ArticlePDF AvailableLiterature Review

A state-of-the-art review on LSD1 and its inhibitors in breast cancer: Molecular mechanisms and therapeutic significance

Frontiers
Frontiers in Pharmacology
Authors:

Abstract

Breast cancer (BC) is a kind of malignant cancer in women, and it has become the most diagnosed cancer worldwide since 2020. Histone methylation is a common biological epigenetic modification mediating varieties of physiological and pathological processes. Lysine-specific demethylase 1 (LSD1), a first identified histone demethylase, mediates the removal of methyl groups from histones H3K4me1/2 and H3K9me1/2 and plays a crucial role in varieties of cancer progression. It is also specifically amplified in breast cancer and contributes to BC tumorigenesis and drug resistance via both demethylase and non-demethylase manners. This review will provide insight into the overview structure of LSD1, summarize its action mechanisms in BC, describe the therapeutic potential of LSD1 inhibitors in BC, and prospect the current opportunities and challenges of targeting LSD1 for BC therapy.
A state-of-the-art review on
LSD1 and its inhibitors in breast
cancer: Molecular mechanisms
and therapeutic signicance
Guan-Jun Yang
1
,
2
,
3
, Yan-Jun Liu
2
, Li-Jian Ding
4
, Fan Tao
2
,
Ming-Hui Zhu
2
, Zhen-Yuan Shi
2
, Juan-Ming Wen
2
,
Meng-Yao Niu
2
, Xiang Li
2
, Zhan-Song Xu
2
, Wan-Jia Qin
2
,
Chen-Jie Fei
1
,
2
,
3
and Jiong Chen
1
,
2
,
3
*
1
State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-
products, Ningbo University, Ningbo, Zhejiang, China,
2
Laboratory of Biochemistry and Molecular
Biology, School of Marine Sciences, Ningbo University, Ningbo, China,
3
Key Laboratory of Aquacultural
Biotechnology Ministry of Education, Ningbo University, Ningbo, China,
4
Li Dak Sum Yip Yio Chin
Kenneth Li Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of
Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
Breast cancer (BC) is a kind of malignant cancer in women, and it has become
the most diagnosed cancer worldwide since 2020. Histone methylation is a
common biological epigenetic modication mediating varieties of physiological
and pathological processes. Lysine-specic demethylase 1 (LSD1), a rst
identied histone demethylase, mediates the removal of methyl groups from
histones H3K4me1/2 and H3K9me1/2 and plays a crucial role in varieties of
cancer progression. It is also specically amplied in breast cancer and
contributes to BC tumorigenesis and drug resistance via both demethylase
and non-demethylase manners. This review will provide insight into the
overview structure of LSD1, summarize its action mechanisms in BC,
describe the therapeutic potential of LSD1 inhibitors in BC, and prospect the
current opportunities and challenges of targeting LSD1 for BC therapy.
KEYWORDS
LSD1, histone demethylase, breast cancer, inhibitors, H3K4me1/2, H3K9me1/2
1 Introduction
Breast cancer (BC) is a kind of malignant tumour in women occurring in the breast
glandular epithelial tissues, and it has become the most diagnosed cancer worldwide since
2020 (Siegel et al., 2021;Siegel et al., 2022). The incidence of BC is increasing year by year
and patients are getting younger and younger, posing a serious threat to womens health
(Siegel et al., 2022). Although advances in early diagnosis and treatment of BC have
partially alleviated the crisis of some BC patients, there are still a large number of patients
suffering from BC due to their complex pathogenesis, insensitivity to existing drugs, and
easy-to-develop drug resistance (Yang et al., 2019;Yang et al., 2021b). Therefore, there is
an urgent need to nd new and effective targeted therapies for this type of BC.
