
. 2023 Feb 9;2023:5449443. doi: 10.1155/2023/5449443

Hydroxymethylation and Epigenetic Drugs: New Insights into the Diagnosis and Treatment in Epigenetics of Hepatocellular Carcinoma

Wei Ouyang 1,2, Ming-Da Wang 2, Wan-Yin Wang 2,3, Chao Li 2, Lan-Qing Yao 2,3, Hong Zhu 1,, Tian Yang 2,3,4,
PMCID: PMC9934982  PMID: 36816356

Abstract

Hepatocellular carcinoma (HCC) is a highly lethal and heterogeneous malignancy with multiple genetic alternations and complex signaling pathways. The complexity and multifactorial nature of HCC pose a tremendous challenge regarding its diagnosis and treatment. Emerging evidence has indicated an important regulatory role of epigenetic modifications in HCC initiation and progression. Epigenetic modifications are stably heritable gene expression traits caused by changing the accessibility of chromatin structure and genetic activity without alteration in the DNA sequence and have been gradually recognized as a hallmark of cancer. In addition, accumulating data suggest a potential value of altered hydroxymethylation in epigenetic modifications and therapeutics targeting the epigenetically mediated regulation. As such, probing the epigenetic field in the era of precision oncology is a valid avenue for promoting the accuracy of early diagnosis and improving the oncological prognosis of HCC patients. This review focuses on the diagnostic performance and clinical utility of 5-hydroxymethylated cytosine, the primary intermediate product of the demethylation process, for early HCC diagnosis and discusses the promising applications of epigenetic-based therapeutic regimens for HCC.

1. Introduction

Hepatocellular carcinoma (HCC) represents the most frequent type of primary liver cancer and accounts for more than 90% of all liver cancer cases worldwide, with an annually increasing incidence and a dismal long-term prognosis [1, 2]. Epigenetic modifications contribute to the complexity and multifactorial nature of HCC as a significant mechanism, and molecular genetic alterations that affect epigenetic modification were reported to be critical factors in HCC carcinogenesis during the preneoplastic stage [3, 4]. The epigenetic regulation of chromatin consists of DNA methylation, nucleosome histone variants, post-translational histone modifications (PTMs), and noncoding RNAs [5]. Of these, DNA methylation is one of the most predominant research hotspots in epigenetics to date. Meanwhile, aberrant methylation processes and intermediates are also suggested to be essential hallmarks of HCC, with a great potential promise for early HCC diagnosis and therapeutic guidance [6].

Despite improvements in the early diagnosis and treatment of HCC, novel biomarkers for earlier diagnosis and better therapeutic interventions are urgently needed to improve long-term outcomes. 5-Hydroxymethylcytosine (5hmC), an intermediate product of the demethylation of 5-methylcytosine (5 mC) by ten-eleven translocation proteins, serves as an eminent epigenetic modification of DNA in the mammalian cells [7, 8]. It is also known as the “sixth base” of DNA [9, 10] and plays an essential role in gene regulation, cell development, and tumorigenesis [1116]. Facing and considering the unsatisfactory diagnostic accuracy of the traditional serum biomarkers in the early diagnosis of HCC and the suboptimal effectiveness of current systemic therapies for patients with advanced disease, it is of utmost importance to identify novel potential biomarkers and effective therapeutic strategies for different stage HCC [17, 18]. Recent advances in the high-throughput sequencing technologies (e.g., nano-hmC-Seal [19] and hMe-Seal [20]) have made it possible to uncover the genome-wide 5hmC profiling of hematological or solid tumors. The specific genomic distribution pattern of 5hmC revealed that this mark was highly enriched at promoters and enhancers of transcriptionally active genes [21]. An increasing amount of studies have indicated that the level of 5hmC in various solid tumors decreased significantly compared to adjacent tissues [20, 2231], suggesting an essential role of 5hmC in tumorigenesis and progression as well as its potential utility in tumor diagnosis. At the same time, there is an immense promise for exploring novel epigenetic biomarkers for cancer due to the limitations of the current traditional histopathology-based approaches for HCC detection in clinical practice. Moreover, given that epigenetic modifications, for example, reversible enzymatic reactions and specific protein-protein interactions (e.g., DNA methylation and PTM processes) are highly flexible and more susceptible to pharmacological interference, such novel strategies may pave new promising avenues toward therapeutic HCC [32].

Herein, we summarize the most recent progress in the diagnostic applications of 5hmC in HCC and evaluate its latent value of being a promising diagnostic biomarker for HCC, highlighting the emerging strategies of epigenetics-based targeted drugs in the era of HCC treatment.

