ArticlePDF AvailableLiterature Review

Histone Deacetylase Inhibitors: A Promising Therapeutic Alternative for Endometrial Carcinoma

Authors:

Abstract and Figures

Endometrial carcinoma is the most common malignant tumor of the female genital tract in the United States. Epigenetic alterations are implicated in endometrial cancer development and progression. Histone deacetylase inhibitors are a novel class of anticancer drugs that increase the level of histone acetylation in many cell types, thereby inducing cell cycle arrest, differentiation, and apoptotic cell death. This review is aimed at determining the role of histone acetylation and examining the therapeutic potential of histone deacetylase inhibitors in endometrial cancer. In order to identify relevant studies, a literature review was conducted using the MEDLINE and LIVIVO databases. The search terms histone deacetylase, histone deacetylase inhibitor, and endometrial cancer were employed, and we were able to identify fifty-two studies focused on endometrial carcinoma and published between 2001 and 2021. Deregulation of histone acetylation is involved in the tumorigenesis of both endometrial carcinoma histological types and accounts for high-grade, aggressive carcinomas with worse prognosis and decreased overall survival. Histone deacetylase inhibitors inhibit tumor growth, enhance the transcription of silenced physiologic genes, and induce cell cycle arrest and apoptosis in endometrial carcinoma cells both in vitro and in vivo. The combination of histone deacetylase inhibitors with traditional chemotherapeutic agents shows synergistic cytotoxic effects in endometrial carcinoma cells. Histone acetylation plays an important role in endometrial carcinoma development and progression. Histone deacetylase inhibitors show potent antitumor effects in various endometrial cancer cell lines as well as tumor xenograft models. Additional clinical trials are however needed to verify the clinical utility and safety of these promising therapeutic agents in the treatment of patients with endometrial cancer.
This content is subject to copyright. Terms and conditions apply.
Review Article
Histone Deacetylase Inhibitors: A Promising Therapeutic
Alternative for Endometrial Carcinoma
Iason Psilopatis ,
1,2
Alexandros Pergaris ,
1
Constantinos Giaginis ,
3
and Stamatios Theocharis
1
1
First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Bld 10,
Goudi, 11527 Athens, Greece
2
Charité-University School of Medicine, Augustenburger Pl. 1, 13353 Berlin, Germany
3
Department of Food Science and Nutrition, University of Aegean, Lemnos, Greece
Correspondence should be addressed to Stamatios Theocharis; stamtheo@med.uoa.gr
Received 18 August 2021; Revised 19 October 2021; Accepted 30 October 2021; Published 12 November 2021
Academic Editor: Kristina W. Thiel
Copyright © 2021 Iason Psilopatis et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Endometrial carcinoma is the most common malignant tumor of the female genital tract in the United States. Epigenetic
alterations are implicated in endometrial cancer development and progression. Histone deacetylase inhibitors are a novel class
of anticancer drugs that increase the level of histone acetylation in many cell types, thereby inducing cell cycle arrest,
dierentiation, and apoptotic cell death. This review is aimed at determining the role of histone acetylation and examining the
therapeutic potential of histone deacetylase inhibitors in endometrial cancer. In order to identify relevant studies, a literature
review was conducted using the MEDLINE and LIVIVO databases. The search terms histone deacetylase,histone deacetylase
inhibitor, and endometrial cancer were employed, and we were able to identify fty-two studies focused on endometrial
carcinoma and published between 2001 and 2021. Deregulation of histone acetylation is involved in the tumorigenesis of both
endometrial carcinoma histological types and accounts for high-grade, aggressive carcinomas with worse prognosis and
decreased overall survival. Histone deacetylase inhibitors inhibit tumor growth, enhance the transcription of silenced
physiologic genes, and induce cell cycle arrest and apoptosis in endometrial carcinoma cells both in vitro and in vivo. The
combination of histone deacetylase inhibitors with traditional chemotherapeutic agents shows synergistic cytotoxic eects in
endometrial carcinoma cells. Histone acetylation plays an important role in endometrial carcinoma development and
progression. Histone deacetylase inhibitors show potent antitumor eects in various endometrial cancer cell lines as well as
tumor xenograft models. Additional clinical trials are however needed to verify the clinical utility and safety of these promising
therapeutic agents in the treatment of patients with endometrial cancer.
1. Introduction
The nucleosome is the building block of DNA structural orga-
nization and enables the necessary packaging of the genetic
material in a denser form tting within the eukaryotic nucleus.
It refers to a negatively charged DNA strand wrapped around
a positively charged histone octamer, a protein core consisting
of two identical copies of each of the four core histone proteins
(H2A, H2B, H3, and H4) [1, 2]. In this condensed formation,
histones have low levels ofacetylation on the lysine residues of
their aminoterminal tails, thus blocking the assembly of the
basal transcriptional factors to form the preinitiation complex
that allows genetic expression [3, 4]. The post-translational
modication of the NH2-terminal tails of histones by acetyla-
tion neutralizes the positive charge on lysine residues and
reduces the anity of histone for the negatively charged
DNA. As such, DNA strands may uncoil and transcription
may occur [5]. The level of histone acetylation is modulated
by the opposing actions of histone acetylases (HATs) and his-
tone deacetylases (HDACs) [6]. HDACs catalyze the removal
of acetyl groups on the NH2-terminal lysine residues of core
nucleosomal histones, which generally results in transcriptional
Hindawi
Disease Markers
Volume 2021, Article ID 7850688, 9 pages
https://doi.org/10.1155/2021/7850688
repression and silencing of tumor-suppressor genes [7, 8].
Consequently, deregulation of histone acetylation can pro-
mote the development of certain human cancers, as shown
by a great number of researchers who focused on revealing
the link between histone acetylation/deacetylation and carci-
nogenesis [9, 10].
Endometrial carcinoma (EC) is the most common malig-
nant tumor of the female genital tract in the United States.
According to the American Cancer Society, about 66,570
new cases of cancer of the body of the uterus will be diagnosed
and about 12,940 women will die from cancers of the uterine
body in the United States in 2021 [11]. EC primarily aects
postmenopausal women aged 55-64, with the median age at
diagnosis being 63 years [12]. ECs can be divided into two
distinct histopathologic subgroups: type I EC deriving from
atypical endometrial hyperplasia and type II EC of non-
endometrioid histology [13]. Type I EC is directly related to
long-term exposure to increased estrogen levels and is associ-
ated with PTEN inactivation by mutation, microsatellite insta-
bility, and mutations of K-ras,β-catenin,orhMLH1/MSH2.
Type II EC is mostly estrogen-independent, develops from
atrophic endometrium in postmenopausal women, and is
characterized by p53 mutations, display inactivation of p16
and E-cadherin, as well as Her2/neu amplication [14, 15].
While surgery is recommended as a monotherapy for low-
risk ECs, adjuvant chemotherapy should be oered to women
with high-intermediate- and high-risk ECs, as well as advanced
or recurrent disease [16]. Combined chemotherapy with carbo-
platin and paclitaxel is the rst-line regimen, followed by
chemotherapeutic agents such as doxorubicin, cyclophospha-
mide, or cisplatin [17].
Despite the reported high response rates, the duration of
response is only short-lasting, ranging from between four and
eight months [18] and 5-year overall survival amounting to
81%, according to the American Cancer Society [19]. However,
prognosis for patients with advanced disease remains grim,
with 5-year survival rates dropping to 17% when distant metas-
tasis is present [19]. Such statistics render imperative the devel-
opment of innovative agents for the eective treatment of EC.
Histone deacetylase inhibitors (HDACIs) are a novel class
of anticancer drugs that increase the level of histone acetyla-
tion in many cell types, thereby inducing cell cycle arrest,
dierentiation, and apoptotic cell death, thus suppressing
carcinogenesis [20, 21] (Figure 1). Few HDACIs have already
received FDA approval for T-cell lymphoma or multiple mye-
loma, yet there is a great number of current clinical trials
investigating the role of HDACIs (alone or in combination
with other anticancer drugs) in the treatment of numerous
solid cancer entities [22, 23]. Given the genetic alternations
in EC, HDACIs could be considered promising therapeutic
agents.
1.1. Histone-Mediated Epigenetics in EC Clinical Samples and
Cell Lines. Histone-mediated epigenetics plays an established
role in EC development and progression. A large number of
studies have assessed the genetic alternations associated with
histone-mediated epigenetics in population-based cohorts of
EC tumor types [15, 2431] (Table 1).
Histone acetylation is involved in the silencing of human
mutL homolog 1 (hMLH1)/mutS homolog 2 (MSH2), phos-
phatase and tensin homolog (PTEN), and progesterone
receptor (PR), thus resulting in early carcinogenesis, more
aggressive carcinomas, and resistance to hormonal treatment,
respectively [15]. Specically, silencing of hMLH1 and/or
MSH2 causes microsatellite instability, invasive growth, and
acquired resistance to cisplatin in EC [24]. Class I HDACs
(HDAC1, HDAC2, and HDAC3) are expressed in the major-
ity of ECs at high levels, with high-grade serous subtypes
exhibiting overexpression of all three HDACs signicantly
more often than endometrioid subtypes [25]. Notably,
HDAC2 overexpression has been suggested to be involved in
the acquisition of aggressive behavior by EC [26]. Krusche
et al. reported that, compared to normal endometrium, many
ECs showed impaired HDAC1 protein expression in the
epithelial and stromal compartment, which might be indicative
of an impaired epigenetic status of epithelial and stromal cells
within ECs [27]. HDAC6, modulated by miR-206, promotes
EC progression through the PTEN/AKT/mTOR pathway
[28]. Deregulating E-cadherin correlates with focal adhesion
kinase (FAK) signaling axis and HDAC/enhancer of zeste
homolog 2 (EZH2) activity. EZH2, FAK, and phospho-FAK
(pFAK) overexpression is mainly identied in type II ECs and
is associated with worse prognosis and decreased overall sur-
vival [29]. Low forkhead box A1 (FOXA1) protein expression
signicantly correlates with high-grade carcinoma, loss of estro-
gen receptor α(ERα) and PR, and poor survival [30]. The
bromodomain-containing gene ATPase family AAA domain
containing 2 (ATAD2) is a mediator of MYC transcriptional
function and represents a marker of aggressive ECs [31].