OPEN ACCESS
EDITED BY
Chao Han,
China Pharmaceutical University, China
REVIEWED BY
Haibing Zhou,
Wuhan University, China
Yingtang Zhou,
Zhejiang Ocean University, China
*CORRESPONDENCE
Jiong Chen,
jchen1975@163.com,
chenjiong@nbu.edu.cn
SPECIALTY SECTION
This article was submitted to
Pharmacology of Anti-Cancer Drugs,
a section of the journal
Frontiers in Pharmacology
RECEIVED 08 July 2022
ACCEPTED 15 August 2022
PUBLISHED 16 September 2022
CITATION
Yang G-J, Liu Y-J, Ding L-J, Tao F,
Zhu M-H, Shi Z-Y, Wen J-M, Niu M-Y,
Li X, Xu Z-S, Qin W-J, Fei C-J and
Chen J (2022), A state-of-the-art review
on LSD1 and its inhibitors in breast
cancer: Molecular mechanisms and
therapeutic signicance.
Front. Pharmacol. 13:989575.
doi: 10.3389/fphar.2022.989575
COPYRIGHT
© 2022 Yang, Liu, Ding, Tao, Zhu, Shi,
Wen, Niu, Li, Xu, Qin, Fei and Chen. This
is an open-access article distributed
under the terms of the Creative
Commons Attribution License (CC BY).
The use, distribution or reproduction in
other forums is permitted, provided the
original author(s) and the copyright
owner(s) are credited and that the
original publication in this journal is
cited, in accordance with accepted
academic practice. No use, distribution
or reproduction is permitted which does
not comply with these terms.
Frontiers in Pharmacology frontiersin.org01
TYPE Review
PUBLISHED 16 September 2022
DOI 10.3389/fphar.2022.989575
Lysine-specic demethylase 1 (LSD1) is a avin-dependent
lysine-specic histone demethylase rst identied in 2004 and
mediated transcriptional activation or repression via erasing
methyl groups from H3K9me2/1 and H3K4me2/1,
respectively (Figure 1)(Shi et al., 2004;Fang et al., 2020;Kim
et al., 2021;Malagraba et al., 2022;Song et al., 2022). Recently
studies found that LSD1 could also remove the methyl groups
from several non-histone proteins such as ERα, MTA1, HIF-1α,
AGO2, HSP90, MEFD2, and STAT3 and be involved in many
cancer cell events (Majello et al., 2019). LSD1 exhibits its catalytic
mechanism via consuming oxidation of FAD and O
2
and
yielding HCHO and H
2
O
2
in cellulo (Yang et al., 2018a).
Apart from demethylase activity, LSD1 exhibits non-
demethylase activity via interacting with its client proteins
and is involved in physiological and pathological processes
(Gu et al., 2020). LSD1 is also overexpressed in varieties of
cancers and mediates their progression (Yang et al., 2018a;
Fang et al., 2019;Fang et al., 2020). LSD1 is aberrantly
expressed in BC and promotes proliferation and metastasis of
BC cells (Liu et al., 2020a;Zhou et al., 2021a). Moreover,
LSD1 also is involved in regulating resistance of
chemotherapy and immunotherapy in BC (Kim et al., 2013;
Boulding et al., 2018;Yang et al., 2018d;Qin et al., 2019;Verigos
et al., 2019;Tu et al., 2020;Sobczak et al., 2022). Given the
multifaceted functions of LSD1 in BC progression, new
therapeutic strategies targeting LSD1 are constantly being
developed, such as the discovery of novel LSD1 inhibitors,
the development of dual-target inhibitors, and the
combination therapies with chemical agents or
immunomodulators (Kim et al., 2013;Boulding et al., 2018;
Yang et al., 2018d;Qin et al., 2019;Verigos et al., 2019;Tu
et al., 2020;Sobczak et al., 2022). Therefore, LSD1 is a potential
target for BC therapy.
Herein, the structures, functions and the regulatory roles of
LSD1 in tumorigenesis were introduced, the reported
LSD1 inhibitors and their therapeutic mechanisms for BC
treatment were summarized, and the current challenges and
the potential opportunities of LSD1 inhibitors for BC therapy
were prospected.