2. 5hmC Serves as a Promising Early Diagnostic Biomarker in HCC

Paralleling the remaining solid tumors, previous studies have demonstrated that the 5hmC level was significantly decreased in an advanced cirrhosis and early HCC stage and was closely associated with poor prognosis and tumor progression [33, 34]. As such, 5hmC appears to be an impressive biomarker for early diagnosis and prognostic prediction of HCC (The flow diagram of 5hmC for HCC detection is shown in Figure 1). Several studies have proved the good diagnostic accuracy of 5hmC for detecting HCC, suggesting a potential prospect of clinical application. A previous study by Chen et al. [35] utilized a constructed mass spectrometer technique to examine 5hmC levels in HCC and revealed the possibility of 5hmC as a biomarker for early detection and prognosis of HCC. Cai et al. [29] established a 32-genes-based 5hmC diagnostic model using circulating cell-free DNA and exhibited a great performance for distinguishing early-stage HCC from non-HCC (training set: area under curve (AUC) = 0.92, 95% confidence interval (CI): 0.91–0.94; validation set: AUC = 0.88, 95%CI: 0.86–0.91), which appeared to outperform α-fetoprotein (AFP) when detecting an early HCC and may compensate for the plight of those patients with early HCC misdiagnosed due to AFP. Additionally, this model could be used to distinguish the patients with early small tumors (≤2 cm) accurately from high-risk patients with chronic liver disease (validation set: AUC = 0.85, 95%CI: 0.81–0.89), confirming the clinical application potential of 5hmC for the early detection of HCC. Another noninvasive diagnostic approach based on 5hmC signatures of plasma cell-free DNA effectively distinguished patients with HCC from cirrhotic patients and healthy controls with a relatively high AUC of 0.93 [36]. Song et al. [20] constructed a diagnostic model using the cell-free 5hmC signature with success in distinguishing HCC patients from hepatitis B virus infection and healthy controls, as well as monitoring treatment outcome and disease recurrence. Meanwhile, the distinct features of cell-free 5hmC yielded accurate predictions for specific cancer types and tumor stages. Given the limited number of studies regarding the mechanisms by which 5hmC regulates the pathogenesis of HCC [32], further basic research is still needed on HCC-related studies caused by 5hmC. Nevertheless, the current robust results about clinical applications of 5hmC as a molecular biomarker to guide the diagnosis of HCC and even for monitoring prognosis and recurrence are promising and advantageous compared to traditional biomarkers. The Cell-free 5hmC provides a novel dimension of informativeness for liquid biopsy-based diagnosis and surveillance.

Figure 1.

Figure 1

Circulating 5hmC as a novel biomarker of epigenetic modification for the early diagnosis of HCC. Small fragments of tumor DNA exist from the peripheral blood with an early-stage HCC, containing marks such as 5mC and 5hmC that are associated with epigenetic alterations in tumors. 5hmC as stable epigenetic marks associated with gene regulation and tumorigenesis, which is a staple of the demethylation process. Sequencing analysis of 5hmC revealed that 5hmC is markedly enriched in tissue-specific and tumor-specific differentially methylated regions, and discriminates between HCC and non-HCC patients with higher sensitivity and specificity versus traditional tumor biomarkers, suggesting a new strategy for the early diagnosis of tumors including HCC as a novel liquid biopsy modality. HCC, hepatocellular carcinoma; 5hmC, 5-hydroxymethylated cytosine; 5mC, 5-methylcytosine; C, cytosine; DNMT, DNA methyltransferases; TET, ten-eleven translocation.

3. Epidrugs: Targeting Epigenetic Marks in HCC Therapies

Intricate biological processes derived from aberrant gene regulation and epigenetic mutations have participated in developing HCC. It is well established that telomerase reverse transcriptase (TERT), Catenin β1 (CTNNB1), and TP53 are the most commonly mutated genes in association with the HCC development, yet the exploration of targeted therapies against these oncogenic driver genes genetic drivers remains unsuccessful [37, 38], highlighting the importance of developing new targeted therapeutics for patients with HCC. Following the evolution of high-throughput sequencing technologies and the accumulation of knowledge in the field of epigenetics, mutations in epigenetically modified genes have been indicated to be closely correlated with the development and progression of HCC, with up to 50% of tumors harboring relevant mutations [39]. Given this, research on epigenetic drugs (epidrugs) has received much interest and extensive attention in clinical practice [40].

