Histone Deacetylases and Histone Deacetylase Inhibitors: Molecular Mechanisms of Action in Various Cancers
Masumeh Sanaei, Fraidoon Kavoosi
Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
Date of Submission | 25-Jun-2019 |
Date of Acceptance | 23-Sep-2019 |
Date of Web Publication | 31-Oct-2019 |
Correspondence Address: Dr. Fraidoon Kavoosi Jahrom University of Medical Sciences, Jahrom, Postal Code: 74148-46199, Fars Province Iran
 Source of Support: None, Conflict of Interest: None  | Check |
DOI: 10.4103/abr.abr_142_19
Epigenetic modifications such as histone modification play an important role in tumorigenesis. There are several evidence that histone deacetylases (HDACs) play a key role in cancer induction and progression by histone deacetylation. Besides, histone acetylation is being accessed as a therapeutic target because of its role in regulating gene expression. HDAC inhibitors (HDACIs) are a family of synthetic and natural compounds that differ in their target specificities and activities. They affect markedly cancer cells, inducing cell differentiation, cell cycle arrest and cell death, reduction of angiogenesis, and modulation of the immune system. Here, we summarize the mechanisms of HDACs and the HDACIs in several cancers. An online search of different sources such as PubMed, ISI, and Scopus was performed to find available data on mechanisms and pathways of HDACs and HDACIs in different cancers. The result indicated that HDACs induce cancer through multiple mechanisms in various tissues. This effect can be inhibited by HDACIs which affect cancer cell by different pathways such as cell differentiation, cell cycle arrest, and cell death. In conclusion, these findings indicate that the HDACs play a major role in carcinogenesis through various pathways, and HDACIs can inhibit HDAC activity by multiple mechanisms resulting in cell cycle arrest, cell growth inhibition, and apoptosis induction.
Keywords: Cancer, histone deacetylase, histone deacetylase inhibitors
How to cite this article: Sanaei M, Kavoosi F. Histone Deacetylases and Histone Deacetylase Inhibitors: Molecular Mechanisms of Action in Various Cancers. Adv Biomed Res 2019;8:63 |
How to cite this URL: Sanaei M, Kavoosi F. Histone Deacetylases and Histone Deacetylase Inhibitors: Molecular Mechanisms of Action in Various Cancers. Adv Biomed Res [serial online] 2019 [cited 2023 Mar 31];8:63. Available from: https://www.advbiores.net/text.asp?2019/8/1/63/270133 |
Introduction | |  |
Epigenetic modifications, such as histone acetylation and deoxyribonucleic acid (DNA) methylation, play an important role in the tumorigenesis and cancer progression. Among them, the importance of histone deacetylase (HDAC)-mediated epigenetic processes in the carcinogenesis has been highlighted. As a reversible posttranslational modification, histone acetylation plays a major and fundamental role in chromatin structure/function and regulating eukaryotic gene expression. Histone acetylation is regulated by opposing activities of HDACs and histone acetyltransferases (HATs) [Figure 1].[1] HDACs are enzymes that remove an acetyl group from histones and are divided into two major families, including zinc-dependent and NADD-dependent families.[2] Bacteria have HDAC- and HAT-like proteins, which may act as enzymes regulating acetylation of nonhistone proteins.[3] The mammalian HDACs can deacetylate histones and a variety of nonhistone cellular proteins.[4] HATs catalyze the transfer of an acetyl group from acetyl coenzyme A (acetyl-CoA) to lysine residues in proteins.[5] The balance between histone acetylation and deacetylation is often damaged in cancer, resulting in silenced expressions of tumor suppressor genes. HDACs can be divided into two distinct families depending on the pathway and molecular mechanisms of removing the acetyl group and also divided into four classes based on the homology to their yeast analogs.[6] HDAC inhibitors (HDACIs) are a family of synthetic and natural compounds that differ in their target specificities and activities. Based on their structure, they are classified into four main groups including cyclic peptides, hydroxamic acids, benzamides, and short-chain fatty acids.[7] Indeed, HDACIs affect markedly cancer cells, inducing cell differentiation, cell cycle arrest, cell death, reduction of angiogenesis, and modulation of the immune system.[8] Previously, we evaluated the effect of HDACIs and DNA demethylating agents on hepatocellular carcinoma (HCC) which encouraged us to write this article.[9],[10],[11] The current review mainly focuses on the action of HDAC and the effect of HDACIs in several cancers. | Figure 1: Acetylation and deacetylation reactions of lysine catalyzed by histone acetyltransferases and histone deacetylases
Click here to view |
Histone Modification | |  |
Histone acetylation relaxes the chromatin structure by which facilitates gene transcription and expression. The overall level of histone acetylation is controlled by a balance between two opposing enzyme groups including HATs and HDACs. HATs catalyze the transfer of the acetyl group to the ε-amino group of lysine side chains utilizing acetyl-CoA as a common acetyl donor by which abolishes the positive charge of lysine resulting in eliminates the electrostatic bond between DNA and histone [Figure 2].[12],[13] HATs, therefore, open the local region of chromatin structure, rendering it more accessible to transcription factors. HDACs remove the acetyl residues and restore the positive charge of lysine. Consequently, HDACs are associated with condensed chromatin structures and transcriptional repression [Figure 3].[14],[15] | Figure 2: Histone acetylation at the N-terminus lysine by histone acetyltransferases and histone deacetylation by histone deacetylases
Click here to view |
 | Figure 3: Histone acetylation converts chromatin to an open state, it is regulated by the histone acetyltransferase (HAC). Histone deacetylation is regulated by the histone deacetylase which converts chromatin structure to a condensed or transcriptionally repressive state
Click here to view |
Histone Deacetylases | |  |
Histone deacetylases and cancer
According to recent studies, HDAC enzyme dysfunctions and altered acetylation levels are linked to numerous cancers.[16] It has been reported that the expression of HDACs is increased in solid and hematological cancers. HDACs play an important role in the epigenetic regulation of gene transcription and expression through their effects on the chromatin compaction state. Recently, HDACs have become promising therapeutic targets because of their potential to reverse the aberrant epigenetic states associated with carcinogenesis. The overexpression of HDACs have been reported in many cancers.[5]
Histone deacetylases classification
In human, 18 HDAC enzymes have been classified into four groups based on their homology with yeast HDACs. Classes I, II, and IV require a zinc molecule, as a cofactor, in their active site and are inhibited by Zn2+-binding HDACIs. Class III HDACs have similar structural homologous to the yeast Sir2 protein and require NAD+ as a cofactor instead of Zn2+. Therefore, Zn2+-binding HDACIs cannot inhibit them. The role of sirtuins in carcinogenesis is still debatable, because some SIRTs have dual roles as tumor suppressors and oncoproteins.[17]
Class I HDACs include HDACs 1, 2, 3, and 8 and are similar to yeast Rpd3. They are the most abundant HDACs localized in the nucleus. Class II HDACs can shuttle between the nucleus and the cytoplasm and are similar to yeast Hda1 and larger than the other two classes of HDACs, based on sequence and domain organization, Class II HDACs can be further subdivided. Class IIa (HDACs 4, 5, 7, and 9) contains a highly conserved C-terminal deacetylase catalytic domain homologous to Hda1. Class IIb (HDACs 6 and 8) is characterized by having two deacetylase domains.[18],[19] HDAC11 is the sole member of class IV. The classification and structures of HDACs are indicated in [Figure 4].[20] | Figure 4: Classification, structures, and cellular localization of Zn2+-dependent histone deacetylase isoforms
Click here to view |
Histone Acetylases Classification | |  |
HATs contain two major types including nuclear (A-type) and cytoplasmic (B-type). The type-A HATs contains various family classified into at least three separate groups based on functional similarities and structural homologies, including GCN5-related N-acetyltransferases family, Moz-Ybf2/Sas3-Sas2-Tip60 family, and the p300/CREB-binding protein (CBP/CREBBP) family.[21] Type-B HATs is highly conserved and share sequence homology with scHat1. This type acetylates newly synthesized histone H4 at K5 and K12 which is important for deposition of the histones, after which the marks are removed.[22],[23]
Mechanism of the Action of Histone Deacetylase Inhibitors | |  |
HDACIs belong to a diverse family of both natural and synthetic compounds which can be divided into four groups including aliphatic fatty acids, hydroximic acid, benzamides, and cyclic peptides. Several agents are characterized for their potential as HDACIs, first of which identified was n-butyrate, responsible for the accumulation of hyper acetylated histone inside the nucleus. Subsequently, trapoxin A and trichostatin A (TSA) were found to be irreversible and reversible inhibitors of HDACs, respectively. HDACIs increase the level of histone acetylation and the molecular mechanism for this effect is associated with the inhibition of HDAC activity.[24] HDACs have multiple mechanisms and many different cellular and target proteins, including proteins that are involved in cancer progression, apoptosis, cell cycle control, angiogenesis, and cell invasion. Thus, HDACIs exert multiple mechanisms of action such as activation of the apoptotic pathway, cell cycle arrest, apoptotic induction occurs via extrinsic (death receptor) or intrinsic (mitochondrial) pathways, both of which lead to caspase activation and cell death induction. HDACIs can induce cell cycle arrest at G1/S or G2/M transition, resulting in differentiation and/or apoptosis. They increase CDK inhibitor p21WAF1/CIP1 expression leads to cell cycle arrest at G1/S.[25] Together, multiple pathways, by which HDACIs act upon cancer cell are indicated in [Figure 5].[26] Chemical structure of several HDACIs has been indicated in [Figure 6]. | Figure 5: Multiple antitumor pathways activated by histone deacetylase inhibitors. Extrinsic and intrinsic refer to two apoptosis pathways, and homologous recombination and nonhomologous end joining refer to two double strand breaks (DBS) repair pathways
Click here to view |
Histone Deacetylases, Histone Deacetylase Inhibitors and Urogenital Cancer | |  |
Renal cancer
The high expression of class I HDAC isoforms 1 and 2 and low the expression of HDAC3 have been shown in renal cell carcinoma (RCC). These differences in the expression patterns suggest difference regulatory pathways.[27] Experimental studies have indicated that the HDACI MS-275 alone and in combination with interleukin-2 have an antitumor effect in vivo in RCC. The effect is associated with a decreased number of T regulatory cells and the increased antitumor cytotoxicity by splenocytes. The MS-275 has antitumor activity in a human RCC of T-cells (CD4+ CD25+ Foxp3+) that have been associated with self-reactive T-cells suppression.[28] The HDACI MS-275 can reactivate epigenetic silencing of retinoic acid receptor B2 (RARb2) in a human RCC model and has greater antitumor activity in combination with 13-cis-retinoic acid compared with single component.[29] In vitro andin vivo studies have shown that HDACI LBH589 has the potent anticancer effect on renal cancer cells. This agent induces G2-M arrest and cell apoptosis of renal cancer via degradation of Aurora A and B kinases by HDAC3 and HDAC6 inhibition.[30]
