- Review
- Open access
- Published:
Advances in epigenetic treatment of adult T-cell leukemia/lymphoma: a comprehensive review
Clinical Epigenetics volume 17, Article number: 39 (2025)
Abstract
Human T-cell lymphotropic virus type 1 (HTLV-1) infection causes the uncommon and deadly cancer known as adult T-cell leukemia/lymphoma (ATLL), which affects mature T cells. Its clinical appearance is varied, and its prognosis is often miserable. Drug resistance to conventional therapies confers significant therapeutic challenges in the management of ATLL. This review discusses the emerging role of epigenetic medical advances in the treatment of ATLL, focusing on DNA methyltransferase inhibitors, histone deacetylase inhibitors, histone methyltransferase inhibitors, and BET inhibitors. Indeed, several classes of epigenetic therapies currently exhibit trailed efficacy in preclinical and clinical studies: DNA methyltransferase inhibitors like azacitidine and decitabine reexpression of silenced tumor suppressors; histone deacetylase inhibitors like vorinostat and romidepsin induce cell cycle arrest and apoptosis; bromodomain and extra-terminal inhibitors like JQ1 disrupt oncogenic signaling pathways. Whereas preclinical and early clinical data indicate modest to good efficacy for such treatments, significant challenges remain. Here, we discuss the current state of understanding of epigenetic dysregulation in ATLL and appraise the evidence supporting the use of these epi-drugs. However, despite the opened doors of epigenetic treatment, much more research is required with regard to showing the best combinations of drugs and their resistance mechanisms, the minimization of adverse effects, and how this hope will eventually be translated into benefit for the patient with ATLL.
Introduction
The complex retrovirus known as HTLV-1, or human T-lymphotropic virus type 1, mainly infects CD4+ T cells, although it can also infect dendritic cells, endothelial cells, monocytes, and CD8+ cells. Its genome includes pol, env, pro, gag, and a number of essential genes, including those in the pX region. Six viral auxiliary proteins—Basic Zipper Factor (HBZ), p13II/p8, p30II, p12I, Tax, and Rex—are encoded by these genes. HTLV-1 infects many organs and is spread through unprotected intercourse, injectable medication usage, organ transplants, and blood transfusions. Adult T-cell leukemia/lymphoma (ATLL) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP) are two serious illnesses brought on by HTLV-1 (Fig. 1) [1, 2]. There are four subtypes of ATLL, a CD4+ T-cell malignancy: acute, chronic, smoldering, and lymphoma. The most prevalent and severe variety is the acute form. ATLL presents with non-Hodgkin’s lymphoma-like symptoms (e.g., fever, lymphadenopathy, organomegaly, jaundice, fatigue, weight loss, and infections) and skin manifestations. They fall into six groups: purpuric, erythrodermic, patch, plaque, multipapular, and nodulotumoral. These skin symptoms have predictive value and may be the initial indication of the illness (Fig. 1) [3,4,5,6].
Some of the current treatment approaches for ATLL include targeting surface molecules, monoclonal antibodies, interferon therapy, chemotherapy, and stem cell transplantation [1].
Much importance has been given to the study of epigenetic changes in cancer, a field arising from several interdisciplinary interests [7].
Nonetheless, epigenetic changes are reversible heritable changes induced by environmental factors. As such, they play a critical role in tumor development or are promising targets for therapies [8].
External factors drive epigenetic changes and reversible modifications and represent the main driving force underlying tumorigenesis; thus, they constitute an appealing target for therapy. “Epi-drugs” aimed at targeting these alterations have been developed during the last 40 years and reached clinical trials with some success, leading to FDA approval of several types. These treatments can affect energy metabolism, affecting several genes and proteins, and induce cell differentiation, cell cycle arrest, and cell death. Since epigenetic modifications are essentially reversible, they are great therapeutic intervention possibilities [8,9,10,11].
The use of combination treatments of epi-drugs with chemotherapy or immunotherapy has so far resulted in improved tumor remission, reduced chemoresistance, increased life expectancy, and decreased side effects for patients. Epi-drugs are helpful as a single treatment or in combination because they may improve anti-tumor effects, overcome drug resistance, and activate the tumor's immune response. This method can also make low dosages of standard chemotherapy possible because of a further consequent reduction in side effects and an increase in the quality of patients' lives [8,9,10].
Epigenetic therapy efficiently acts as a treatment strategy for various malignancies [7].
After establishing the importance of epigenetic mechanisms in the pathogenesis of diseases, we focus on the therapeutic role of epigenetic drugs in the context of ATLL. Next, certain epigenetic agents that have shown efficiency in targeting ATLL cells and modifying disease outcomes will be discussed.
Epigenetic alternation in ATLL
Epigenetic mechanisms such as DNA methylation, histone modifications, nucleosome positioning, and chromatin looping play pivotal roles in regulating HTLV-1’s transcription and contributing to its pathogenesis [12].
EP300 and its homolog CBP transcriptionally co-activate genes involved in various cellular functions, such as DNA repair, apoptosis, differentiation, and proliferation. Most recently, Northern American individuals with adult T-cell leukemia/lymphoma (ATLL) were found to have somatic hypermutations in EP300 (but not CBP) [13].
Epigenetic regulator mutations are more common in North American ATLL (NA-ATLL), especially in EP300, which is mutated in 20% of patients [14]. Reduced p53 levels are linked to EP300 mutations [15].
These mutations cause ATLL cells to experience replication stress and genomic instability, particularly during S-phase [13].
Additionally, research has shown that ATLL patients had hypermethylation in several cell cycle regulatory genes, such as p15INK4b, p16INK4a, and p14ARF [16].
Epigenetic changes contribute significantly to the development of ATLL by deactivating tumor suppressor genes, which are crucial for preserving genetic and genomic integrity. These changes impair cell adhesion, DNA damage response, apoptosis mechanisms, and control over cellular proliferation [17].
Tax interacts with methyl-CpG-binding domain 2 (MBD2) to stimulate transcription from a highly methylated HTLV-1 LTR [18].
HTLV-1 provirus represents hypomethylation within the plus-strand promoter for the viral proteins that are expressed with Tax [19].