Epidrugs are well-characterized small molecule inhibitors that mainly target epigenetic genes or enzymes and are divided into three categories: writers, readers, and erasers [32, 41]. Writers are enzymes that add covalent modifications to DNA and histones. They include DNA methyltransferases (DNMTs), which transfer methyl groups from S-adenosyl methionine (SAM) to cytosine bases of CpG dinucleotides at gene promoters and regulatory regions [42]. Histones are methylated on lysine and arginine residues to develop complex PTM. Catalytic enzymes in histone methylation involve histone methyltransferases (HMTs) and histone acetyltransferases (HATs), which exert a crucial impact on chromatin remodeling and gene expression [32]. Shanmugam et al. [43] further illustrated the link between aberrant epigenetic histone modifications and carcinogenesis and assessed their possible impacts on clinical outcomes of patients with HCC. Erasers (e.g., histone demethylases (HDMs) or histone deacetylases (HDACs), however, regulate DNA demethylation to reverse writers' functions. Moreover, epigenetic modifications are recognized by the third group of proteins named readers, the unique structural domains endowed with specific covalent modifications that function as effector proteins (e.g., methyl-binding domain proteins or Bromo- and extra-terminal (BETs) domain proteins) (Figure 2). The modification process described above emphasizes the complexity and reciprocal interaction of epigenetic regulatory mechanisms that underline the promising epidrugs.

Figure 2.

Figure 2

Schematic diagram: the regulation mechanism of DNA epigenetic modifiers on gene expression and epigenetic drugs. Epigenetic modifiers include epigenetic writers, readers, and erasers. HCC, hepatocellular carcinoma; 5hmC, 5-hydroxymethylated cytosine; 5mC, 5-methylcytosine; Epidrugs, epigenetic drugs; DNMTs, DNA methyltransferases; HATs, histone acetyltransferases; HMTs, histone methyltransferases; HDMs, histone demethylases; HDACs, histone deacetylases; BETs, Bromo- and extra-terminal; DNMTi, DNMT inhibitors; HDACi, HDAC inhibitors.

3.1. DNA Methylation Inhibitors

With epidrugs being recognized as a promising targeted therapeutic approach for treating and reversing cancer drug resistance, particularly notable for therapies with DNA methylation inhibitors and histone acetylation inhibitors, the following section emphasizes the previous methods. The current epidrugs have mainly been applied in hematological malignancies and exerted an anti-tumor effect via the inhibition of DNMTs [44] and HDACs [45], while seldom used to treat solid tumors due to the high rates of acquired drug resistance and lack of specific therapeutic targets. Either the first-generation DNMT inhibitors (DNMTi) (e.g., azacitidine [46] and Decitabine) or the second-generation DNMTi (e.g., guadecitabine (SGI-110)) developed to improve stability and overcome short-halflives, and HDAC inhibitors (HDACi) (e.g., vorinostat and panobinostat) appear to be widely applied in hematological tumors with the U.S. Food and Drug Administration approved. Additionally, the combination of other drugs in solid tumors has yielded greater anticancer effects than that induced by either drug alone [47], although there remain several mild adverse effects [48]. Emerging evidence suggests that DNMTi can be successfully applied in managing HCCs. Liu et al. [49] demonstrated that DNMTi significantly inhibited the colony formation of sorafenib-resistant HCC cells, indicating a therapeutic effect on resistant HCCs to sorafenib. Mei et al. [50] and Fan et al. [51] revealed that low-dose decitabine was effective in resensitizing resistant HCC cells to sorafenib alone or in combination with chemotherapy or immunotherapy in treating advanced HCC. Similarly, second-generation DNMTi is equally effective as low-dose guadecitabine alone or combined with oxaliplatin [52] or sorafenib [53], Gailhouste et al. [46] Also, DNMTi has therapeutic implications for HCC by promoting the reactivation of aberrantly silenced tumor suppressor genes, thereby enhancing sensitivity to sorafenib in HCC cells. As for combined therapies, DNMTi improved the efficacy of treatments such as chemotherapy, and equally, the combined immunotherapy modality holds advantages in treating HCC, benefiting from immunotherapy and improving outcomes [54]. Recently, a phase Ib clinical trial (NCT03257761) was conducted to evaluate the efficacy of guadecitabine in combination with durvalumab for treating gastrointestinal tumors, including HCC, and suggested the potential benefit of the combined therapy in selected patients. Furthermore, CM-272, a novel targeted dual-acting small molecule inhibitor of HMTs and DNMTs, exhibited potent anti-tumor activity against HCC cell lines by synergistically downregulating the expressions of DNMT1 and G9a [55], yet further clinical trials are needed to demonstrate its effectiveness and safety.

The above study found that low doses of DNMTi were effective in reducing the incidence of drug-related toxic effects, but common adverse events observed in the study were neutropenia, thrombocytopenia, anemia, nausea, and fatigue. Besides, neurological toxicity has been reported in a nonsmall cell lung cancer study with decitabine in combination with valproic acid [56].