2 The overview of LSD1
2.1 The structure of LSD1
LSD1 is a FAD-dependent demethylase encoding a peptide
with 852 amino acid residues. LSD1 consists of highly conserved
three distinct domains: a SWI3/Rac8/Moira (SWIRM) domain, a
tower domain (TD), and a catalytic amine oxidase domain
(AOD) (Figures 1A,B)(Forneris et al., 2008;Yang et al.,
2018a). The SWIRM domain is an indispensable domain for
LSD1-mediated histone modication and chromatin remodeling
(Metzger et al., 2005). The TD is a special domain with two
antiparallel helices, and it can bind to RCOR1 and form the
CoREST complex (Pilotto et al., 2015;Marabelli et al., 2016). The
AOD domain is the catalytic domain of LSD1, and it consists of
two well-dened motifs: the substrate-recognition motif and the
FAD-binding motif (Yang et al., 2018a). The latter is highly
conserved and responsible for binding sites of some reported
LSD1 inhibitors. The two motifs assemble into a big cavity
containing the interface of enzyme activity centre (Forneris
et al., 2008). In the active state of LSD1, the second lobe of
the AOD domain could form a hydrophobic binding pocket
SWIRM domain (Yoneyama et al., 2007). This binding pocket
mediates LSD1 binding to histone H3 and is also chosen as the
binding pocket for developing LSD1 inhibitors (Zhou et al.,
2015).
2.2 The functions and regulations of LSD1
LSD1 plays heterogeneous roles via transcriptionally
modulating its downstream genes in demethylase-dependent or
-independent modesin varieties of cancers(Song et al., 2022;Yang
FIGURE 1
Structure and function of LSD1. (A) Distribution of domains of LSD1. (B) LSD1-mediated transcriptional modulation. (C) Overview structure of
LSD1.
Frontiers in Pharmacology frontiersin.org02
Yang et al. 10.3389/fphar.2022.989575
et al., 2022). It acts as an oncogene in some cancers, while
functioning as a cancer-suppressor gene in the other cancers
(Yang et al., 2018a;Hu et al., 2019). In addition, LSD1 is also
regulated by multiple epigenetic regulators in BC.
2.2.1 LSD1 as a transcription co-repressor
Upregulating of H3K4me2/1 often contributes to the
transcriptional activation (Figure 1C). LSD1 removes methyl
groups from active Histone 3 via assembling into different co-
repressor complexes with several distinct proteins and shaping
chromatin into a repressive conformation. For example,
LSD1 was found to form a co-repressor complex with SIN3A/
HDAC and maintain sensitivity to chemotherapy via reducing
inhibiting several genes such as TGFB2, CASP7, TERT,MDM2,
and HIF1αin BC (Yang et al., 2018d). It also could assemble into
LSD1/CoREST/BRMS1 and inhibit the metastasis of BC cells via
reduced levels of Vimentin,COL5A2,INSIG2,MRPL33,SLC1A1,
KLK11, and OLFML3 (Qiu et al., 2018).
2.2.2 LSD1 as a transcription co-activator
LSD1 also works as a transcriptional co-activator via
demethylating H3K9me2/1 (Figure 1C). It can promote
estrogen transcription in breast cancer cells through
interacting with estrogen receptor (ER) (Bennesch et al.,
2016). Additionally, LSD1 also regulates chromatin events
such as DNA replication, heterochromatin, and imprinting
propagation (Zhu et al., 2012;Yang et al., 2018a).
2.2.3 The epigenetic regulation of LSD1
The function of LSD1 has also been found to be regulated by
many epigenetic components. For example, miR-708 inhibits BC
proliferation and invasion via directly binding to the 3UTR of
LSD1 and reducing its level (Ma et al., 2016). In addition,
epigenetic modications such as phosphorylation (Peng et al.,
2015;Feng et al., 2016;Zhou et al., 2016), acetylation (Luo et al.,
2016), methylation (Liu et al., 2020a), and ubiquitination (Wu
et al., 2013;Yi et al., 2016;Zhou et al., 2016;Gong et al., 2021) also
contribute to the function of LSD1 (Figure 2). PKCαcan
phosphorylate LSD1 at S112, activating its demethylase
activity and enhancing the occupancy on E-cadherin
promoter, and nally promote EMT and metastasis in BC
(Feng et al., 2016). Phosphorylated modication is also found
to regulate LSD1-medatied DNA damage (Peng et al., 2015). To
be specic, LSD1 was di-phosphorylated at S131 and S137 by
CK2 and Wip1, respectively, which promoted its interaction with
RNF168 and RNF168-dependent 53BP1 ubiquitination and
subsequent recruitment to the DNA damage sites. CK1αwas
also found to phosphorylate LSD1 at S687 and then induce
S683 phosphorylation of LSD1 by nuclear GSK3β(Zhou et al.,
2016). The di-phosphorylated LSD1 would be deubiquitylated by
USP22 and induced stabilization itself. MOF, a lysine
acetyltransferase rst found acetylated histone H4 at
K16 residue, also could catalyze the acetylation of LSD1 at
K432, K433, and K436 and suppress LSD1-medaited EMT in
epithelial cells (Luo et al., 2016). Arginine methyltransferase 4
(PRMT4) was also found to mediate deubiquitination and
stabilization of LSD1 via dimethylating it at R838 and
promoting its deubiquitination by USP7 (Liu et al., 2020a).