Histone modifications, such as H3K4me3, H3K9Ac, and H3K27Ac, are dynamic and highly correlated with the expression of HTLV-1 plus-strand. These histone modifications are all linked to transcriptional repression and chromatin condensation, contributing to the silencing of genes that would typically limit cell proliferation, promote apoptosis, or maintain genomic stability; therefore, these histone modifications participate in the regulation of viral transcription [12, 19, 20].
One of the modifications often reprogrammed in ATLL cells is the trimethylation at histone H3Lys27 via polycomb-repressive complex 2, which influences regulation at the level of gene expression and progresses the disease [21].
Epigenetic treatment
The field of pharmaco-epigenomics is also developing at present, considering the targeting of epigenetic marks for therapy with implications in cancer [9].
This therapeutic strategy can be used either alone or in conjunction with other therapies, such as immunotherapy or chemotherapy, to improve the anti-tumor effect, overcome medication resistance, and stimulate immune responses [10, 22].
Patients with ATLL in North America have a unique genomic landscape with frequent epigenetic alterations, especially in EP300, which are linked to a poor prognosis [14].
In preclinical and clinical trials, epigenetic treatments for ATLL and ALL have shown promise, including DNA methyltransferase inhibitors and histone deacetylase inhibitors (HDIs) [14, 23].
Histone methylation, histone demethylation, histone deacetylation, and DNA methylation inhibitors are examples of epi-drugs [24].
DNMT inhibitors (DNMTIs)
Tumour tissues frequently overexpress DNA methyltransferases (DNMT), particularly DNMT1, DNMT3A, and DNMT3B, which aid in the expression of tumor suppressor genes [25]. Establishing and maintaining the DNA methylation pattern is the responsibility of DNMT1, DNMT3A, and DNMT3B [26]. The downregulation of KLF4 expression has been linked to the hypermethylation of the KLF4 promoter, which is mediated by DNA methyltransferases (DNMTs), specifically DNMT1 [27]. Also, DNMTs can cause hypermethylation in the EGR3 gene [28].
Hypermethylation of KLF4 and EGR3, among other specific genes in ATLL, increases throughout disease development. The silencing of these genes through methylation allows the cells of ATLL to evade apoptosis and activation-induced cell death [29]. Inhibition of DNMTs has transformed into one of the most promising methods in cancer treatment, and different DNMT inhibitors were developed [30]. Those inhibitors can be classified into two groups: nucleoside analogs and non-nucleoside inhibitors [31].
Nucleoside analogs like Decitabine and zebularine integrate into DNA and interfere with methylation processes through various mechanisms [32]. Non-nucleoside inhibitors are synthetic compounds such as hydralazine and RG108 as well as natural compounds including curcumin and genistein that inhibit DNMTs through multiple mechanisms [30]. Of late, paradoxically, in some CpG sites, DNMTi exhibits increases in methylation, which indeed affects cancerous cell proliferation and pathways of induced apoptosis [33]. Zebularine is a more stable cytidine analog that has shown potential, but there has been no recent clinical data [34]. These medications work by blocking DNMTs, which causes previously inactive tumor suppressor genes to become active again [30]. The mechanisms of action include epigenetic changes and induction of apoptosis [35]. Other DNMT inhibitors include nucleoside analogs SGI-110 and CP-4200 and non-nucleoside inhibitors, synthetic hydralazine, RG108, and natural curcumin, EGCG [30, 36] (Tables 1, 2).
Learning their pharmacological properties, cellular interactions, and mode of action are essential in optimizing clinical efficacy [35].
To date, two FDA-approved DNMTi, azacitidine and Decitabine, show clinical efficacy in hematologic malignancies with limited activity against solid tumors [68, 69].
Azacytidine and Decitabine are two cytosine analogous molecules applied in the epigenetic treatments of cancers. They work mainly as DNA methyltransferase inhibitors. Both agents can indeed induce DNA hypomethylation, but their modes of action differ [35]. Decitabine’s effects more clearly relate to DNA demethylation and cell cycle advancement, particularly in S-phase [70]. AZA acts more globally, inhibiting RNA methylation on DNMT2 target sites, which could impact RNA metabolism [71].
Between the two, AZA demonstrated greater potency in non-small cell lung cancer cell lines, inducing more pronounced DNA damage and apoptotic markers. The drugs also exhibited distinct effects on gene expression patterns and cell cycle dynamics, with AZA leading to sub-G1 phase accumulation and DAC(decitabine) promoting an increase in G2/M phase cells [72]. Such differences emphasize the complex and multifaceted nature through which these drugs exert their anticancer effects.
In general, the pathophysiology of ATLL may be significantly influenced by DNA hypermethylation. By adding DNA hypomethylating agents, the hypermethylated subgroups in T-ALL and ATLL have generated groups with poor outcomes [73, 74]. Two studies demonstrate frequent genomic loss of p16INK4a (CDKN2A) in Adult T-cell Leukemia/Lymphoma (ATLL). The first investigated CDKN2 gene alterations in ATLL, explicitly focusing on homozygous deletions, finding these alterations more frequent in patients with acute ATLL compared to indolent subtypes. The second study, using SNP array karyotyping on 426 samples, identified significantly recurrent focal losses, including those in CDKN2A. These findings support the frequent loss of function of the tumor suppressor gene p16INK4a in ATLL [75, 76].
DNA demethylating agents such as 5-azacytidine and Decitabine promoted gene re-expression and inhibited tumor growth in treating hypermethylated T-ALL and ATLL [37, 73].
Novel oral demethylating agents, such as OR-2100, have demonstrated anti-ATLL activity with less hematotoxicity compared to Decitabine. These agents target the aberrant DNA methylation that affects genes in T-cell receptor signaling and other critical pathways for the tumorigenesis of ATLL [41].
Decitabine and its new prodrugs, OR-1200 and OR-2100 (OR21), show promise for ATLL treatment by targeting abnormal DNA hypermethylation, which contributes to the growth of HTLV-1-infected cells [41]. These compounds reduce cell growth through global DNA hypomethylation in xenograft tumors, with OR21 causing less hematotoxicity compared to Decitabine [41, 74].
It has been reported that DNA methylation in hypermethylated sites strongly correlates with the development and progression of ATLL [41]. Decitabine has a selective effect in the EP300-mutated ATLL samples, therefore showing a targeted therapy [14].