3.2. HDAC Inhibitors

As aberrant histone deacetylation causes silencing of tumor suppressors in many of the known cancers, and research has shown aberrant expression of HDAC in HCC [57, 58], thus HDACi offers a promising approach to treat HCC. The pan-HDACi panobinostat [59] and pan-HDACi belinostat [60] have been proved to be effective treatment strategies for HCC by inhibiting the proliferative effect of HCC [61]. Other epigenetic therapies, such as Trichostatin (TSA) [62, 63] and Reminostat, are currently approved for clinical use and exhibit excellent anti-tumor effects in the HCC treatment [64]. Recent evidence demonstrated that targeting epigenetic modification strategies is capable of enhancing immune recognition of tumor cells hence the combinations of immunotherapy yield synergistic effects and induce robust anti-tumor responses [54, 65]. Immuno-combination therapies are a rapidly expanding field in targeting anti-tumor therapies, including HCC. The combinations of pan-HDACi belinostat with anti-CTLA-4 and anti-PD-1 antibodies have been studied to improve the anti-tumor efficacy of immune checkpoint inhibitors in a murine HCC model [66]. Correspondingly a multitude of clinical trials of epidrugs in conjunction with immune checkpoint inhibitors by HDACi are ongoing and are expected to yield implications for the clinical practice of immune conjugation strategies against HCC. Besides, therapeutic strategies including HDACi combined with other analogs also have been suggested to have anti-HCC potential and warrant further validation (A summary of the clinical stages of the different epidrugs in the treatment of HCC is presented in Table 1).

Table 1.

List of the various clinical phases of epigenetic drugs in the treatment of HCC.

Epigenetic drugs Preclinical stage Clinical trials Ref./clinical trial number Clinical trial start
Mechanism of action
DNMT inhibitors
 Azacitidine [46]
 Decitabine Phase I/II (NCT01799083) Dec 2012
 CM-272 [55]
 Guadecitabine (SGI-110) Phase II (NCT01752933) Dec 2012
Phase I (NCT03257761) Feb 2018
HDAC inhibitors
 Vorinostat Phase I (NCT01075113) Aug 2010
 Belinostat Phase I/II (NCT00321594) May 2006
 Panobinostat Phase I (NCT00873002) Mar 2009
Phase 1 (NCT00823290) Jan 2009
 Trichostatin [61, 62]
 Reminostat Phase II (NCT00943449) Jul 2009
Phase I/II (NCT02400788) Apr 2013

Abbreviations; DNMT : DNA methyltransferases; HDAC: histone deacetylases.

Regardless of the positive anti-tumor efficacy of HDACi against HCC, adverse side effects associated with HDACi deserve to be taken into account, notably when HDACi is used in combination with a variety of therapeutic drugs. Hepatic impairment mainly due to cumulative dose toxicity of the drug has been observed in clinical trials, including hyperbilirubinemia, elevated liver enzymes, and other dominant toxicities comprising fatigue, abdominal pain, anemia, and vomiting.

4. Conclusion

Aberrant epigenetic alterations are implicated in the pathogenesis of HCC. Epigenetic modifications include DNA methylation, hydroxymethylation, histone modifications, which can exert the differential expression of the genome and chromatin at the cellular transcriptome level. The development of high-throughput sequencing technologies has revealed a genome-wide map of 5hmC and low levels of 5hmC in the context of an early-stage HCC and associated with HCC progression, exploring it as a biomarker to serve in the field of diagnosis. Identification of 5hmC levels by liquid biopsy improves the diagnostic accuracy of HCC, making it possible to detect HCC earlier in large high-risk populations. Furthermore, with the growing establishment of epigenetic markers for the diagnosis and prognosis of solid tumors, epigenomic-targeted therapies may provide more combination strategies for treating HCC in the near future. Particularly, DNMTi and HDACi have been well-tested alone or in combination with other categories of drugs for treating HCC. Subsequent exploration of epigenetic modifications, including abnormal DNA methylations and histone modifications, is warranted to ascertain potential biomarkers for HCC diagnosis and formulate effective combined treatment strategies on the basis of epigenetic modification inhibitors in an attempt to overcome adverse effects and improve anti-tumor efficacy with better pharmacodynamics. However, it is notable that the lack of approved epidrugs available in the domain of HCC to date, hopefully the solutions offered to address the limitations referred therein will yield the optimal results in future. In conclusion, it is worthwhile to work towards a better comprehension of the mechanisms of epigenetic modifications and the interactions of epigenetic modifiers as a means to develop other more effective biomarkers and epigenomic therapies in the clinical setting.

Acknowledgments

This study was supported by the National Natural Science Foundation of China (No. 81972726, 81871949, and 81572345), Dawn Project Foundation of Shanghai (No. 21SG36), and Adjunct Talent Fund of Zhejiang Provincial People's Hospital (No. 2021-YT), and Jiangsu Six Talent Peaks Project (WSN-102), and Shanghai Science and Technology Committee Rising-Star Program (No. 22QA1411600).