Bladder cancer
High expression levels of HDAC-1 and HDAC-2 have been reported in bladder cancer. Similarly, overexpression of HDAC-1 to HDAC-3 has been reported in this cancer.[31] Expression array data from another study has been shown the overexpression of HDAC-1 in bladder cancer compared to normal bladder tissue.[32] Clinical studies have indicated a high level of HDAC1 mRNA expression in human bladder cancer specimens. The immunohistochemical study has shown that HDAC1 is expressed in the cytoplasm and nucleus in the bladder specimens.[33]
The potential efficacy of HDACI TSA and sodium butyrate (NaB) against bladder cancer cells has been reported. Experimental studies have indicated that TSA inhibits the growth of BIU-87 bladder cancer cells through cell cycle arrest in G1 phase and induces apoptosis. This pathway is controlled by protein p21WAF1, since increased expression of this gene has been reported in TSA-treated cells. It should be noted that p21WAF1 is one of the most commonly induced genes by HDACIs such as suberoylanilide hydroxamic acid (SAHA), TSA, and sodium butyrate.[34],[35] Experimental studies have been demonstrated the potential preventive efficacy of valproic acid (VPA) on N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN) in bladder cancer.[36],[37] VPA-induced inhibition can be attributed to increased levels of the cyclin-dependent kinase inhibitor p21 WAF1, which can lead to the arrest of cells in the G1 phase.[38]
Prostate cancer
Prostate cancer has been reported as the second most frequently diagnosed cancer, and the third most common cause of cancer-related death in men. The cancer is a heterogeneous disease, the etiology of which appears to be related to a complex range of risk factors, such as genetic factors and epigenetic modifications. HDAC upregulation has been established in most human cancers.[39] The overexpression of various Class I and Class II HDACs in PC-3, DU145, and LNCaP human prostate cancer cell lines have been indicated. All HDAC isoforms are presented in prostate cancer at various levels. HDAC1 protein is abundantly presented in normal and malignant epithelial cell of the prostate tissue. HDAC5 and HDAC8 have not been detected in prostate tissues.[40] Expression of the Class I HDAC in the epithelial and stromal cells, and the prominent cytosolic distribution of HDAC8 in epithelial cells suggest that the various HDAC isoforms may play an important role in the prostate cancer induction and progression. The other studies have shown strong expression of HDAC1, HDAC2, and HDAC3 in the prostate cancer and the expression of HDAC2 as a highly significant prognostic value. HDAC1 expression is increased in premalignant and malignant lesions. HDAC4 is predominantly localized in the cytoplasm of benign prostate hyperplasia cells and primary prostate cancer cells.[41]
HDACIs induce dose-dependent inhibition of Class I or Class II HDACs leading to G1 or G2 cell cycle. Some HDACIs increase Ku70 acetylation, a crucial agent of the DNA repair machinery, resulted in decreased DNA-binding affinity.[42] Some compounds are potentially effective for both chemoprevention and cancer therapy.[43] HDACIs selectively reactivate tumor suppressor genes, a therapeutic effect that is not induced by traditional chemotherapy. Five classes of HDACIs have been recognized. The response of prostate cancer cells to HDACIs is not uniform, as shown in [Table 1].[44] | Table 1: Several histone deacetylase inhibitors studied in prostate cancer
Click here to view |
Histone Deacetylases, Histone Deacetylase Inhibitors, and Reproductive Cancer | |  |
Ovarian cancer
It has been reported that Class I HDACs (HDAC 1, 2, and 3) promote ovarian cancer induction and progression. The overexpression of this class play a critical role in ovarian cancer[45] and increases gradually from a benign state to borderline, and malignant ovarian tumors. The expression level of Class I HDAC is markedly different in various ovarian cancer subtypes and the most positive in mucinous subtypes, followed by high-grade serous, clear cell, and endometrioid subtypes. Strongly tumor cell proliferation exhibits increased Class I HDAC expression which is an independent risk factor for poor malignant ovarian tumor prognosis.[46] The specific mechanisms of Class I HDACs in the ovarian carcinogenesis is the suppression of the promoter region of RGS2.[47] RGS2 is a regulator of G-protein signaling 2 and an inhibitor of G-protein coupled receptors through accelerating the deactivation of heterotrimeric G-proteins. It has been reported that HDAC1 enhances cell proliferation via Cyclin A promotion. In ovarian epithelial cancer cells, HDAC2 remodels chromatin structure in response to platinum-based chemical therapies. HDAC3 facilitates cell migration by suppressing the E-cadherin expression.[48] HDACIs have been reported to decrease cancer cell proliferation, induce apoptosis, and promote cell differentiation. A wide variety of agents that can function as HDACIs include organic hydroxamic acids, short-chain fatty acids, benzamides, cyclic tetrapeptides, and sulfonamides.[49] Of the current HDACIs, three have been tested in ovarian cancer including VPA, SAHA, and romidepsin. The other HDACIs have also recently indicated for the potential treatment of ovarian cancer including M344 and TSA. The M344 is specific for HDAC6 and promotes cell growth inhibition, cell cycle arrest, and cellular apoptosis[50] and also TSA specifically inhibits Class I and II mammalian HDAC families, resulting in increases p73 gene expression and promote Bax-dependent apoptosis in cisplatin-resistant ovarian cancer cells.[51] The other investigators have reported the effects of a wide array of HDACIs (VPA, SAHA, TSA, and NaB) on OVCAR-3, SK-OV-3, TOV-21G, TOV-112D, OV-90, OVCA429, OVCA420, OVCA432, and OVCA433, nine ovarian cell lines.[52] The mechanism of HDACIs has been depicted in [Figure 7].[53] | Figure 7:. The mechanism of histone deacetylase inhibitorsHDACIs against ovarian cancer
Click here to view |
Endometrial cancer
Endometrial cancer is the seventh most common carcinoma among women worldwide. Strong HDAC1 protein expression has been reported with poor prognosis of endometrial carcinoma.[46] Overexpression of HDAC2 has been shown in this cancer too.[54] Several studies have indicated the proapoptotic or the antiproliferative effects of HDACIs on endometrial cancer cells. In endometrial cancer cells, HDACIs markedly increase the expression level of E-cadherin exhibit antiproliferative activity in these cancer cells. They can alter the degree of the acetylation of nonhistone effector molecules by which increase or decrease the transcription of genes such E2F1, EKLF, FEN 1, ACTR, cMyb, GATA, NFκ B, PCNA, HNF-4, HSP90, Runx, SF1 Sp3, Ku70, p53, RB, STAT, TFIIE, TCF, YY1, and so forth.[55] Different classes of HDACIs studied in endometrial cancer have been shown in [Table 2].[56]
Cervical cancer
The expression of HDAC6 has been demonstrated in different cell lines of cervical cancer such as HeLa, A549, HCT11, K562, and MDAMB 231. There is no significant change in the expression of HDAC6 in between HeLa, HEK 293 T, and HCT11 cell lines, when compared to K562, A549, and MDA-MB 231. Whereas, the overexpression of HDAC8 has been reported in HeLa, HCT11, A549, and MDA-MB 231.[57] A positive correlation between histone H3 acetylation and the tumor suppressor expression RARb2 and E-cadherin has been reported in cervical squamous cell carcinoma specimens. Recently, the studies have indicated that the combination of local histone deacetylation and CpG island methylation results in the strong epigenetic silence of E-cadherin and RARb2. Besides, a direct correlation between RARb2 and E-cadherin expression has been shown in cervical cancer.[58] Other investigators have demonstrated that HDAC1 and 2 are overexpressed in cervical cancer. The suppression of HDAC2 resulting in apoptosis induction is associated with an increased p53-independent expression of p21Cip1/WAF1.[59]
Cervical cancer is the second most frequent cancer in women. Recently, significant interest in epigenetic modifications of tumor suppressor genes has catalyzed the investigation of novel treatment methods using histone HDACIs. HDACs remove the acetyl groups resulting in chromatin compaction and tumor suppressor genes silenced in various malignancies. In cervical cancer, VPA acts as a specific inhibitor of class I HDACs and induces proteaosomal degradation of HDAC2, leading to cell growth arrestin vitro and in vivo.[60]
Breast cancer
The initiation and progression of breast cancer are secondary to the accumulation of genetic and epigenetic alternations which lead to aberrant cellular function. The more recent studies have reported reversible alterations in histone proteins and DNA which leads to carcinogenesis. Epigenetic alterations including histone deacetylation are prevalent in breast cancers. In this cancer, HDAC1 expression is associated with estrogen receptor (ER) and progesterone receptor (PR) expression, an earlier stage of disease at diagnosis.[61] HDAC6 is more frequently expressed in breast cancer ER and PR-positive.[62] The HDAC family is divided into two groups including zinc-dependent enzymes (Classes I, IIa, IIb, and IV) and zinc-independent enzymes (class III also called sirtuins).[63] Based on their chemical structures, they are divided into four groups, including hydroxamic acids, short-chain fatty acids, cyclic tetrapeptides, and benzamides.[64] Most of the HDACIs have been designed to target primarily the zinc cofactor at the active site of the HDACs and to exhibit their inhibitory effects in the nanomolar or micromolar range. HDACIs inhibit HDAC activity result in ER alpha and PR gene reactivation in ER-negative breast cancer cells.[65] It has been indicated that inhibition of Class III HDAC SIRT1 using a splitomicin or siRNA reactivates silenced SFRP1, SFRP2, CRBP1 genes, and E-cadherin in human breast cancer cells.[66]
Another HDACI vorinostat has been evaluated in several Phase II trials in breast cancer cells, including combination therapy of vorinostat with standard components (for example, paclitaxel), novel targeted therapy (trastuzumab, bevacizumab), and endocrine therapy (tamoxifen). Other HDACIs such as LBH-589 (panobinostat) and MS-275 (entinostat) are in Phase I/II study in combination with other components, such as trastuzumab, in women with metastatic HER2-positive breast cancer.[67]
Histone Deacetylases, Histone Deacetylase Inhibitors, and Gastrointestinal and Associated Glands Cancer | |  |
Colon cancer
HDACs alterations are found in many cancers including colorectal cancer (CRC). In colon cancer, the expression of HDAC1, HDAC2, HDAC3, and HDAC8 has been reported.[68] A tumor suppressor gene Rb represses gene expression by modulating the chromatin architecture. This gene recruits HDAC to E2F and cooperates with HDAC1 to suppress E2F regulated promoter of genes encoding cyclin E as a cell cycle protein. HDAC1 removes the highly charged acetyl groups from core histones, preventing transcription factor from accessing to DNA.[69] The role of HDAC1, HDAC2, HDAC3, HDAC5, and HDAC7 upregulation has been demonstrated by other investigators in human CRC.