Resistance to DNA demethylating agents in ATLL cells can arise from the misregulation of enzymes critical to pyrimidine metabolism, such as deoxycytidine kinase and uridine cytidine kinase 2 [77]. From that perspective, targeting aberrant DNA methylation is still considered one of the promising therapeutic approaches for ATLL, thus opening new horizons for novel treatment of this aggressive hematological malignancy [41, 78].
Histone deacetylase inhibitors (HDACIs)
Histone deacetylase inhibitors (HDACIs) have demonstrated potential as effective anticancer agents by reactivating tumor suppressor genes and promoting selective apoptosis in cancer cells [79, 80]. HDACIs induce the acetylation of histones, which results in a decompaction of the chromatin structure and increased gene transcription (Fig. 2) [81]. These compounds also affect non-histone proteins, thereby modulating genes associated with cell cycle, apoptosis, and angiogenesis (Fig. 2) [80]. Importantly, HDACIs have minimal toxicity in normal tissues, hence carrying great potential as therapeutic agents [82]. Further research has been directed at several combination regimens of HDACIs with other anti-cancer drugs to enhance their therapeutic potential [81, 83].
HDAC inhibitor and its function in HTLV-1 infection (created with biorender). Adapted from Zhang et al. [83]
HDACIs are an exciting new class of anticancer therapeutics and can broadly be divided into five classes: hydroxamic acids, cyclic tetrapeptides, short-chain fatty acids, benzamides, and electrophilic ketones [84]. Hydroxamic acid-based HDACIs, such as vorinostat, chelate the zinc ion in the HDAC catalytic sites through the hydroxamic acid moiety in a way that provides very effective inactivation of these enzymes [85]. Such compounds have been helpful against both hematologic cancers and solid tumors, with a number currently in clinical trials and some already FDA-approved [86]. All these HDACIs are being studied either individually or in combination with other anticancer therapies for their potential in treating various hematological cancers and solid tumors [86, 87]. A summary of HDAC inhibitors is shown in Table 3.
Histone methyltransferases inhibitors
EZH2 inhibitors
The enhancer of zeste homolog 2 (EZH2)-containing polycomb repressive complex 2 (PRC2) catalyzes the trimethylation of histone H3 at lysine 27 (H3K27me3). This alteration may cause target genes' CpG islands to become methylated, which would silence transcription and possibly aid in the development of cancer. Several malignancies, notably adult T-cell leukemia/lymphoma (ATLL), have poor prognoses and tumor progression associated with elevated EZH2 expression and elevated H3K27me3. EZH2 overexpression and the resulting H3K27 trimethylation may play a critical role in ATLL initiation and progression. Dysregulation of signal transduction pathways contributes to altered EZH2 expression in HTLV-1-driven ATLL carcinogenesis. When EZH2 is inhibited or transcriptionally silenced, H3K27me3 decreases, and tumor suppressor gene expression is restored in HTLV-1-infected T- and ATLL cells. Histone modification and promoter methylation may be essential factors in inactivating tumor suppressor genes, such as NDRG2 [110].
Target genes are transcriptionally repressed when EZH2, a histone methyltransferase and the catalytic member of the polycomb repressive complex 2 (PRC2), trimethylates lysine 27 on histone H3 (H3K27me3) [111, 112]. This epigenetic modulator is crucial for the development, chemoresistance, and carcinogenesis of numerous cancer types [112, 113]. Overexpression of EZH2 and its mutations are related to tumor malignancy for various kinds of cancers [111]; therefore, given its multifaceted properties in cancer, EZH2 has become one of the promising therapeutic targets, and several inhibitors are presently under investigation in various preclinical and clinical studies [113, 114].
EZH2 inhibitors indeed hold promise in several cancer therapies. Tazemetostat (EPZ-6438), GSK2816126, and CPI-1205 have reached clinical trials for hematologic malignancies and solid tumors [115].
Recent studies disclosed EZH2 as a putative target in ATLL. The polycomb repressive complex 2 component EZH2 is involved in the epigenetic reprogramming of ATLL cells, which often leads to the downregulation and silence of tumor suppressors. Pharmacological inhibition has shown great promise in the targeted eradication of HTLV-1-infected and leukemic cells [21]. The EZH2 inhibitor DZNep induces apoptosis in adult T-cell leukemia/lymphoma cells by BCL2 suppression via regulation of Mir-181a [116].
Nowadays, valemetostat is one of the most effective dual inhibitors of EZH1 and EZH2, showing promise as an antitumor agent in lymphomas, mainly diffuse large B-cell lymphoma and ATLL. In Japan, it was authorized in 2022 to treat ATLL that had relapsed or was refractory [117]. Although valemetostat has promise, cytopenias are a common adverse effect that can typically be controlled without stopping medication [118].
Bromodomain and extra terminal inhibitors (BETIs)
Each member of the BET protein family—BRDT, BRD2, BRD3, and BRD4—has an extra-terminal domain at the C-terminal and two bromodomains at the N-terminal [119, 120]. Through their interactions with acetylated histones and transcription factors, activation of transcriptional machinery, and activation of RNA polymerase II, BET proteins control the transcription of genes [120].
In conclusion, preclinical research shows that BET inhibitors, especially JQ1, work by targeting BRD4 to prevent the growth of Tax-positive HTLV-1-infected cells. By interfering with BRD4’s association with acetylated RelA, an NF-κB subunit, JQ1 prevents Tax-induced NF-κB activation and carcinogenesis in HTLV-1-infected cells [121]. Cell cycle arrest, diminished S-phase entry, and induction of apoptosis were observed via E2f1 downregulation [122]. It is also noteworthy to mention that the BET inhibitors dislodge the HTLV-I-regulated BATF3/IRF4 transcriptional circuitry, which acts at the heart of the propagation of ATLL, due to the collapse of the HBZ-driven transcriptional program included in MYC expression [123]. These findings identified a therapeutic potential of BET inhibitors in the treatment of ATLL.
Arsenic-based epigenetic therapies
Arsenicals are outdated but virulent and have been applied in the treatment of a wide range of diseases since ancient times. Their luster was lost after the discovery of antibiotics in the 1940s. There has been some revival of the use of medications containing arsenic in the treatment of virus-related cancers. Treatment with arsenic trioxide combined with IFN-α and nucleoside reverse-transcriptase inhibitor (NRTI) gave a superior cytokine gene expression profile in adults with ATLL associated with human T-cell leukemia virus type 1. Further, it altered the patient from initial immunosuppression to a more immunocompetent state. This is likely due to an improved immune response that assists in the elimination of ATLL cells and the control of infections caused by opportunistic pathogens. In clinical studies, there is a possible anti-leukemia effect in patients with poor prognosis of ATLL, which needs further optimization [124].