Contributor Information

Hong Zhu, Email: zhuhong_jasmine@suda.edu.cn.

Tian Yang, Email: yangtianehbh@smmu.edu.cn.

Data Availability

The data supporting this mini-review are from previously reported studies and datasets, which have been cited. The processed data are available from the corresponding author.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors' Contributions

Writing-original draft preparation was contributed by Wei Ouyang; writing-review and editing were contributed by Ming-Da Wang and Wan-Yin Wang; figures were prepared by Chao Li and Lan-Qing Yao; supervision was conducted by Hong Zhu and Tian Yang; Wei Ouyang and Ming-Da Wang are equal contributors and co-first authors.

References

  • 1.Forner A., Reig M., Bruix J. Hepatocellular carcinoma. The Lancet . 2018;391(10127):1301–1314. doi: 10.1016/s0140-6736(18)30010-2. [DOI] [PubMed] [Google Scholar]
  • 2.Villanueva A. Hepatocellular carcinoma. New England Journal of Medicine . 2019;380(15):1450–1462. doi: 10.1056/nejmra1713263. [DOI] [PubMed] [Google Scholar]
  • 3.Erkekoglu P., Oral D., Chao M. W., Kocer-Gumusel B. Hepatocellular carcinoma and possible chemical and biological causes: a review. Journal of Environmental Pathology, Toxicology and Oncology . 2017;36(2):171–190. doi: 10.1615/jenvironpatholtoxicoloncol.2017020927. [DOI] [PubMed] [Google Scholar]
  • 4.Bouyahya A., Mechchate H., Oumeslakht L., et al. The role of epigenetic modifications in human cancers and the use of natural compounds as epidrugs: mechanistic pathways and pharmacodynamic actions. Biomolecules . 2022;12(3):p. 367. doi: 10.3390/biom12030367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cavalli G., Heard E. Advances in epigenetics link genetics to the environment and disease. Nature . 2019;571(7766):489–499. doi: 10.1038/s41586-019-1411-0. [DOI] [PubMed] [Google Scholar]
  • 6.Arechederra M., Recalde M., Gárate-Rascón M., Fernández-Barrena M. G., Ávila M. A., Berasain C. Epigenetic biomarkers for the diagnosis and treatment of liver disease. Cancers . 2021;13(6):p. 1265. doi: 10.3390/cancers13061265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Tahiliani M., Koh K. P., Shen Y., et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science . 2009;324(5929):930–935. doi: 10.1126/science.1170116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ito S., Shen L., Dai Q., et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science . 2011;333(6047):1300–1303. doi: 10.1126/science.1210597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wu X., Zhang Y. TET-mediated active DNA demethylation: mechanism, function and beyond. Nature Reviews Genetics . 2017;18(9):517–534. doi: 10.1038/nrg.2017.33. [DOI] [PubMed] [Google Scholar]
  • 10.Li W., Xu L. Epigenetic function of TET family, 5-methylcytosine, and 5-hydroxymethylcytosine in hematologic malignancies. Oncology research and treatment . 2019;42(6):309–318. doi: 10.1159/000498947. [DOI] [PubMed] [Google Scholar]
  • 11.Portela A., Esteller M. Epigenetic modifications and human disease. Nature Biotechnology . 2010;28(10):1057–1068. doi: 10.1038/nbt.1685. [DOI] [PubMed] [Google Scholar]
  • 12.Haffner M. C., Chaux A., Meeker A. K., et al. Global 5-hydroxymethylcytosine content is significantly reduced in tissue stem/progenitor cell compartments and in human cancers. Oncotarget . 2011;2(8):627–637. doi: 10.18632/oncotarget.316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Song C. X., Clark T. A., Lu X. Y., et al. Sensitive and specific single-molecule sequencing of 5-hydroxymethylcytosine. Nature Methods . 2011;9(1):75–77. doi: 10.1038/nmeth.1779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Song C. X., Szulwach K. E., Fu Y., et al. Selective chemical labeling reveals the genome-wide distribution of 5-hydroxymethylcytosine. Nature Biotechnology . 2011;29(1):68–72. doi: 10.1038/nbt.1732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Prins G. S., Ye S. H., Birch L., et al. Prostate cancer risk and DNA methylation signatures in aging rats following developmental bpa exposure: a dose-response analysis. Environmental Health Perspectives . 2017;125(7) doi: 10.1289/ehp1050.077007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wan J. C. M., Massie C., Garcia-Corbacho J., et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nature Reviews Cancer . 2017;17(4):223–238. doi: 10.1038/nrc.2017.7. [DOI] [PubMed] [Google Scholar]