Apart from USP7 and USP22, USP28 also could remove
ubiquitin modications from LSD1, maintain its stability, and
thus confer stemness to BC cells (Wu et al., 2013). Recently,
OTUD7B was also found to mediate deubiquitination of LSD1 at
K226/277 residues, which stabilized LSD1 and promoted its
assembly into co-repressor complex (Gong et al., 2021).
3 The role of LSD1 in breast cancer
3.1 Role of LSD1 in breast cancer
progression
LSD1 is overexpressed in several subtypes of BC and
functions as an oncogene mediating proliferation,
differentiation, invasion, and metastasis of BC cells (Figure 3;
Tables 13)(Ma et al., 2016;Feng et al., 2017;Yang et al., 2018a;
Hu et al., 2019;Zhou et al., 2021a;Ji et al., 2021). When normal
human mammary epithelial cells are exposed to carcinogens,
their LSD1 levels would be upregulated and promote the
carcinogenesis via reducing p57
kip2
level (Bradley et al., 2007).
LSD1 exhibits its oncogene functions via interacting with distinct
ligands in different BC subtypes. As a major biomarker of ER-
positive BC, ERαand its transcriptional activity are regulated by
LSD1 via assembling into complex with different ligands to
mediate BC proliferation (Lim et al., 2010;Pollock et al.,
2012;Zhu et al., 2012;Andresen et al., 2017). For example,
CAC1 antagonized LSD1-mediated ERαactivation and
suppressed the proliferation of BC cells (Kim et al., 2013),
while ASXL2 promoted proliferation of BC cells via forming a
complex ASXL2/LSD1/UTX/MLL to activate ERαactivity (Park
et al., 2016). In addition, LSD1 reduces tumor suppressor gene
Lefty1 via interacting with β-catenin in BC cells (Yakulov et al.,
2013), and suppresses BC cell growth through binding to histone
deacetylases (HDACs) (Huang et al., 2012;Vasilatos et al., 2013).
It also sensitizes BC cells to chemotherapy via assembling into a
complex with SIN3A/HDAC and inhibits BC proliferation and
metastasis via interacting with HDAC5 (Cao et al., 2017;Cao
et al., 2018;Yang et al., 2018d). Further studies found that
LSD1 promotes BC metastasis via H3K4me2 demethylase
occupying the gene promoters of Snail and Slug and reducing
their levels (Lin et al., 2010;Wu et al., 2012;Lin et al., 2014;
Phillips and Kuperwasser, 2014;Bai et al., 2017). Interestingly,
androgen receptor (AR) is also involved in BC metastasis via
interacting with LSD1 to reduce E-cadherin and upregulate
Vimentin (Feng et al., 2017).
