Skip to main content

Update on histone deacetylase inhibitors in peripheral T-cell lymphoma (PTCL)

Abstract

Peripheral T-cell lymphomas (PTCLs) are a group of highly aggressive malignancies with generally poor prognoses, and the first-line chemotherapy of PTCL has limited efficacy. Currently, several novel targeted agents, including histone deacetylase inhibitors (HDACis), have been investigated to improve the therapeutic outcome of PTCLs. Several HDACis, such as romidepsin, belinostat, and chidamide, have demonstrated favorable clinical efficacy and safety in PTCLs. More novel HDACis and new combination therapies are undergoing preclinical or clinical trials. Mutation analysis based on next-generation sequencing may advance our understanding of the correlation between epigenetic mutation profiles and relevant targeted therapies. Multitargeted HDACis and HDACi-based prodrugs hold promising futures and offer further directions for drug design.

Introduction

Peripheral T-cell lymphomas (PTCLs) are derived from mature T-cells or NK/T-cells and represent a group of non-Hodgkin lymphomas. Mature T-cell and NK cell neoplasms are classified into 30 subtypes, and among them, peripheral T-cell lymphoma not otherwise specified (PTCL-NOS), nodal T-follicular helper cell lymphoma (T-FHCL), and anaplastic large cell lymphoma (ALCL) are common subtypes of PTCL [1].

The CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) regimen is the first-line chemotherapy for PTCLs. However, CHOP regimen research was previously based on practice with aggressive B-cell lymphomas [2]. Although CHOP regimen has a remission rate of 50%–65% in PTCL, brief response and high risk of relapse are the two primary problems [3]. In one of the largest international prospective T-cell projects, out of 937 PTCL patients who received an active first-line treatment, 633 (68%) were identified as refractory or relapsed patients with a 3-year overall survival (OS) rate not reaching 30% [3]. High-dose chemotherapy and hematopoietic stem cell transplantation can improve prognosis as consolidation in first or second remission, yet strictly limited to patients with tolerable body status and chemotherapy sensitivity [3]. As a result, for patients with PTCL, the CHOP regimen exhibits an unmet need for effective treatment. No optimal standard of treatment for PTCLs has been widely accepted, so effective novel therapies are urgently warranted. With further insights into molecular pathogenesis, targeted therapy becomes an alternative therapeutic option for PTCLs.

Due to the aberrant activity and expression of histone deacetylases (HDACs) in tumor cell growth, HDACs are widely studied as therapeutic targets for PTCLs. HDACs mainly function in transcription regulation by removing acetyl groups from the ε-amino groups of the lysine residues of histone tails [4, 5]. Reduced positivity of histone tails contributes to chromatin condensation, hindering the accessibility of transcription factors to DNA. As transcriptional corepressors, HDACs induce a closed status of the nucleosome, resulting in gene suppression. Besides histone substrates, HDACs also regulate the stability and activity of non-histones via post-translational deacetylation, as shown in Fig. 1. Chaperone proteins, transcription factors, structural proteins, and steroid receptors are the principally non-histone substrates of HDACs, including nuclear factor κB (NF-κB), p53, GATA1, GATA2, STAT3, and heat shock protein 90 (Hsp90) [4, 5]. Non-histone protein deacetylation plays a vital role in physiology and pathogenic cellular processes, such as gene transcription, signal transduction, protein folding, autophagy, DNA repair, cell proliferation, and metabolism [6]. Histone acetyltransferases (HATs) reverse these processes by adding acetyl groups [7, 8]. However, HATs and their inhibitors are paid less attention because of their inadequate medical value for PTCLs.

Fig. 1
figure 1

The histone or non-histone substrates for and the integrated biological effects of HDACs. The acetylation of histone substrates modulates the chromatin structure to reduce the accessibility to transcriptional regulatory proteins and subsequent gene expression. For non-histone substrates, HDACs have an impact on their activity by acetylating. In general, HDACs contribute to proliferative effects. Moreover, HDACs play a pivotal role in hematopoiesis and T-cell development. Abnormal expression or activity of HDACs is involved in the malignant transformation of PTCL in multiple ways, including intracellular derangements, protumorigenic microenvironment, and virus-mediated transformation

Regarding mammals, 18 kinds of HDACs have been subdivided into four classes based on their homology to yeast HDACs—sequence motifs, cellular location, tissue specificity, and enzymatic activity, as summarized in Table 1. In Class I, HDAC1, 2, and 3 are ubiquitously distributed in human tissues, while HDAC8 is located in smooth muscles and affects their contractility [9]. Of note, PD-L1 nuclear translocation has been reported as being HDAC2-dependent, providing a hint for research to identify targeted agents [10]. HDAC3 uniquely localizes to the mitotic spindle during mitotic progression to maintain proper kinetochore-microtubule attachment and chromosome alignment [11]. In addition, HDAC3 is essential for inflammatory response during host defense against bacterial infection by facilitating TNFα-mediated NF-κB activation [12]. Furthermore, HDAC8 has been found to have an anti-apoptotic effect via repressing transcription of the proapoptotic protein Bcl-2-modifying factor [13]. Due to different sequence homology and domain organization, Class II HDACs were further subdivided into IIa and IIb subclasses [5]. Subclass IIa (HDAC4, 5, 7, and 9) is characteristic of signal-dependent nucleocytoplasmic shuttling and tissue-specific expression [5, 14]. Subclass IIb (HDAC6 and 10) has two catalytic HDAC domains and is distributed in limited tissue types with a cytoplasmic localization [5]. HDAC6 is well known for its deacetylating function on specific cytosolic non-histone substrates that participate in the tumorous genesis, development, and metastasis. The common substrate types are α-tubulin, cortactin, peroxiredoxin, Hsp90, and heat shock transcription factor-1 (HSF-1) [15]. Unlike HDAC6 as an acetyllysine deacetylase, HDAC10 is an N8-acetylspermidine deacetylase, which is associated with dysregulated polyamine metabolism and relevant neoplastic diseases, such as colon cancer, prostate cancer and neuroblastoma [16]. Several studies demonstrated that HDAC10 promotes cell survival through autophagy in response to chemotherapeutic drugs and pathogen infection [16, 17]. Hence, suppression of HDAC10 autophagy may be a novel strategy to sustain the cytotoxicity of cancer chemotherapy, especially for the treatment of advanced-stage neuroblastoma [16]. HDAC11 is the only class IV HDAC due to a unique sequence motif [5, 18]. HDAC11 has deacetylase activity and more efficient defatty-acylase activity, which is significant for lipid metabolism [18]. It is promising for HDAC11 to be an emerging therapeutic target for chronic metabolic diseases [19]. A pan-cancer analysis found that HDAC11 is not a pure oncogenic factor but plays a protective prognostic role in specific cancers (e.g., kidney renal clear cell carcinoma and rectum adenocarcinoma) [20]. The sirtuins (SIRT1-7) comprise the Class III HDACs, taking NAD2+ rather than Zn2+ as its cofactor to facilitate deacetylase activity [5, 21,22,23,24]. In particular, SIRT3-5 is located in the mitochondrial matrix and is associated with metabolic control [22]. Through affecting reactive oxygen species (ROS) and even reactive nitrogen species (RNS), SIRTs regulate the metabolism, cellular reproduction, aging, autophagy, and mitophagy, which are considered promising candidate targets for the exploitation of antitumoral therapies [22,23,24].

Table 1 Classification and features of HDACs

HDACs in hematopoiesis, T-cell development, and the pathogenesis of PTCL

HDACs involve in hematopoietic multilineage development. In Class I, HDAC1 and 2 are regarded as necessary for hematopoietic stem cell (HSC) formation, survival, and homeostasis [25]. In addition, HDAC1 exhibits dynamic expression changes and is involved in lymphoid lineage commitment determination during hematopoiesis and cell differentiation [25]. HDAC1 and 2 repress the Runx3-CBFβ complexes that induce CD8 lineage programs in CD4+ T-cells, leading to the maintenance of CD4 lineage integrity. Loss of either HDAC1 or HDAC2 alone has little effect, but dual inactivation at the early stage results in developmental arrest and reduction in thymocyte numbers and peripheral T-cells, without response to TCR signaling [25,26,27]. HDAC3 is a negative regulator in normal human HSC expansion and development. During T-cell development, HDAC3 is involved in multiple biological processes, such as CD4 and CD8 lineage commitment, positive selection, and peripheral T-cell maturation [25]. Knockdown of HDAC3 in mice blocked hematopoietic progenitor differentiation toward lymphoid lineages and hinder DNA replication [25, 28]. HDAC3-deficient Peripheral T-cells without HDAC3 have an intrinsic defect in their ability to produce TNF efficiently after TCR/CD28 stimulation [25]. HDAC8, upregulated in long-term HSCs and multipotent progenitor cells, is essential for the quiescence, maintenance, and functional integrity of long-term HSCs by modulating p53 activity [25, 29, 30]. In Class II, HDAC5 was reported as a negative regulator for HSC homing and engraftment via regulating p65 deacetylation and CXCR4 surface expression [31]. Besides, Class II HDACs play a pivotal role in the regulation of TCR-mediated apoptosis during T-cell negative selection through interaction with the transcript factor MEF2D [8]. HDAC7 deletion triggers a truncated repertoire of TCR Jα segments, promotes thymocyte apoptosis, and causes inefficient positive selection [25, 32]. In Class III, SIRT1-7 facilitate the maintenance of HSC homeostasis and protect HSCs against aging through different signaling pathways [25]. In Class IV, antagonistic to HDAC6, HDAC11 serves as a transcription repressor of IL-10 expression in antigen-presenting cells, which is essential for T-cell activation [33]. A murine model revealed that activation of resting T-cells triggered lower expression of HDAC11, which enhanced proliferation, proinflammatory cytokine production, and effector molecule expression [34].