Several in vitro studies have examined the role of
histone-mediated epigenetics in EC cell lines as well.
Mitogen-inducible gene 6 (MIG6) mRNA levels are
lower in cell lines derived from high-grade ECs than in
low-grade EC cell lines. MIG6 is an essential downstream
component of PR-mediated growth suppression [32]. Aber-
rant expression of miRNAs including miR-200b, miR130a/b,
miR-625, and miR-222 is associated with tumorigenesis and
metastasis in EC cell lines [33].
All of the aforementioned genetic alternations in ECs are
strongly inuenced by histone-mediated epigenetics.
1.2. In Vitro Eects of HDACIs on EC Cell Lines. There are
ve identied classes of HDACIs including organic hydroxa-
mic acids, short-chain fatty acids, benzamides, cyclic tetra-
peptides, and sulfonamide anilides [14, 34]. Dierent
in vitro studies have investigated the eects of various HDA-
CIs on genetic alternations in EC cell lines associated with
histone-mediated epigenetics (Table 2). The reported HDA-
CIs seem to have a profound eect on cell viability by inhi-
bitingcell proliferation and inducing cell death in EC. The
specic chemical structures of HDACIs used in EC-related
studies are depicted in Figure 2.
Apicidin. Apicidin is a fungal metabolite shown to exert
antiparasitic activity by the inhibition of HDAC [35]. In EC
cell lines, Apicidin results in the upregulation of acetylated
H3 and H4, p21, p27, and E-cadherin and the downregula-
tion of cyclin A, cyclin D1, cyclin E, CDK2, CDK4, p53,
2 Disease Markers
HDAC3, and HDAC4. As a result, Apicidin induces morpho-
logical changes, increases the proportion of cells in the G1
phase, and decreases the number of cells in the S phase [18,
36, 37]. Moreover, Apicidin increases the level of PARP cleav-
age and caspase-3 activity, induces cytoplasmic localization of
cytochromec, and causes the downregulation ofthe antiapop-
totic gene, Bcl-2, and upregulation of the proapoptotic gene,
Bax, thus inducing apoptotic cell death [18, 36]. Concerning
estrogen-dependent cancers, Apicidin suppresses transcrip-
tion of 17β-hydroxy steroid dehydrogenase type 1 in EC cells,
which is responsible for intratumoral estrone to 17βestradiol
conversion [38].
Trichostatin A (TSA). TSA, an antifungal antibiotic initially
isolated from Streptomyces hygroscopicus, is a potent and spe-
cic HDACI [39]. TSA increases the levels of acetyl H3, acetyl
H4, acetyl tubulin, p21, p27, miR-130b, DICER1, BIM,
L1CAM, FOXA1, glycodelin, and E-cadherin and decreases
the levels of cyclin A, cyclin D1 and D2, MMP2, MMP9,
G0 phase
G1
G2S
M
Cell cycle arrest
Dierentiation
Upregulation of tumor
suppressor genes and PR
Neoangiogenesis
(through VEGF downregulation)
Cell proliferation and
tumor growth
Transcription of oncomirRs
(miR-200b, miR-130a/b,
miR-625, miR-222)
Mitosis
(through MIG6 upregulation) Apoptosis
HDACs
HDAC inhibitors
(resting)
Figure 1: HDAC inhibitors exert their tumor-suppressive role through various mechanisms. Green arrows: procedures enhanced by HDAC
inhibitors. Red arrows: procedures blocked by HDAC inhibitors (created with http://Biorender.com). HDAC: histone deacetylase; PR:
progesterone receptor; MIG6: mitogen-inducible gene 6; VEGF: vascular endothelial growth factor.
Table 1: Genetic alternations in ECs associated with histone-mediated epigenetics.
Genetic alternations in ECs Impact on EC development and progression Reference
Silencing of hMLH1/MSH2, PTEN, and PR Early carcinogenesis, more aggressive carcinomas, resistance to hormonal
treatment [15]
Silencing of hMLH1 and/or MSH2 Microsatellite instability, invasive growth, acquired resistance to cisplatin [24]
Overexpression of class I HDACs Signicantly more often in high-grade serous subtypes [25]
Overexpression of HDAC2 Acquisition of aggressive behavior [26]
Impaired HDAC1 protein expression Impaired epigenetic status of epithelial and stromal cells [27]
miR-206 modulation of HDAC6 Progression through the PTEN/AKT/mTOR pathway [28]
Overexpression of EZH2, FAK, and pFAK Worse prognosis, decreased overall survival [29]
Low FOXA1 protein expression High-grade carcinomas, loss of ERαand PR, poor survival [30]
ATAD2 expression Aggressive carcinomas [31]
Low MIG6 mRNA levels High-grade carcinomas, failure of PR-mediated growth suppression [32]
Aberrant expression of miRNAs Tumorigenesis, metastasis [33]
3Disease Markers
Table 2: In vitro eects of HDACIs on EC cell lines.
HDACI Upregulatory eects Downregulatory eects Synergetic eects References
Apicidin Acetylated H3 and H4, p21, p27,
E-cadherin, PARP, caspase-3, cytochrome c, Bax
Cyclin A, cyclin D1, cyclin E, CDK2,
CDK4, p53, HDAC3,
HDAC4, Bcl-2, 17β-hydroxysteroid-dehydrogenase
type 1
n/a [18, 3638]
TSA
Acetylated H3, H4, and tubulin, p21, p27, miR-
130b, DICER1, BIM, L1CAM, FOXA1, glycodelin,
E-cadherin, PARP, caspase-3
Cyclin A, cyclin D1 and D2, MMP2,
MMP9, DNMT3B mRNA, ERα,
MCM7 mRNA, MYC,
miR-106b-93-25
Aza-deoxycytidine: PR-B upregulation
High-glucose condition: degradation of
CLDN-2
Paclitaxel: cell death induction
[26, 30, 31, 33,
4050]
SAHA
Acetylated H3 and H4 bound to either Tig1 or C/
ebpa gene, caspase-8 and caspase-9, glycodelin,
FOXA1, E-cadherin, p21, p27, insulin-like growth
factor-I receptor
Cyclin D1 and D2, Bcl-2, FLIP mRNA and protein
levels, AURKA n/a [30, 40, 41, 5358]
LBH589 PR mRNA, MIG6 MYC
Death receptor ligand TRAIL: cell
death induction after knockdown of
metadherin
Proteasome: overcomes the impact of
gain-of-function p53 mutations
[32, 6063]
NaB Acetylated H3 and H4, p21, p27, ROS, phospho-
p38 mitogen-activated protein kinase, γH2AX ERαAdriamycin: high human telomerase
reverse transcriptase expression [41, 42, 6568]
VPA E-cadherin Bcl-2 VE465: PARP cleavage induction [56, 70, 71]
OBP-801/YM753 n/a n/a LY294002: BIM increase with
accumulation of ROS [72]
Oxamatin PARP, caspase-8 and caspase-9 n/a n/a [15]
Scriptaid Acetylated H3 and H4, p21, p27, E-cadherin Cyclin A, Bcl-2 n/a [75]
FK228 Acetylated H3 and H4, p21, p53, caspase-3,
caspase-7, and caspase-8, PARP n/a n/a [77]
PsA Acetylated H3 and H4, p21 p53, pRb, cyclins, CDKs n/a [78]
MHY2256 p53 SIRT1 enzyme activity, SIRT protein levels, MDM2 n/a [79]
4 Disease Markers
DNMT3B mRNA, ERα, and MCM7 mRNA [26, 30, 33,
4046]. After treatment with TSA, cleavage of PARP and
caspase-3 was observed, indicating its apoptotic eects [26,
46]. TSA inhibits cell proliferation by arrest in the G1 and/or
G2 phases of the cell cycle [33, 46]. Raeder et al. suggested that
dependency on MYC predicts dependency on ATAD2 and
response to TSA in EC [31], while Zhao et al. demonstrated that
the downregulation of MYC in the presence of TSA resulted in
the reduction of miR-106b-93-25 cluster [46]. TSA acts syner-
gistically with aza-deoxycytidine and results in a robust and sus-
tainable PR-B upregulation [47]. High-glucose condition and
TSA induce degradation of CLDN-2 in Sawano cells [48].
TSA in combination with paclitaxel induces synergistic cell
death, results in signicant morphologic changes, induces
activation of the intrinsic mitochondria-dependent apoptotic
pathway, and stabilizes microtubules [49, 50].
Suberoylanilide bis hydroxamine (SAHA, Vorinostat).
Vorinostat is a HDACI that reacts with and blocks the cata-
lytic site of HDACs [51, 52]. SAHA induces the activation of
caspase-8 and caspase-9, results in the upregulation of glyco-
delin and acetylated H3 and H4 bound to either Tig1 or
C/ebpa gene, downregulates the expression of Bcl-2, cyclin
D1, and D2, increases the levels of FOXA1, E-cadherin,
p21, and p27, causes a dramatic decrease of FLIP mRNA
and protein levels, and induces apoptosis in EC [30, 40, 41,
5356]. Sarfstein et al. examined SAHAs mechanism of
action in type I and type II EC cell lines in the presence or
absence of IGF-I and found out that Vorinostat exhibits a
potent apoptotic and antiproliferative eect in both type I
and II EC cells through interaction with the insulin-like
growth factor signaling pathway [57]. SAHA is also eective
at reducing AURKA expression in EC, a cell-cycle-regulated
kinase that functions in spindle formation and chromosome
segregation during the M phase of the cell cycle [58].