Interestingly, LSD1 also exert its function as a tumor
suppressor gene via forming different complexes with distinct
Frontiers in Pharmacology frontiersin.org03
Yang et al. 10.3389/fphar.2022.989575
ligand proteins (Li et al., 2017). LSD1 was also found to inhibit
proliferation, invasion, and metastasis in vitro and in vivo via
assembling into LSD1/NuRD complex (Wang et al., 2009;Li
et al., 2017). This complex exhibits its heterogeneous tumor
suppressor functions dependent of the different subunits in
varieties of BC cells (Wang et al., 2009;Li et al., 2017). In
MCF-7 cells, zinc-nger protein 516 (ZNF516) inhibited
EGFR transcription, and thus reduced the proliferation and
invasion of BC in vitro and in vivo via targeting CtBP/LSD1/
CoREST complex (Li et al., 2017). Breast carcinoma metastasis
suppressor 1 (BRMS1) is another gene coordinating with LSD1/
NuRD complex to inhibit the metastasis of MCF7 cells (Qiu et al.,
2018). In MDA-MB-231 cells, LSD1/NuRD complex suppressed
BC tumorigenesis and metastasis via recruiting the homeotic
protein SIX3 (Zheng et al., 2018). CC chemokine ligand 14
(CCL14) is a chemokine promoting angiogenesis in viral
infection and tumor progression (Mortier et al., 2008;Li et al.,
2011). KDM5B reduce CCL14 transcription to impede metastasis
via targeting LSD1/NuRD complex (Li et al., 2011). In addition,
in luminal BC, LSD1 suppressed invasion, migration, and
metastasis BC cells via raising GATA3 and repressing
TRIM37 (Hu et al., 2019).
Apart from as a subunit of many complexes mediating BC
progression, the function of LSD1 was also regulated by several
epigenetic enzymes in BC (Feng et al., 2016;Liu et al., 2020a;
Gong et al., 2021). Feng et al. (2016) highlighted the PKCα-
mediated phosphorylation of the S112 residue of LSD1 which
was crucial for epithelial-mesenchymal transition (EMT) and
FIGURE 2
Residue sites were modied by varieties of posttranslational modications in LSD1.
FIGURE 3
Role of LSD1 in breast cancer progression, angiogenesis, tumor microenvironment, and drug resistance.
Frontiers in Pharmacology frontiersin.org04
Yang et al. 10.3389/fphar.2022.989575
metastasis of BC cells. Liu et al. (2020a) found that
PRMT4 methylated and stabilized LSD1 via promoting it and
it binding to deubiquitinase USP7 in BC cells. Recently, Gong
et al. (2021) revealed that OTUD7B could remove the Poly-Ub
Chains of LSD1 at K226/277 residues, maintain the integrity of
LSD1/CoREST/HDACs co-repressor complexes, and inhibit BC
metastasis.
3.2 Role of LSD1 in tumor angiogenesis
Angiogenesis is a pivotal process for BC growth and metastasis
(Ayoub et al., 2022). LSD1 is also involved in this process via
modulating several pathways (Table 2)(Li et al., 2011;Lee et al.,
2017). Li et al. (2011) found that LSD1 worked as a co-repressor and
suppressed angiogenesis and metastasis of BC cells via assembling
TABLE 1 Roles of LSD1 in BC progression.
Substrates Complexes/
pathways
Target genes Functions References
H3K4me2/me1 p57kip2 Promoting BC initiation Bradley et al. (2007)
H3K4me2/me1 CAC1/LSD1/ERαERα-target genes Inhibiting proliferation Kim et al. (2013)
H3K9me2/me1 ASXL2/LSD1/UTX/
MLL2
ERαPromoting proliferation Park et al. (2016)
H3K4me2/1 LSD1/β-catenin Lefty1 Promoting proliferation Yakulov et al. (2013)
H3K4me2/1 LSD1/SIN3A/HDAC CASP7, TGFB2, CDKN1A(p21), HIF1A,
TERT, and MDM
Sensitizing BC cells to
chemotherapy
Yang et al. (2018d)
H3K4me1/me2 LSD1/HDAC5 p21 and CLDN7 Hindering BC proliferation and
invasion
Cao et al. (2017),Cao et al. (2018)
H3K4me2 Snail E-cadherin, PTEN Promoting EMT Lin et al., 2010,Lin et al. (2014)
H3K4me2 Slug BRCA1, ESR1 Inhibiting invasion Bai et al., (2017),Phillips and
Kuperwasser, (2014)
H3K9me2, H3K4me2 AR E-cadherin and Vimentin Promoting progression and
metastasis
Feng et al. (2017)
H3K4me2 LSD1/CoREST IGF1R, RHOA, and TGFB1 Inhibiting metastasis Wang et al. (2009)
H3K4me2 ZNF516/CtBP/LSD1/
CoREST
EGFR Inhibiting proliferation and
metastasis
Li et al. (2017)
H3K4me1/me2 BRMS1/LSD1/CoREST Vimentin Suppressing metastasis and
invasion
Qiu et al. (2018)
H3K4me1/me2 LSD1/NuRD/SIX3 WNT1 and FOXC2 Zheng et al. (2018)
H3K4me2, H3K4me3 LSD1/NuRD/KDM5B CCL14 Li et al. (2011)
H3K4me2 LSD1/GATA3 GATA3,TRIM37 Hu et al. (2019)
H3K4me1/me2,
H3K9me2
OTUD7B/LSD1 Snail, CDK6, ... Promoting metastasis Gong et al. (2021)
H3K4me1/me2 PKCα/LSD1 E-cadherin Promoting EMT and metastasis Feng et al. (2016)
H3K4me1/me2 PRMT4/LSD1/USP7 E-cadherin and Vimentin Promoting invasion and
metastasis
Liu et al. (2020a)
TABLE 2 Regulatory roles of LSD1 in BC angiogenesis and microenvironment.