HDACs also play a significant role in the malignant transformation of hematologic diseases [25]. Abnormal expression or activity of HDACs has been found in several hematologic malignancies, such as diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and chronic lymphocytic leukemia (CLL), contributing to the epigenetic silencing of tumor suppressor genes and oncogene activation [8, 35]. In a murine model, HDAC1 or 2 haploinsufficiency triggers T-cell lymphomas with global histone acetylation and chromosomal instability [25, 27]. Low levels of HDAC7 and HDAC1 or 2 activity are essential for T-cell lymphoma development [35]. High expression of HDAC1, 2, and 6 frequently occurs in cases with PTCL [36]. Moreover, HDAC6 overexpression in PTCL is associated with poor outcomes [35]. The intrinsic alters of HDACs based on molecular subtypes of PTCL are temporarily under-reported. The pathogenesis of PTCL is summarized as the following three aspects: (i) intracellular derangements, (ii) protumorigenic microenvironment, and (iii) virus-mediated transformation [37]. The involvement of HDACs in the pathogenesis of PTCL from the above aspects is discussed as follows.

Intracellular derangements

HDACs are extensively involved in the regulation of downstream gene networks and signaling pathways, mainly relying on the deacetylation of non-histone substrates such as transcription factors and signaling mediators. For instance, HDAC1, 2, and 3 inhibit transcription of STAT3 target genes in the JAK/STAT pathway, contributing to epigenetic tumor suppressor gene silencing and decreased induction of cell growth arrest or apoptosis [5, 35]. In addition, HDAC1 induces the deacetylation of p53 to repress its function and reduce the level of apoptosis signaling [5]. The acetylation of the K382 site induced by HDACi leads to the decreased degradation-promoting activity of mouse double minute 2 homolog (MDM2) and increased p53 stabilization [38]. In terms of p53 mutants, HDAC1 and 2 are still able to integrate their expression, which was reported in murine pancreatic cancer cells [39]. Moreover, HDAC3 participates in the NF-κB activation via mediating TNF expression in the TCR/CD3 pathway [12, 25, 37].

HDACs promote protein degradation, due to direct competition between acetylation and ubiquitylation for modification of the same lysine residues [6]. TET2 gene mutation frequently occurs in the early stage of the pathogenesis of PTCL. HDAC1 and 2 mediate the deacetylation of the TET2 protein and cause its degradation via the ubiquitin–proteasome pathway [40]. Interestingly, TET2 proteins in breast cancer cells can recruit HDACs to the PD-L1 gene promoter to suppress its transcription, which may be independent of DNA demethylation [41].

Protumorigenic microenvironment

Regulatory T-cells (Tregs) can be selectively recruited by PTCL to dampen the antitumor immune response and promote tumor survival [37]. Different categories of Tregs have been recorded in PTCL subtypes. Suppressor or malignant Tregs can be found in PTCL-NOS and ALCL. Incompetent Tregs, typically with a resting phenotype, emerge into AITL [37]. FOXP3, as a vital transcription factor of Treg, is regulated by various post-translational modifications, including lysine acetylation induced by HDACs [42]. HDAC5 plays a pivotal role in Treg homeostasis [43]. Tregs bereft of HDAC3 or 5 display reduced suppressive function and decreased FOXP3 [25, 44]. Deficiencies in HDAC3 or HDAC5 impair the ability of conventional T-cells to convert into induced Tregs [25, 44]. As opposed to HDAC3 and 5, HDAC6, 9, 10, or 11-defect Tregs exhibit enhanced immunosuppressive capacity and higher expression or acetylation of FOXP3 [20, 42, 43]. Furthermore, HDAC11-defect T-cells are less susceptible to suppression by Tregs in vitro [34]. A meta-analysis based on subgroups of lymphomas demonstrated that there was a significant association between high Tregs and longer OS in Hodgkin lymphoma (HL), DLBCL, and NK/TCL, but it was not discovered in TCL or FL [45].

Cytokine milieu, as one of the critical contributors to lymphomagenesis, plays a pivotal role in defining the T-cell phenotype, modulating gene expression, and integrating T-cell plasticity [37, 46]. Deletion of HDAC1 in Th1 cells and CD8+ T-cells showed increased production of IFN-γ [43]. HDAC2 and 3 are recruited by Kruppel-like factor 4 (KLF-4) at the vascular endothelial growth factor (VEGF) promoter to participate in transcriptional repression of VEGF, which has been first reported in breast cancer [47]. Loss of KLF-4 or HDAC2 and 3 may contribute to the angiogenesis of the development of tumors [47]. DAC3-deficient conventional T-cells produced large amounts of IL-2, IL-6, and IL-17 [44]. Deficiency in HDAC5 caused reduced IFN-γ in CD8+ T-cells but did not affect the proliferation or cytokine expression of CD4+ T-cells [25, 43]. CD4+ T-cells lacking HDAC7 produced elevated levels of IL-2 and IFN-γ [43]. Analogously, T-cells missing HDAC11 promoted cell proliferation and proinflammatory cytokine production (e.g., IL-2 and IFN-γ) and suppressed lymphomatous progression in the murine model [25]. The correlation between serum cytokines and the prognosis of PTCL subtypes was explored by a study, which measured 34 cytokines in serum samples from 121 PTCL patients (PTCL-NOS, n = 55; AITL, n = 44; ALK ALCL, n = 22) [48]. In the AITL group, only IL-10 showed the prognostic value and has been suggested to promote lymphomagenesis through dysregulation of monocyte differentiation, aberrant activation of the JAK2 pathway, and down-regulation of antigen presentation inducing immune escape [48]. In the ALK ALCL group, higher expression of IFN-γ, IL-8, IL-10, IL-17, IL-23, IP-10, MCP-1, MIP-1β, and RANTES were associated with shorter OS, while IFN-γ, IL-8, and RANTES had a further association with a lower CR rate, which indicated that the recruitment of tumor-associated macrophages (TAMs) may be involved in the lymphomagenesis [48]. Cytokines including IFN-γ, IL-7, and IL-23 are associated with poor prognosis in the PTCL-NOS group [48].

Tumor-associated macrophages (TAMs) are regarded as contributors to the poor prognosis of malignancy and are often associated with immune escape and early local or metastatic relapse [49]. One of the VAV1 fusion oncogenes, Vav1-Myo1f was found functioning in inducing recruitment and enrichment of TAMs to the protumorigenic microenvironment in PTCL [50]. Another study revealed that in melanoma, breast cancer, and lung cancer, low-dose pan-HDACi TSA may abrogate the functions of pro-tumoral TAMs to facilitate tumor regression both in vitro and in vivo, with decreased M2-like macrophages and increased M1-like macrophages, which can be a clue for the therapeutic intervention of PTCL [49].

Virus-mediated transformation

Epstein-Barr Virus (EBV) is a γ-herpesvirus and is regarded as an etiological factor in the tumor progression of multiple human malignancies, including lymphoma, gastric carcinoma, and nasopharyngeal carcinoma (NPC) [51]. EBV was reported a frequency of 30–100% in PTCL [52]. Except for occasional PTCL-NOS, EBV infects bystander B cells rather than the neoplastic T lymphocytes in PTCL, which means that EBV plays an indirect role in the pathogenesis of PTCL [37]. A study showed that only AITL was strongly associated with EBV and assigned a score of 3 (defined as “with positive large CD30+ B-cells”) among all subtypes of T-cell lymphoma enrolled [53]. Besides, EBER (EBV-encoded RNA) positivity in the T-cell lymphoma group was statistically and significantly associated with relapse (p < 0·01) [53]. However, up to now, how EBV induces the occurrence or development of AITL has remained unclear and the role of involved HDACs also has not been fully understood [51, 53]. Notably, the HDAC-related EBV infection of other cell lines, such as Burkitt lymphoma [54] and NPC [55], has been reported. In Burkitt lymphoma, Class I HDACi romidepsin potently induces EBV lytic cycle and mediates enhanced cell death with ganciclovir through inhibiting HDAC1, 2, and 3 and upregulating p21 [56]. In the NPC cell lines, EBV latent protein LMP1 upregulates STAT5A and recruits HDAC1/2 to the locus of the CEBPA gene, which is involved in neoplastic plasticity regulation and cellular dedifferentiation [55]. In addition, HDACis (romidepsin and chidamide) have been confirmed their effects in restoring CEBPA expression and reversing cellular dedifferentiation in vitro in EBV+ NPC [55]. Human T-cell leukemia virus type 1 and Kaposi’s sarcoma-associated herpesvirus were reported in isolated PTCL cases [51]. However, the relationship between HDACs and oncogenic viruses excluding EBV has remained a mystery in PTCL.

Histone deacetylase inhibitors

Deregulation of HDACs plays a pivotal role in oncogenesis, and HDACis exert an antitumor effect by facilitating apoptosis, autophagy, and other cell damage processes [21]. HDACis have been confirmed as a class of available targeted agents, used alone or in combination, for various cancers, such as hematological, breast, colorectal, pancreatic, gastric, liver, bladder, and lung cancers, as well as malignant melanoma [15]. There are totally 5 approved HDACis by FDA, but only three are approved agents (romidepsin, belinostat, and chidamide) in clinical application for PTCLs. More details on correlated trials can be found in Table 2 and Fig. 2.