Furthermore, HDAC2 upregulation has been reported as the earliest events in CRC.
The universality of HDAC2 upregulation suggests that HDAC2 upregulation may serve as a CRC biomarker.[70] HDACI SAHA can reduce the expression of active glucose transporter (SGLT1), and thereby suppressed the glucose uptake of colon cancer cells. Besides, it can induce the dissociation of SP1/CBP/HDAC3 from the regions around epidermal growth factor receptor (EGFR) transcription start site, the region in which the histones became hypoacetylated. Furthermore, SAHA can serve as a single agent to block EGFR and HDAC, two important factors in CRC.[71] Experimental studies have indicated that vorinostat and LBH589 can rapidly induce histone acetylation, cell growth inhibition, and cell cycle arrest in both HCT116 and HT29 colon cancer cells.[72]
Hepatocellular carcinoma
Recent investigations have shown that HDAC1 and HDAC2 play different roles during HCC progression. These enzymes are expressed in HCC, and the expression of both is associated with mortality from HCC. HDAC1 expression is correlated with moderately and poorly differentiated tumors. Another research has demonstrated that high HDAC2 expression is correlated with poor survival in early-stage HCC.[73]
HDAC3 plays an important role in HCC formation. It is expressed in liver cancer stem cells and is required for the self-renewal of these cells. HDAC3 downregulation decreases the expression of stem cell markers, including OCT4, Nanog, and SOX2.[74]
HDAC2 has been reported as an independent predictor of survival in HCC. Overexpression of HDACs 1, 2, 3, and 7 have been reported in primary HCC by Several studies. HDACs 1, 2, and 3 upregulation are highly related to the growth of tumor grades. A high level of HDAC2 is also associated with poor survival in low-grade and early-stage tumors.[73] HDAC activity is suppressed by TSA and sodium butyrate in HCC lead to the inhibition of invasion and metastasis by upregulation of early growth response claudin-3 and gene-1.[75] HDACI sodium butyrate performs its anticancer effect by HDAC4 inhibition on HCC SMMC-7721 and HepG2 cells. The high concentrations of sodium butyrate significantly inhibit the HCC cell growth in various states including apoptosis, cell cycle arrest, and inhibition of cell migration/invasion. Other HDACIs SAHA and OSU-HDAC42 induce autophagy through downregulation of Akt/mTOR signaling and ER stress response induction in HCC HepG2, Hep3B, and Huh7 cell lines.[76] Preclinical studies have shown that treatment with HDACI belinostat can induce apoptosis in HCC cell lines.[77]
Cholangiocarcinoma
Several HDACIs targeting chromatin remodeling has been approved by the FDA such as vorinostat and romidepsin. These inhibitors have increased therapeutic utility in cholangiocarcinoma (CCA). MS-275 treatment potently inhibits the cell proliferation of EGI-1 and TFK-1 CCA cells by inducing cell cycle arrest and apoptosis.[78] The apoptotic pathway is characterized by activation of caspase-3, downregulation of Bcl-2, and upregulation of Bax. The cell cycle is predominantly arrested at the G1/S checkpoint, which is associated with the cyclin-dependent kinase inhibitor p21Waf/CIP1 induction.[79] It has been shown that HDAC3-specific inhibitor MI192 can inhibit the deacetylase activity of HDAC3 in CCA. Immunochemistry study has been indicated that HDAC3 is upregulated in CCA tissues compared with adjacent normal tissues. Taken together, MI192 targets HDAC3 and induces cell apoptosis in human CCA cells.[80] Several experimental studies have demonstrated that administration of the cisplatin in combination with HDACIs TSA, and SAHA resulted in cell growth inhibition and apoptosis induction in the CCA KKU-100 and KKU-M214 cell lines.[81] The expression of HDAC isoforms HDAC 1 and 2 is upregulated in the CCA HuCCT-1 and TFK-1 cell lines. HDACIs SAHA treatment causes significant cell number decline in three cell lines.[82]
Gallbladder carcinoma
Gallbladder carcinoma is an aggressive disease affecting older people. Unfortunately, it is difficult to detect this cancer in the early stage, because of lacking characteristic signs or symptoms. Another study has indicated that HDAC 1 ⁄ 2 is expressed in the nuclei of gallbladder carcinoma cells.[82]
Among HDACIs, SAHA is one of the most advanced in clinical fields as an anticancer drug.[83] SAHA treatment inhibits gallbladder carcinoma cell proliferation. It activates tumor suppressor genes p21 in gallbladder carcinoma cells. Other works have shown that SAHA and TSA reduce gallbladder carcinoma SGC-996 cells viability and arrest the cell cycle at the G1 phase. Furthermore, they promote apoptosis of these cells, downregulate the expression of c-Myc, cyclin D1, and Bmi1, and decrease the phosphorylation of mTOR p70S6K1, AKT, S6, and 4E-BP1.[82] Besides, SAHA treatment induces a significant inhibition of cell viability in gallbladder carcinoma TGBC2TKB cells.[84] Clinical and experimental studies have shown that HDACI PCI-24781 has an inhibitory effect on human gallbladder carcinoma and BK5.erbB2 in mice. This effect is associated with downregulation of erbB2 mRNA and erbB2 protein/activity and upregulation of acetylated tubulin and acetylated histone. Treatment with this agent results in decreased expression of Muc4, an intramembrane ligand for erbB2, in human gallbladder carcinoma cells.[85]
Pancreatic cancer
Pancreatic ductal adenocarcinoma (PDAC) is ranked fourth and fifth to the sixth leading cause of cancer death. It has been demonstrated that HDAC gene expression, particularly HDAC7, could be a possible marker of pancreatic cancer.[86]
Overexpression of class I HDACs, HDAC2, and HDAC7 has been reported in PDAC. The function and expression of individual HDACs in PDAC are shown in [Table 3].[86] | Table 3: Function and expression of individual histone deacetylases in pancreatic ductal adenocarcinoma
Click here to view |
TSA and SAHA have been demonstrated to induce apoptosis in human pancreatic adenocarcinoma cell. Other studies have indicated that TSA can synergize with the proteasome inhibitor PS-341 or gemcitabine[87] to induce cell apoptosis in human pancreatic cancer cell lines. Furthermore, HDAC Class I and II inhibitors such as TSA can induce apoptosis in tumor cell lines.[88] Other investigators have reported that Class III HDACis, such as sirtinol and nicotinamide, can induce apoptosis in the pancreatic cancer cells.[89] TSA and SAHA, as HDACIs, can induce apoptosis in pancreatic cancer cell lines IMIM-PC-2, IMIM-PC-1, and RWP-1. TSA and SK-7041both induce apoptosis and G2-M cell cycle arrest in the pancreatic cancer cell lines. They increase H4 histone acetylation and also suppress the expression of the anti-apoptotic proteins Bcl-XL, and Mcl-1 but do not affect either Bcl-2 or the pro-apoptotic Bak and Bax proteins.[90] Apoptotic effect of TSA in PDAC cell lines correlates with overexpression of mRNA expression of the pro-apoptotic BH3-only protein BIM together with attenuation of the anti-apoptotic BCL2 family members BCLW and BCLXL [Figure 8].[91] | Figure 8: Characterized mechanisms of histone deacetylasesHDACs in pancreatic ductal adenocarcinomaPDAC. Three histone deacetylasesHDAC pathways are demonstrated. Right part: histone deacetylasesHDACs control expression of the CDKI p21Cip1/Waf1 and cyclin B1 to control the G1/S-phase or G2/M-phase or the cell cycle. Middle part: histone deacetylasesHDACs contribute to the imbalanced expression of the anti-apoptotic (BCLw, MCL1, BCLXL, and c-Flip) and pro-apoptotic (BIM, BAX, and NOXA) genes. Left part: histone deacetylases HDAC1 and 2 containing repressor complex is recruited to the E-box of the E-cadherin promoter by the transcription factor SNAIL