Challenges and restrictions of epigenetic treatment
Epi-drugs have very bright prospects for cancer treatment, but there are still a lot of obstacles ahead. Driver genes need to be identified versus those that are stimulated; also, potential side effects may limit the widespread application of these genes. Some epi-drugs received FDA approval, but their optimal use and clinical validation remain to be done. Epigenetic modifications may vary with the context and stages of the disease. Clinical trials are often expensive and may take an extended period. Despite all the challenges, targeting epigenetic modifications still has a place in personalized cancer therapy, mainly when used in combination with other approaches. However, key epigenetic alterations are yet to be identified, and the development of effective strategies for targeting these modifications is of utmost importance for further progress [8, 10, 125, 126].
Availability of data and materials
Not applicable.
References
Letafati A, Soheili R, Norouzi M, Soleimani P, Mozhgani S-H. Therapeutic approaches for HTLV-1-associated adult T-cell leukemia/lymphoma: a comprehensive review. Med Oncol. 2023;40(10):295.
Brites C, Grassi MF, Quaresma JAS, Ishak R, Vallinoto ACR. Pathogenesis of HTLV-1 infection and progression biomarkers: an overview. Braz J Infect Dis. 2021;25:101594.
Bangham CR. HTLV-1 infections. J Clin Pathol. 2000;53(8):581–6.
Cook LB, Elemans M, Rowan AG, Asquith B. HTLV-1: persistence and pathogenesis. Virology. 2013;435(1):131–40.
Tokura Y, Sawada Y, Shimauchi T. Skin manifestations of adult T-cell leukemia/lymphoma: clinical, cytological and immunological features. J Dermatol. 2014;41(1):19–25.
Zhang Y, Chen H, Sun J-F. Cutaneous manifestations and treatment advances of adult T-cell leukemia/lymphoma. Int J Dermatol Venereol. 2022;5(01):40–4.
Ilango S, Paital B, Jayachandran P, Padma PR, Nirmaladevi R. Epigenetic alterations in cancer. Front Biosci Landmark. 2020;25(6):1058–109.
Fardi M, Solali S, Hagh MF. Epigenetic mechanisms as a new approach in cancer treatment: an updated review. Genes Dis. 2018;5(4):304–11.
Miranda Furtado CL, Dos Santos Luciano MC, Silva Santos RD, Furtado GP, Moraes MO, Pessoa C. Epidrugs: targeting epigenetic marks in cancer treatment. Epigenetics. 2019;14(12):1164–76.
Lu Y, Chan Y-T, Tan H-Y, Li S, Wang N, Feng Y. Epigenetic regulation in human cancer: the potential role of epi-drug in cancer therapy. Mol Cancer. 2020;19:1–16.
Bojang P Jr, Ramos KS. The promise and failures of epigenetic therapies for cancer treatment. Cancer Treat Rev. 2014;40(1):153–69.
Ratner L. Epigenetic regulation of human T-cell leukemia virus gene expression. Microorganisms. 2021;10(1):84.
Barreto-Galvez A, Madireddy A, Ye BH, Gagliardi J, Juwarwala A, Pradeep A, Niljikar M. The cause and consequence of replication stress in adult T-cell leukemia. Blood. 2022;140(Supplement 1):11512.
Shah UA, Chung EY, Giricz O, Pradhan K, Kataoka K, Gordon-Mitchell S, et al. North American ATLL has a distinct mutational and transcriptional profile and responds to epigenetic therapies. Blood J Am Soc Hematol. 2018;132(14):1507–18.
Ye BH, Chung E, Pradhan K, Acuna-Villaorduna A, Wang Y, Shastri A, Madireddy A, Verma A, Sica RA, Janakiram M. S-Phase progression of North American ATLL cells is critically regulated by the proto-oncogene BCL6 and targetable by PARP inhibition. Blood. 2019;134:3779.
Hofmann WK, Tsukasaki K, Takeuchi N, Takeuchi S, Koeffler HP. Methylation analysis of cell cycle control genes in adult T-cell leukemia/lymphoma. Leuk Lymphoma. 2001;42(5):1107–9.
Nicot C. Tumor suppressor inactivation in the pathogenesis of adult T-cell leukemia. J Oncol. 2015;2015(1):183590.
Ego T, Tanaka Y, Shimotohno K. Interaction of HTLV-1 Tax and methyl-CpG-binding domain 2 positively regulates the gene expression from the hypermethylated LTR. Oncogene. 2005;24(11):1914–23.
Miura M, Miyazato P, Satou Y, Tanaka Y, Bangham C. Spontaneous HTLV-1 transcription is accompanied by distinct epigenetic changes in the 5′ and 3′ long terminal repeats. 2018.
Audia JE, Campbell RM. Histone modifications and cancer. Cold Spring Harb Perspect Biol. 2016;8(4):a019521.
Fujikawa D, Nakagawa S, Hori M, Kurokawa N, Soejima A, Nakano K, et al. Polycomb-dependent epigenetic landscape in adult T-cell leukemia. Blood J Am Soc Hematol. 2016;127(14):1790–802.
Castro-Muñoz LJ, Vázquez Ulloa E, Sahlgren C, Lizano M, De La Cruz-Hernández E, Contreras-Paredes A. Modulating epigenetic modifications for cancer therapy. Oncol Rep. 2023;49(3):59.
Mummery A, Narendran A, Lee KY. Targeting epigenetics through histone deacetylase inhibitors in acute lymphoblastic leukemia. Curr Cancer Drug Targets. 2011;11(7):882–93.
Yang T, Yang Y, Wang Y. Predictive biomarkers and potential drug combinations of epi-drugs in cancer therapy. Clin Epigenet. 2021;13(1):113.
Zhou Z, Li H-Q, Liu F. DNA methyltransferase inhibitors and their therapeutic potential. Curr Top Med Chem. 2018;18(28):2448–57.