Table 2 Clinical trials for HDACis in PTCLs
Fig. 2
figure 2

Five approved HDACis. To date, five HDACis have been approved by the FDA for the treatment of various cancers. However, only three approved HDACis, including romidepsin, belinostat, and chidamide, are in clinical application for PTCLs

Romidepsin

Romidepsin (FK228), a selective HDAC1 and 2 inhibitor, has been a research hotspot since it was approved by the US Food and Drug Administration (FDA) for the treatment of PTCLs in patients who have received at least one prior therapy [57]. This approval was mainly based on a phase II trial for single-agent romidepsin in relapsed/refractory (R/R) PTCL, with an objective response rate (ORR) of 25% (33/130), a confirmed/unconfirmed complete response (CR/CRu) rate of 15% (19/130), and a 28-month median duration of response (mDOR) for all responders [57, 58]. The most common grade 3/4 adverse events (AEs) were thrombocytopenia (24%), neutropenia (20%), and infections (19%) [58]. In another phase II in Japanese patients with R/R PTCL, a 43% (17/40) ORR, a 25% (10/40) complete response rate (CRR) and a 5.6-month median progression-free survival (mPFS) were responded [59]. Furthermore, the two major PTCL subtypes demonstrated similar ORRs [AITL, 44% (8/18) vs. PTCL-NOS, 41% (7/17)] [59]. The most common treatment-emergent grade 3/4 AEs were lymphopenia (74%) and neutropenia (54%) [59].

Romidepsin induces cell cycle arrest by raising the expression of the p21 tumor-suppressor gene, which is more sensitive to tumor cells [60]. However, romidepsin mediates apoptosis preferentially in terms of cell lines with reduced p21 expression [60]. In addition, romidepsin induces hyperacetylation of the chaperone protein HSP90, thus degrading the oncoproteins that require HSP90, including mutant p53, RAF-1, and BCR-ABL [60]. Moreover, romidepsin has been found to downregulate the expression of angiogenic-stimulating factors and block the migration and adhesion of endothelial cells to inhibit neovascularization and tumor expansion [60].

Romidepsin exhibits single-agent activity in PTCLs, and the efficacy and safety of this compound in various combination regimens have been widely evaluated. Of note, in a randomized phase III study for previously untreated PTCLs, the therapeutic strategy of romidepsin plus CHOP (Ro-CHOP, n = 211) did not achieve marked advantages in efficacy compared to CHOP (n = 210), with evaluations in mPFS (12.0 vs. 10.2 months), median overall survival (mOS) (51.8 vs. 42.9 months), ORR (63% vs. 60%), and CR/CRu rate (41% vs. 37%) [61]. Additionally, Ro-CHOP increased the frequency of grade ≥ 3 treatment-emergent AEs, including thrombocytopenia (50% vs. 10%), neutropenia (49% vs. 33%), anemia (47% vs. 17%), and leukopenia (32% vs. 20%) [61]. Combination regimens for other novel agents with romidepsin in PTCLs have also been explored: 1) The multiple mutations in epigenetic modifier genes (e.g., TET2, IDH2, and DNMT3A) and TCR-related genes (e.g., RHOA and FYN) may impair immunogenicity of PTCL and facilitate immune escape [62]. In a phase II trial (NCT03278782) of pembrolizumab (a check-point PD-1 inhibitor) plus romidepsin in R/R TCL, a 50% (7/14) ORR and a 35.7% (5/14) CRR were recorded but no responses were achieved in the 3 patients with mycosis fungoides (MF) [62]. A higher level of PD-L1 was found in CRs than in PR or SD [62]. 2) A strong synergistic interaction between DNA methyltransferase (DNMT) inhibitor and HDACis has been found in TCL models, through the down-regulation of genes involved in protein and lipid biosynthesis and up-regulation of molecules involved in protein kinase cascade and cell cycle arrest [63]. Markedly, combined romidepsin and 5-azacytidine (an oral DNMTi) demonstrated favorable therapeutic outcomes in patients with T-FHCL, a subtype of PTCL especially vulnerable to epigenetic modifiers, exhibiting an ORR of 80% (12/15), a CRR of 60% (9/15), and lower toxicity [64]. 3) Aurora A kinase (AAK) inhibitor alisertib (MLN8237) and romidepsin appear to be highly synergistic in the TCL model through modulation of mitotic proteins and cytokinesis failure, which has an association with HDAC3 function [65]. In a phase I trial (NCT01897012) in R/R BCL and TCL, the combination of alisertib and romidepsin resulted in an ORR of 28% (7/25) and a CRR of 12% (3/25) [66]. Only one case of AITL was enrolled and achieved no response [66]. 4) A cytotoxic drug bendamustine functions in the promotion of p53-dependent apoptosis with DNA breaks and generation of mitochondrial-mediated ROS and apoptosis in a non-p53-dependent way, which is similar to romidepsin [67]. A study in 7 patients with R/R PTCL showed that romidepsin plus bendamustine attained 2 CRs, 1 PR, and an mPFS of 7 months with nausea and vomiting as the most predominant AE [67]. 5) PI3Ki has been found synergistic effects with HDACi in vitro and in vivo in non-Hodgkin lymphoma (NHL), via induction of DNA damage, downregulation of Mcl-1, and upregulation of Bim protein [68]. Duvelisib, a PI3K-δ,γ inhibitor, combined with romidepsin was evaluated in R/R PTCL and cutaneous T-cell lymphoma (CTCL) in a phase I trial (NCT02783625) [69]. For patients with PTCL, this regimen achieved a 47.2% (17/36) ORR and a 25.0% (9/36) CRR with a lower rate of transaminitis than single-agent duvelisib [69]. Of note, there was a grade 5 AE, Stevens-Johnson syndrome, which was considered possibly treatment-related [69]. A phase I/II study was designed to evaluate a new combination regimen of romidepsin plus tenalisib (RP6530, a highly selective PI3K δ/γ and SIK3 inhibitor) in R/R PTCL and CTCL [70]. Twelve evaluable R/R PTCL patients responded with an ORR of 75% (9/12), a CRR of 50 (6/12) and a 5.03-month median duration of response (mDOR) [70]. 6) The novel antifolate dihydrofolate reductase (DHFR) inhibitor pralatrexate was the first drug approved for patients with R/R PTCL in 2009 [71]. The synergistic effect of pralatrexate and romidepsin was attributed to the inhibition of DNA synthesis and repair, which was studied in vitro in a murine model of human TCL [72]. This combination regimen led to an ORR of 71% (10/14) and a CRR of 29% (4/14) in patients with R/R PTCL in a phase I study (NCT01947140) [71]. For the R/R PTCL group, the median DOR, PFS, and OS were 4.29, 4.4, and 12.4 months, respectively [71]. 7) Cereblon (CRBN) inhibitor lenalidomide has been approved by FDA for various hematologic malignancies with antineoplastic effects of repressing angiogenesis and intensifying immune responses [73]. Lenalidomide and romidepsin had a synergistic or additive effect in specific TCL cell lines via various induced ways, such as induction of apoptosis, increased production of reactive oxygen species, and promotion of endoplasmic reticulum stress [74]. In addition, carfilzomib is an irreversible proteasome inhibitor approved for the treatment of multiple myeloma (MM) [73]. In a combined analysis of two phase I studies (NCT01947140 and NCT01755975), two regimens (A: romidepsin and lenalidomide; B: romidepsin, lenalidomide, and carfilzomib) were carefully compared in every respect [73]. In the R/R PTCL group, ORRs were similar in two regimens (A: 53%, 8/15 vs. B: 54%, 7/13), but regimen B had a superior CRR than regimen A (A: 13%, 2/15 vs. B: 39%, 5/13) [73]. Compared to regimen A, regimen B showed lower chemotherapeutic toxicity with fewer AEs [73]. However, the AEs of the R/R PTCL group were not acquired. 8) Gemcitabine, as a pyrimidine anti-metabolite, demonstrated single-agent activity in R/R PTCL with an ORR of up to 51% (20/39) and a CRR of 23% (9/39) [75, 76]. However, in a phase II study (NCT01822886), the GEMRO regimen (romidepsin plus gemcitabine) did not show additional advantages on efficacy or safety over single agent romidepsin with a 30% (6/20) ORR and a 15% (3/20) CRR [75]. Another combined regimen of romidepsin plus GDP (gemcitabine, dexamethasone, and cisplatin) was explored in R/R PTCL and DLBCL in a phase I trial (NCT01846390) [77]. Among the PTCL patients, the ORR, 1-year PFS, 1-year OS, mPFS, and mOS were 60% (6/10), 30%, 52%, 5.45 months, and 15.08 months, respectively [77]. The AEs occurring in PTCL were not acquired with a further illustration. 9) Anthracycline antibiotics doxorubicin and romidepsin were reported synergistic in growth inhibition and apoptosis induction in both CTCL cell lines and patient-derived primary CTCL cells [78]. Compared to doxorubicin, liposomal doxorubicin (LD) shows lower cardiotoxicity through reduced myocardial drug accumulation and induction of IFN-related DNA damage resistance [79]. In a phase I trial (NCT01902225), the combination of romidepsin and LD was evaluated in patients with R/R CTCL or PTCL [78]. This regimen demonstrated superior efficacy in CTCL than PTCL in terms of ORR (70% vs. 27%), mPFS (6.9 months vs. 2.1 months), mOS (not reached vs. 17.5 months), the median time to response (mTTR) (2 months vs. 3.5 months), and mDOR (5.1 months vs. 4.2 months) [78]. Moreover, grade 3/4 hematologic AEs occurred more frequently in the PTCL cohort [78]. Markedly, there were no cardiac-related AEs despite high anthracycline exposure [78].

Belinostat

As a broad-spectrum HDACi and a second-generation analog of vorinostat, belinostat (PXD101) shows antiangiogenic properties and preferential cytotoxicity toward tumor cells, resulting in the apoptosis and cell cycle arrest of some transformed cells [80, 81]. Belinostat was approved by the FDA for the single-agent treatment of R/R PTCL in 2014, based on a phase II single-arm BELIEF trial (NCT00865969) including 120 evaluable cases with R/R PTCL [82,83,84]. Monotherapy with belinostat yielded an ORR of approximately 25%, also reported in another early phase II trial (NCT00274651) with a CRR of 8.3% (2/24) in the R/R PTCL cohort [83, 85]. In the former, severe AEs occurred in 47.3% (61/129) of patients, recording a higher frequency in nonhematologic AEs [83]. However, one case that died from toxic liver failure without complications was attributed to belinostat [83]. The rare hepatotoxicity of belinostat was presumed to be a result of the sulfonamide-like hepatic reaction due to its chemical properties of sulfonamide hydroxyamide [86].