Click here to view |
Histone Deacetylases, Histone Deacetylase Inhibitors, and Respiratory System Cancer | |  |
Lung cancer
Increased HDAC1 mRNA levels are more common in advanced stages of lung cancer, suggesting a role of HDAC in more aggressive tumors. The patients with lung cancer with high level of HDAC3 have significantly shorter survivals than the patients with low HDAC3 expression.[92] Similarly, it has been reported that overexpression of HDAC1 mRNA or protein expression is closely associated with the differentiation grade of lung cancer.[93] Compared to normal lung cells, lung cancer cells display aberrant histone H4 modification patterns with hypoacetylation of H4K12/H4K16, hyperacetylation of H4K5/H4K8, and loss of H4K20 trimethylation. The modifications of histone H4 is known as a potential biomarker for the detection and therapeutic approaches of lung cancer.[94] Overexpression of HDAC1 appears to be correlated with lung cancer progression and also overexpression of HDAC1 and HDAC3 correlates with a poor prognosis in pulmonary AdC patients. The histological evaluation has been shown the elevation of HDAC3 in SqCC.[95] Epigenetic aberrations involving lung cancer has been shown by many studies. HDACIs TSA and vorinostat both display antitumor activities in nonsmall cell lung cancer (NSCLC).[96] LBH589 is a novel inhibitor of Class I and II HDACs in two human NSCLC cell lines (H23 and H460) by several pathways including Bax, Bak, p53, caspase-3, caspase-8, caspase-9, Bid, and Bad. They repress anti-apoptotic genes such as survivin, Bcl-2, C-FLIP, and NF-jB.[97] It has been reported that HDACI FR901228 effectively inhibits lung cancer cells. In this cancer, FR901228 induce caspase-dependent apoptosis via the mitochondrial pathway rather than the death receptor pathway.[98] Experimental studies have shown that HDACI SAHA alone and in combination with BAY-11-7085 induce significant apoptosis and cell death in five tumorigenic NSCLC cell lines including H157, A549, H358, H460, and H1299.[99] The main classes of HDAIs have been shown in [Table 4].
Conclusions | |  |
In the past decade, it has been an increasing report in the field of HDACs inhibition by HDACI. Right now, we have extensive studies that have reported the mechanism of HDACs and HDACIs. In the current review, we summarized recent studies on the classification and molecular mechanisms of action of HDACs and HDACIs in several cancers. These findings indicate that the HDACs play a major role in carcinogenesis through various pathways and HDACIs can inhibit HDAC activity by multiple mechanisms resulting in cell cycle arrest, cell growth inhibition, and apoptosis induction. These compounds can upregulate global histone acetylation levels, by which regulate the expression of genes that are involved in diverse biological pathways. The changes in histone acetylation levels at gene promoters tend to correlate with the changes in gene expression levels in most cases. Together, a better understanding of the molecular mechanism and pathway of HDACs and HDACIs leads a new window toward treating patients who have cancer.
Acknowledgments
We appreciate the adjutancy of Research of Jahrom Medical University, Iran.
Financial support and sponsorship
This article was supported by the adjutancy of Research of Jahrom Medical University, Iran.
Conflicts of interest
There are no conflicts of interest.
References | |  |
1. | Yoshida M, Kudo N, Kosono S, Ito A. Chemical and structural biology of protein lysine deacetylases. Proc Jpn Acad Ser B Phys Biol Sci 2017;93:297-321. |
2. | Khochbin S, Verdel A, Lemercier C, Seigneurin-Berny D. Functional significance of histone deacetylase diversity. Curr Opin Genet Dev 2001;11:162-6. |
3. | Jones JD, O'Connor CD. Protein acetylation in prokaryotes. Proteomics 2011;11:3012-22. |
4. | Allis CD, Berger SL, Cote J, Dent S, Jenuwien T, Kouzarides T, et al. New nomenclature for chromatin-modifying enzymes. Cell 2007;131:633-6. |
5. | Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 2006;5:769-84. |
6. | Hrabeta J, Stiborova M, Adam V, Kizek R, Eckschlager T. Histone deacetylase inhibitors in cancer therapy. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 158:161-9. |
7. | Hull EE, Montgomery MR, Leyva KJ. HDAC inhibitors as epigenetic regulators of the immune system: Impacts on cancer therapy and inflammatory diseases. Biomed Res Int 2016; 10: 1-15. |
8. | Ceccacci E, Minucci S. Inhibition of histone deacetylases in cancer therapy: Lessons from leukaemia. Br J Cancer 2016; 114:605-11. |
9. | Kavoosi F, Sanaei M. Comparative analysis of the effects of valproic acid and tamoxifen on proliferation, and apoptosis of human hepatocellular carcinoma WCH 17 celllin. Iran J Peadiatr Hematol Oncol 2018;8:12-20. |
10. | Sanaei M, Kavoosi F, Atashpour S, Haghighat S. Effects of genistein and synergistic action in combination with tamoxifen on the hepG2 human hepatocellular carcinoma cell line Asian Pac J Cancer Prev 2017;18:2381-5. |
11. | Sanaei M, Kavoosi F, Pourahmadi M, Moosavi SN. Effect of genistein and 17-β estradiol on the viability and apoptosis of human hepatocellular carcinoma hepG2 cell line. Adv Biomed Res 2017;6:163.  [ PUBMED] [Full text] |
12. | Singh AK, Bishayee A, Pandey AK. Targeting histone deacetylases with natural and synthetic agents: An emerging anticancer strategy. Nutrients 2018;10. pii: E731. |
13. | Shahbazian MD, Grunstein M. Functions of site-specific histone acetylation and deacetylation. Annu Rev Biochem 2007;76:75-100. |
14. | Dawson MA, Kouzarides T. Cancer epigenetics: From mechanism to therapy. Cell 2012;150:12-27. |
15. | Cress WD, Seto E. Histone deacetylases, transcriptional control, and cancer. J Cell Physiol 2000;184:1-6. |
16. | Senese S, Zaragoza K, Minardi S, Muradore I, Ronzoni S, Passafaro A, et al. Role for histone deacetylase 1 in human tumor cell proliferation. Mol Cell Biol 2007;27:4784-95. |
17. | Firestein R, Blander G, Michan S, Oberdoerffer P, Ogino S, Campbell J, et al. The SIRT1 deacetylase suppresses intestinal tumorigenesis and colon cancer growth. PLoS One 2008;3:e2020. |
18. | Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases: Versatile regulators. Trends Genet 2003;19:286-93. |
19. | Kim HJ, Bae SC. Histone deacetylase inhibitors: Molecular mechanisms of action and clinical trials as anti-cancer drugs. Am J Transl Res 2011;3:166-79. |
20. | Yang Q, Yang Y, Zhou N, Tang K, Lau WB, Lau B, et al. Epigenetics in ovarian cancer: Premise, properties, and perspectives. Mol Cancer 2018;17:109. |
21. | Hodawadekar SC, Marmorstein R. Chemistry of acetyl transfer by histone modifying enzymes: Structure, mechanism and implications for effector design. Oncogene 2007;26:5528-40. |
22. | Parthun MR. Hat1: The emerging cellular roles of a type B histone acetyltransferase. Oncogene 2007;26:5319-28. |
23. | Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res 2011;21:381-95. |
24. | Siu LL, Pili R, Duran I, Messersmith WA, Chen EX, Sullivan R, et al. Phase I study of MGCD0103 given as a three-times-per-week oral dose in patients with advanced solid tumors. J Clin Oncol 2008;26:1940-7. |
25. | Zhang Z, Hao C, Wang L, Liu P, Zhao L, Zhu C, et al. Inhibition of leukemic cells by valproic acid, an HDAC inhibitor, in xenograft tumors. Onco Targets Ther 2013;6:733-40. |