Panobinostat (LBH589). LBH589 is a potent pan-
deacetylase inhibitor [59]. Treatment with LBH589 induces
a profound upregulation of PR mRNA and MIG6, cell cycle
arrest in G1, and a downregulation of the oncogene MYC
[32, 60, 61]. Knockdown of metadherin sensitizes EC cells
to cell death induction by death receptor ligand TRAIL
and LBH589 co-treatment [62] while the combination of
proteasome and LBH589 overcomes the impact of gain-of-
function p53 mutations [63].
Sodium butyrate (NaB). NaB is a part of the metabolic
fatty acid fuel cycle that also acts as a HDACI [64]. NaB induces
upregulation of p21, p27, acetyl H3, and H4 and inhibition of
transcription from multiple ERαpromoters, cell cycle arrest,
and apoptosis [41, 42, 65]. The addition of NaB signicantly
enhances adriamycin cytotoxicity for the primary EC cells with
high human telomerase reverse transcriptase expression [66].
NaB has been also reported to inhibit the self-renewal capacity
of endometrial tumor side-population cells by promoting the
production of intracellular ROS and by upregulating the
expression of the phospho-p38 mitogen-activated protein
kinase, γH2AX, acetyl H3, p21, and p27 [67, 68].
Valproic acid (VPA). VPA is a HDACI approved for the
treatment of epilepsy [24, 69]. VPA inhibits proliferation,
induces cell cycle arrest, enhances the apoptotic index in EC
cell lines, upregulates E-cadherin mRNA and protein levels,
and downregulates Bcl-2 mRNA levels [56]. Moreover, VPA
enhances the action of antiestrogens in ERα-positive breast
cancer cells and blocks tamoxifen-induced proliferation of
uterine cells [70]. Cotreatment with VPA and the Aurora
CH3
CH3
O
N
O
NH
O
HN
ONH
N
O
CH3
O
CH3
Apicidin
CH3CH3
O
NH
OH
O
N
CH3
CH3
Trichostatin A (TSA)
NH
O
O
NH OH
Suberoylanilide bishydroxamine (SAHA)
CH3
NH
NH
O
NH
HO
Panobinostat (LBH589)
Na+(I)
CH3O
Sodium Butyrate (NaB)
CH3
CH3
O
O
O
Valproic acid (VPA)
CH3O
HN S
S
ONH
O
O
NH
O
CH3
CH3
HO
OBP-801/YM753
SO
O
NH
O
NH OH
ONO
O
HN
OH
Scriptaid
CH3
O
NH
O
O
O
NH
O
NH
S
S
ONH
CH3
CH3
CH3
CH3
Romidepsin (FK228)
N
OH
O
NH
S
S
NH
O
N
OH
OH
Br
Br OH
Psammaplin A (PsA)
NH
NH
S
O
O
OH
MHY2256
Figure 2: Chemical structures of HDACIs used in EC treatment
studies.
5Disease Markers
kinase inhibitor VE465 induces enhanced apoptosis, cleaved
PARP, and cytotoxic eects in EC cells [71].
OBP-801/YM753. Combination of the novel HDAC inhib-
itor OBP-801/YM753 and the PI3K inhibitor LY294002
synergistically induces apoptosis in human EC cells due to
increase of BIM with accumulation of ROS [72].
Oxamatin. Oxamatinis is a HDACI that induces tran-
scriptional activation of jun D and morphological reversion
in v-Kras-transformed NIH3T3 cells [73]. Administration
of Oxamatin causes morphologic changes, loss of mito-
chondrial membrane potentials, and cleavage of PARP,
caspase-8, and caspase-9, conrming the activation of apo-
ptotic cascades in EC cells [15].
Scriptaid. Scriptaid is a potent HDACI with a >100-fold
increase in histone acetylation, with relatively low toxicity
[74]. Exposure to Scriptaid decreases the proportion of cells
in the S phase, increases the proportion in the G0/G1 and/or
G2/M phases of the cell cycle, upregulates the expression of
E-cadherin, acetyl-H3 and acetyl-H4, p21, and p27, downre-
gulates the expression of cyclin A and Bcl-2, and induces
apoptosis in EC cells [75].
Romidepsin (FK228). FK228 is a HDACI which has been
conrmed as a useful anticancer agent [76]. In EC cell lines,
FK228 induces apoptosis and cell cycle arrest at G0/G1
phase, increases the mRNA and protein expressions of p53,
p21, cleaved caspases such as 3, 7, and 8, and PARP, and
upregulates the acetylation of H3 and H4 [77].
Psammaplin A (PsA). PsA is a natural bromotyrosine
derivative from a two-sponge association, Poecillastra sp.
and Jaspis sp., which was rst isolated from the Psammaply-
silla sponge. PsA induces the expression of acetylated H3
and H4 histone proteins, upregulates the expression of
cyclin-dependent kinase inhibitors and p21, and downregu-
lates the expression of p53, pRb, cyclins, and CDKs, which
lead to induce cell cycle arrest [78].
MHY2256. MHY2256 is a novel HDACI that inhibits
class III HDAC sirtuin (SIRT). MHY2256 reduces both
SIRT1 enzyme activity and SIRT protein levels in EC cells,
inhibits cell cycle distribution, increases p53 levels, reduces
the expression of mouse double minute 2 (MDM2), and
induces apoptotic/autophagic cell death [79].
Takai et al. have summarized the half maximal inhibi-
tory concentrations (IC50) of the dierent classes of HDA-
CIs which indicate how much of each HDACI is needed to
inhibit in vitro cell growth in EC cell lines by 50% [14].
1.3. In Vivo Impact of HDACI Use in EC. Several studies
have examined the anti-tumor eect of HDACIs on human
EC cells in mouse models (Table 3).
Apicidin downregulates HDAC3 and HDAC4 and sup-
presses the tumor growth of transplanted Ishikawa cells,
the expression of proliferative cell nuclear antigen (PCNA),
and vascular endothelial growth factor (VEGF) in tumor
xenograft model, respectively [37].
Co-treatment with TSA and paclitaxel results in a signi-
cant reduction in tumor weight, increases microtubule stabili-
zation, and induces apoptosis as well as tubulin acetylation in
mouse xenograft models [50].
Combination of Vorinostat and caspase-8 inhibition causes
a nearly complete inhibition of tumor xenograft growth [53].
NaB results in marked suppression of tumor growth and
SA-β-gal activity in tumor xenograft models [65].
VPA and MHY2256 signicantly inhibit human uterine
tumor growth without toxic side eects in mouse models
[41, 79]. Notably, VPA inhibits tumor growth, upregulates
CDH1 mRNA, and downregulates Bcl-2 mRNA levels
in vivo [56]. Yoshioka et al. showed that combined treatment
with OBP-801/YM753 and LY294002 signicantly suppressed
tumor growth compared to the control in vivo [72].
In a surgical window trial of women with newly diag-
nosed endometrioid EC, co-treatment with medroxyproges-
terone acetate and the HDACI Entinostat resulted in the
reduction of PR H-scores and Ki-67 levels [80].
2. Conclusions
The present review summarizes the important role of
HDACs in EC development and progression and highlights
the potent antitumor eects of various HDACIs on EC cell
lines both in vitro and in vivo. HDACs seem to be involved
in the tumorigenesis of both EC tumor types and account for
high-grade, aggressive carcinomas with worse prognosis and
Table 3: Antitumor eects of HDACIs on human EC cells in mouse models.
HDACI Upregulatory
eects
Downregulatory
eects Synergetic eects References
Apicidin n/a
HDAC3, HDAC4,
PCNA, VEGF,
Tumor growth
n/a [37]
TSA n/a n/a Paclitaxel: reduction in tumor weight, increase in microtubule
stabilization, apoptosis induction, tubulin acetylation induction [50]
SAHA n/a n/a Caspase-8 inhibition: tumor growth inhibition [53]
NaB n/a SA-β-gal activity,
Tumor growth n/a [65]
VPA CDH1
mRNA
Bcl-2,
Tumor growth n/a [41, 56, 79]
MHY2256 n/a Tumor growth n/a [41, 79]
OBP-801/YM753 n/a n/a LY294002: tumor growth inhibition [72]
6 Disease Markers
decreased overall survival. HDACIs represent promising
therapeutic agents that inhibit tumor growth, enhance the
transcription of silenced physiologic genes, and induce cell
cycle arrest and apoptosis in EC cells. Notably, the combina-
tion of HDACIs with traditional chemotherapeutic agents
shows synergistic cytotoxic eects in EC cells. Nevertheless,
clinical trials are needed to verify the clinical utility and
safety of HDACIs in the treatment of women with EC, to
investigate possible adverse side eects following their
administration to patients and to assure their eectiveness
depending on HDAC expression by EC cells.
Conflicts of Interest
The authors declare no conict of interest.
AuthorsContributions
Literature analysis and conceptualization were contributed
by I.P., A.P., C.G., and S.T.; original draft preparation and
writing were performed by I.P.; art work was done by A.P.;
review and supervision were contributed by S.T. and C.G.
All authors have read and agreed to the submitted version
of the manuscript.
References
[1] K. Luger, A. W. Mäder, R. K. Richmond, D. F. Sargent, and
T. J. Richmond, Crystal structure of the nucleosome core par-
ticle at 2.8 Å resolution,Nature, vol. 389, no. 6648, pp. 251
260, 1997.
[2] C. A. Davey, D. F. Sargent, K. Luger, A. W. Maeder, and T. J.
Richmond, Solvent Mediated Interactions in the Structure
of the Nucleosome Core Particle at 1.9 Å Resolution,Journal
of Molecular Biology, vol. 319, no. 5, pp. 10971113, 2002.
[3] G. Felsenfeld, Chromatin as an essential part of the transcrip-
tional mechanim,Nature, vol. 355, no. 6357, pp. 219224,
1992.