The exploration of combinatorial therapy for belinostat is still underway. In a phase I trial for newly diagnosed PTCLs (NCT01839097), the addition of belinostat to CHOP (Bel-CHOP) achieved 86% (6/7, 12/14) ORRs in both cohorts with different doses of belinostat and the same dose of CHOP [87]. Of the cases, 43% experienced severe AEs, with febrile neutropenia (17%) occurring most [87]. Synergistic effects between belinostat and proteasome inhibitor ixazomib [88] or dihydrofolate reductase inhibitor pralatrexate [89] have been demonstrated in T-cell lymphoma (TCL) in vitro, providing clues for combination drug trials. In addition, a scale liposomal subcutaneous delivery system of belinostat has been successfully designed for PTCLs to prolong the short half-life of 1.1 h and improve the burst effect and pharmacokinetic properties [90]. However, this novel system temporarily lacks test data in the human body.

Chidamide

Chidamide (Tucidinostat), a novel selective inhibitor targeting HDAC1, 2, 3, and 10, has demonstrated broad-spectrum antitumor activity. Previous research suggests that chidamide causes tumor cell growth arrest and apoptosis, promotes cellular antitumor immunity, and reverses epithelial–mesenchymal transitions and drug resistance [91]. It acquired approval from the China Food and Drug Administration in 2014 for the treatment of R/R PTCL, based on a multicenter, pivotal phase II trial for R/R PTCL, in which a 28% (22/79) ORR and a 14% (11/79) CR/CRu rate were reported [91]. Angioimmunoblastic T-cell lymphoma (AITL) cases appeared to be more susceptive, with a higher ORR (50%, 5/10) and a CR/CRu rate (40%, 4/10) [91]. For adult T-cell leukemia/lymphoma (ATLL), a 30.4% (7/23) ORR, a 4.3% (1/23) CRR, and an mPFS of 1.7 months were achieved in a phase IIb trial (NCT02955589) of single-agent chidamide [92].

Chidamide-based combined regimens for untreated PTCLs have been studied in recent years. A phase I trial (NCT02809573) and a phase II trial (NCT02987244) evaluated the addition of chidamide to CHOP and CHOEP (CHOP with etoposide) regimens in untreated PTCLs, respectively, with ORRs of 89.3% and 60.2% and CRRs of 57.1% and 40.7% [93, 94]. However, in research on the Chi-CHOEP regimen, patients with AITL showed significantly inferior outcomes with an mPFS of 9.6 months, compared to anaplastic lymphoma kinase-negative anaplastic large cell lymphoma (ALKALCL) (26.0 months) and PTCL-NOS (19.4 months) [94]. Leukopenia and neutropenia were the most common grade 3/4 hematological AEs in both trials [93, 94]. Another chemotherapy regimen, the CPET regimen (chidamide with prednisone, etoposide, and thalidomide), was evaluated among AITL patients in a multicenter phase II trial (NCT03273452). The ORR and CRR of 51 evaluable cases were 90.2% and 54.9%, respectively [40]. The most common grade 3/4 AE was neutropenia (32.3%), while the other AEs were mainly mild and reversible [40].

Panobinostat

Panobinostat (LBH-589), a potent oral pan-HDACi, has been reported to have a highly synergistic effect with proteasome inhibitor bortezomib. Their synergy is seemingly attributed to the concurrent inhibition of proteasome and HDAC6, contributing to the accumulation of polyubiquitinated proteins and increased cell stress and apoptosis. In an open-label, multicenter phase II trial (NCT00901147), this combination regimen was evaluated in 25 cases with R/R PTCL, with the majority subtypes being PTCL-NOS (9, 36%) and AITL (8, 32%) [95]. A 43.5% (10/23) ORR and a 21.7% (5/23) CRR were recorded in assessable patients, while two cases discontinued treatment due to peripheral neuropathy, diarrhea, and acute coronary syndrome [95].

Vorinostat

Vorinostat (SAHA) is an oral pan-HDACi approved for the treatment of R/R CTCL by the FDA in 2006 [82]. In vitro, vorinostat triggers growth arrest and caspase-dependent apoptotic and caspase-independent autophagic cell death [96]. A phase I trial (NCT00787527) evaluated the safety and efficacy of vorinostat combined with six cycles of CHOP in newly diagnosed PTCLs. In addition to 2 cases with premature treatment termination, the remaining 12 all achieved complete remission, whereas 4 experienced disease recurrence [97]. The most common toxicity related to vorinostat was estimated to be diarrhea [97]. In another phase I/II trial (NCT00972842), the regimen of vorinostat in combination with lenalidomide and dexamethasone did not demonstrate satisfactory results, with one complete remission and one partial remission [98]. The regimen of vorinostat plus alisertib was studied in lymphoid malignancies in a phase I trial (NCT01567709) [99]. An 11.8% (4/34) ORR and two CRs only with DLBCL were reported among all cohorts with different dose levels [99]. Two cases of PTCL were enrolled but achieved no response [99]. More vorinostat-based combination therapies for PTCLs are temporarily absent.

Other potential HDACis for PTCL

Generally, the development of novel agent appeared to be a long-term and intricate procedure. There are many potential HDACis remaining non-approved and not acquiring comprehensive clinical evaluation in PTCL, as shown in Table 3 and Fig. 3.

Table 3 Other potential non-approved HDACis for PTCL
Fig. 3
figure 3

6 potential non-approved HDACis for PTCL. These potential HDACis remaining non-approved and not acquiring comprehensive clinical evaluation in PTCL

Nanatinostat (VRx-3996), selectively targeting Class-I HDACs, can induce cell apoptosis and inhibition of viral and cellular DNA synthesis in EBV+ tumor cells, via upregulating the lytic BGLF4 EBV protein kinase and activating ganciclovir [52]. In the phase Ib/II VT3996-201 study (NCT03397706) with 55 patients enrolled (PTCL-NOS, n = 5; AITL, n = 6), Nanatinostat in combined with valganciclovir (VGCV) achieved a 40% (17/43) ORR and a 19% (8/43) CRR in 43 evaluable patients with histologically confirmed EBV+ lymphomas [52]. The most common grade 3/4 AEs across all histologic types included neutropenia (27%), thrombocytopenia (20%), anemia (20%), and lymphopenia (14%) [52].

Ricolinostat (ACY-1215), an oral selective HDAC6 inhibitor with a weak potency against HDAC1 and 2, can disrupt protein homeostasis via the unfolded protein response to induce programmed cell death [100]. In a single-agent phase Ib/II study of ricolinostat (NCT02091063), no complete or partial response was recorded in 16 evaluable R/R lymphoma patients enrolled, despite no dose-limiting toxicities being observed [100]. A combined regimen dually targeting protein degradation pathways, ricolinostat plus proteasome inhibitor bortezomib, was reported profound synergism in both cell-based and in vivo studies [101]. Citarinostat (ACY-241) is a second-generation selective HDAC6 inhibitor, yet with greater inhibition of Class I HDACs due to higher serum concentrations and a form of tablet formulation [100]. Citarinostat plus momelotinib, a JAK1/2 inhibitor, was reported anticancer effects in vitro in hematological malignant cell lines (including FL, MCL, CTCL, ALCL, HL, MM, and CLL) [102]. Multiple cell death mechanisms were recorded, such as activating mitochondrial apoptosis pathways, elevating ROS production, facilitating ER stress, and modulating cell cycle perturbation [102]. Most cell lines showed well susceptibilities to this combined regimen excluding two specific cell lines (Granta-519 and L-1236) [102].

Selective HDAC8 inhibitor PCI-34051 induces caspase-dependent apoptosis in T-cell lymphomas or leukemias cell lines [103]. Deficiency in phospholipase C-γ1 (PLCγ1) rather than in TCR signaling may impair the sensitivity to PCI-34051 [103]. In addition, the tumor-suppressive effect of PCI-34051 was reported as T-cell-dependent and calcium-induced, increasing tumor-infiltrating CD8 T-cells in a preclinical model of hepatocellular carcinoma (HCC) [104]. CD8 T-cell depletion, regulatory T-cell adoptive transfer, Ca2+ chelators may cause PCI-34051 to fail in antitumor effect [104]. Without any obvious indications of toxicity, PCI-34051 enhanced the removal of existing hepatomas by anti-PD-L1 therapy [104]. Besides, the synergistic effect of ALK inhibitor and PCI-34051 was reported both in vitro and in vivo in neuroblastoma via blocking the activation of growth receptor survival signaling and shifting the cell cycle arrest [105]. Despite multiple preclinical tests, no clinical trial of PCI-34051 in any malignancy has been registered.

Natural HDAC1 and 8 inhibitor baicalein has been studied in multiple diseases, mainly exhibits antitumor, anti-inflammatory, and anti-fibrotic activities [38]. Baicalein was observed to induce a dose dependent cell death in TCL in vitro via inhibition of thioredoxin system [106]. In another preclinical test in CTCL cell lines, baicalein significantly induced cell apoptosis with a p53 wild type via dramatically raising the level of higher level of acetylation of p53 and proteasome-dependent degradation of HDAC1 [38]. Baicalein appeared superior to traditional HDACis in CTCL due to no influence on the expression of ATP-binding cassette transporter genes [38]. However, existing clinical trials of baicalein have been limited to study its role in influenza.