Tajima S, Suetake I, Takeshita K, Nakagawa A, Kimura H, Song J. Domain structure of the Dnmt1, Dnmt3a, and Dnmt3b DNA methyltransferases. DNA Methyltransferases-Role Funct. 2022:45–68.
Xiao X, Tang W, Yuan Q, Peng L, Yu P. Epigenetic repression of Krüppel-like factor 4 through Dnmt1 contributes to EMT in renal fibrosis. Int J Mol Med. 2015;35(6):1596–602.
Charostad J, Astani A, Goudarzi H, Faghihloo E. DNA methyltransferases in virus-associated cancers. Rev Med Virol. 2019;29(2):e2022.
Yasunaga J-I, Taniguchi Y, Nosaka K, Yoshida M, Satou Y, Sakai T, et al. Identification of aberrantly methylated genes in association with adult T-cell leukemia. Cancer Res. 2004;64(17):6002–9.
Singh V, Sharma P, Capalash N. DNA methyltransferase-1 inhibitors as epigenetic therapy for cancer. Curr Cancer Drug Targets. 2013;13(4):379–99.
Biswas S, Rao CM. Epigenetic tools (The Writers, The Readers and The Erasers) and their implications in cancer therapy. Eur J Pharmacol. 2018;837:8–24.
Gowher H, Jeltsch A. Mechanism of inhibition of DNA methyltransferases by cytidine analogs in cancer therapy. Cancer Biol Ther. 2004;3(11):1062–8.
Giri AK, Aittokallio T. DNMT inhibitors increase methylation in the cancer genome. Front Pharmacol. 2019;10:385.
Gnyszka A, Jastrzębski Z, Flis S. DNA methyltransferase inhibitors and their emerging role in epigenetic therapy of cancer. Anticancer Res. 2013;33(8):2989–96.
Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer. 2008;123(1):8–13.
Yu N, Wang M. Anticancer drug discovery targeting DNA hypermethylation. Curr Med Chem. 2008;15(14):1350–75.
Uenogawa K, Hatta Y, Arima N, Hayakawa S, Sawada U, Aizawa S, et al. Azacitidine induces demethylation of p16INK4a and inhibits growth in adult T-cell leukemia/lymphoma. Int J Mol Med. 2011;28(5):835–9.
Pechalrieu D, Etievant C, Arimondo PB. DNA methyltransferase inhibitors in cancer: from pharmacology to translational studies. Biochem Pharmacol. 2017;129:1–13.
Christman JK. 5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene. 2002;21(35):5483–95.
Sutherland MK, Yu C, Anderson M, Zeng W, van Rooijen N, Sievers EL, Grewal IS, Law CL. 5-azacytidine enhances the anti-leukemic activity of lintuzumab (SGN-33) in preclinical models of acute myeloid leukemia. In: MAbs, vol. 2, no. 4. Taylor & Francis; 2010. pp. 440–448.
Watanabe T, Yamashita S, Ureshino H, Kamachi K, Kurahashi Y, Fukuda-Kurahashi Y, et al. Targeting aberrant DNA hypermethylation as a driver of ATL leukemogenesis by using the new oral demethylating agent OR-2100. Blood J Am Soc Hematol. 2020;136(7):871–84.
Hackanson B, Daskalakis M. Decitabine. Small Mol Oncol. 2014:269–97.
Chowdhury B, McGovern A, Cui Y, Choudhury SR, Cho IH, Cooper B, Chevassut T, Lossie AC, Irudayaraj J. The hypomethylating agent decitabine causes a paradoxical increase in 5-hydroxymethylcytosine in human leukemia cells. Sci Rep. 2015;5(1):9281.
Wilson VL, Jones PA, Momparler RL. Inhibition of DNA methylation in L1210 leukemic cells by 5-aza-2′-deoxycytidine as a possible mechanism of chemotherapeutic action. Can Res. 1983;43(8):3493–6.
Griffiths EA, Choy G, Redkar S, Taverna P, Azab M, Karpf AR. SGI-110: DNA methyltransferase inhibitor oncolytic. Drugs Future. 2013;38(8):535.
Stewart DJ, Donehower RC, Eisenhauer EA, Wainman N, Shah AK, Bonfils C, MacLeod AR, Besterman JM, Reid GK. A phase I pharmacokinetic and pharmacodynamic study of the DNA methyltransferase 1 inhibitor MG98 administered twice weekly. Ann Oncol. 2003;14(5):766–74.
Beaulieu N, Fournel M, Macleod A. Antitumor activity of MG98, an antisense oligodeoxynucleotide targeting DNA methyltransferase-1 (DNMT1). In: Clinical cancer research, vol. 7, no. 11. Birmingham: American Association Cancer Research; 2001. pp. 3800S–3800S.
Nakamura K, Nakabayashi K, Htet Aung K, Aizawa K, Hori N, Yamauchi J, Hata K, Tanoue A. DNA methyltransferase inhibitor zebularine induces human cholangiocarcinoma cell death through alteration of DNA methylation status. PLoS ONE. 2015;10(3):e0120545.
Cheng JC, Matsen CB, Gonzales FA, Ye W, Greer S, Marquez VE, Jones PA, Selker EU. Inhibition of DNA methylation and reactivation of silenced genes by zebularine. J Natl Cancer Inst. 2003;95(5):399–409.
García-Domínguez P, Dell’Aversana C, Alvarez R, Altucci L, de Lera AR. Synthetic approaches to DNMT inhibitor SGI-1027 and effects on the U937 leukemia cell line. Bioorg Med Chem Lett. 2013;23(6):1631–5.
Datta J, Ghoshal K, Denny WA, Gamage SA, Brooke DG, Phiasivongsa P, Redkar S, Jacob ST. RETRACTED: a new class of quinoline-based DNA hypomethylating agents reactivates tumor suppressor genes by blocking DNA methyltransferase 1 activity and inducing its degradation. Can Res. 2009;69(10):4277–85.
Chen S, Wang Y, Zhou W, Li S, Peng J, Shi Z, Hu J, Liu YC, Ding H, Lin Y, Li L. Identifying novel selective non-nucleoside DNA methyltransferase 1 inhibitors through docking-based virtual screening. J Med Chem. 2014;57(21):9028–41.