26. | Mottamal M, Zheng S, Huang TL, Wang G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules 2015;20:3898-941. |
27. | Fritzsche FR, Weichert W, Röske A, Gekeler V, Beckers T, Stephan C, et al. Class I histone deacetylases 1, 2 and 3 are highly expressed in renal cell cancer. BMC Cancer 2008;8:381. |
28. | Kato Y, Yoshimura K, Shin T, Verheul H, Hammers H, Sanni TB, et al. Synergistic in vivo antitumor effect of the histone deacetylase inhibitor MS-275 in combination with interleukin 2 in a murine model of renal cell carcinoma. Clin Cancer Res 2007;13:4538-46. |
29. | Wang XF, Qian DZ, Ren M, Kato Y, Wei Y, Zhang L, et al. Epigenetic modulation of retinoic acid receptor beta2 by the histone deacetylase inhibitor MS-275 in human renal cell carcinoma. Clin Cancer Res 2005;11:3535-42. |
30. | Cha TL, Chuang MJ, Wu ST, Sun GH, Chang SY, Yu DS, et al. Dual degradation of aurora A and B kinases by the histone deacetylase inhibitor LBH589 induces G2-M arrest and apoptosis of renal cancer cells. Clin Cancer Res 2009;15:840-50. |
31. | Poyet C, Jentsch B, Hermanns T, Schweckendiek D, Seifert HH, Schmidtpeter M, et al. Expression of histone deacetylases 1, 2 and 3 in urothelial bladder cancer. BMC Clin Pathol 2014;14:10. |
32. | Wild PJ, Herr A, Wissmann C, Stoehr R, Rosenthal A, Zaak D, et al. Gene expression profiling of progressive papillary noninvasive carcinomas of the urinary bladder. Clin Cancer Res 2005;11:4415-29. |
33. | Ozawa A, Tanji N, Kikugawa T, Sasaki T, Yanagihara Y, Miura N, et al. Inhibition of bladder tumour growth by histone deacetylase inhibitor. BJU Int 2010;105:1181-6. |
34. | Gui CY, Ngo L, Xu WS, Richon VM, Marks PA. Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci U S A 2004;101:1241-6. |
35. | Chopin V, Toillon RA, Jouy N, Le Bourhis X. P21(WAF1/CIP1) is dispensable for G1 arrest, but indispensable for apoptosis induced by sodium butyrate in MCF-7 breast cancer cells. Oncogene 2004;23:21-9. |
36. | Jones J, Juengel E, Mickuckyte A, Hudak L, Wedel S, Jonas D, et al. The histone deacetylase inhibitor valproic acid alters growth properties of renal cell carcinoma in vitro and in vivo. J Cell Mol Med 2009;13:2376-85. |
37. | Sami S, Höti N, Xu HM, Shen Z, Huang X. Valproic acid inhibits the growth of cervical cancer both in vitro and in vivo. J Biochem 2008;144:357-62. |
38. | Lagger G, O'Carroll D, Rembold M, Khier H, Tischler J, Weitzer G, et al. Essential function of histone deacetylase 1 in proliferation control and CDK inhibitor repression. EMBO J 2002;21:2672-81. |
39. | Nakagawa M, Oda Y, Eguchi T, Aishima S, Yao T, Hosoi F, et al. Expression profile of class I histone deacetylases in human cancer tissues. Oncol Rep 2007;18:769-74. |
40. | Waltregny D, North B, Van Mellaert F, de Leval J, Verdin E, Castronovo V. Screening of histone deacetylases (HDAC) expression in human prostate cancer reveals distinct class I HDAC profiles between epithelial and stromal cells. Eur J Histochem 2004;48:273-90. |
41. | Weichert W, Röske A, Gekeler V, Beckers T, Stephan C, Jung K, et al. Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer 2008;98:604-10. |
42. | Chen CS, Wang YC, Yang HC, Huang PH, Kulp SK, Yang CC, et al. Histone deacetylase inhibitors sensitize prostate cancer cells to agents that produce DNA double-strand breaks by targeting ku70 acetylation. Cancer Res 2007;67:5318-27. |
43. | Jung M, Kozikowski A, Dritschilo A. Rational design and development of radiation-sensitizing histone deacetylase inhibitors. Chem Biodivers 2005;2:1452-61. |
44. | Abbas A, Gupta S. The role of histone deacetylases in prostate cancer. Epigenetics 2008;3:300-9. |
45. | Khabele D, Son DS, Parl AK, Goldberg GL, Augenlicht LH, Mariadason JM, et al. Drug-induced inactivation or gene silencing of class I histone deacetylases suppresses ovarian cancer cell growth: Implications for therapy. Cancer Biol Ther 2007;6:795-801. |
46. | Weichert W, Denkert C, Noske A, Darb-Esfahani S, Dietel M, Kalloger SE, et al. Expression of class I histone deacetylases indicates poor prognosis in endometrioid subtypes of ovarian and endometrial carcinomas. Neoplasia 2008;10:1021-7. |
47. | Cacan E. Epigenetic regulation of RGS2 (Regulator of G-protein signaling 2) in chemoresistant ovarian cancer cells. J Chemother 2017;29:173-8. |
48. | Hayashi A, Horiuchi A, Kikuchi N, Hayashi T, Fuseya C, Suzuki A, et al. Type-specific roles of histone deacetylase (HDAC) overexpression in ovarian carcinoma: HDAC1 enhances cell proliferation and HDAC3 stimulates cell migration with downregulation of E-cadherin. Int J Cancer 2010;127:1332-46. |
49. | Takai N, Narahara H. Histone deacetylase inhibitor therapy in epithelial ovarian cancer. J Oncol 2010; 5: 1-6. |
50. | Weberpals JI, O'Brien AM, Niknejad N, Garbuio KD, Clark-Knowles KV, Dimitroulakos J. The effect of the histone deacetylase inhibitor M344 on BRCA1 expression in breast and ovarian cancer cells. Cancer Cell Int 2011;11:29. |
51. | Muscolini M, Cianfrocca R, Sajeva A, Mozzetti S, Ferrandina G, Costanzo A, et al. Trichostatin A up-regulates p73 and induces bax-dependent apoptosis in cisplatin-resistant ovarian cancer cells. Mol Cancer Ther 2008;7:1410-9. |
52. | Takai N, Kawamata N, Gui D, Said JW, Miyakawa I, Koeffler HP. Human ovarian carcinoma cells: Histone deacetylase inhibitors exhibit antiproliferative activity and potently induce apoptosis. Cancer 2004;101:2760-70. |
53. | Takai N, Narahara H. Human endometrial and ovarian cancer cells: Histone deacetylase inhibitors exhibit antiproliferative activity, potently induce cell cycle arrest, and stimulate apoptosis. Curr Med Chem 2007;14:2548-53. |
54. | Cuppens T, Tuyaerts S, Amant F. Potential therapeutic targets in uterine sarcomas. Sarcoma 2015; 3: 1-14. |
55. | Marchion D, Münster P. Development of histone deacetylase inhibitors for cancer treatment. Expert Rev Anticancer Ther 2007;7:583-98. |
56. | Takai N, Narahara H. Preclinical studies of chemotherapy using histone deacetylase inhibitors in endometrial cancer. Obstet Gynecol Int 2010; 6: 1-8. |
57. | Vanaja GR, Ramulu HG, Kalle AM. Overexpressed HDAC8 in cervical cancer cells shows functional redundancy of tubulin deacetylation with HDAC6. Cell Commun Signal 2018;16:20. |
58. | Feng D, Wu J, Tian Y, Zhou H, Zhou Y, Hu W, et al. Targeting of histone deacetylases to reactivate tumour suppressor genes and its therapeutic potential in a human cervical cancer xenograft model. PLoS One 2013;8:e80657. |
59. | Huang BH, Laban M, Leung CH, Lee L, Lee CK, Salto-Tellez M, et al. Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1. Cell Death Differ 2005;12:395-404. |
60. | Atmaca A, Al-Batran SE, Maurer A, Neumann A, Heinzel T, Hentsch B, et al. Valproic acid (VPA) in patients with refractory advanced cancer: A dose escalating phase I clinical trial. Br J Cancer 2007;97:177-82. |
61. | Krusche CA, Wülfing P, Kersting C, Vloet A, Böcker W, Kiesel L, et al. Histone deacetylase-1 and -3 protein expression in human breast cancer: A tissue microarray analysis. Breast Cancer Res Treat 2005;90:15-23. |
62. | Zhang Z, Yamashita H, Toyama T, Sugiura H, Omoto Y, Ando Y, et al. HDAC6 expression is correlated with better survival in breast cancer. Clin Cancer Res 2004;10:6962-8. |
63. | Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 2006;6:38-51. |
64. | Dokmanovic M, Marks PA. Prospects: Histone deacetylase inhibitors. J Cell Biochem 2005;96:293-304. |