[4] A. P. Wole, Transcription: in tune with the histones,Cell,
vol. 77, no. 1, pp. 1316, 1994.
[5] T. Kouzarides, Chromatin modications and their function,
Cell, vol. 128, no. 4, pp. 693705, 2007.
[6] N. J. McKenna and B. W. O'Malley, Combinatorial control of
gene expression by nuclear receptors and coregulators,Cell,
vol. 108, no. 4, pp. 465474, 2002.
[7] M. Grunstein, Histone acetylation in chromatin structure and
transcription,Nature, vol. 389, no. 6649, pp. 349352, 1997.
[8] P. A. Marks, V. M. Richon, and R. A. Rifkind, Histone deacet-
ylase inhibitors: inducers of dierentiation or apoptosis of
transformed cells,Journal of the National Cancer Institute,
vol. 92, no. 15, pp. 12101216, 2000.
[9] S. Y. Archer and R. A. Hodint, Histone acetylation and can-
cer,Current Opinion in Genetics & Development, vol. 9,
no. 2, pp. 171174, 1999.
[10] S. Ropero and M. Esteller, The role of histone deacetylases
(HDACs) in human cancer,Molecular Oncology,vol. 1,
no. 1, pp. 1925, 2007.
[11] American Cancer Society, Facts & gures 2021, American
Cancer Society, Atlanta, Ga, 2021.
[12] National Cancer Institute, Cancer Stat Facts: Uterine Can-
cer,2021, April 2021, https://seer.cancer.gov/statfacts/html/
corp.html.
[13] M. E. Sherman, M. E. Bur, and R. J. Kurman, p53 in endome-
trial cancer and its putative precursors: evidence for diverse
pathways of tumorigenesis,Human Pathology, vol. 26,
no. 11, pp. 12681274, 1995.
[14] N. Takai and H. Narahara, Preclinical studies of chemother-
apy using histone deacetylase inhibitors in endometrial can-
cer,Obstetrics and Gynecology International, vol. 2010,
Article ID 923824, 8 pages, 2010.
[15] S. Jiang, S. C. Dowdy, X. W. Meng et al., Histone deacetylase
inhibitors induce apoptosis in both type I and type II endome-
trial cancer cells,Gynecologic Oncology, vol. 105, no. 2,
pp. 493500, 2007.
[16] N. Concin, X. Matias-Guiu, I. Vergote et al., ESGO/ES-
TRO/ESP guidelines for the management of patients with
endometrial carcinoma,International Journal of Gynecologi-
cal Cancer, vol. 31, no. 1, pp. 1239, 2021.
[17] C. M. Bestvina and G. F. Fleming, Chemotherapy for endo-
metrial cancer in adjuvant and advanced disease settings,
The Oncologist, vol. 21, no. 10, pp. 12501259, 2016.
[18] T. Ueda, N. Takai, M. Nishida, K. Nasu, and H. Narahara,
Apicidin, a novel histone deacetylase inhibitor, has profound
anti-growth activity in human endometrial and ovarian cancer
cells,International Journal of Molecular Medicine, vol. 19,
no. 2, pp. 301308, 2007.
[19] American Cancer Society, Survival rates for endometrial can-
cer, American Cancer Society, Atlanta, Ga, 2021.
[20] P. A. Marks, V. M. Richon, T. Miller, and W. K. Kelly, His-
tone deacetylase inhibitors,Advances in Cancer Research,
vol. 91, pp. 137168, 2004.
[21] J. Tasoulas, C. Giaginis, E. Patsouris, E. Manolis, and
S. Theocharis, Histone deacetylase inhibitors in oral squa-
mous cell carcinoma treatment,Expert Opinion on Investiga-
tional Drugs, vol. 24, no. 1, pp. 6978, 2015.
[22] T. Eckschlager, J. Plch, M. Stiborova, and J. Hrabeta, Histone
deacetylase inhibitors as anticancer drugs,International Jour-
nal of Molecular Sciences, vol. 18, no. 7, p. 1414, 2017.
[23] R. Jenke, N. Reßing, F. K. Hansen, A. Aigner, and T. Büch,
Anticancer therapy with HDAC inhibitors: mechanism-
based combination strategies and future perspectives,Can-
cers, vol. 13, no. 4, p. 634, 2021.
[24] N. Garmpis, C. Damaskos, A. Garmpi et al., Targeting histone
deacetylases in endometrial cancer: a paradigm-shifting thera-
peutic strategy?,European Review for Medical and Pharmaco-
logical Sciences, vol. 22, no. 4, pp. 950960, 2018.
[25] W. Weichert, C. Denkert, A. Noske et al., Expression of class I
histone deacetylases indicates poor prognosis in endometrioid
subtypes of ovarian and endometrial carcinomas,Neoplasia,
vol. 10, no. 9, pp. 10211027, 2008.
[26] H. Fakhry, T. Miyamoto, H. Kashima et al., Immunohisto-
chemical detection of histone deacetylases in endometrial car-
cinoma: involvement of histone deacetylase 2 in the
proliferation of endometrial carcinoma cells,Human Pathol-
ogy, vol. 41, no. 6, pp. 848858, 2010.
[27] C. A. Krusche, A. J. Vloet, I. Classen-Linke, U. von Rango,
H. M. Beier, and J. Alfer, Class I histone deacetylase expres-
sion in the human cyclic endometrium and endometrial ade-
nocarcinomas,Human Reproduction, vol. 22, no. 11,
pp. 29562966, 2007.
7Disease Markers
[28] Y. Zheng, X. Yang, C. Wang et al., HDAC6, modulated by
miR-206, promotes endometrial cancer progression through
the PTEN/AKT/mTOR pathway,Scientic Reports, vol. 10,
no. 1, p. 3576, 2020.
[29] J. Zhou, J. W. Roh, S. Bandyopadhyay et al., Overexpression
of enhancer of zeste homolog 2 (EZH2) and focal adhesion
kinase (FAK) in high grade endometrial carcinoma,Gyneco-
logic Oncology, vol. 128, no. 2, pp. 344348, 2013.
[30] I. L. Tangen, C. Krakstad, M. K. Halle et al., Switch in FOXA1
status associates with endometrial cancer progression,PLoS
One, vol. 9, no. 5, article e98069, 2014.
[31] M. B. Raeder, E. Birkeland, J. Trovik et al., Integrated genomic
analysis of the 8q24 amplication in endometrial cancers iden-
ties ATAD2 as essential to MYC-dependent cancers,PLoS
One, vol. 8, no. 2, article e54873, 2013.
[32] H. Ando, T. Miyamoto, H. Kashima et al., Panobinostat
enhances growth suppressive eects of progestin on endome-
trial carcinoma by increasing progesterone receptor and
mitogen-inducible gene-6,Hormones and Cancer, vol. 8,
no. 4, pp. 257267, 2017.
[33] B. L. Li, W. Lu, C. Lu et al., CpG island hypermethylation-
associated silencing of microRNAs promotes human endome-
trial cancer,Cancer Cell International,vol.13,no.1,p.44,2013.
[34] G. Kouraklis and S. Theocharis, Histone deacetylase inhibi-
tors and anticancer therapy,Current Medicinal Chemistry.
Anti-Cancer Agents, vol. 2, no. 4, pp. 477484, 2002.
[35] S. J. Darkin-Rattray, A. M. Gurnett, R. W. Myers et al., Apici-
din: a novel antiprotozoal agent that inhibits parasite histone
deacetylase,Proceedings of the National Academy of Sciences
of the United States of America, vol. 93, no. 23, pp. 13143
13147, 1996.
[36] M. Y. Ahn, J. Lee, Y. J. Na et al., Mechanism of apicidin-
induced cell cycle arrest and apoptosis in Ishikawa human
endometrial cancer cells,Chemico-Biological Interactions,
vol. 179, no. 2-3, pp. 169177, 2009.
[37] M. Y. Ahn, H. Y. Chung, W. S. Choi, B. M. Lee, S. Yoon, and
H. S. Kim, Anti-tumor eect of apicidin on Ishikawa human
endometrial cancer cells both in vitro and in vivo by blocking
histone deacetylase 3 and 4,International Journal of Oncol-
ogy,vol. 36, no. 1, pp. 125131, 2010.
[38] E. Keleş, M. Lianeri, and P. P. Jagodziński, Apicidin sup-
presses transcription of 17β-hydroxysteroid dehydrogenase
type 1 in endometrial adenocarcinoma cells,Molecular Biol-
ogy Reports, vol. 38, no. 5, pp. 33553360, 2011.
[39] N. Tsuji, M. Kobayashi, K. Nagashima, Y. Wakisaka, and
K. Koizumi, A new antifungal antibiotic, trichostatin,Jour-
nal of Antibiotics, vol. 29, no. 1, pp. 16, 1976.
[40] H. Uchida, T. Maruyama, T. Nagashima, H. Asada, and
Y. Yoshimura, Histone deacetylase inhibitors induce dieren-
tiation of human endometrial adenocarcinoma cells through
up-regulation of glycodelin,Endocrinology, vol. 146, no. 12,
pp. 53655373, 2005.
[41] N. Takai, J. C. Desmond, T. Kumagai et al., Histone deacetyl-
ase inhibitors have a profound antigrowth activity in endome-
trial cancer cells,Clinical Cancer Research, vol. 10, no. 3,
pp. 11411149, 2004.
[42] W. Rocha, R. Sanchez, J. Deschênes et al., Opposite eects of
histone deacetylase inhibitors on glucocorticoid and estrogen
signaling in human endometrial Ishikawa cells,Molecular
Pharmacology, vol. 68, no. 6, pp. 18521862, 2005.
[43] R. Margueron, V. Duong, S. Bonnet et al., Histone deacetylase
inhibition and estrogen receptor alpha levels modulate the
transcriptional activity of partial antiestrogens,Journal of
Molecular Endocrinology, vol. 32, no. 2, pp. 583594, 2004.