AR-42, as a pan-HDACi targeting Class I and IIb HDAC, shows antitumor activity in in vitro and in vivo numerous models of solid tumors and hematologic malignancies [107]. In multiple preclinical lymphoma models, AR-42 exhibits a more potent activity than vorinostat [107]. In a phase I trial (NCT01129193) of AR-42, the best response in MM was the minimal response in 17.6% (3/17) patients, while In patients with relapsed lymphoma was a stable disease [107]. Only one PTCL patient that was diagnosed as EBV+ AITL was enrolled and achieved progressive disease [107].

Discussion and future perspectives

HDACs are one of the keys to epigenetic regulation involved in tumorigenesis and tumor maintenance, being promising targets for antitumor agent discovery and development. HDACis have been confirmed to be targeted and available for various tumors, including PTCLs, to improve the poor therapeutic outcomes of the historical standard frontline approach, such as CHOP or CHOP-like regimens. Accumulating evidence suggests that AITL and other T follicular cell-originated PTCL subtypes characteristic of epigenetic disruption have a unique vulnerability to epigenetic inhibitors, including HDACis. However, no additional benefit has been observed by adding chidamide to the CHOEP regimen in untreated AITL, though the high single-agent activity of chidamide has been validated in R/R AITL (50% ORR and 40% CRR) [91, 94]. Hence, it is considered that AITL cases might not significantly profit from intensive chemotherapy.

With the advanced application of next-generation sequencing (NGS), the correlation between mutation profiles, especially in epigenetic genes, and targeted drug therapeutic effects deserves much attention. In a multicenter phase II study (NCT01998035) of R/R PTCL, mutations of genes involved in DNA methylation, histone methylation, or histone acetylation were found more frequently in patients responding to 5-azacytidine plus romidepsin [64]. Clinical responses were more seen in TET2 gene-mutated patients (n = 16; ORR 69%; CRR 53%) compared to wild-type mutations (n = 5; ORR 40%; CRR 20%), with no statistically significant differences due to small sample size [64]. Whereas, mutation analysis based on NGS may serve as a promising and sensitive biomarker for predicting response and estimating the vulnerability to targeted therapy in patients with PTCL.

Despite the approval of several HDACis for specific cancer treatments, the outcomes of HDACi monotherapies in PTCLs are unsatisfactory and sporadically drug-resistant. Thus, combination therapies of HDACi are still being actively explored to overcome drug resistance and compensatory pathways caused by single target as well as to minimize the side effects. The addition of different kinds of HDACis to CHOP or CHOP-like regimens is not completely extra-beneficial for PTCLs or specific subtypes, as mentioned above. HDACi-based regimens plus other novel targeted agents, such as pembrolizumab [62], 5-azacitidine [64], bendamustine [67], duvelisib [69], tenalisib [70], pralatrexate [71], lenalidomide [73], and carfilzomib [73], hold promising futures for more effective and safer therapeutic outcomes. Combining HDACis with radiotherapy and phototherapy provides new strategies for antitumor therapies due to the inhibition of DNA repair with HDACis [108].

Dual inhibitors, which simultaneously aim at two or more targets, may offer more excellent therapeutic benefits over single-acting agents in overcoming drug resistance and amplifying synergistic effects. The discovery and development of HDACi-based multitarget antitumor agents have achieved initial success. Several dual HDAC inhibitors have been accomplished in preclinical tests or early clinical trials (as shown in Fig. 4), such as bromodomain and extra-terminal (BET)/HDAC inhibitors [109], various kinase/HDAC inhibitors [110, 111], STAT3/HDAC inhibitors [112], and Nicotinamide Phosphoribosyltransferase (NAMPT)/HDAC inhibitors [113]. For instance, a novel Janus Kinase (JAK)/HDAC dual inhibitor targeting both JAK2 and HDAC6 has exhibited improved antiproliferative and proapoptotic activities over vorinostat and ruxolitinib in several hematological cell lines [111]. Moreover, a dual PI3K/HDAC inhibitor BEBT-908 can promote a proinflammatory tumor microenvironment and induce immunogenic ferroptosis to synergize with immunotherapy [114]. Of note, BEBT-908 has been further studied in virous clinical trials in China, such as R/R PTCL (Phase II, CTR20210170), R/R DLBCL (Phase II, CTR20200035), advanced non-small cell lung cancer (Phase Ib/II, CTR20213331), and advanced recurrent or metastatic HR+/HER2 breast cancer (Phase Ib/II, CTR20213267). More related details can be found on www.chinadrugtrials.org.cn. In general, the therapeutic potential of novel dual HDACis urgently needs to be further confirmed in clinical trials.

Fig. 4
figure 4

Five kinds of dual HDAC inhibitors in preclinical tests or early clinical trials. Dual HDAC inhibitors may offer more excellent therapeutic benefits in overcoming drug resistance and amplifying synergistic effects. However, the therapeutic potential of novel dual HDACis is urgently warranted further confirmation in clinical trials

Apart from restricted combination regimens and underway multitargeted development, the potential clinical benefits of HDACis are also limited by the insufficient physicochemical properties, selectivity, and potency, which results in inconstant off-target effects and undesirable side effects. HDACi-based prodrugs, an inactive form needing biochemical transformations, are considered an optimistic way to improve medication performance. HDACi-based prodrug strategies are often utilized to enhance targeted location accessibility of drug delivery and overcome deficiencies in the physicochemical properties, such as stability, aqueous solubility, lipophilicity, and oral bioavailability [115]. Of note, romidepsin has prodrug properties and favorable pharmacokinetic properties, making it inactive and stable in blood circulation but active after uptake into tumor cells and resulting in intracellular reduction [115]. Despite plenty of novel HDACi-based prodrugs having been developed, most of the reported ones still need to be thoroughly investigated in terms of in vivo characteristics.

Conclusions

HDACis have been extensively studied in the treatment of PTCLs, used either alone or in combination. The currently approved HDACis usually demonstrate moderate single-agent activity and controllable incidence of hematological AEs. However, more effective and safe agents or combination regimens are urgently needed to improve the poor prognosis of PTCL patients. Mutation analysis based on NGS may help to predict the efficacy of targeted therapy in PTCL. The development of multitargeted HDACis and HDACi-based prodrugs is a promising strategy, but their efficacy and safety deserve more clinical data to allow verification.

References

  1. Alaggio R, Amador C, Anagnostopoulos I, Attygalle AD, Araujo IBO, Berti E, et al. The 5th edition of the World Health Organization classification of haematolymphoid tumours: lymphoid neoplasms. Leukemia. 2022;36(7):1720–48.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Savage KJ. Therapies for peripheral T-cell lymphomas. Hematology. 2011;2011(1):515–24.

    Article  PubMed  Google Scholar 

  3. Bellei M, Foss FM, Shustov AR, Horwitz SM, Marcheselli L, Kim WS, et al. The outcome of peripheral T-cell lymphoma patients failing first-line therapy: a report from the prospective, International T-cell project. Haematologica. 2018;103(7):1191–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Carpio LR, Bradley EW, McGee-Lawrence ME, Weivoda MM, Poston DD, Dudakovic A, et al. Histone deacetylase 3 supports endochondral bone formation by controlling cytokine signaling and matrix remodeling. Sci Signal. 2016;9(440):ra79.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Chun P. Histone deacetylase inhibitors in hematological malignancies and solid tumors. Arch Pharmacal Res. 2015;38(6):933–49.

    Article  CAS  Google Scholar 

  6. Narita T, Weinert BT, Choudhary C. Functions and mechanisms of non-histone protein acetylation. Nat Rev Mol Cell Biol. 2019;20(3):156–74.

    Article  CAS  PubMed  Google Scholar 

  7. New M, Olzscha H, La Thangue NB. HDAC inhibitor-based therapies: can we interpret the code? Mol Oncol. 2012;6(6):637–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Haery L, Thompson RC, Gilmore TD. Histone acetyltransferases and histone deacetylases in B- and T-cell development, physiology and malignancy. Genes Cancer. 2015;6(5–6):184–213.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Waltregny D, Glénisson W, Tran SL, North BJ, Verdin E, Colige A, et al. Histone deacetylase HDAC8 associates with smooth muscle α-actin and is essential for smooth muscle cell contractility. FASEB J. 2005;19(8):966–8.

    Article  CAS  PubMed  Google Scholar 

  10. Gao Y, Nihira NT, Bu X, Chu C, Zhang J, Kolodziejczyk A, et al. Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy. Nat Cell Biol. 2020;22(9):1064–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ishii S, Kurasawa Y, Wong J, Yu-Lee LY. Histone deacetylase 3 localizes to the mitotic spindle and is required for kinetochore-microtubule attachment. Proc Natl Acad Sci U S A. 2008;105(11):4179–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yang L, Chen S, Xia J, Zhou Y, Peng L, Fan H, et al. Histone deacetylase 3 facilitates TNFα-mediated NF-κB activation through suppressing CTSB induced RIP1 degradation and is required for host defense against bacterial infection. Cell Biosci. 2022;12(1):81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kang Y, Nian H, Rajendran P, Kim E, Dashwood WM, Pinto JT, et al. HDAC8 and STAT3 repress BMF gene activity in colon cancer cells. Cell Death Dis. 2014;5(10):e1476–576.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Edwards AJ, Pender SL. Histone deacetylase inhibitors and their potential role in inflammatory bowel diseases. Biochem Soc Trans. 2011;39(4):1092–5.

    Article  CAS  PubMed  Google Scholar 

  15. Pulya S, Amin SA, Adhikari N, Biswas S, Jha T, Ghosh B. HDAC6 as privileged target in drug discovery: a perspective. Pharmacol Res. 2021;163:105274.