Substrates Ligand
proteins
Complexes/
pathways
Target genes Functions References
HIFαK391 NuRD LSD1/NuRD VEGF Promoting angiogenesis Lee et al. (2017)
H3K4me2,
H3K4me3
KDM5B and
NuRD
KDM5B/LSD1/
NuRD
CCL14 Suppressing angiogenesis Li et al. (2011)
H3K4me2/1 PKC-θLSD1S111/PKC-θJUNB, KLF10, KLF6,
and CCL2
Promoting mesenchymal and stem-like signature,
and reducing M1 macrophage
Boulding et al. (2018)
H3K4me2 —— CCL5, CXCL -9, -10,
and PD-L1
Reducing CD8
+
T cell inltration Bennani-Baiti, (2012),Shen
et al. (2022)
H3K4me2,
H3K9me2
CoREST LSD1/CoREST Macrophage
polarization genes
Inhibiting Mvtoward a M1-like phenotype in
the TME
Benedetti et al. (2019)
Frontiers in Pharmacology frontiersin.org05
Yang et al. 10.3389/fphar.2022.989575
into a complex with KDM5B and NuRD and reducing the
transcription of CCL4. Hypoxia-inducible factor alpha (HIF1α),
a transcription factor promoting breast cancer angiogenesis, is also
found to be regulated by LSD1/NuRD complex. To be specic, this
complex demethylated HIF1αto stabilize it, and then stabilized
HIF1αwould upregulate the vascular endothelial growth factor
(VEGF) via cooperating with CBP and metastasis-associated
antigen 1, and induce angiogenesis in BC (Lee et al., 2017).
3.3 Role of LSD1 in the breast cancer
microenvironment
Tumor microenvironment refers to the internal and external
environment of tumor cells where they survive, grow, and
metastasize (Xiao and Yu, 2021). Tumor stroma consists of
several heterogeneous cells such as cancer-associated
broblasts (CAFs) and macrophages (Mv), which promote
tumorigenesis via the secretion of varieties of chemokines,
cytokines, and growth factors (Disis, 2010). While
LSD1 increases CAF burden and reducing innate M1 Mv
inltration at the primary tumor site in BC (Boulding et al.,
2018). In TNBC, LSD1 also mediated CD8
+
lymphocyte
trafcking to the tumor microenvironment and reducing Mv
polarization toward M1-like phenotype (Qin et al., 2019;Tan
et al., 2019;Shen et al., 2022).
3.4 Role of LSD1 in drug resistance of
breast cancer
Drug resistance is one of the major causes that leads to
distant metastasis, poor prognosis, and death of BC (Wen et al.,
2020). LSD1 is widely involved in the resistance to chemotherapy
(Kim et al., 2013;Boulding et al., 2018;Verigos et al., 2019;
Sobczak et al., 2022), hormone therapy (Bennani-Baiti, 2012;
Cortez et al., 2012;Benedetti et al., 2019;Sukocheva et al., 2020),
immunotherapy (Qin et al., 2019;Tu et al., 2020), and targeted
therapy (Strachowska et al., 2021;Liu et al., 2022)ofBC(Table 3).