Medicament PF. Centre National de la Recherche Scientifique (CNRS). Quinazoline derivatives and their use as DNA methyltransferase inhibitors. WO2015040169. 2015.
Erdmann A, Menon Y, Gros C, Molinier N, Novosad N, Samson A, Gregoire JM, Long C, Ausseil F, Halby L, Arimondo PB. Design and synthesis of new non nucleoside inhibitors of DNMT3A. Bioorg Med Chem. 2015;23(17):5946–53.
Halby L, Champion C, Sénamaud-Beaufort C, Ajjan S, Drujon T, Rajavelu A, Ceccaldi A, Jurkowska R, Lequin O, Nelson WG, Guy A. Rapid synthesis of new DNMT inhibitors derivatives of procainamide. ChemBioChem. 2012;13(1):157–65.
Mora FP, Salazar YI, Margalef MD, Caffarena MR, Cáneva SV, Ansa DO, Arévalo EA, Larzabal ES, Olascoaga AZ, inventors; Euskal Herriko Unibertsitatea, IKERCHEM SL, assignee. Indole derivatives, pharmaceutical compositions containing such indoles and their use as DNA methylation modulators. United States patent application US 14/441,086. 2015.
Castellano S, Kuck D, Viviano M, Yoo J, López-Vallejo F, Conti P, Tamborini L, Pinto A, Medina-Franco JL, Sbardella G. Synthesis and biochemical evaluation of Δ2-isoxazoline derivatives as DNA methyltransferase 1 inhibitors. J Med Chem. 2011;54(21):7663–77.
Castellano S, Kuck D, Sala M, Novellino E, Lyko F, Sbardella G. Constrained analogues of procaine as novel small molecule inhibitors of DNA methyltransferase-1. J Med Chem. 2008;51(7):2321–5.
Ceccaldi A, Rajavelu A, Ragozin S, Sénamaud-Beaufort C, Bashtrykov P, Testa N, Dali-Ali H, Maulay-Bailly C, Amand S, Guianvarch D, Jeltsch A. Identification of novel inhibitors of DNA methylation by screening of a chemical library. ACS Chem Biol. 2013;8(3):543–8.
Asgatay S, Champion C, Marloie G, Drujon T, Senamaud-Beaufort C, Ceccaldi A, Erdmann A, Rajavelu A, Schambel P, Jeltsch A, Lequin O. Synthesis and evaluation of analogues of N-phthaloyl-l-tryptophan (RG108) as inhibitors of DNA methyltransferase 1. J Med Chem. 2014;57(2):421–34.
Suzuki T, Tanaka R, Hamada S, Nakagawa H, Miyata N. Design, synthesis, inhibitory activity, and binding mode study of novel DNA methyltransferase 1 inhibitors. Bioorg Med Chem Lett. 2010;20(3):1124–7.
Kilgore JA, Du X, Melito L, Wei S, Wang C, Chin HG, Posner B, Pradhan S, Ready JM, Williams NS. Identification of DNMT1 selective antagonists using a novel scintillation proximity assay. J Biol Chem. 2013;288(27):19673–84.
Kuck D, Caulfield T, Lyko F, Medina-Franco JL. Nanaomycin A selectively inhibits DNMT3B and reactivates silenced tumor suppressor genes in human cancer cells. Mol Cancer Ther. 2010;9(11):3015–23.
Fagan RL, Cryderman DE, Kopelovich L, Wallrath LL, Brenner C. Laccaic acid A is a direct, DNA-competitive inhibitor of DNA methyltransferase 1. J Biol Chem. 2013;288(33):23858–67.
Weng JR, Lai IL, Yang HC, Lin CN, Bai LY. Identification of kazinol Q, a natural product from Formosan plants, as an inhibitor of DNA methyltransferase. Phytother Res. 2014;28(1):49–54.
Ceccaldi A, Rajavelu A, Champion C, Rampon C, Jurkowska R, Jankevicius G, Sénamaud-Beaufort C, Ponger L, Gagey N, Dali Ali H, Tost J. C5-DNA methyltransferase inhibitors: from screening to effects on zebrafish embryo development. ChemBioChem. 2011;12(9):1337–45.
Machijima Y, Ishikawa C, Sawada S, Okudaira T, Uchihara J-N, Tanaka Y, et al. Anti-adult T-cell leukemia/lymphoma effects of indole-3-carbinol. Retrovirology. 2009;6:1–13.
Yang AS, Yang BJ. The failure of epigenetic combination therapy for cancer and what it might be telling us about DNA methylation inhibitors. Taylor & Francis; 2016. p. 9–12.
Gonzalez-Fierro A, Dueñas-González A. Emerging DNA methylation inhibitors for cancer therapy: challenges and prospects. Expert Rev Precis Med Drug Dev. 2019;4(1):27–35.
Al-Salihi M, Yu M, Burnett DM, Alexander A, Samlowski WE, Fitzpatrick FA. The depletion of DNA methyltransferase-1 and the epigenetic effects of 5-aza-2′deoxycytidine (decitabine) are differentially regulated by cell cycle progression. Epigenetics. 2011;6(8):1021–8.
Schaefer M, Hagemann S, Hanna K, Lyko F. Azacytidine inhibits RNA methylation at DNMT2 target sites in human cancer cell lines. Can Res. 2009;69(20):8127–32.
Nguyen AN, Hollenbach PW, Richard N, Luna-Moran A, Brady H, Heise C, et al. Azacitidine and decitabine have different mechanisms of action in non-small cell lung cancer cell lines. Lung Cancer Targets Therapy 2010:119–40.
Touzart A, Mayakonda A, Smith C, Hey J, Toth R, Cieslak A, et al. Epigenetic analysis of patients with T-ALL identifies poor outcomes and a hypomethylating agent-responsive subgroup. Sci Transl Med. 2021;13(595):eabc4834.
Watanabe T, Ureshino H, Kurahashi Y, Fukuda-Kurahashi Y, Yamashita S, Ushijima T, et al. Aberrant regional DNA hypermethylation as a preventive and therapeutic target for adult T-cell leukemia-lymphoma. Blood. 2018;132:4175.
Uchida T, Kinoshita T, Murate T, Saito H, Hotta T. CDKN2 (MTS1/p16INK4A) gene alterations in adult T-cell leukemia/lymphoma. Leuk Lymphoma. 1998;29(1–2):27–35.