    Article  CAS  PubMed  Google Scholar 

  16. Hai Y, Shinsky SA, Porter NJ, Christianson DW. Histone deacetylase 10 structure and molecular function as a polyamine deacetylase. Nat Commun. 2017;8(1):15368.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhou W, Wang J, Wang X, Wang B, Zhao Z, Fu J, et al. Degradation of HDAC10 by autophagy promotes IRF3-mediated antiviral innate immune responses. Sci Signal. 2022;15(765):eabo4356.

    Article  CAS  PubMed  Google Scholar 

  18. Liu S-S, Wu F, Jin Y-M, Chang W-Q, Xu T-M. HDAC11: a rising star in epigenetics. Biomed Pharmacother. 2020;131:110607.

    Article  CAS  PubMed  Google Scholar 

  19. Chen H, Xie C, Chen Q, Zhuang S. HDAC11, an emerging therapeutic target for metabolic disorders. Front Endocrinol (Lausanne). 2022;13:989305.

    Article  PubMed  Google Scholar 

  20. Li R, Wu X, Zhao P, Xue K, Li J. A pan-cancer analysis identifies HDAC11 as an immunological and prognostic biomarker. FASEB J. 2022;36(7):e22326.

    Article  CAS  PubMed  Google Scholar 

  21. Movafagh S, Munson A. Chapter 4 - histone deacetylase inhibitors in cancer prevention and therapy. In: Bishayee A, Bhatia D, editors. Epigenetics of cancer prevention, vol. 8. Cambridge: Academic Press; 2019. p. 75–105.

    Chapter  Google Scholar 

  22. Ji Z, Liu GH, Qu J. Mitochondrial sirtuins, metabolism, and aging. J Genet Genomics. 2022;49(4):287–98.

    Article  PubMed  Google Scholar 

  23. Aventaggiato M, Vernucci E, Barreca F, Russo MA, Tafani M. Sirtuins’ control of autophagy and mitophagy in cancer. Pharmacol Ther. 2021;221:107748.

    Article  CAS  PubMed  Google Scholar 

  24. Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N. The role of sirtuins in antioxidant and redox signaling. Antioxid Redox Signal. 2018;28(8):643–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang P, Wang Z, Liu J. Role of HDACs in normal and malignant hematopoiesis. Mol Cancer. 2020;19(1):1–21.

    CAS  Google Scholar 

  26. Wada T, Kikuchi J, Nishimura N, Shimizu R, Kitamura T, Furukawa Y. Expression levels of histone deacetylases determine the cell fate of hematopoietic progenitors. J Biol Chem. 2009;284(44):30673–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Dovey OM, Foster CT, Conte N, Edwards SA, Edwards JM, Singh R, et al. Histone deacetylase 1 and 2 are essential for normal T-cell development and genomic stability in mice. Blood. 2013;121(8):1335–44.

    Article  CAS  PubMed  Google Scholar 

  28. Summers AR, Fischer MA, Stengel KR, Zhao Y, Kaiser JF, Wells CE, et al. HDAC3 is essential for DNA replication in hematopoietic progenitor cells. J Clin Invest. 2013;123(7):3112–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hua W-K, Qi J, Cai Q, Carnahan E, Li L, Ngo VN, et al. HDAC8 regulates long-term hematopoietic stem cell quiescence and maintenance. Blood. 2016;128(22):1468–568.

    Article  Google Scholar 

  30. Hua W-K, Qi J, Cai Q, Carnahan E, Ramirez MA, Li L, et al. HDAC8 regulates long-term hematopoietic stem cell maintenance under stress by modulating p53 activity. Blood. 2017;130(Supplement 1):1146–246.

    Google Scholar 

  31. Huang X, Guo B, Liu S, Wan J, Broxmeyer HE. Neutralizing negative epigenetic regulation by HDAC5 enhances human haematopoietic stem cell homing and engraftment. Nat Commun. 2018;9(1):2741.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Kasler HG, Lee IS, Lim HW, Verdin E. Histone deacetylase 7 mediates tissue-specific autoimmunity via control of innate effector function in invariant Natural killer T cells. Elife. 2018;7:e32109.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Cheng F, Lienlaf M, Perez-Villarroel P, Wang HW, Lee C, Woan K, et al. Divergent roles of histone deacetylase 6 (HDAC6) and histone deacetylase 11 (HDAC11) on the transcriptional regulation of IL10 in antigen presenting cells. Mol Immunol. 2014;60(1):44–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Woods DM, Woan KV, Cheng F, Sodré AL, Wang D, Wu Y, et al. T cells lacking HDAC11 have increased effector functions and mediate enhanced alloreactivity in a murine model. Blood. 2017;130(2):146–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhang P, Zhang M. Epigenetic alterations and advancement of treatment in peripheral T-cell lymphoma. Clin Epigenet. 2020;12(1):169.

    Article  CAS  Google Scholar 

  36. Marquard L, Poulsen CB, Gjerdrum LM, de Nully BP, Christensen IJ, Jensen PB, et al. Histone deacetylase 1, 2, 6 and acetylated histone H4 in B- and T-cell lymphomas. Histopathology. 2009;54(6):688–98.

    Article  PubMed  Google Scholar 

  37. Pizzi M, Margolskee E, Inghirami G. Pathogenesis of peripheral T cell lymphoma. Annu Rev Pathol. 2018;13(1):293–320.

    Article  CAS  PubMed  Google Scholar 

  38. Yu X, Li H, Zhu M, Hu P, Liu X, Qing Y, et al. Involvement of p53 acetylation in growth suppression of cutaneous T-cell lymphomas induced by HDAC inhibition. J Investig Dermatol. 2020;140(10):2009-22.e4.

    Article  CAS  PubMed  Google Scholar 

  39. Stojanovic N, Hassan Z, Wirth M, Wenzel P, Beyer M, Schäfer C, et al. HDAC1 and HDAC2 integrate the expression of p53 mutants in pancreatic cancer. Oncogene. 2017;36(13):1804–15.

    Article  CAS  PubMed  Google Scholar 

  40. Wang Y, Zhang M, Song W, Cai Q, Zhang L, Sun X, et al. Chidamide plus prednisone, etoposide, and thalidomide for untreated angioimmunoblastic T-cell lymphoma in a Chinese population: a multicenter phase II trial. Am J Hematol. 2022;97(5):623–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Shen Y, Liu L, Wang M, Xu B, Lyu R, Shi YG, et al. TET2 inhibits PD-L1 gene expression in breast cancer cells through histone deacetylation. Cancers. 2021;13(9):2207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Dahiya S, Beier UH, Wang L, Han R, Jiao J, Akimova T, et al. HDAC10 deletion promotes Foxp3+ T-regulatory cell function. Sci Rep. 2020;10(1):424.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ellmeier W, Seiser C. Histone deacetylase function in CD4+ T cells. Nat Rev Immunol. 2018;18(10):617–34.

    Article  CAS  PubMed  Google Scholar 

  44. Wang L, Liu Y, Han R, Beier UH, Bhatti TR, Akimova T, et al. FOXP3+ regulatory T cell development and function require histone/protein deacetylase 3. J Clin Investig. 2015;125(3):1111–23.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Peng F, Qin Y, Mu S, Li J, Ai L, Hu Y. Prognostic role of regulatory T cells in lymphoma: a systematic review and meta-analysis. J Cancer Res Clin Oncol. 2020;146(12):3123–35.

    Article  CAS  PubMed  Google Scholar 

  46. Fiore D, Cappelli LV, Broccoli A, Zinzani PL, Chan WC, Inghirami G. Peripheral T cell lymphomas: from the bench to the clinic. Nat Rev Cancer. 2020;20(6):323–42.

    Article  CAS  PubMed  Google Scholar 

  47. Ray A, Alalem M, Ray B. Loss of epigenetic Kruppel-like factor 4 histone deacetylase (KLF-4-HDAC)-mediated transcriptional suppression is crucial in increasing vascular endothelial growth factor (VEGF) expression in breast cancer. J Biol Chem. 2013;288:27232–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yi JH, Ryu KJ, Ko YH, Kim WS, Kim SJ. Profiles of serum cytokines and their clinical implications in patients with peripheral T-cell lymphoma. Cytokine. 2019;113:371–9.

    Article  CAS  PubMed  Google Scholar 

  49. Li X, Su X, Liu R, Pan Y, Fang J, Cao L, et al. HDAC inhibition potentiates anti-tumor activity of macrophages and enhances anti-PD-L1-mediated tumor suppression. Oncogene. 2021;40(10):1836–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Cortes JR, Filip I, Albero R, Patiño-Galindo JA, Quinn SA, Lin W-HW, et al. Oncogenic Vav1-Myo1f induces therapeutically targetable macrophage-rich tumor microenvironment in peripheral T cell lymphoma. Cell Rep. 2022;39(3):110695.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Nakhoul H, Lin Z, Wang X, Roberts C, Dong Y, Flemington E. High-throughput sequence analysis of peripheral T-cell lymphomas indicates subtype-specific viral gene expression patterns and immune cell microenvironments. mSphere. 2019;4(4):e00248–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Haverkos BM, Alpdogan O, Baiocchi R, Brammer JE, Feldman TA, Capra M, et al. Nanatinostat (Nstat) and Valganciclovir (VGCV) in relapsed/refractory (R/R) Epstein-Barr virus-positive (EBV +) lymphomas: final results from the phase 1b/2 VT3996-201 study. Blood. 2021;138(Supplement 1):623–723.

    Article  Google Scholar 

  53. Donzel M, Bonjour M, Combes J-D, Broussais F, Sesques P, Traverse-Glehen A, et al. Lymphomas associated with Epstein-Barr virus infection in 2020: results from a large, unselected case series in France. eClinicalMedicine. 2022;54:101674.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Li S, Yang L, Li Y, Yue W, Xin S, Li J, et al. Epstein-Barr virus synergizes with BRD7 to conquer c-Myc-mediated viral latency maintenance via chromatin remodeling. Microbiol Spectr. 2023;11(2):e0123722.