[44] Y. Xiong, S. C. Dowdy, K. C. Podratz et al., Histone deacetyl-
ase inhibitors decrease DNA methyltransferase-3B messenger
RNA stability and down-regulate de novo DNA methyltrans-
ferase activity in human endometrial cells,Cancer Research,
vol. 65, no. 7, pp. 26842689, 2005.
[45] U. Schirmer, H. Fiegl, M. Pfeifer et al., Epigenetic regulation
of L1CAM in endometrial carcinoma: comparison to cancer-
testis (CT-X) antigens,BMC Cancer, vol. 13, no. 1, p. 156,
2013.
[46] Z. N. Zhao, J. X. Bai, Q. Zhou et al., TSA suppresses miR-
106b-93-25 cluster expression through downregulation of
MYC and inhibits proliferation and induces apoptosis in
human EMC,PLoS One, vol. 7, no. 9, article e45133, 2012.
[47] Y. Xiong, S. C. Dowdy, J. Gonzalez Bosquet et al., Epigenetic-
mediated upregulation of progesterone receptor B gene in
endometrial cancer cell lines,Gynecologic Oncology, vol. 99,
no. 1, pp. 135141, 2005.
[48] T. Okada, T. Konno, T. Kohno et al., Possibility of targeting
claudin-2 in therapy for human endometrioid endometrial
carcinoma,Reproductive Sciences, vol. 27, no. 11, pp. 2092
2103, 2020.
[49] Y. N. Yang, Y. Wang, X. G. Wang, and S. J. Jiang, Eects of
trichostatin A and paclitaxel on apoptosis and mitochondrial
membrane potential of human endometrial carcinoma Ark2
cells,Ai Zheng, vol. 27, no. 8, pp. 816821, 2008.
[50] S. C. Dowdy, S. Jiang, X. C. Zhou et al., Histone deacetylase
inhibitors and paclitaxel cause synergistic eects on apoptosis
and microtubule stabilization in papillary serous endometrial
cancer cells,Molecular Cancer Therapeutics, vol. 5, no. 11,
pp. 27672776, 2006.
[51] P. A. Marks and R. Breslow, Dimethyl sulfoxide to vorinostat:
development of this histone deacetylase inhibitor as an anti-
cancer drug,Nature Biotechnology, vol. 25, no. 1, pp. 8490,
2007.
[52] N. Tandon, V. Ramakrishnan, and S. K. Kumar, Clinical use
and applications of histone deacetylase inhibitors in multiple
myeloma,Clinical Pharmacology, vol. 8, pp. 3544, 2016.
[53] L. Bergadà, A. Sorolla, A. Yeramian et al., Combination of
Vorinostat and caspase-8 inhibition exhibits high anti-
tumoral activity on endometrial cancer cells,Molecular
Oncology, vol. 7, no. 4, pp. 763775, 2013.
[54] S. R. Teixeira, C. M. Abreu, L. Parkes et al., Direct monitoring
of breast and endometrial cancer cell epigenetic response to
DNA methyltransferase and histone deacetylase inhibitors,
Biosensors & Bioelectronics, vol. 141, article 111386, 2019.
[55] N. Takai, N. Kawamata, C. S. Walsh et al., Discovery of epige-
netically masked tumor suppressor genes in endometrial can-
cer,Molecular Cancer Research, vol. 3, no. 5, pp. 261269,
2005.
[56] T. Z. Yi, J. Li, X. Han et al., DNMT inhibitors and HDAC
inhibitors regulate E-cadherin and Bcl-2 expression in endo-
metrial carcinoma in vitro and in vivo,Chemotherapy,
vol. 58, no. 1, pp. 1929, 2012.
[57] R. Sarfstein, I. Bruchim, A. Fishman, and H. Werner, The
mechanism of action of the histone deacetylase inhibitor vor-
inostat involves interaction with the insulin-like growth factor
8 Disease Markers
signaling pathway,PLoS One, vol. 6, no. 9, article e24468,
2011.
[58] M. H. Townsend, Z. E. Ence, A. M. Felsted et al., Potential
new biomarkers for endometrial cancer,Cancer Cell Interna-
tional, vol. 19, no. 1, p. 19, 2019.
[59] H. M. Prince, M. J. Bishton, and R. W. Johnstone, Panobino-
stat (LBH589): a potent pan-deacetylase inhibitor with prom-
ising activity against hematologic and solid tumors,Future
Oncology, vol. 5, no. 5, pp. 601612, 2009.
[60] S. Yang, X. Xiao, Y. Jia et al., Epigenetic modication restores
functional PR expression in endometrial cancer cells,Current
Pharmaceutical Design, vol. 20, no. 11, pp. 18741880, 2014.
[61] T. Kavlashvili, Y. Jia, D. Dai et al., Inverse relationship
between progesterone receptor and Myc in endometrial can-
cer,PLoS One, vol. 11, no. 2, article e0148912, 2016.
[62] X. Meng, P. Brachova, S. Yang et al., Knockdown of MTDH
sensitizes endometrial cancer cells to cell death induction by
death receptor ligand TRAIL and HDAC inhibitor LBH589
co-treatment,PLoS One, vol. 6, no. 6, article e20920, 2011.
[63] X. Meng, S. Yang, Y. Li et al., Combination of proteasome and
histone deacetylase inhibitors overcomes the impact of gain-
of-function p53 mutations,Disease Markers, vol. 2018, Arti-
cle ID 3810108, 7 pages, 2018.
[64] K. Steliou, M. S. Boosalis, S. P. Perrine, J. Sangerman, and D. V.
Faller, Butyrate histone deacetylase inhibitors,BioResearch
Open Access, vol. 1, no. 4, pp. 192198, 2012.
[65] Y. Terao, J. Nishida, S. Horiuchi et al., Sodium butyrate
induces growth arrest and senescence-like phenotypes in gyne-
cologic cancer cells,International Journal of Cancer, vol. 94,
no. 2, pp. 257267, 2001.
[66] M. Yu, H. Kong, Y. Zhao et al., Enhancement of adriamycin
cytotoxicity by sodium butyrate involves hTERT
downmodulation-mediated apoptosis in human uterine can-
cer cells,Molecular Carcinogenesis, vol. 53, no. 7, pp. 505
513, 2014.
[67] K. Kato, A. Kuhara, T. Yoneda et al., Sodium butyrate inhibits
the self-renewal capacity of endometrial tumor side-
population cells by inducing a DNA damage response,Molec-
ular Cancer Therapeutics, vol. 10, no. 8, pp. 14301439, 2011.
[68] K. Kato, Endometrial cancer stem cells: a new target for can-
cer therapy,Anticancer Research, vol. 32, no. 6, pp. 2283
2293, 2012.
[69] K. T. Thurn, S. Thomas, A. Moore, and P. N. Munster, Ratio-
nal therapeutic combinations with histone deacetylase inhibi-
tors for the treatment of cancer,Future Oncology, vol. 7,
no. 2, pp. 263283, 2011.
[70] L. Hodges-Gallagher, C. D. Valentine, S. E. Bader, and P. J.
Kushner, Inhibition of histone deacetylase enhances the
anti-proliferative action of antiestrogens on breast cancer cells
and blocks tamoxifen-induced proliferation of uterine cells,
Breast Cancer Research and Treatment, vol. 105, no. 3,
pp. 297309, 2007.
[71] Y. Li, T. Liu, C. Ivan et al., Enhanced cytotoxic eects of com-
bined valproic acid and the Aurora kinase inhibitor VE465 on
gynecologic cancer cells [published correction appears in
Front Oncol. 2018 Feb 09; 8: 9],Frontiers in Oncology,
vol. 3, p. 58, 2013.
[72] T. Yoshioka, S. Yogosawa, T. Yamada, J. Kitawaki, and
T. Sakai, Combination of a novel HDAC inhibitor OBP-
801/YM753 and a PI3K inhibitor LY294002 synergistically
induces apoptosis in human endometrial carcinoma cells due
to increase of Bim with accumulation of ROS,Gynecologic
Oncology, vol. 129, no. 2, pp. 425432, 2013.
[73] Y. B. Kim, K. H. Lee, K. Sugita, M. Yoshida, and S. Horinouchi,
Oxamatin is a novel antitumor compound that inhibits
mammalian histone deacetylase,Oncogene, vol. 18, no. 15,
pp. 24612470, 1999.
[74] G. H. Su, T. A. Sohn, B. Ryu, and S. E. Kern, A novel histone
deacetylase inhibitor identied by high-throughput transcrip-
tional screening of a compound library,Cancer Research,
vol. 60, no. 12, pp. 31373142, 2000.
[75] N. Takai, T. Ueda, M. Nishida, K. Nasu, and H. Narahara, A
novel histone deacetylase inhibitor, Scriptaid, induces growth
inhibition, cell cycle arrest and apoptosis in human endome-
trial cancer and ovarian cancer cells,International Journal
of Molecular Medicine, vol. 17, no. 2, pp. 323329, 2006.
[76] E. Pojani and D. Barlocco, Romidepsin (FK228), A histone
deacetylase Inhibitor and its analogues in cancer chemother-
apy,Current Medicinal Chemistry, vol. 28, no. 7, pp. 1290
1303, 2021.
[77] L. H. Li, P. R. Zhang, P. Y. Cai, and Z. C. Li, Histone deacet-
ylase inhibitor, Romidepsin (FK228) inhibits endometrial can-
cer cell growth through augmentation of p53-p21 pathway,
Biomedicine & Pharmacotherapy, vol. 82, pp. 161166, 2016.
[78] M. Y. Ahn, J. H. Jung, Y. J. Na, and H. S. Kim, A natural his-
tone deacetylase inhibitor, Psammaplin A, induces cell cycle
arrest and apoptosis in human endometrial cancer cells,
Gynecologic Oncology, vol. 108, no. 1, pp. 2733, 2008.