Kataoka K, Nagata Y, Kitanaka A, Shiraishi Y, Shimamura T, Yasunaga JI, Totoki Y, Chiba K, Sato-Otsubo A, Nagae G, Ishii R. Integrated molecular analysis of adult T cell leukemia/lymphoma. Nat Genet. 2015;47(11):1304–15.
Yoshida-Sakai N, Watanabe T, Yamamoto Y, Ureshino H, Kamachi K, Kurahashi Y, et al. Adult T-cell leukemia-lymphoma acquires resistance to DNA demethylating agents through dysregulation of enzymes involved in pyrimidine metabolism. Int J Cancer. 2022;150(7):1184–97.
Marino-Merlo F, Mastino A, Grelli S, Hermine O, Bazarbachi A, Macchi B. Future perspectives on drug targeting in adult T cell leukemia-lymphoma. Front Microbiol. 2018;9:925.
Kim T-Y, Bang Y-J, Robertson KD. Histone deacetylase inhibitors for cancer therapy. Epigenetics. 2006;1(1):15–24.
Shankar S, Srivastava RK. Histone deacetylase inhibitors: mechanisms and clinical significance in cancer: HDAC inhibitor-induced apoptosis. Adv Exp Med Biol. 2008;615:261–98.
Lane AA, Chabner BA. Histone deacetylase inhibitors in cancer therapy. J Clin Oncol. 2009;27(32):5459–68.
Vigushin DM, Coombes RC. Histone deacetylase inhibitors in cancer treatment. Anticancer Drugs. 2002;13(1):1–13.
Zhang Q, Wang S, Chen J, Yu Z. Histone deacetylases (HDACs) guided novel therapies for T-cell lymphomas. Int J Med Sci. 2019;16(3):424.
Tan Y-M, Huang W-Y, Yu N. Structure-activity relationships of histone deacetylase inhibitors. Yao xue xue bao= Acta Pharmaceutica Sinica. 2009;44(10):1072–83.
Marks PA, Richon VM, Kelly WK, Chiao JH, Miller T, editors. Histone deacetylase inhibitors: development as cancer therapy. In: Reversible protein acetylation: Novartis Foundation symposium, vol. 259. Wiley Online Library; 2004.
Cappellacci L, Perinelli DR, Maggi F, Grifantini M, Petrelli R. Recent progress in histone deacetylase inhibitors as anticancer agents. Curr Med Chem. 2020;27(15):2449–93.
Lemoine M, Younes A. Histone deacetylase inhibitors in the treatment of lymphoma. Discov Med. 2010;10(54):462–70.
Marks PA. HDAC inhibitors: much to learn about effective therapy. Oncology. 2010;24(2):185.
Duvic M, Vu J. Vorinostat: a new oral histone deacetylase inhibitor approved for cutaneous T-cell lymphoma. Expert Opin Investig Drugs. 2007;16(7):1111–20.
Le VKH, Pham TPD, Truong DH. Delivery systems for vorinostat in cancer treatment: an updated review. J Drug Deliv Sci Technol. 2021;61:102334.
Zhang C, Richon V, Ni X, Talpur R, Duvic M. Selective induction of apoptosis by histone deacetylase inhibitor SAHA in cutaneous T-cell lymphoma cells: relevance to mechanism of therapeutic action. J Investig Dermatol. 2005;125(5):1045–52.
Salama BM, Helmy MW, Fouad H, Shamaa MM, Houssen ME. The synergistic antitumor effect of decitabine and vorinostat combination on HepG2 human hepatocellular carcinoma cell line via epigenetic modulation of autophagy-apoptosis molecular crosstalk. Curr Issues Mol Biol. 2023;45(7):5935–49.
Ding JD, Ri M, Narita T, Masaki A, Mori F, Ito A, Kusumoto S, Ishida T, Komatsu H, Shiotsu Y, Niimi A. Reduced expression of HDAC3 contributes to the resistance against HDAC inhibitor, Vorinostat (SAHA) in mature lymphoid malignancies. Blood. 2012;120(21):1342.
VanderMolen KM, McCulloch W, Pearce CJ, Oberlies NH. Romidepsin (Istodax, NSC 630176, FR901228, FK228, depsipeptide): a natural product recently approved for cutaneous T-cell lymphoma. J Antibiot. 2011;64(8):525–31.
Harrison SJ, Bishton M, Bates SE, Grant S, Piekarz RL, Johnstone RW, et al. A focus on the preclinical development and clinical status of the histone deacetylase inhibitor, romidepsin (depsipeptide, Istodax®). Epigenomics. 2012;4(5):571–89.
Piekarz RL, Frye R, Prince HM, Kirschbaum MH, Zain J, Allen SL, et al. Phase 2 trial of romidepsin in patients with peripheral T-cell lymphoma. Blood J Am Soc Hematol. 2011;117(22):5827–34.
Yu P, Petrus MN, Ju W, Zhang M, Conlon KC, Nakagawa M, et al. Augmented efficacy with the combination of blockade of the Notch-1 pathway, bortezomib and romidepsin in a murine MT-1 adult T-cell leukemia model. Leukemia. 2015;29(3):556–66.
Mukhi N, Verma V, Ahmed A, Lim H, Gonsky J, Sidhu G. Romidepsin in relapsed/refractory HTLV-1 associated adult T-cell lymphoma/leukemia: a case series. Blood. 2015;126(23):5113.
Toomey N, Barber G, Ramos JC. Preclinical efficacy of belinostat in combination with zidovudine in adult T-cell leukemia-lymphoma. Retrovirology. 2015;12(Suppl 1):P73.
Duenas-Gonzalez A, Candelaria M, Perez-Plascencia C, Perez-Cardenas E, de la Cruz-Hernandez E, Herrera LA. Valproic acid as epigenetic cancer drug: preclinical, clinical and transcriptional effects on solid tumors. Cancer Treat Rev. 2008;34(3):206–22.
Detich N, Bovenzi V, Szyf M. Valproate induces replication-independent active DNA demethylation. J Biol Chem. 2003;278(30):27586–92.
Gurvich N, Tsygankova OM, Meinkoth JL, Klein PS. Histone deacetylase is a target of valproic acid-mediated cellular differentiation. Can Res. 2004;64(3):1079–86.