    Article  PubMed  Google Scholar 

  55. Xie J, Wang Z, Fan W, Liu Y, Liu F, Wan X, et al. Targeting cancer cell plasticity by HDAC inhibition to reverse EBV-induced dedifferentiation in nasopharyngeal carcinoma. Signal Transduct Target Ther. 2021;6(1):333.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hui KF, Cheung AK, Choi CK, Yeung PL, Middeldorp JM, Lung ML, et al. Inhibition of class I histone deacetylases by romidepsin potently induces Epstein-Barr virus lytic cycle and mediates enhanced cell death with ganciclovir. Int J Cancer. 2016;138(1):125–36.

    Article  CAS  PubMed  Google Scholar 

  57. Foss F, Horwitz S, Pro B, Prince HM, Sokol L, Balser B, et al. Romidepsin for the treatment of relapsed/refractory peripheral T cell lymphoma: prolonged stable disease provides clinical benefits for patients in the pivotal trial. J Hematol Oncol. 2016;9(1):1–8.

    Article  Google Scholar 

  58. Coiffier B, Pro B, Prince HM, Foss F, Sokol L, Greenwood M, et al. Results from a pivotal, open-label, phase II study of romidepsin in relapsed or refractory peripheral T-cell lymphoma after prior systemic therapy. J Clin Oncol. 2012;30(6):631–6.

    Article  CAS  PubMed  Google Scholar 

  59. Maruyama D, Tobinai K, Ogura M, Uchida T, Hatake K, Taniwaki M, et al. Romidepsin in Japanese patients with relapsed or refractory peripheral T-cell lymphoma: a phase I/II and pharmacokinetics study. Int J Hematol. 2017;106(5):655–65.

    Article  CAS  PubMed  Google Scholar 

  60. Smolewski P, Robak T. The discovery and development of romidepsin for the treatment of T-cell lymphoma. Expert Opin Drug Discov. 2017;12(8):859–73.

    CAS  PubMed  Google Scholar 

  61. Bachy E, Camus V, Thieblemont C, Sibon D, Casasnovas R-O, Ysebaert L, et al. Romidepsin plus CHOP versus CHOP in patients with previously untreated peripheral T-cell lymphoma: results of the Ro-CHOP phase III study (conducted by LYSA). J Clin Oncol. 2022;40(3):242–51.

    Article  CAS  PubMed  Google Scholar 

  62. Iyer SP, Xu J, Becnel MR, Nair R, Steiner R, Feng L, et al. A phase II study of pembrolizumab in combination with romidepsin demonstrates durable responses in relapsed or refractory T-cell lymphoma (TCL). Blood. 2020;136(Supplement 1):40–1.

    Article  Google Scholar 

  63. Marchi E, Zullo KM, Amengual JE, Kalac M, Bongero D, McIntosh CM, et al. The combination of hypomethylating agents and histone deacetylase inhibitors produce marked synergy in preclinical models of T-cell lymphoma. Br J Haematol. 2015;171(2):215–26.

    Article  CAS  PubMed  Google Scholar 

  64. Falchi L, Ma H, Klein S, Lue JK, Montanari F, Marchi E, et al. Combined oral 5-azacytidine and romidepsin are highly effective in patients with PTCL: a multicenter phase 2 study. Blood. 2021;137(16):2161–70.

    Article  CAS  PubMed  Google Scholar 

  65. Zullo KM, Guo Y, Cooke L, Jirau-Serrano X, Mangone M, Scotto L, et al. Aurora A kinase inhibition selectively synergizes with histone deacetylase inhibitor through cytokinesis failure in T-cell lymphoma. Clin Cancer Res. 2015;21(18):4097–109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Strati P, Nastoupil LJ, Davis RE, Fayad LE, Fowler N, Hagemeister FB, et al. A phase 1 trial of alisertib and romidepsin for relapsed/refractory aggressive B-cell and T-cell lymphomas. Haematologica. 2020;105(1):e26–8.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Nachmias B, Shaulov A, Lavie D, Goldschmidt N, Gural A, Saban R, et al. Romidepsin-bendamustine combination for relapsed/refractory T cell lymphoma. Acta Haematol. 2019;141(4):216–21.

    Article  CAS  PubMed  Google Scholar 

  68. Rahmani M, Aust MM, Benson EC, Wallace L, Friedberg J, Grant S. PI3K/mTOR inhibition markedly potentiates HDAC inhibitor activity in NHL cells through BIM- and MCL-1-dependent mechanisms in vitro and in vivo. Clin Cancer Res. 2014;20(18):4849–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Horwitz SM, Moskowitz AJ, Jacobsen ED, Mehta-Shah N, Khodadoust MS, Fisher DC, et al. The combination of duvelisib, a PI3K-δ, γ Inhibitor, and romidepsin is highly active in relapsed/refractory peripheral T-cell lymphoma with low rates of transaminitis: results of parallel multicenter, phase 1 combination studies with expansion cohorts. Blood. 2018;132(Supplement 1):683–783.

    Article  Google Scholar 

  70. Iyer SP, Huen A, Ai WZ, Jagadeesh D, Lechowicz MJ, Okada C, et al. Safety and efficacy of tenalisib given in combination with Romidepsin in patients with relapsed/refractory T-cell lymphoma: final results from a phase I/II open label multi-center study. Blood. 2021;138(Supplement 1):1365–465.

    Article  Google Scholar 

  71. Amengual JE, Lichtenstein R, Lue J, Sawas A, Deng C, Lichtenstein E, et al. A phase 1 study of romidepsin and pralatrexate reveals marked activity in relapsed and refractory T-cell lymphoma. Blood. 2018;131(4):397–407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Jain S, Jirau-Serrano X, Zullo KM, Scotto L, Palermo CF, Sastra SA, et al. Preclinical pharmacologic evaluation of pralatrexate and romidepsin confirms potent synergy of the combination in a murine model of human T-cell lymphoma. Clin Cancer Res. 2015;21(9):2096–106.

    Article  CAS  PubMed  Google Scholar 

  73. Mehta-Shah N, Lunning MA, Moskowitz AJ, Boruchov AM, Ruan J, Lynch P, et al. Romidepsin and lenalidomide-based regimens have efficacy in relapsed/refractory lymphoma: Combined analysis of two phase I studies with expansion cohorts. Am J Hematol. 2021;96(10):1211–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Cosenza M, Civallero M, Fiorcari S, Pozzi S, Marcheselli L, Bari A, et al. The histone deacetylase inhibitor romidepsin synergizes with lenalidomide and enhances tumor cell death in T-cell lymphoma cell lines. Cancer Biol Ther. 2016;17(10):1094–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Pellegrini C, Dodero A, Chiappella A, Monaco F, Degl’Innocenti D, Salvi F, et al. A phase II study on the role of gemcitabine plus romidepsin (GEMRO regimen) in the treatment of relapsed/refractory peripheral T-cell lymphoma patients. J Hematol Oncol. 2016;9(1):1–7.

    Article  Google Scholar 

  76. Zinzani PL, Venturini F, Stefoni V, Fina M, Pellegrini C, Derenzini E, et al. Gemcitabine as single agent in pretreated T-cell lymphoma patients: evaluation of the long-term outcome. Ann Oncol. 2010;21(4):860–3.

    Article  CAS  PubMed  Google Scholar 

  77. Reiman T, Savage KJ, Crump M, Cheung MC, Macdonald D, Buckstein R, et al. A phase I study of romidepsin, gemcitabine, dexamethasone and cisplatin combination therapy in the treatment of peripheral T-cell and diffuse large B-cell lymphoma; the Canadian cancer trials group LY.15 study†. Leuk Lymphoma. 2019;60(4):912–9.

    Article  CAS  PubMed  Google Scholar 

  78. Vu K, Wu C-H, Yang C-Y, Zhan A, Cavallone E, Berry W, et al. Romidepsin plus liposomal doxorubicin is safe and effective in patients with relapsed or refractory T-cell lymphoma: results of a phase I dose-escalation study. Clin Cancer Res. 2020;26(5):1000–8.

    Article  CAS  PubMed  Google Scholar 

  79. Gyöngyösi M, Lukovic D, Zlabinger K, Spannbauer A, Gugerell A, Pavo N, et al. Liposomal doxorubicin attenuates cardiotoxicity via induction of interferon-related DNA damage resistance. Cardiovasc Res. 2020;116(5):970–82.

    PubMed  Google Scholar 

  80. Reimer P. New developments in the treatment of peripheral T-cell lymphoma–role of Belinostat. Cancer Manag Res. 2015;7:145–51.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Lee H-Z, Kwitkowski VE, Del Valle PL, Ricci MS, Saber H, Habtemariam BA, et al. FDA approval: belinostat for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma. Clin Cancer Res. 2015;21(12):2666–70.

    Article  CAS  PubMed  Google Scholar 

  82. Wolska-Washer A, Smolewski P, Robak T. Advances in the pharmacotherapeutic options for primary nodal peripheral T-cell lymphoma. Expert Opin Pharmacother. 2021;22(9):1203–15.

    Article  CAS  PubMed  Google Scholar 

  83. O’Connor OA, Horwitz S, Masszi T, Van Hoof A, Brown P, Doorduijn J, et al. Belinostat in patients with relapsed or refractory peripheral T-cell lymphoma: results of the pivotal phase II BELIEF (CLN-19) study. J Clin Oncol. 2015;33(23):2492–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Kenny RG, Ude Z, Docherty JR, Marmion CJ. Vorinostat and Belinostat, hydroxamate-based anti-cancer agents, are nitric oxide donors. J Inorg Biochem. 2020;206:110981.