65. | Zhou Q, Atadja P, Davidson NE. Histone deacetylase inhibitor LBH589 reactivates silenced estrogen receptor alpha (ER) gene expression without loss of DNA hypermethylation. Cancer Biol Ther 2007;6:64-9. |
66. | Pruitt K, Zinn RL, Ohm JE, McGarvey KM, Kang SH, Watkins DN, et al. Inhibition of SIRT1 reactivates silenced cancer genes without loss of promoter DNA hypermethylation. PLoS Genet 2006;2:e40. |
67. | Pathiraja TN, Stearns V, Oesterreich S. Epigenetic regulation in estrogen receptor positive breast cancer – Role in treatment response. J Mammary Gland Biol Neoplasia 2010;15:35-47. |
68. | Zhu P, Martin E, Mengwasser J, Schlag P, Janssen KP, Göttlicher M. Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell 2004;5:455-63. |
69. | Dashwood RH, Ho E. Dietary histone deacetylase inhibitors: From cells to mice to man. Semin Cancer Biol 2007;17:363-9. |
70. | Stypula-Cyrus Y, Damania D, Kunte DP, Cruz MD, Subramanian H, Roy HK, et al. HDAC up-regulation in early colon field carcinogenesis is involved in cell tumorigenicity through regulation of chromatin structure. PLoS One 2013;8:e64600. |
71. | Chou CW, Wu MS, Huang WC, Chen CC. HDAC inhibition decreases the expression of EGFR in colorectal cancer cells. PLoS One 2011;6:e18087. |
72. | LaBonte MJ, Wilson PM, Fazzone W, Groshen S, Lenz HJ, Ladner RD. DNA microarray profiling of genes differentially regulated by the histone deacetylase inhibitors vorinostat and LBH589 in colon cancer cell lines. BMC Med Genomics 2009;2:67. |
73. | Quint K, Agaimy A, Di Fazio P, Montalbano R, Steindorf C, Jung R, et al. Clinical significance of histone deacetylases 1, 2, 3, and 7: HDAC2 is an independent predictor of survival in HCC. Virchows Arch 2011;459:129-39. |
74. | Liu C, Liu L, Shan J, Shen J, Xu Y, Zhang Q, et al. Histone deacetylase 3 participates in self-renewal of liver cancer stem cells through histone modification. Cancer Lett 2013;339:60-9. |
75. | Kim SO, Choi BT, Choi IW, Cheong J, Kim GY, Kwon TK, et al. Anti-invasive activity of histone deacetylase inhibitors via the induction of egr-1 and the modulation of tight junction-related proteins in human hepatocarcinoma cells. BMB Rep 2009;42:655-60. |
76. | Liu YL, Yang PM, Shun CT, Wu MS, Weng JR, Chen CC. Autophagy potentiates the anti-cancer effects of the histone deacetylase inhibitors in hepatocellular carcinoma. Autophagy 2010;6:1057-65. |
77. | Yeo W, Chung HC, Chan SL, Wang LZ, Lim R, Picus J, et al. Epigenetic therapy using belinostat for patients with unresectable hepatocellular carcinoma: A multicenter phase I/II study with biomarker and pharmacokinetic analysis of tumors from patients in the mayo phase II consortium and the cancer therapeutics research group. J Clin Oncol 2012;30:3361-7. |
78. | Rizvi S, Gores GJ. Emerging molecular therapeutic targets for cholangiocarcinoma. J Hepatol 2017;67:632-44. |
79. | Baradari V, Höpfner M, Huether A, Schuppan D, Scherübl H. Histone deacetylase inhibitor MS-275 alone or combined with bortezomib or sorafenib exhibits strong antiproliferative action in human cholangiocarcinoma cells. World J Gastroenterol 2007;13:4458-66. |
80. | Yin Y, Zhang M, Dorfman RG, Li Y, Zhao Z, Pan Y, et al. Histone deacetylase 3 overexpression in human cholangiocarcinoma and promotion of cell growth via apoptosis inhibition. Cell Death Dis 2017;8:e2856. |
81. | Asgar MA, Senawong G, Sripa B, Senawong T. Synergistic anticancer effects of cisplatin and histone deacetylase inhibitors (SAHA and TSA) on cholangiocarcinoma cell lines. Int J Oncol 2016;48:409-20. |
82. | Yamaguchi J, Sasaki M, Sato Y, Itatsu K, Harada K, Zen Y, et al. Histone deacetylase inhibitor (SAHA) and repression of EZH2 synergistically inhibit proliferation of gallbladder carcinoma. Cancer Sci 2010;101:355-62. |
83. | Marks PA, Breslow R. Dimethyl sulfoxide to vorinostat: Development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol 2007;25:84-90. |
84. | Zhang P, Guo Z, Wu Y, Hu R, Du J, He X, et al. Histone deacetylase inhibitors inhibit the proliferation of gallbladder carcinoma cells by suppressing AKT/mTOR signaling. PLoS One 2015;10:e0136193. |
85. | Kitamura T, Connolly K, Ruffino L, Ajiki T, Lueckgen A, DiGiovanni J, et al. The therapeutic effect of histone deacetylase inhibitor PCI-24781 on gallbladder carcinoma in BK5.erbB2 mice. J Hepatol 2012;57:84-91. |
86. | Ouaïssi M, Sielezneff I, Silvestre R, Sastre B, Bernard JP, Lafontaine JS, et al. High histone deacetylase 7 (HDAC7) expression is significantly associated with adenocarcinomas of the pancreas. Ann Surg Oncol 2008;15:2318-28. |
87. | Bai J, Demirjian A, Sui J, Marasco W, Callery MP. Histone deacetylase inhibitor trichostatin A and proteasome inhibitor PS-341 synergistically induce apoptosis in pancreatic cancer cells. Biochem Biophys Res Commun 2006;348:1245-53. |
88. | Ouaïssi M, Cabral S, Tavares J, da Silva AC, Mathieu Daude F, Mas E, et al. Histone deacetylase (HDAC) encoding gene expression in pancreatic cancer cell lines and cell sensitivity to HDAC inhibitors. Cancer Biol Ther 2008;7:523-31. |
89. | Ouaïssi M, Giger U, Sielezneff I, Pirrò N, Sastre B, Ouaissi A. Rationale for possible targeting of histone deacetylase signaling in cancer diseases with a special reference to pancreatic cancer. J Biomed Biotechnol 2011; 8: 1-8. |
90. | Koutsounas I, Giaginis C, Patsouris E, Theocharis S. Current evidence for histone deacetylase inhibitors in pancreatic cancer. World J Gastroenterol 2013;19:813-28. |
91. | Moore PS, Barbi S, Donadelli M, Costanzo C, Bassi C, Palmieri M, et al. Gene expression profiling after treatment with the histone deacetylase inhibitor trichostatin A reveals altered expression of both pro- and anti-apoptotic genes in pancreatic adenocarcinoma cells. Biochim Biophys Acta 2004;1693:167-76. |
92. | Jakopovic M, Thomas A, Balasubramaniam S, Schrump D, Giaccone G, Bates SE. Targeting the epigenome in lung cancer: Expanding approaches to epigenetic therapy. Front Oncol 2013;3:261. |
93. | Cao LL, Song X, Pei L, Liu L, Wang H, Jia M. Histone deacetylase HDAC1 expression correlates with the progression and prognosis of lung cancer: A meta-analysis. Medicine (Baltimore) 2017;96:e7663. |
94. | Van Den Broeck A, Brambilla E, Moro-Sibilot D, Lantuejoul S, Brambilla C, Eymin B, et al. Loss of histone h4k20 trimethylation occurs in preneoplasia and influences prognosis of non–small cell lung cancer. Clin. Cancer. Res 2008;14:7237-45. |
95. | Bartling B, Hofmann HS, Boettger T, Hansen G, Burdach S, Silber RE, et al. Comparative application of antibody and gene array for expression profiling in human squamous cell lung carcinoma. Lung Cancer 2005;49:145-54. |
96. | Ansari J, Shackelford RE, El-Osta H. Epigenetics in non-small cell lung cancer: From basics to therapeutics. Transl Lung Cancer Res 2016;5:155-71. |
97. | Geng L, Cuneo KC, Fu A, Tu T, Atadja PW, Hallahan DE. Histone deacetylase (HDAC) inhibitor LBH589 increases duration of gamma-H2AX foci and confines HDAC4 to the cytoplasm in irradiated non-small cell lung cancer. Cancer Res 2006;66:11298-304. |
98. | Doi S, Soda H, Oka M, Tsurutani J, Kitazaki T, Nakamura Y, et al. The histone deacetylase inhibitor FR901228 induces caspase-dependent apoptosis via the mitochondrial pathway in small cell lung cancer cells. Mol Cancer Ther 2004;3:1397-402. |
99. | Rundall BK, Denlinger CE, Jones DR. Combined histone deacetylase and NF-kappaB inhibition sensitizes non-small cell lung cancer to cell death. Surgery 2004;136:416-25. |
[Figure 1], [Figure 2], [Figure 3], [Figure 4], [Figure 5], [Figure 6], [Figure 7], [Figure 8]
[Table 1], [Table 2], [Table 3], [Table 4]