Briey, LSD1 promotes chemoresistance via functioning as a co-
activator through interacting with different ligand proteins (Kim
et al., 2013;Boulding et al., 2018;Verigos et al., 2019;
Strachowska et al., 2021). It mediates resistance to hormone
therapy via activating ER transcriptional activity (Cortez et al.,
2012;Benedetti et al., 2019). In addition, LSD1 is also involved in
resistance to PD-1 antibody treatment and BRD4 inhibitors via
its transcriptional inhibitory activity against multiple oncogenes
(Qin et al., 2019;Tu et al., 2020;Liu et al., 2022).
4 Targeting LSD1 for breast cancer
therapy
Considering the crucial roles of LSD1 in BC progression, it
has the potential as a therapeutic target for BC treatment.
Currently, tens of LSD1 inhibitors have been documented
with anti-BC activity with some of these having entered
clinical trials. Herein, they were classied into seven
subcategories based on their structural characteristics: PCPA-
based LSD1 inhibitors, polyamine analogues, natural products,
propargylamine derivatives, benzohydrazide derivatives, phenyl
oxazole derivatives, and dual-target inhibitors.
4.1 PCPA-based LSD1 inhibitors
Since LSD1 and monoamine oxidases (MAOs) share high
similarity in their catalytic domains, several LSD1 inhibitors have
TABLE 3 Roles of LSD1 in drug resistance of BC.
Resistant agents Complexes/
pathways
Target genes Functions References
Chemotherapy Paclitaxel CAC1/LSD1/ER TFF1, TERB Promoting paclitaxel Kim et al. (2013)
LSD1S111/PKC-θJUNB,KLF10,KLF6,
and CCL2
Resistance Boulding et al. (2018)
Doxorubicin —— Enhancing BC stemness Verigos et al. (2019)
Doxorubicin, cisplatin,
daunorubicin, and methotrexate
CBP/LSD1 ABCC1 and ABCC10 Promoting drug efux Strachowska et al., (2021);
Sobczak et al., (2022)
Hormone
therapy
Tamoxifen LSD1/PELP1/ER GREB1C,Aromatase Promoting BC hormone resistance Cortez et al. (2012)
LSD1/ER ER Activating ER transcriptional activity Benedetti et al. (2019)
Immunotherapy PD-1 antibody CCL5,CXCL9,
CXCL10, and PD-L1
Reducing efcacy of PD-1 antibody Qin et al. (2019)
EOMES/LSD1 Mvpolarization
genes
Reducing immune Cell inltration
and increasing checkpoint markers
Qin et al., (2019);Tu et al.,
(2020)
Targeted therapy BRD4 inhibitors BRD4/LSD1/NuRD GNA13 and PDPK1 Promoting JQ1 resistance Liu et al. (2022)
Frontiers in Pharmacology frontiersin.org06
Yang et al. 10.3389/fphar.2022.989575
been discovered based on reported MAO inhibitors via in silicon
screening and chemical structure optimizations (Yang et al.,
2007;Yang et al., 2018a). Tranylcypromine hydrochloride (2-
PCPA, 1), an irreversible monoamine oxidase (MAO) inhibitor
with half maximal inhibitory concentration (IC
50
) of 11.5 and
7.0 μM for MAO A and MAO B in vitro, respectively, has also
exhibited inhibitory activity against LSD1 (IC
50
= 22.3 μM)
(Figure 4)via covalently binding to its FAD-binding motif (Ji
et al., 2017;Cao et al., 2018). Further study veried that
compound 1also inhibited migration, invasion, and metastasis
of TNBC cell lines BT-549 and MDA-MB-231 cells and tumor
bone metastasis in vivo. In the mechanism, 1blocked the
interaction between LSD1 and slug, and thus upregulated
suppressor E-cadherin and reduced epithelial markers
(Ferrari-Amorotti et al., 2014).To improve the potency, and
selectivity of 2-PCPA, some 2-PCPA derivatives have been
designed and synthesized based on multiple optimization
strategies. GlaxoSmithKline lnc. has designed 2 PCPA-based
LSD1 inhibitors 2and 3with IC
50
of 1.7 μM, and 0.016 μM,
respectively, (Figure 4)(Tu et al., 2020;Zhou et al., 2021b).