[79] U. de, J. Y. Son, R. Sachan et al., A new synthetic histone
deacetylase inhibitor, MHY2256, induces apoptosis and
autophagy cell death in endometrial cancer cells via p53 acet-
ylation,International Journal of Molecular Sciences, vol. 19,
no. 9, p. 2743, 2018.
[80] L. R. Duska, V. L. Filiaci, J. L. Walker et al., A surgical window
trial evaluating medroxyprogesterone acetate with or without
Entinostat in patients with endometrial cancer and validation
of biomarkers of cellular response,Clinical Cancer Research,
vol. 27, no. 10, pp. 27342741, 2021.
9Disease Markers
... On the contrary, global histone acetylation levels in H2AK5, H3K9, and H4K8, experience a profound decline during the late secretory phase [24]. Except for their expression in the normal cyclic endometrium, HDACs seem to play an important role in endometrial pathologies, as well [25][26][27][28]. The present review of the literature aims at closely investigating the potential impact of histone modifications and their therapeutic targeting in endometriosis. ...
... In the field of gynecology and obstetrics, HDACIs have long been proposed as potent alternative drug agents for the treatment of diverse gynecological cancer entities including uterine, cervical, ovarian, and breast cancer [26,[74][75][76]. As far as benign gynecological health conditions are concerned, Olaniyi et al. have, for instance, suggested that the short chain fatty acid acetate successfully restores the ovarian function in experimentally induced polycystic ovarian syndrome (PCOS) rat models [77]. ...
Article
Full-text available
Endometriosis is a chronic disorder of the female reproductive system which afflicts a great number of women worldwide. Histone deacetylases (HDACs) prevent the relaxation of chromatin, thereby positively or negatively modulating gene transcription. The current review aims at studying the impact of histone modifications and their therapeutic targeting in endometriosis. In order to identify relevant studies, a literature review was conducted using the MEDLINE and LIVIVO databases. The current manuscript represents the most comprehensive, up-to-date review of the literature focusing on the particular role of HDACs and their inhibitors in the context of endometriosis. HDAC1, HDAC2, HDAC3, Sirtuin 1, and Sirtuin 3, are the five most studied HDAC enzymes which seem to, at least partly, influence the pathophysiology of endometriosis. Both well-established and novel HDACIs could possibly represent modern, efficacious anti-endometriotic drug agents. Altogether, histone modifications and their therapeutic targeting have been proven to have a strong impact on endometriosis.
... Interestingly, ovarian and uterine cancers are usually due to inefficient DNA repair [49]. The standard of care for these cancer types is mainly based on neoadjuvant chemotherapy and several assays on cell lines identified drugs targeting apoptosis or cell cycle regulators as being effective [50][51][52][53][54][55][56][57][58][59][60][61][62]. ...
Article
Full-text available
Background Intra-tumour heterogeneity (ITH) presents a significant obstacle in formulating effective treatment strategies in clinical practice. Single-cell RNA sequencing (scRNA-seq) has evolved as a powerful instrument for probing ITH at the transcriptional level, offering an unparalleled opportunity for therapeutic intervention. Results Drug response prediction at the single-cell level is an emerging field of research that aims to improve the efficacy and precision of cancer treatments. Here, we introduce DREEP (Drug Response Estimation from single-cell Expression Profiles), a computational method that leverages publicly available pharmacogenomic screens from GDSC2, CTRP2, and PRISM and functional enrichment analysis to predict single-cell drug sensitivity from transcriptomic data. We validated DREEP extensively in vitro using several independent single-cell datasets with over 200 cancer cell lines and showed its accuracy and robustness. Additionally, we also applied DREEP to molecularly barcoded breast cancer cells and identified drugs that can selectively target specific cell populations. Conclusions DREEP provides an in silico framework to prioritize drugs from single-cell transcriptional profiles of tumours and thus helps in designing personalized treatment strategies and accelerating drug repurposing studies. DREEP is available at https://github.com/gambalab/DREEP .
... To date, the Food and Drug Administration has approved some HDACis such as vorinostat (SAHA), belinostat (PXD-101), panobinostat , and romidepsin (FK-228) for the treatment of cancer (Squarzoni et al., 2022). Clinical and preclinical studies have also shown that these compounds can be used as adjuvants to traditional chemotherapeutics in different types of cancer (Suraweera et al., 2018;Psilopatis et al., 2021;Pramanik et al., 2022). More recently, it has been shown that epigenetic targeting of colon cancer based on combined HDACis with DNA methyltransferase (DNMT) inhibitors has revealed clinical relevance (Tang et al., 2023). ...
Article
Full-text available
Introduction: Long non-coding RNA H19 (lncH19) is highly expressed in colorectal cancer (CRC) and plays critical roles in tumor development, proliferation, metastasis, and drug resistance. Indeed, the expression of lncH19 usually affects the outcomes of chemo-, endocrine, and targeted therapies. ITF2357 (givinostat) is a histone deacetylase inhibitor (HDACi) that revealed a significant anti-tumor action by inducing apoptosis in different tumor models, including leukemia, melanoma, and glioblastoma. However, no data are present in the literature regarding the use of this compound for CRC treatment. Here, we investigate the role of lncH19 in ITF2357-induced apoptosis in CRC cells. Methods: The HCT-116 CRC cell line was stably silenced for H19 to investigate the role of this lncRNA in ITF2357-induced cell death. Cell viability assays and flow cytometric analyses were performed to assess the anti-proliferative and pro-apoptotic effects of ITF2357 in CRC cell lines that are silenced or not for lncH19. RT-PCR and Western blot were used to study the effects of ITF2357 on autophagy and apoptosis markers. Finally, bioinformatics analyses were used to identify miRNAs targeting pro-apoptotic factors that can be sponged by lncH19. Results: ITF2357 increased the expression levels of H19 and reduced HCT-116 cell viability, inducing apoptosis, as demonstrated by the increase in annexin-V positivity, caspase 3 cleavage, and poly (ADP-ribose) polymerase (PARP-1) degradation. Interestingly, the apoptotic effect of ITF2357 was much less evident in lncH19-silenced cells. We showed that lncH19 plays a functional role in the pro-apoptotic activity of the drug by stabilizing TP53 and its transcriptional targets, NOXA and PUMA. ITF2357 also induced autophagy in CRC cells, which was interpreted as a pro-survival response not correlated with lncH19 expression. Furthermore, ITF2357 induced apoptosis in 5-fluorouracil-resistant HCT-116 cells that express high levels of lncH19. Conclusion: This study shows that lncH19 expression contributes to ITF2357-induced apoptosis by stabilizing TP53. Overall, we suggest that lncH19 expression may be exploited to favor HDACi-induced cell death and overcome 5-fluorouracil chemoresistance.
... In spite of the reported high response rates to chemotherapy, the duration of response only ranges between four and eight months [8,9], whereas the 5-year overall survival amounts to 84%, according to the American Cancer Society [10]. Nevertheless, the prognosis for women with advanced disease remains grim, with 5-year survival rates dropping to 20%, in cases of distant metastasis [10]. ...
Article
Full-text available
Endometrial carcinoma is the most common malignant tumor of the female genital tract in the United States. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptor proteins which regulate gene expression. In order to investigate the role of PPARs in endometrial cancer, we conducted a literature review using the MEDLINE and LIVIVO databases and were able to identify 27 relevant studies published between 2000 and 2023. The PPARα and PPARβ/δ isoforms seemed to be upregulated, whereas PPARγ levels were reported to be significantly lower in endometrial cancer cells. Interestingly, PPAR agonists were found to represent potent anti-cancer therapeutic alternatives. In conclusion, PPARs seem to play a significant role in endometrial cancer.
... Most study groups have employed the human breast cancer cell lines MCF-7 and MDA-MB-231 and discovered that the bitter melon extract might efficiently inhibit the in vitro progression of both estrogen receptor-positive and estrogen receptor-negative breast cancer cells. Of note, its bioactive components seem to interact with histone deacetylases and PPARs, which have been proposed to play a significant role in breast and gynecologic cancer development and progression [50][51][52][53][54][55]. In addition, bitter gourd extract enhanced the cytotoxic effects of standard chemotherapy on breast cancer cells, hence endorsing the assumption of potent synergistic antitumor effects. ...
Article
Full-text available
Phytotherapy has long represented a widely accepted treatment alternative to conventional therapy. Bitter melon is a vine with potent antitumor effects against numerous cancer entities. To date, no review article has, however, been published on the role of bitter melon in breast and gynecological cancer prevention and therapy. The current work constitutes the most comprehensive, up-to-date review of the literature, which highlights the promising anticancer effects of bitter melon on breast, ovarian, and cervical cancer cells and discusses future research recommendations.
... Of note, HDACs catalyze the removal of acetyl groups on the NH2-terminal lysine residues, thereby repressing transcription and tumor-suppressor gene activation [21][22][23][24]. As a consequence, the deregulation of histone acetylation potentially justifies the genesis of diverse human cancer entities [25][26][27]. ...
Article
Full-text available
Simple Summary Histone deacetylase inhibitors (HDACIs) are a relatively new drug class with important effects on the epigenetic regulation in cancer, inducing cancer cell death, apoptosis induction, and cell cycle arrest. Even though HDACIs have, to date, received approval for mainly hematologic malignancies, there are plentiful studies in cervical cancer setting with encouraging results. The present review summarizes all studies with HDACIs in cervical cancer from the emerging data in labor research to the possible applicability in clinical practice. Abstract Cervical carcinoma is one of the most common cancers among women globally. Histone deacetylase inhibitors (HDACIs) constitute anticancer drugs that, by increasing the histone acetylation level in various cell types, induce differentiation, cell cycle arrest, and apoptosis. The aim of the current review is to study the role of HDACIs in the treatment of cervical cancer. A literature review was conducted using the MEDLINE and LIVIVO databases with a view to identifying relevant studies. By employing the search terms “histone deacetylase” and “cervical cancer”, we managed to identify 95 studies published between 2001 and 2023. The present work embodies the most up-to-date, comprehensive review of the literature centering on the particular role of HDACIs as treatment agents for cervical cancer. Both well-established and novel HDACIs seem to represent modern, efficacious anticancer drugs, which, alone or in combination with other treatments, may successfully inhibit cervical cancer cell growth, induce cell cycle arrest, and provoke apoptosis. In summary, histone deacetylases seem to represent promising future treatment targets in cervical cancer.