This article has been cited by | 1 |
The role of viruses in cancer development versus cancer therapy: An oncological perspective |
|
| Hossein Javid, Alireza Sharbaf Mashhad, Shaghayegh Yazdani, Mahsa Akbari Oryani, Sanaz Akbari, Nastaran Rezagholinejad, Mahboubeh Tajaldini, Mehdi Karimi-Shahri | | Cancer Medicine. 2023; | | [Pubmed] | [DOI] | | 2 |
Epigenetics and immune cells in medulloblastoma |
|
| Francesca Gorini, Marco Miceli, Pasqualino de Antonellis, Stefano Amente, Massimo Zollo, Veronica Ferrucci | | Frontiers in Genetics. 2023; 14 | | [Pubmed] | [DOI] | | 3 |
The First-In-Class Anti-AXL×CD3e Pronectin™-Based Bispecific T-Cell Engager Is Active in Preclinical Models of Human Soft Tissue and Bone Sarcomas |
|
| Nicoletta Polerŕ, Antonia Mancuso, Caterina Riillo, Daniele Caracciolo, Stefania Signorelli, Katia Grillone, Serena Ascrizzi, Craig A. Hokanson, Francesco Conforti, Nicoletta Staropoli, Luigia Gervasi, Maria Teresa Di Martino, Mariamena Arbitrio, Giuseppe Nisticň, Roberto Crea, Pierosandro Tagliaferri, Giada Juli, Pierfrancesco Tassone | | Cancers. 2023; 15(6): 1647 | | [Pubmed] | [DOI] | | 4 |
Anticancer effects of the HDAC inhibitor, 3ß,6ß-dihydroxyurs-12-en-27-oic acid, in MCF-7 breast cancer cells via the inhibition of Akt/mTOR pathways |
|
| Jong Lim, So Kyung, Yukyoung Jeon, In Kim, Jong Kwak, Hyung Kim | | Oncology Reports. 2023; 49(2) | | [Pubmed] | [DOI] | | 5 |
The Role of HDACs in the Response of Cancer Cells to Cellular Stress and the Potential for Therapeutic Intervention |
|
| Rahma K. Alseksek, Wafaa S. Ramadan, Ekram Saleh, Raafat El-Awady | | International Journal of Molecular Sciences. 2022; 23(15): 8141 | | [Pubmed] | [DOI] | | 6 |
Recent Advances in the Management of Relapsed and Refractory Peripheral T-Cell Lymphomas |
|
| Zachary Braunstein, Miguel Ruiz, Walter Hanel, Polina Shindiapina, John C. Reneau, Jonathan E. Brammer | | Journal of Personalized Medicine. 2022; 12(6): 964 | | [Pubmed] | [DOI] | | 7 |
Clinical Implications of Necroptosis Genes Expression for Cancer Immunity and Prognosis: A Pan-Cancer Analysis |
|
| Xin-yu Li, Li-xin Su, Wen-Xue Chen, Hui Liu, Lu-yu Zhang, Yu-Chen Shen, Jian-Xiong You, Jing-Bing Wang, Liming Zhang, Deming Wang, Ming-Zhe Wen, Zhenfeng Wang, Yu-hao Shao, De-Hu Chen, Xi-tao Yang | | Frontiers in Immunology. 2022; 13 | | [Pubmed] | [DOI] | | 8 |
Potential of histone deacetylase inhibitors in the control and regulation of prostate, breast and ovarian cancer |
|
| Siddhartha Das Pramanik, Amit Kumar Halder, Ushmita Mukherjee, Dharmendra Kumar, Yadu Nandan Dey, Mogana R | | Frontiers in Chemistry. 2022; 10 | | [Pubmed] | [DOI] | | 9 |
DNTTIP1 is a Prognostic Biomarker Correlated With Immune Infiltrates in Hepatocellular Carcinoma: A Study Based on The Cancer Genome Atlas Data |
|
| Changyu Zhu, Rongsheng Tong, Xiaolei Jiang, Hua Xiao, Jianmei Guan, Jianchen Shu | | Frontiers in Genetics. 2022; 12 | | [Pubmed] | [DOI] | | 10 |
Potential of histone deacetylase inhibitors for the therapy of ovarian cancer |
|
| Fengyi Guo, Hongjing Wang | | Frontiers in Oncology. 2022; 12 | | [Pubmed] | [DOI] | | 11 |
Review: Neurological Complications From Therapies for Pediatric Brain Tumors |
|
| Thien Nguyen, Sabine Mueller, Fatema Malbari | | Frontiers in Oncology. 2022; 12 | | [Pubmed] | [DOI] | | 12 |
HDAC8 suppresses the epithelial phenotype and promotes EMT in chemotherapy-treated basal-like breast cancer |
|
| Garyfallia Pantelaiou-Prokaki, Iga Mieczkowska, Geske E. Schmidt, Sonja Fritzsche, Evangelos Prokakis, Julia Gallwas, Florian Wegwitz | | Clinical Epigenetics. 2022; 14(1) | | [Pubmed] | [DOI] | | 13 |
Repression of the PRELP gene is relieved by histone deacetylase inhibitors through acetylation of histone H2B lysine 5 in bladder cancer |
|
| Kanto Shozu, Syuzo Kaneko, Norio Shinkai, Ai Dozen, Hirofumi Kosuge, Makoto Nakakido, Hidenori Machino, Ken Takasawa, Ken Asada, Masaaki Komatsu, Kouhei Tsumoto, Shin-Ichi Ohnuma, Ryuji Hamamoto | | Clinical Epigenetics. 2022; 14(1) | | [Pubmed] | [DOI] | | 14 |
Selective Inhibitors of Histone Deacetylase 10 (HDAC-10) |
|
| Eftiola Pojani, Daniela Barlocco | | Current Medicinal Chemistry. 2022; 29(13): 2306 | | [Pubmed] | [DOI] | | 15 |
Histone deacetylase 4 and 5 translocation elicited by microsecond pulsed electric field exposure is mediated by kinase activity |
|
| Zahra Safaei, Gary L. Thompson | | Frontiers in Bioengineering and Biotechnology. 2022; 10 | | [Pubmed] | [DOI] | | 16 |
Synthesis and Anticancer Potential of New Hydroxamic Acid Derivatives as Chemotherapeutic Agents |
|
| Isil Nihan Korkmaz, Hasan Özdemir | | Applied Biochemistry and Biotechnology. 2022; | | [Pubmed] | [DOI] | | 17 |
Novel pyridine-containing histone deacetylase inhibitors strongly arrest proliferation, induce apoptosis and modulate miRNAs in cancer cells |
|
| Elisabetta Di Bello, Veronica Sian, Giulio Bontempi, Clemens Zwergel, Rossella Fioravanti, Beatrice Noce, Carola Castiello, Stefano Tomassi, Davide Corinti, Daniela Passeri, Roberto Pellicciari, Ciro Mercurio, Mario Varasi, Lucia Altucci, Marco Tripodi, Raffaele Strippoli, Angela Nebbioso, Sergio Valente, Antonello Mai | | European Journal of Medicinal Chemistry. 2022; : 115022 | | [Pubmed] | [DOI] | | 18 |
KDM1A/LSD1 as a promising target in various diseases treatment by regulating autophagy network |
|
| Ting Ma, Anqi Li, Yueyang Guo, Shaotong Li, Meng Li, Siqi Feng, Hongmin Liu | | Biomedicine & Pharmacotherapy. 2022; 148: 112762 | | [Pubmed] | [DOI] | | 19 |
Design, synthesis and biological evaluation of novel hybrids targeting mTOR and HDACs for potential treatment of hepatocellular carcinoma |
|
| Shiyang Zhai,Huimin Zhang,Rui Chen,Jiangxia Wu,Daiqiao Ai,Shunming Tao,Yike Cai,Ji-Quan Zhang,Ling Wang | | European Journal of Medicinal Chemistry. 2021; : 113824 | | [Pubmed] | [DOI] | | 20 |
A Phase 2a cohort expansion study to assess the safety, tolerability, and preliminary efficacy of CXD101 in patients with advanced solid-organ cancer expressing HR23B or lymphoma |
|
| Stephen W. Booth,Toby A. Eyre,John Whittaker,Leticia Campo,Lai Mun Wang,Elizabeth Soilleux,Daniel Royston,Gabrielle Rees,Murali Kesavan,Catherine Hildyard,Farasat Kazmi,Nick La Thangue,David Kerr,Mark R. Middleton,Graham P. Collins | | BMC Cancer. 2021; 21(1) | | [Pubmed] | [DOI] | | 21 |
How can exposure to engineered nanomaterials influence our epigenetic code? A review of the mechanisms and molecular targets |
|
| Luciana Moreira,Carla Costa,Joana Pires,Joăo Paulo Teixeira,Sónia Fraga | | Mutation Research/Reviews in Mutation Research. 2021; 788: 108385 | | [Pubmed] | [DOI] | | 22 |
Mitochondrial Proteins in Heart Failure: The Role of Deacetylation by SIRT3 |
|
| Chunfang Wang,Yating Wang,Li Shen | | Pharmacological Research. 2021; : 105802 | | [Pubmed] | [DOI] | | 23 |
Molecular mechanisms underpinning sarcomas and implications for current and future therapy |
|
| Victoria Damerell,Michael S. Pepper,Sharon Prince | | Signal Transduction and Targeted Therapy. 2021; 6(1) | | [Pubmed] | [DOI] | | 24 |
Characterizing binding intensity and energetic features of histone deacetylase inhibitor pracinostat towards class I HDAC isozymes through futuristic drug designing strategy |
|
| Shabir Ahmad Ganai | | In Silico Pharmacology. 2021; 9(1) | | [Pubmed] | [DOI] | | 25 |
Targeting the signal pathways triggered by cyclic peptides: A potential therapeutic strategy for alleviation of cancer |
|
| Yiyuan Yang,Hui Mao,Linxi Chen,Lanfang Li | | Archives of Biochemistry and Biophysics. 2021; : 108776 | | [Pubmed] | [DOI] | | 26 |