  • 17.Altekruse S. F., McGlynn K. A., Dickie L. A., Kleiner D. E. Hepatocellular carcinoma confirmation, treatment, and survival in surveillance, epidemiology, and end results registries, 1992-2008. Hepatology . 2012;55(2):476–482. doi: 10.1002/hep.24710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ayoub W. S., Steggerda J., Yang J. D., Kuo A., Sundaram V., Lu S. C. Current status of hepatocellular carcinoma detection: screening strategies and novel biomarkers. Therapeutic advances in medical oncology . 2019;11 doi: 10.1177/1758835919869120.1758835919869120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Han D., Lu X., Shih A. H., et al. A highly sensitive and robust method for genome-wide 5hmC profiling of rare cell populations. Molecular Cell . 2016;63(4):711–719. doi: 10.1016/j.molcel.2016.06.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Song C. X., Yin S., Ma L., et al. 5-Hydroxymethylcytosine signatures in cell-free DNA provide information about tumor types and stages. Cell Research . 2017;27(10):1231–1242. doi: 10.1038/cr.2017.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Smeets E., Lynch A. G., Prekovic S., et al. The role of TET-mediated DNA hydroxymethylation in prostate cancer. Molecular and Cellular Endocrinology . 2018;462:41–55. doi: 10.1016/j.mce.2017.08.021. [DOI] [PubMed] [Google Scholar]
  • 22.Jin S. G., Jiang Y., Qiu R., et al. 5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations. Cancer Research . 2011;71(24):7360–7365. doi: 10.1158/0008-5472.can-11-2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lian C. G., Xu Y., Ceol C., et al. Loss of 5-hydroxymethylcytosine is an epigenetic hallmark of melanoma. Cell . 2012;150(6):1135–1146. doi: 10.1016/j.cell.2012.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yang H., Liu Y., Bai F., et al. Tumor development is associated with decrease of TET gene expression and 5-methylcytosine hydroxylation. Oncogene . 2013;32(5):663–669. doi: 10.1038/onc.2012.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pfeifer G. P., Xiong W., Hahn M. A., Jin S. G. The role of 5-hydroxymethylcytosine in human cancer. Cell and Tissue Research . 2014;356(3):631–641. doi: 10.1007/s00441-014-1896-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Park J. L., Kim H. J., Seo E. H., et al. Decrease of 5hmC in gastric cancers is associated with TET1 silencing due to with DNA methylation and bivalent histone marks at TET1 CpG island 3’-shore. Oncotarget . 2015;6(35):37647–37662. doi: 10.18632/oncotarget.6069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chen K., Zhang J., Guo Z., et al. Loss of 5-hydroxymethylcytosine is linked to gene body hypermethylation in kidney cancer. Cell Research . 2016;26(1):103–118. doi: 10.1038/cr.2015.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bonvin E., Radaelli E., Bizet M., et al. TET2-Dependent hydroxymethylome plasticity reduces melanoma initiation and progression. Cancer Research . 2019;79(3):482–494. doi: 10.1158/0008-5472.can-18-1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cai J., Chen L., Zhang Z., et al. Genome-wide mapping of 5-hydroxymethylcytosines in circulating cell-free DNA as a non-invasive approach for early detection of hepatocellular carcinoma. Gut . 2019;68(12):2195–2205. doi: 10.1136/gutjnl-2019-318882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kristensen G., Strand S. H., Røder M. A., et al. 5hmC level predicts biochemical failure following radical prostatectomy in prostate cancer patients with ERG negative tumors. International Journal of Molecular Sciences . 2019;20(5):p. 1025. doi: 10.3390/ijms20051025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang J., Su Y., Tian Y., Ding Y., Wang X. Characterization of DNA hydroxymethylation profile in cervical cancer. Artificial Cells, Nanomedicine, and Biotechnology . 2019;47(1):2706–2714. doi: 10.1080/21691401.2019.1634578. [DOI] [PubMed] [Google Scholar]
  • 32.Fernández-Barrena M. G., Arechederra M., Colyn L., Berasain C., Avila M. A. Epigenetics in hepatocellular carcinoma development and therapy: the tip of the iceberg. JHEP Reports . 2020;2(6) doi: 10.1016/j.jhepr.2020.100167.100167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hlady R. A., Sathyanarayan A., Thompson J. J., et al. Integrating the epigenome to identify drivers of hepatocellular carcinoma. Hepatology . 2019;69(2):639–652. doi: 10.1002/hep.30211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Liu J., Jiang J., Mo J., et al. Global DNA 5-hydroxymethylcytosine and 5-formylcytosine contents are decreased in the early stage of hepatocellular carcinoma. Hepatology . 2019;69(1):196–208. doi: 10.1002/hep.30146. [DOI] [PubMed] [Google Scholar]