    Article  CAS  PubMed  Google Scholar 

  85. Foss F, Advani R, Duvic M, Hymes KB, Intragumtornchai T, Lekhakula A, et al. A Phase II trial of Belinostat (PXD101) in patients with relapsed or refractory peripheral or cutaneous T-cell lymphoma. Br J Haematol. 2015;168(6):811–9.

    Article  CAS  PubMed  Google Scholar 

  86. Belinostat. In: LiverTox: Clinical and research information on drug-induced liver injury. National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda. 2012.

  87. Johnston PB, Cashen AF, Nikolinakos PG, Beaven AW, Barta SK, Bhat G, et al. Belinostat in combination with standard cyclophosphamide, doxorubicin, vincristine and prednisone as first-line treatment for patients with newly diagnosed peripheral T-cell lymphoma. Exp Hematol Oncol. 2021;10(1):15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Passero FC, Ravi D, McDonald JT, Beheshti A, David KA, Evens AM. Combinatorial ixazomib and belinostat therapy induces NFE2L2-dependent apoptosis in Hodgkin and T-cell lymphoma. Br J Haematol. 2020;188(2):295–308.

    Article  CAS  PubMed  Google Scholar 

  89. Peters GJ, van Gemert FPA, Kathmann I, Reddy G, Cillessen SAGM, Jansen G. Schedule-dependent synergy between the histone deacetylase inhibitor belinostat and the dihydrofolate reductase inhibitor pralatrexate in T-and B-cell lymphoma cells in vitro. Front Cell Dev Biol. 2020;8:577215.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Cheng M-H, Weng J-Y, Chuang C-H, Liao W-T, Lai Y-F, Liu J-Y, et al. Prolonging the half-life of histone deacetylase inhibitor belinostat via 50 nm scale liposomal subcutaneous delivery system for peripheral T-cell lymphoma. Cancers. 2020;12(9):2558.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Shi Y, Dong M, Hong X, Zhang W, Feng J, Zhu J, et al. Results from a multicenter, open-label, pivotal phase II study of chidamide in relapsed or refractory peripheral T-cell lymphoma. Ann Oncol. 2015;26(8):1766–71.

    Article  CAS  PubMed  Google Scholar 

  92. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Gui L, Cao J, Ji D, Zhang H, Fan Q, Zhu J, et al. Chidamide combined with cyclophosphamide, doxorubicin, vincristine and prednisone in previously untreated patients with peripheral T-cell lymphoma. Chin J Cancer Res. 2021;33(5):616–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Zhang W, Su L, Liu L, Gao Y, Wang Q, Su H, et al. The combination of chidamide with the CHOEP regimen in previously untreated patients with peripheral T-cell lymphoma: a prospective, multicenter, single arm, phase 1b/2 study. Cancer Biol Med. 2021;18(3):841–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Tan D, Phipps C, Hwang WY, Tan SY, Yeap CH, Chan YH, et al. Panobinostat in combination with bortezomib in patients with relapsed or refractory peripheral T-cell lymphoma: an open-label, multicentre phase 2 trial. Lancet Haematol. 2015;2(8):e326–33.

    Article  PubMed  Google Scholar 

  96. Ma H, O’Connor OA, Marchi E. New directions in treating peripheral T-cell lymphomas (PTCL): leveraging epigenetic modifiers alone and in combination. Expert Rev Hematol. 2019;12(3):137–46.

    Article  PubMed  Google Scholar 

  97. Oki Y, Younes A, Copeland A, Hagemeister F, Fayad LE, McLaughlin P, et al. Phase I study of vorinostat in combination with standard CHOP in patients with newly diagnosed peripheral T-cell lymphoma. Br J Haematol. 2013;162(1):138–41.

    Article  CAS  PubMed  Google Scholar 

  98. Hopfinger G, Nösslinger T, Lang A, Linkesch W, Melchardt T, Weiss L, et al. Lenalidomide in combination with vorinostat and dexamethasone for the treatment of relapsed/refractory peripheral T cell lymphoma (PTCL): report of a phase I/II trial. Ann Hematol. 2014;93(3):459–62.

    Article  CAS  PubMed  Google Scholar 

  99. Siddiqi T, Frankel P, Beumer JH, Kiesel BF, Christner S, Ruel C, et al. Phase 1 study of the Aurora kinase A inhibitor alisertib (MLN8237) combined with the histone deacetylase inhibitor vorinostat in lymphoid malignancies. Leuk Lymphoma. 2020;61(2):309–17.

    Article  CAS  PubMed  Google Scholar 

  100. Amengual JE, Lue JK, Ma H, Lichtenstein R, Shah B, Cremers S, et al. First-in-class selective HDAC6 inhibitor (ACY-1215) has a highly favorable safety profile in patients with relapsed and refractory lymphoma. Oncologist. 2021;26(3):184-e366.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Amengual JE, Johannet P, Lombardo M, Zullo K, Hoehn D, Bhagat G, et al. Dual targeting of protein degradation pathways with the selective HDAC6 inhibitor ACY-1215 and bortezomib is synergistic in lymphoma. Clin Cancer Res. 2015;21(20):4663–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Cosenza M, Civallero M, Marcheselli L, Sacchi S, Pozzi S. Citarinostat and Momelotinib co-target HDAC6 and JAK2/STAT3 in lymphoid malignant cell lines: a potential new therapeutic combination. Apoptosis. 2020;25(5–6):370–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Balasubramanian S, Ramos J, Luo W, Sirisawad M, Verner E, Buggy JJ. A novel histone deacetylase 8 (HDAC8)-specific inhibitor PCI-34051 induces apoptosis in T-cell lymphomas. Leukemia. 2008;22(5):1026–34.

    Article  CAS  PubMed  Google Scholar 

  104. Yang W, Feng Y, Zhou J, Cheung OK, Cao J, Wang J, et al. A selective HDAC8 inhibitor potentiates antitumor immunity and efficacy of immune checkpoint blockade in hepatocellular carcinoma. Sci Transl Med. 2021;13(588):eaaz6804.

    Article  CAS  PubMed  Google Scholar 

  105. Shen J, Najafi S, Stäble S, Fabian J, Koeneke E, Kolbinger FR, et al. A kinome-wide RNAi screen identifies ALK as a target to sensitize neuroblastoma cells for HDAC8-inhibitor treatment. Cell Death Differ. 2018;25(12):2053–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Patwardhan RS, Pal D, Checker R, Sharma D, Sandur SK. Baicalein induces cell death in murine T cell lymphoma via inhibition of thioredoxin system. Int J Biochem Cell Biol. 2017;91(Pt A):45–52.

    Article  CAS  PubMed  Google Scholar 

  107. Sborov DW, Canella A, Hade EM, Mo X, Khountham S, Wang J, et al. A phase 1 trial of the HDAC inhibitor AR-42 in patients with multiple myeloma and T- and B-cell lymphomas. Leuk Lymphoma. 2017;58(10):2310–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Zhou M, Yuan M, Zhang M, Lei C, Aras O, Zhang X, et al. Combining histone deacetylase inhibitors (HDACis) with other therapies for cancer therapy. Eur J Med Chem. 2021;226:113825.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. He S, Dong G, Li Y, Wu S, Wang W, Sheng C. Potent dual BET/HDAC inhibitors for efficient treatment of pancreatic cancer. Angew Chem Int Ed Engl. 2020;59(8):3028–32.

    Article  CAS  PubMed  Google Scholar 

  110. Luan Y, Li J, Bernatchez JA, Li R. Kinase and histone deacetylase hybrid inhibitors for cancer therapy. J Med Chem. 2019;62(7):3171–83.

    Article  CAS  PubMed  Google Scholar 

  111. Liang X, Zang J, Li X, Tang S, Huang M, Geng M, et al. Discovery of novel janus kinase (JAK) and histone deacetylase (HDAC) dual inhibitors for the treatment of hematological malignancies. J Med Chem. 2019;62(8):3898–923.

    Article  CAS  PubMed  Google Scholar 

  112. Ren Y, Li S, Zhu R, Wan C, Song D, Zhu J, et al. Discovery of STAT3 and histone deacetylase (HDAC) dual-pathway inhibitors for the treatment of solid cancer. J Med Chem. 2021;64(11):7468–82.

    Article  CAS  PubMed  Google Scholar 

  113. Dong G, Chen W, Wang X, Yang X, Xu T, Wang P, et al. Small molecule inhibitors simultaneously targeting cancer metabolism and epigenetics: discovery of novel nicotinamide phosphoribosyltransferase (NAMPT) and histone deacetylase (HDAC) dual inhibitors. J Med Chem. 2017;60(19):7965–83.

    Article  CAS  PubMed  Google Scholar 

  114. Fan F, Liu P, Bao R, Chen J, Zhou M, Mo Z, et al. A dual PI3K/HDAC inhibitor induces immunogenic ferroptosis to potentiate cancer immune checkpoint therapy. Can Res. 2021;81(24):6233–45.

    Article  CAS  Google Scholar 

  115. Fan W, Zhang L, Jiang Q, Song W, Yan F, Zhang L. Histone deacetylase inhibitor based prodrugs. Eur J Med Chem. 2020;203:112628.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are indebted to the editors of MDPI Author Services for editing and reviewing this manuscript for English language.

Funding

This research received no external funding.

Author information

Authors and Affiliations

Authors

Contributions

SJ wrote original draft and did visualization; S.L. and J.D. contributed to writing—review and editing; GL, YG, LW, JY, WM and JD supervised this study. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Weiwei Mou or Jun Du.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, G., Jin, S., Lin, S. et al. Update on histone deacetylase inhibitors in peripheral T-cell lymphoma (PTCL). Clin Epigenet 15, 124 (2023). https://doi.org/10.1186/s13148-023-01531-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13148-023-01531-8

Keywords