Epigenetic Enzymes: A Role in Aging and Prospects for Pharmacological Targeting |
|
| Elena G. Pasyukova,Alexander V. Symonenko,Olga Y. Rybina,Alexander M. Vaiserman | | Ageing Research Reviews. 2021; : 101312 | | [Pubmed] | [DOI] | | 27 |
Regulation of SIRT2 by Wnt/ß-catenin signaling pathway in colorectal cancer cells |
|
| Chang Li,Yuning Zhou,Ji Tae Kim,Tomoko Sengoku,Michael C. Alstott,Heidi L. Weiss,Qingding Wang,B. Mark Evers | | Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 2021; 1868(4): 118966 | | [Pubmed] | [DOI] | | 28 |
Histone Deacetylase Sirtuin 1 Promotes Loss of Primary Cilia in Cholangiocarcinoma |
|
| Kishor Pant,Estanislao Peixoto,Seth Richard,Aalekhya Biswas,M. Gerard O’Sullivan,Nasra Giama,Yeonjung Ha,Jun Yin,Pietro Carotenuto,Massimiliano Salati,Yanan Ren,Rendong Yang,Brunella Franco,Lewis R. Roberts,Sergio A. Gradilone | | Hepatology. 2021; | | [Pubmed] | [DOI] | | 29 |
Quisinostat mediated autophagy is associated with differentiation in neuroblastoma SK-N-SH cells |
|
| Vamsi Krishna Kommalapati,Dinesh Kumar,Anjana Devi Tangutur | | Molecular Biology Reports. 2021; | | [Pubmed] | [DOI] | | 30 |
Post-translational lysine ac(et)ylation in health, ageing and disease |
|
| Anna-Theresa Blasl,Sabrina Schulze,Chuan Qin,Leonie G. Graf,Robert Vogt,Michael Lammers | | Biological Chemistry. 2021; 0(0) | | [Pubmed] | [DOI] | | 31 |
Anticancer Therapy with HDAC Inhibitors: Mechanism-Based Combination Strategies and Future Perspectives |
|
| Robert Jenke,Nina Reßing,Finn K. Hansen,Achim Aigner,Thomas Büch | | Cancers. 2021; 13(4): 634 | | [Pubmed] | [DOI] | | 32 |
Oncolytic Viruses in Combination Therapeutic Approaches with Epigenetic Modulators: Past, Present, and Future Perspectives |
|
| Annalisa Chianese,Biagio Santella,Annalisa Ambrosino,Debora Stelitano,Luca Rinaldi,Massimiliano Galdiero,Carla Zannella,Gianluigi Franci | | Cancers. 2021; 13(11): 2761 | | [Pubmed] | [DOI] | | 33 |
A Novel ZIP4-HDAC4-VEGFA Axis in High-Grade Serous Ovarian Cancer |
|
| Qipeng Fan,Lihong Li,Tian-Li Wang,Robert E. Emerson,Yan Xu | | Cancers. 2021; 13(15): 3821 | | [Pubmed] | [DOI] | | 34 |
Roles of Histone Deacetylases in Acute Myeloid Leukemia With Fusion Proteins |
|
| Juan Zhang,Xuefeng Gao,Li Yu | | Frontiers in Oncology. 2021; 11 | | [Pubmed] | [DOI] | | 35 |
Role of HDAC6 and Its Selective Inhibitors in Gastrointestinal Cancer |
|
| Bingyi Zhou, Deliang Liu, Yuyong Tan | | Frontiers in Cell and Developmental Biology. 2021; 9 | | [Pubmed] | [DOI] | | 36 |
8a, a New Acridine Antiproliferative and Pro-Apoptotic Agent Targeting HDAC1/DNMT1 |
|
| Qiting Zhang,Ziyan Wang,Xinyuan Chen,Haoxiang Qiu,Yifan Gu,Ning Wang,Tao Wang,Ze Wang,Huabin Ma,Yufen Zhao,Bin Zhang | | International Journal of Molecular Sciences. 2021; 22(11): 5516 | | [Pubmed] | [DOI] | | 37 |
Las propiedades epigenéticas y anticáncer del ácido valproico |
|
| Ismael Vásquez-Moctezuma,Estefanía Fernández-Navarrete,Gabriela Rebeca Luna-Palencia | | Revista Mexicana de Mastología. 2020; 10(2): 54 | | [Pubmed] | [DOI] | | 38 |
Role of Histone Deacetylases in Carcinogenesis: Potential Role in Cholangiocarcinoma |
|
| Kishor Pant,Estanislao Peixoto,Seth Richard,Sergio A. Gradilone | | Cells. 2020; 9(3): 780 | | [Pubmed] | [DOI] | | 39 |
Bladder Cancer Metastasis Induced by Chronic Everolimus Application Can Be Counteracted by Sulforaphane In Vitro |
|
| Saira Justin,Jochen Rutz,Sebastian Maxeiner,Felix K.-H. Chun,Eva Juengel,Roman A. Blaheta | | International Journal of Molecular Sciences. 2020; 21(15): 5582 | | [Pubmed] | [DOI] | | 40 |
A Toxicity Prediction Tool for Potential Agonist/Antagonist Activities in Molecular Initiating Events Based on Chemical Structures |
|
| Kota Kurosaki,Raymond Wu,Yoshihiro Uesawa | | International Journal of Molecular Sciences. 2020; 21(21): 7853 | | [Pubmed] | [DOI] | | 41 |
Epigenetic Research in Stem Cell Bioengineering—Anti-Cancer Therapy, Regenerative and Reconstructive Medicine in Human Clinical Trials |
|
| Claudia Dompe,Krzysztof Janowicz,Greg Hutchings,Lisa Moncrieff,Maurycy Jankowski,Mariusz J. Nawrocki,Malgorzata Józkowiak,Paul Mozdziak,Jim Petitte,Jamil A. Shibli,Marta Dyszkiewicz-Konwinska,Malgorzata Bruska,Hanna Piotrowska-Kempisty,Bartosz Kempisty,Michal Nowicki | | Cancers. 2020; 12(4): 1016 | | [Pubmed] | [DOI] | | 42 |
Recent Progress in Histone Deacetylase Inhibitors as Anticancer Agents |
|
| Loredana Cappellacci,Diego R. Perinelli,Filippo Maggi,Mario Grifantini,Riccardo Petrelli | | Current Medicinal Chemistry. 2020; 27(15): 2449 | | [Pubmed] | [DOI] | | 43 |
Histone Deacetylase Inhibitors in Pediatric Brain Cancers: Biological Activities and Therapeutic Potential |
|
| Alexandre Perla,Lívia Fratini,Paula S. Cardoso,Carolina Nör,André T. Brunetto,Algemir L. Brunetto,Caroline Brunetto de Farias,Mariane Jaeger,Rafael Roesler | | Frontiers in Cell and Developmental Biology. 2020; 8 | | [Pubmed] | [DOI] | | 44 |
Recent Update of HDAC Inhibitors in Lymphoma |
|
| I-Chung Chen,Bidyadhar Sethy,Jing-Ping Liou | | Frontiers in Cell and Developmental Biology. 2020; 8 | | [Pubmed] | [DOI] | | 45 |
Construction prognosis model based on autophagy-related gene signatures in hepatocellular carcinoma |
|
| Jiangtao Wang,Yandong Miao,Juntao Ran,Yuan Yang,Quanlin Guan,Denghai Mi | | Biomarkers in Medicine. 2020; 14(13): 1229 | | [Pubmed] | [DOI] | | 46 |
A novel HDAC inhibitor chidamide combined with imatinib synergistically targets tyrosine kinase inhibitor resistant chronic myeloid leukemia cells |
|
| Bailin He,Qiang Wang,Xiaoli Liu,Ziyuan Lu,Jiaochan Han,Chengyun Pan,Bing Z. Carter,Qifa Liu,Na Xu,Hongsheng Zhou | | Biomedicine & Pharmacotherapy. 2020; 129: 110390 | | [Pubmed] | [DOI] | | 47 |
Molecular docking-based virtual drug screening revealing an oxofluorenyl benzamide and a bromonaphthalene sulfonamido hydroxybenzoic acid as HDAC6 inhibitors with cytotoxicity against leukemia cells |
|
| Mona Dawood,Mohamed Elbadawi,Madeleine Böckers,Gerhard Bringmann,Thomas Efferth | | Biomedicine & Pharmacotherapy. 2020; 129: 110454 | | [Pubmed] | [DOI] | | 48 |
Class IIa HDAC inhibitor TMP195 alleviates lipopolysaccharide-induced acute kidney injury |
|
| Wei Zhang,Yinjie Guan,George Bayliss,Shougang Zhuang | | American Journal of Physiology-Renal Physiology. 2020; 319(6): F1015 | | [Pubmed] | [DOI] | | 49 |
Genome-wide identification and transcriptional modulation of histone variants and modification related genes in the low pH-exposed marine rotifer Brachionus koreanus |
|
| Young Hwan Lee,Min-Sub Kim,Haksoo Jeong,Atsushi Hagiwara,Jae-Seong Lee | | Comparative Biochemistry and Physiology Part D: Genomics and Proteomics. 2020; 36: 100748 | | [Pubmed] | [DOI] | | 50 |
Gelated vorinostat with inner-lysosome triggered release for tumor-targeting chemotherapy |
|
| Changyong Guo,Qirong Wang,Xingjie Zhang,Fengkun Lu,Miao Sun,Peiyu Zeng,Linhong Sun,Lan She,Bingkai Wang,Yunchang Zhang,Chen Wang,Zhiqiang Ma,Feng Yang | | Colloids and Surfaces B: Biointerfaces. 2020; : 111144 | | [Pubmed] | [DOI] | | 51 |
The Development Process: from SAHA to Hydroxamate HDAC Inhibitors with Branched CAP Region and Linear Linker |
|
| Feifei Yang,Na Zhao,Yunjie Hu,Cheng-Shi Jiang,Hua Zhang | | Chemistry & Biodiversity. 2019; | | [Pubmed] | [DOI] | | 52 |
Epigenetic Control of Autophagy in Cancer Cells: A Key Process for Cancer-Related Phenotypes |
|
| Paul Peixoto,Céline Grandvallet,Jean-Paul Feugeas,Michaël Guittaut,Eric Hervouet | | Cells. 2019; 8(12): 1656 | | [Pubmed] | [DOI] | |
|
 |
 |
|