... Additionally, sodium butyrate upregulated p21 and p27 protein expression [68][69][70]. Furthermore, TSA causes G2/M-phase arrest and G0/G1 arrest in hepatoma cells [71,72]. The fact that apoptosis plays an important role in tumor development makes it an obvious target for cancer therapy. ...
Article
Full-text available
Human diseases such as cancer can be caused by aberrant epigenetic regulation. Polyphenols play a major role in mammalian epigenome regulation through mechanisms and proteins that remodel chromatin. In fruits, seeds, and vegetables, as well as food supplements, polyphenols are found. Compounds such as these ones are powerful anticancer agents and antioxidants. Gallic acid, kaempferol, curcumin, quercetin, and resveratrol, among others, have potent anti-tumor effects by helping reverse epigenetic changes associated with oncogene activation and tumor suppressor gene inactivation. The role dietary polyphenols plays in restoring epigenetic alterations in cancer cells with a particular focus on DNA methylation and histone modifications was summarized. We also discussed how these natural compounds modulate gene expression at the epigenetic level and described their molecular targets in cancer. It highlights the potential of polyphenols as an alternative therapeutic approach in cancer since they modulate epigenetic activity.
Article
Full-text available
The most common malignant gynecologic diseases are cervical, uterine, ovarian, vaginal, and vulvar cancer. Among them, ovarian cancer causes more deaths than any other cancer of the female reproductive system. A great number of women suffer from endometriosis, uterine fibroids (UFs), adenomyosis, dysmenorrhea, and polycystic ovary syndrome (PCOS), which are widespread benign health problems causing troublesome and painful symptoms and significantly impairing the quality of life of affected women, and they are some of the main causes of infertility. In addition to the available surgical and pharmacological options, the effects of supporting standard treatment with naturally occurring compounds, mainly polyphenols, are being studied. Catechins are responsible for the majority of potential health benefits attributed to green tea consumption. Epigallocatechin gallate (EGCG) is considered a non-toxic, natural compound with potential anticancer properties. Antioxidant action is its most common function, but attention is also drawn to its participation in cell division inhibition, apoptosis stimulation and epigenetic regulation. In this narrative review, we describe the role of EGCG consumption in preventing the development of benign reproductive disorders such as UF, endometriosis, and PCOS, as well as malignant gynecologic conditions. We discuss possible epigenetic mechanisms that may be related to the action of EGCG.
Article
Full-text available
Endometrial cancer and uterine sarcoma represent the two major types of uterine cancer. In advanced stages, both cancer entities are challenging to treat and correlate with a meagre survival and prognosis. Hyperthermic Intraperitoneal Chemotherapy (HIPEC) is a form of localized chemotherapy that is heated to improve the chemotherapeutic effect on peritoneal metastases. The aim of the current review is to study the role of HIPEC in the treatment of uterine cancer. A literature review was conducted using the MEDLINE and LIVIVO databases with a view to identifying relevant studies. By employing the search terms “hyperthermic intraperitoneal chemotherapy”, “uterine cancer”, “endometrial cancer”, and/or “uterine sarcoma”, we managed to identify 26 studies published between 2004 and 2023. The present work embodies the most up-to-date, comprehensive review of the literature centering on the particular role of HIPEC as treatment modality for peritoneally metastasized uterine cancer. Patients treated with cytoreductive surgery, alongside HIPEC, seem to profit from not only higher survival but also lower recurrence rates. Factors such as the completeness of cytoreductive surgery, the peritoneal cancer index, the histologic subtype, or the applied chemotherapeutic agent, all influence HIPEC therapy effectiveness. In summary, HIPEC seems to represent a promising treatment alternative for aggressive uterine cancer.
Preprint
Full-text available
Drug response prediction at the single cell level is an emerging field of research that aims to improve the efficacy and precision of cancer treatments. Here, we introduce DREEP (Drug Response Estimation from single-cell Expression Profiles), a computational method that leverages publicly available pharmacogenomic screens and functional enrichment analysis to predict single cell drug sensitivity from transcriptomic data. We extensively tested DREEP on several independent single-cell datasets with over 200 cancer cell lines and showed its accuracy and robustness. Additionally, we also applied DREEP to molecularly barcoded breast cancer cells and identified drugs that can selectively target specific cell populations. DREEP provides an in-silico framework to prioritize drugs from single-cell transcriptional profiles of tumours and thus helps in designing personalized treatment strategies and accelerate drug repurposing studies. DREEP is available at https://github.com/gambalab/DREEP.
Article
Full-text available
A European consensus conference on endometrial carcinoma was held in 2014 to produce multidisciplinary evidence-based guidelines on selected questions. Given the large body of literature on the management of endometrial carcinoma published since 2014, the European Society of Gynaecological Oncology (ESGO), the European SocieTy for Radiotherapy & Oncology (ESTRO) and the European Society of Pathology (ESP) jointly decided to update these evidence-based guidelines and to cover new topics in order to improve the quality of care for women with endometrial carcinoma across Europe and worldwide. ESGO/ESTRO/ESP nominated an international multidisciplinary development group consisting of practicing clinicians and researchers who have demonstrated leadership and expertise in the care and research of endometrial carcinoma (27 experts across Europe). To ensure that the guidelines are evidence-based, the literature published since 2014, identified from a systematic search was reviewed and critically appraised. In the absence of any clear scientific evidence, judgment was based on the professional experience and consensus of the development group. The guidelines are thus based on the best available evidence and expert agreement. Prior to publication, the guidelines were reviewed by 191 independent international practitioners in cancer care delivery and patient representatives. The guidelines comprehensively cover endometrial carcinoma staging, definition of prognostic risk groups integrating molecular markers, pre- and intra-operative work-up, fertility preservation, management for early, advanced, metastatic, and recurrent disease and palliative treatment. Principles of radiotherapy and pathological evaluation are also defined.
Article
Full-text available
Simple Summary Beyond mutations, epigenetic changes have been described as drivers for cancer as well. While leaving the overall DNA structure intact, they can be responsible for tumor malignancy by mediating the transcriptional upregulation of oncogenes. This provides the basis for “epigenetic therapies” in cancer. Histone deacetylases (HDACs) are major players in epigenetic reprogramming. HDAC inhibitors (HDACis), either with broad-spectrum activity on various HDAC isoforms or with specific subtype specificity, have shown promising anticancer efficacies. The tremendous number of genes potentially affected creates the possibility for the parallel targeting of multiple disease-relevant pathways. Here, we give a comprehensive overview of various preclinical and clinical studies on HDACis. A particular focus is placed on the detailed description of promising strategies based on the combination of HDACis with other drugs. This also includes the development of new bifunctional inhibitors as well as novel approaches for HDAC degradation, rather than inhibition, via PROteolysis-TArgeting Chimeras (PROTACs). Abstract The increasing knowledge of molecular drivers of tumorigenesis has fueled targeted cancer therapies based on specific inhibitors. Beyond “classic” oncogene inhibitors, epigenetic therapy is an emerging field. Epigenetic alterations can occur at any time during cancer progression, altering the structure of the chromatin, the accessibility for transcription factors and thus the transcription of genes. They rely on post-translational histone modifications, particularly the acetylation of histone lysine residues, and are determined by the inverse action of histone acetyltransferases (HATs) and histone deacetylases (HDACs). Importantly, HDACs are often aberrantly overexpressed, predominantly leading to the transcriptional repression of tumor suppressor genes. Thus, histone deacetylase inhibitors (HDACis) are powerful drugs, with some already approved for certain hematological cancers. Albeit HDACis show activity in solid tumors as well, further refinement and the development of novel drugs are needed. This review describes the capability of HDACis to influence various pathways and, based on this knowledge, gives a comprehensive overview of various preclinical and clinical studies on solid tumors. A particular focus is placed on strategies for achieving higher efficacy by combination therapies, including phosphoinositide 3-kinase (PI3K)-EGFR inhibitors and hormone- or immunotherapy. This also includes new bifunctional inhibitors as well as novel approaches for HDAC degradation via PROteolysis-TArgeting Chimeras (PROTACs).
Article
Full-text available
A European consensus conference on endometrial carcinoma was held in 2014 to produce multi-disciplinary evidence-based guidelines on selected questions. Given the large body of literature on the management of endometrial carcinoma published since 2014, the European Society of Gynaecological Oncology (ESGO), the European SocieTy for Radiotherapy and Oncology (ESTRO), and the European Society of Pathology (ESP) jointly decided to update these evidence-based guidelines and to cover new topics in order to improve the quality of care for women with endometrial carcinoma across Europe and worldwide.
Article
Full-text available
Endometrial cancer (EC) is the sixth most common cancer in women. Since early EC has a good prognosis, identifying methods for early diagnosis is valuable. Here, we aimed to study the role of HDAC6, which has been indicated important in many kinds of cancers, in EC diagnosis and therapy. First, the expression levels of HDAC6 in EC tissues and cells were measured by qRT-PCR and Western blotting, and through bioinformatics and dual luciferase assays, HDAC6 was found to be a direct target of miR-206. Then, CCK-8, colony formation, wound healing, and Transwell assays were performed; these results indicated that HDAC6 promoted EC cell proliferation, metastasis and invasion, while miR-206 produced the opposite effects. In addition, rescue assays verified that the effect of miR-206 could be reversed by HDAC6, and global gene expression analysis confirmed the relationship between miR-206 and HDAC6. Finally, we measured the levels of PTEN, p-AKT and p-mTOR and other key molecules and speculated that miR-206 might target HDAC6 to suppress EC progression via the PTEN/AKT/mTOR pathway. In conclusion, downregulation of miR-206 and upregulation of HDAC6 in EC may predict poor prognosis, and as the target gene of miR-206, HDAC achieves its carcinogenic effect through the PTEN/AKT/mTOR pathway.