  • 35.Chen M. L., Shen F., Huang W., et al. Quantification of 5-methylcytosine and 5-hydroxymethylcytosine in genomic DNA from hepatocellular carcinoma tissues by capillary hydrophilic-interaction liquid chromatography/quadrupole TOF mass spectrometry. Clinical Chemistry . 2013;59(5):824–832. doi: 10.1373/clinchem.2012.193938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Cai Z., Zhang J., He Y., et al. Liquid biopsy by combining 5-hydroxymethylcytosine signatures of plasma cell-free DNA and protein biomarkers for diagnosis and prognosis of hepatocellular carcinoma. ESMO open . 2021;6(1) doi: 10.1016/j.esmoop.2020.100021.100021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zucman-Rossi J., Villanueva A., Nault J. C., Llovet J. M. Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology . 2015;149(5):1226–1239.e4. doi: 10.1053/j.gastro.2015.05.061. [DOI] [PubMed] [Google Scholar]
  • 38.Dhanasekaran R., Nault J. C., Roberts L. R., Zucman-Rossi J. Genomic medicine and implications for hepatocellular carcinoma prevention and therapy. Gastroenterology . 2019;156(2):492–509. doi: 10.1053/j.gastro.2018.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Schulze K., Imbeaud S., Letouzé E., et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nature Genetics . 2015;47(5):505–511. doi: 10.1038/ng.3252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ganesan A., Arimondo P. B., Rots M. G., Jeronimo C., Berdasco M. The timeline of epigenetic drug discovery: from reality to dreams. Clinical Epigenetics . 2019;11(1):p. 174. doi: 10.1186/s13148-019-0776-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cossío F. P., Esteller M., Berdasco M. Towards a more precise therapy in cancer: exploring epigenetic complexity. Current Opinion in Chemical Biology . 2020;57:41–49. doi: 10.1016/j.cbpa.2020.04.008. [DOI] [PubMed] [Google Scholar]
  • 42.Stallcup M. R. Role of protein methylation in chromatin remodeling and transcriptional regulation. Oncogene . 2001;20(24):3014–3020. doi: 10.1038/sj.onc.1204325. [DOI] [PubMed] [Google Scholar]
  • 43.Shanmugam M. K., Arfuso F., Arumugam S., et al. Role of novel histone modifications in cancer. Oncotarget . 2018;9(13):11414–11426. doi: 10.18632/oncotarget.23356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Nagaraju G. P., Dariya B., Kasa P., Peela S., El-Rayes B. F. Epigenetics in hepatocellular carcinoma. Seminars in Cancer Biology . 2022;86:622–632. doi: 10.1016/j.semcancer.2021.07.017. [DOI] [PubMed] [Google Scholar]
  • 45.Rodríguez-Paredes M., Esteller M. Cancer epigenetics reaches mainstream oncology. Nature medicine . 2011;17(3):330–339. doi: 10.1038/nm.2305. [DOI] [PubMed] [Google Scholar]
  • 46.Gailhouste L., Liew L. C., Yasukawa K., et al. Differentiation therapy by epigenetic reconditioning exerts antitumor effects on liver cancer cells. Molecular Therapy . 2018;26(7):1840–1854. doi: 10.1016/j.ymthe.2018.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pili R., Liu G., Chintala S., et al. Combination of the histone deacetylase inhibitor vorinostat with bevacizumab in patients with clear-cell renal cell carcinoma: a multicentre, single-arm phase I/II clinical trial. British Journal of Cancer . 2017;116(7):874–883. doi: 10.1038/bjc.2017.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Falchook G. S., Fu S., Naing A., et al. Methylation and histone deacetylase inhibition in combination with platinum treatment in patients with advanced malignancies. Investigational New Drugs . 2013;31(5):1192–1200. doi: 10.1007/s10637-013-0003-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Liu J., Liu Y., Meng L., Liu K., Ji B. Targeting the PD-L1/DNMT1 axis in acquired resistance to sorafenib in human hepatocellular carcinoma. Oncology Reports . 2017;38(2):899–907. doi: 10.3892/or.2017.5722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Mei Q., Chen M., Lu X., et al. An open-label, single-arm, phase I/II study of lower-dose decitabine based therapy in patients with advanced hepatocellular carcinoma. Oncotarget . 2015;6(18):16698–16711. doi: 10.18632/oncotarget.3677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Fan H., Lu X., Wang X., et al. Low-dose decitabine-based chemoimmunotherapy for patients with refractory advanced solid tumors: a phase I/II report. Journal of immunology research . 2014;2014:14. doi: 10.1155/2014/371087.371087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kuang Y., El-Khoueiry A., Taverna P., Ljungman M., Neamati N. Guadecitabine (SGI-110) priming sensitizes hepatocellular carcinoma cells to oxaliplatin. Molecular oncology . 2015;9:1799–1814. doi: 10.1016/j.molonc.2015.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Jueliger S., Lyons J., Cannito S., et al. Efficacy and epigenetic interactions of novel DNA hypomethylating agent guadecitabine (SGI-110) in preclinical models of hepatocellular carcinoma. Epigenetics . 2016;11(10):709–720. doi: 10.1080/15592294.2016.1214781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Villanueva L., Álvarez-Errico D., Esteller M. The contribution of epigenetics to cancer immunotherapy. Trends in Immunology . 2020;41(8):676–691. doi: 10.1016/j.it.2020.06.002. [DOI] [PubMed] [Google Scholar]