Yves P, Stephane M, Rishika B, Christine D, Gérard P. Characteristics of adult T-cell leukemia/lymphoma patients with long survival: prognostic significance of skin lesions and possible beneficial role of valproic acid. Leukemia Res Treat. 2015;2015(1):476805.
Zimmerman B, Sargeant A, Landes K, Fernandez SA, Chen C-S, Lairmore MD. Efficacy of novel histone deacetylase inhibitor, AR42, in a mouse model of, human T-lymphotropic virus type 1 adult T cell lymphoma. Leuk Res. 2011;35(11):1491–7.
Pongas G, Toomey NL, Reis IM, Komanduri KV, Chapman-Fredricks JR, Ramos JC. Safety and efficacy of belinostat trial with zidovudine plus interferon for HTLV-1 related adult T-cell leukemia-lymphoma: interim results. Blood. 2022;140(Supplement 1):9448–9.
Khwaja S, Kumar K, Das R, Negi AS. Microtubule associated proteins as targets for anticancer drug development. Bioorg Chem. 2021;116:105320.
Nishioka C, Ikezoe T, Yang J, Komatsu N, Bandobashi K, Taniguchi A, et al. Histone deacetylase inhibitors induce growth arrest and apoptosis of HTLV-1-infected T-cells via blockade of signaling by nuclear factor κB. Leuk Res. 2008;32(2):287–96.
Utsunomiya A, Izutsu K, Jo T, Yoshida S, Tsukasaki K, Ando K, et al. Oral histone deacetylase inhibitor tucidinostat (HBI-8000) in patients with relapsed or refractory adult T-cell leukemia/lymphoma: Phase IIb results. Cancer Sci. 2022;113(8):2778–87.
Hasegawa H, Bissonnette RP, Gillings M, Sasaki D, Taniguchi H, Kitanosono H, et al. Induction of apoptosis by HBI-8000 in adult T-cell leukemia/lymphoma is associated with activation of Bim and NLRP3. Cancer Sci. 2016;107(8):1124–33.
Ichikawa T, Nakahata S, Fujii M, Iha H, Shimoda K, Morishita K. The regulation of NDRG2 expression during ATLL development after HTLV-1 infection. Biochimica et Biophysica Acta (BBA) Mol Basis Dis. 2019;1865(10):2633–46.
Li L-Y. EZH2: novel therapeutic target for human cancer. Biomedicine. 2014;4(1):1.
Yamaguchi H, Hung M-C. Regulation and role of EZH2 in cancer. Cancer Res Treat Off J Korean Cancer Assoc. 2014;46(3):209–22.
Italiano A. Role of the EZH2 histone methyltransferase as a therapeutic target in cancer. Pharmacol Ther. 2016;165:26–31.
Gan L, Yang Y, Li Q, Feng Y, Liu T, Guo W. Epigenetic regulation of cancer progression by EZH2: from biological insights to therapeutic potential. Biomark Res. 2018;6:1–10.
Gulati N, Béguelin W, Giulino-Roth L. Enhancer of zeste homolog 2 (EZH2) inhibitors. Leuk Lymphoma. 2018;59(7):1574–85.
Daisuke S, Hiroo H, Hiroaki T, Imaizumi Y, Naoki U, Yoshitomo M, et al. EZH2 inhibitor DZNep induces apoptosis in adult T-cell leukemia/lymphoma cells by BCL2 suppression via regulation of Mir-181a. Blood. 2013;122(21):4265.
Dou F, Tian Z, Yang X, Li J, Wang R, Gao J. Valemetostat: First approval as a dual inhibitor of EZH1/2 to treat adult T-cell leukemia/lymphoma. Drug Discov Ther. 2022;16(6):297–9.
Horwitz SM, Izutsu K, Mehta-Shah N, Cordoba R, Barta SK, Bachy E, et al. Efficacy and safety of valemetostat monotherapy in patients with relapsed or refractory peripheral T-Cell lymphomas: primary results of the phase 2 VALENTINE-PTCL01 study. Blood. 2023;142:302.
Taniguchi Y. The bromodomain and extra-terminal domain (BET) family: functional anatomy of BET paralogous proteins. Int J Mol Sci. 2016;17(11):1849.
Cheung KL, Kim C, Zhou M-M. The functions of BET proteins in gene transcription of biology and diseases. Front Mol Biosci. 2021;8:728777.
Wu X, Qi J, Bradner JE, Xiao G, Chen L-F. Bromodomain and extraterminal (BET) protein inhibition suppresses human T cell leukemia virus 1 (HTLV-1) Tax protein-mediated tumorigenesis by inhibiting nuclear factor κB (NF-κB) signaling. J Biol Chem. 2013;288(50):36094–105.
Shi X, Liu C, Liu B, Chen J, Wu X, Gong W. JQ1: a novel potential therapeutic target. Die Pharmazie Int J Pharm Sci. 2018;73(9):491–3.
Nakagawa M, Shaffer AL, Ceribelli M, Zhang M, Wright G, Xiao W, et al. Targeting the HTLV-I-regulated BATF3/IRF4 transcriptional network in adult T cell leukemia/lymphoma. Cancer Cell. 2018;34(2):286–97.
Paul NP, Galván AE, Yoshinaga-Sakurai K, Rosen BP, Yoshinaga M. Arsenic in medicine: past, present and future. Biometals. 2023;36(2):283–301.
Day JJ, Sweatt JD. Epigenetic treatments for cognitive impairments. Neuropsychopharmacology. 2012;37(1):247–60.
Ghasemi S. Cancer’s epigenetic drugs: Where are they in the cancer medicines? Pharmacogenomics J. 2020;20(3):367–79.
Acknowledgements
Not applicable.
Funding
None.
Author information
Authors and Affiliations
Contributions
AL, SHM: Conceptualization, supervision. ZT, BM, SS, AVF, MN, SK, AZ, MM, SF, MN, FK: writing original draft, tables. OSA, TF, FK, MR: Investigation, validation, review and editing.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
All authors agree.
Competing interests
The authors declare that they have no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Letafati, A., Mehdigholian Chaijani, R., Edalat, F. et al. Advances in epigenetic treatment of adult T-cell leukemia/lymphoma: a comprehensive review. Clin Epigenet 17, 39 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13148-025-01841-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13148-025-01841-z