Article
Full-text available
Background Incidence of endometrial cancer are rising both in the United States and worldwide. As endometrial cancer becomes more prominent, the need to develop and characterize biomarkers for early stage diagnosis and the treatment of endometrial cancer has become an important priority. Several biomarkers currently used to diagnose endometrial cancer are directly related to obesity. Although epigenetic and mutational biomarkers have been identified and have resulted in treatment options for patients with specific aberrations, many tumors do not harbor those specific aberrations. A promising alternative is to determine biomarkers based on differential gene expression, which can be used to estimate prognosis. Methods We evaluated 589 patients to determine differential expression between normal and malignant patient samples. We then supplemented these evaluations with immunohistochemistry staining of endometrial tumors and normal tissues. Additionally, we used the Library of Integrated Network-based Cellular Signatures to evaluate the effects of 1826 chemotherapy drugs on 26 cell lines to determine the effects of each drug on HPRT1 and AURKA expression. Results Expression of HPRT1, Jag2, AURKA, and PGK1 were elevated when compared to normal samples, and HPRT1 and PGK1 showed a stepwise elevation in expression that was significantly related to cancer grade. To determine the prognostic potential of these genes, we evaluated patient outcome and found that levels of both HPRT1 and AURKA were significantly correlated with overall patient survival. When evaluating drugs that had the most significant effect on lowering the expression of HPRT1 and AURKA, we found that Topo I and MEK inhibitors were most effective at reducing HPRT1 expression. Meanwhile, drugs that were effective at reducing AURKA expression were more diverse (MEK, Topo I, MELK, HDAC, etc.). The effects of these drugs on the expression of HPRT1 and AURKA provides insight into their role within cellular maintenance. Conclusions Collectively, these data show that JAG2, AURKA, PGK1, and HRPT1 have the potential to be used independently as diagnostic, prognostic, or treatment biomarkers in endometrial cancer. Expression levels of these genes may provide physicians with insight into tumor aggressiveness and chemotherapy drugs that are well suited to individual patients.
Article
Full-text available
Mutations in the “guardian of the genome” TP53 predominate in solid tumors. In addition to loss of tumor suppressor activity, a specific subset of missense mutations confers additional oncogenic properties. These “gain-of-function” (GOF) mutations portend poor prognosis across cancer types regardless of treatment. Our objective in this study was to identify novel therapeutic opportunities to overcome the deleterious effects of GOF TP53 mutants. Using gynecologic cancer cell lines with known TP53 mutational status, we established that treatment with a proteasome inhibitor induced cell death in cells with two recurrent GOF TP53 mutations (R175H and R248Q), and addition of a histone deacetylase inhibitor (HDACi) enhanced this effect. By contrast, p53-null cancer cells were relatively resistant to the combination. Proteasome inhibition promoted apoptosis of cells with TP53 GOF mutations, potentially through induction of the unfolded protein response. In line with the reported hyperstabilization of GOF p53 protein, cells treated with HDACi exhibited reduced levels of p53 protein. Together, these data form the basis for future clinical studies examining therapeutic efficacy in a preselected patient population with GOF TP53 mutations.
Article
Purpose: This surgical window of opportunity (window) study assessed the short-term effect of medroxyprogesterone acetate (MPA) alone versus MPA plus the histone deacetylase (HDAC) inhibitor entinostat on regulation of progesterone receptor (PR) in women with newly diagnosed endometrioid endometrial adenocarcinoma. Patients and methods: This multisite, randomized, open-label surgical window study treated women intramuscularly on day 1 with 400 mg MPA. Entinostat given 5 mg by mouth on days 1, 8, and 15 was randomly assigned with equal probability. Surgery followed on days 21-24. Pretreatment and posttreatment tissue was assessed for PR H-scores, Ki-67 levels, and histologic response. Results: Fifty patients were accrued in 4 months; 22 and 20 participants had PR evaluable pretreatment and posttreatment slides in the MPA and MPA/entinostat arms, respectively. Median posttreatment PR H-scores were significantly lower than pretreatment H-scores in both arms but did not differ significantly (MPA: 247 vs. 27, MPA/entinostat 260 vs. 23, respectively, P = 0.87). Decreased Ki-67 was shown in 90% treated with MPA/entinostat compared with 68% treated with MPA alone (P = 0.13). Median PR H-score decreases were larger when Ki-67 was decreased (208) versus not decreased (45). The decrease in PR pretreatment versus posttreatment was associated with loss of Ki-67 nuclear staining, consistent with reduced cellular proliferation (P < 0.008). Conclusions: This surgical window trial rapidly accrued in a multisite setting and evaluated PR as its primary endpoint and Ki-67 as secondary endpoint. Despite no immediate effect of entinostat on PR in this short-term study, lessons learned can inform future window and treatment trials.
Article
Claudin-2 (CLDN-2) is a leaky-type tight junction protein, and its overexpression increases tumorigenesis of some types of cancer cells. In the present study, to examine the possibility of targeting CLDN-2 in the therapy for endometrioid endometrial adenocarcinoma, we investigated the regulation and role of CLDN-2 in endometriosis and endometrioid endometrial adenocarcinoma. In endometrioid endometrial adenocarcinoma tissues, marked upregulation of CLDN-2 was observed together with malignancy, while in endometriosis tissues, a change in the localization of CLDN-2 was observed. In cells of the endometrial adenocarcinoma cell line Sawano, which highly express CLDN-2, downregulation of CLDN-2 induced by the siRNA upregulated the epithelial barrier and inhibited cell migration. Furthermore, the downregulation of CLDN-2 affected the cell cycle and inhibited cell proliferation. In Sawano cells cultured with high-glucose medium, CLDN-2 expression was downregulated at the mRNA and protein levels. The high-glucose medium upregulated the epithelial barrier, cell proliferation, and migration, and inhibited cell invasion. The histone deacetylase (HDAC) inhibitor tricostatin A (TSA), which has antitumor effects, downregulated CLDN-2 expression, cell proliferation, invasion, and migration, and upregulated the epithelial barrier. The mitochondrial respiration level, an indicator of cancer metabolism, was downregulated by CLDN-2 knockdown and upregulated by the high-glucose condition. Taken together, these results indicated that overexpression of CLDN-2 closely contributed to the malignancy of endometrioid endometrial adenocarcinoma. Downregulation of CLDN-2 via the changes of the glucose concentration and treatment with HDAC inhibitors may be important in the therapy for endometrial cancer.
Article
Background: Human HDACs represent a group of enzymes able to modify histone and non-histone proteins, which interact with DNA to generate chromatin. The correlation between irregular covalent modification of histones and tumor development has been proven over the last decades. Therefore, HDAC inhibitors are considered as potential drugs in cancer treatment. Romidepsin (FK228), Belinostat (PXD-101), Vorinostat (SAHA), Panobinostat (LBH-589) and Chidamide were approved by FDA as novel antitumor agents. Objective: The aim of this review article is to highlight the structure-activity relationships of several FK228 analogues as HDAC inhibitors. In addition, the synergistic effects of a dual HDAC/PI3K inhibition by some derivatives have been investigated. Materials and methods: PubMed, MEDLINE, CAPLUS, SciFinder Scholar database were considered by selecting articles which fulfilled the objectives of this review, dating from 2015 till present time. Results: HDAC inhibitors have a significant role in cancer pathogenesis and evolution. Class I HDAC isoformrs are expressed in many tumor types, therefore, potent and selective Class I HDAC inhibitors are of great interest as candidate therapeutic agents with limited side effects. By structure-based optimization, several FK228 analogues [15 (FK-A5), 22, 23, 26 (FK-A11)] were identified, provided with significant activity against Class I HDAC enzymes and dose dependent antitumor activity. Compound 26 was recognized as an interesting HDAC/PI3K dual inhibitor (IC50 against p110α of 6.7 µM while for HDAC1 inhibitory activity IC50 was 0.64 nM). Conclusion: Romidepsin analogues HDAC inhibitors have been confirmed as useful anticancer agents. In addition, dual HDAC/PI3K inhibition showed by some of them exhibited synergistic effects in inducing apoptosis in human cancer cells. Further studies on FK228 analogues may positively contribute to the availability of potent agents in tumor treatment.
Article
DNA methylation and histone deacetylation are key epigenetic processes involved in normal cellular function and tumorigenesis. Therapeutic strategies based on DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors are currently in use and under development for the treatment of cancers. Genome-wide DNA methylation profiling has been proposed for use in disease diagnosis, and histone modification profiling for disease stratification will follow suit. However, whether epigenome sequencing technologies will be feasible for rapid clinic diagnosis and patient treatment monitoring remains to be seen, and alternative detection technologies will almost certainly be needed. Here we used electrochemical impedance spectroscopy (EIS) employing a graphene-based screen-printed electrode system to directly measure global DNA methylation and histone H3 acetylation to compare non-cancer and breast cancer cell lines. We demonstrated that whilst global methylation was not useful as a differential marker in the cellular systems tested, histone H3 acetylation was effective at higher chromatin levels. Using breast and endometrial cancer cell models, EIS was then used to monitor cellular responses to the DNMT and HDAC inhibitors 5-Aza-2′-deoxycytidine and suberoylanilide hydroxamic acid in vitro, and proved very effective at detecting global cellular responses to either treatment, indicating that this approach could be useful in following treatment response to epigenetic drugs. Moreover, this work reports the first combined analysis of two epigenetic markers using a unified graphene-based biosensor platform, demonstrating the potential for multiplex analysis of both methylation and acetylation on the same sample.