  • 55.Bárcena-Varela M., Caruso S., Llerena S., et al. Dual targeting of histone methyltransferase G9a and DNA-methyltransferase 1 for the treatment of experimental hepatocellular carcinoma. Hepatology . 2019;69(2):587–603. doi: 10.1002/hep.30168. [DOI] [PubMed] [Google Scholar]
  • 56.Chu B. F., Karpenko M. J., Liu Z., et al. Phase I study of 5-aza-2’-deoxycytidine in combination with valproic acid in non-small-cell lung cancer. Cancer Chemotherapy and Pharmacology . 2013;71(1):115–121. doi: 10.1007/s00280-012-1986-8. [DOI] [PubMed] [Google Scholar]
  • 57.Haberland M., Montgomery R. L., Olson E. N. The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nature Reviews Genetics . 2009;10(1):32–42. doi: 10.1038/nrg2485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ler S. Y., Leung C. H. W., Khin L. W., et al. HDAC1 and HDAC2 independently predict mortality in hepatocellular carcinoma by a competing risk regression model in a Southeast Asian population. Oncology Reports . 2015;34(5):2238–2250. doi: 10.3892/or.2015.4263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Gahr S., Mayr C., Kiesslich T., et al. The pan-deacetylase inhibitor panobinostat affects angiogenesis in hepatocellular carcinoma models via modulation of CTGF expression. International Journal of Oncology . 2015;47(3):963–970. doi: 10.3892/ijo.2015.3087. [DOI] [PubMed] [Google Scholar]
  • 60.Yeo W., Chung H. C., Chan S. L., et al. Epigenetic therapy using belinostat for patients with unresectable hepatocellular carcinoma: a multicenter phase I/II study with biomarker and pharmacokinetic analysis of tumors from patients in the Mayo Phase II Consortium and the Cancer Therapeutics Research Group. Journal of Clinical Oncology . 2012;30(27):3361–3367. doi: 10.1200/jco.2011.41.2395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Toh T. B., Lim J. J., Chow E. K. H. Epigenetics of hepatocellular carcinoma. Clinical and Translational Medicine . 2019;8(1):p. 13. doi: 10.1186/s40169-019-0230-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kong W. Y., Yee Z. Y., Mai C. W., Fang C. M., Abdullah S., Ngai S. C. Zebularine and trichostatin A sensitized human breast adenocarcinoma cells towards tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-induced apoptosis. Heliyon . 2019;5(9) doi: 10.1016/j.heliyon.2019.e02468.e02468 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sanaei M., Kavoosi F. Effect of zebularine in comparison to trichostatin A on the intrinsic and extrinsic apoptotic pathway, cell viability, and apoptosis in hepatocellular carcinoma SK-hep 1, human colorectal cancer SW620, and human pancreatic cancer PaCa-44 cell lines. Iranian Journal of Pharmaceutical Research: Iranian Journal of Pharmaceutical Research . 2021;20(3):310–323. doi: 10.22037/ijpr.2021.115097.15196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tsilimigras D. I., Ntanasis-Stathopoulos I., Moris D., Spartalis E., Pawlik T. M. Histone deacetylase inhibitors in hepatocellular carcinoma: a therapeutic perspective. Surgical oncology . 2018;27(4):611–618. doi: 10.1016/j.suronc.2018.07.015. [DOI] [PubMed] [Google Scholar]
  • 65.Cao J., Yan Q. Cancer epigenetics, tumor immunity, and immunotherapy. Trends in cancer . 2020;6(7):580–592. doi: 10.1016/j.trecan.2020.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Llopiz D., Ruiz M., Villanueva L., et al. Enhanced anti-tumor efficacy of checkpoint inhibitors in combination with the histone deacetylase inhibitor Belinostat in a murine hepatocellular carcinoma model. Cancer Immunology, Immunotherapy . 2019;68:379–393. doi: 10.1007/s00262-018-2283-0. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data supporting this mini-review are from previously reported studies and datasets, which have been cited. The processed data are available from the corresponding author.


Articles from Journal of Oncology are provided here courtesy of Wiley

RESOURCES