N-alkylated 2-PCPA derivative 2, a selective and orally bioavailable
for LSD1 inhibitor induced IFN-γ/TNF-α-expressing CD8 T cell
inltration into the tumors of 4T1 immunotherapy-resistant mice
and sensitized to immunotherapy via aLSD1-EOMESswitch.
Interestingly, 2 showed much potent anti-BC activity than PD-L1
antibody (Tu et al., 2020). Compound 3promoted the antigen
presentation and enhanced the tumor-killing activity of tumor-
specic cytotoxic T-cells in 4T1 mouse model (Zhou et al., 2021b).
Compounds 4and 5(Figure 4) are two PCPA-4-hydroxytamoxifen
conjugates, which released 4-hydroxytamoxifen catalyzing by
LSD1 in vitro and in cellulo and exhibited anti-proliferative
activity against MCF-7 cells at concentrations as low as 0.1 μM.
In addition, both of the two conjugates have better in cellulo anti-
proliferative activity than their parent compounds (Ota et al., 2016).
NCD38 (6), a selective LSD1 inactivator optimized from PCPA with
IC
50
of 0.59 μM(Sugino et al., 2017), could reduce the stemness of
TNBC cells and tumor growth in vitro (Zhou et al., 2021a). ORY-
1001 (7), a PCPA derivative in phase II clinical trial for acute
myelocytic leukemia, could inhibit TNBC cells and HER2-positive
BC cells in distinct mechanisms (Figure 4)(Cuyàs et al., 2020;Wang
et al., 2022). Compound 7inhibited HER2-positive BC via reducing
SOX2-driven breast cancer stem cells. In the mechanism, compound
7disturbed the assembly between LSD1 and co-repressor RCOR1/
CoREST via blocking the binding between LSD1 and FAD cofactor,
and thus enhanced transcriptional repression of SOX2 (Cuyàs et al.,
2020). In TNBC, compound 7suppressed the proliferation of TNBC
cells via devitalizing androgen receptor (Wang et al., 2022).
4.2 Polyamine analogues
Polyamine analogues, previously identied as the FAD-
dependent spermine oxidase inhibitors, were also found to
inhibit LSD1 demethylase activity in 2007 (Huang et al.,
2007). Bisguanidine 8and biguanide 9exhibited demethylase
inhibitory activity by over 50% at 1 μM but both of them lack
selectivity among MAOs (Nowotarski et al., 2015). Four (bis)-
thioureidopropyldiamine compounds (1013) showed improved
selectivity compared with their lead compounds 8and 9
(Figure 5). Among them,compound 13 exhibited the best
selectivity and potency with IC
50
values of 5.0 and 4.8 μM
in vitro, respectively. In fact, 13 also showed much more
potent anticancer activity against MCF7 cells than 2-PCPA
(Nowotarski et al., 2015). Further study showed that
compounds 1013 suppressed the proliferation of MCF-7 cells
via increasing H3K4me2 levels, which signicantly upregulated
tumor suppressor genes p16,GATA4,HCAD, and SFRP2. The
docking assay indicated that 11 could form three hydrogen bonds
with residues N535 and A539, and FAD within the LSD catalytic
pocket. In addition, the hydrophobic interactions between
hydrophobic residues in lining the LSD1 pocket and 11 also
contributed to their binding ability.
4.3 Natural products
Natural products are one of the major sources in drug
discovery due to their diverse chemical scaffolds and activity
proles (Fang et al., 2020;Yang et al., 2020;Yang et al., 2021a;
Cheng et al., 2022b;Song et al., 2022). Many natural products and
their derivatives have been found with in vitro inhibitory activity
against LSD1.
Kongs group identied six avonoid compounds (Figure 6,
1419) with inhibitory activity against LSD1 from Scutellaria
baicalensis Georgi using countercurrent chromatography (CCC)
(Han et al., 2018). Among them, compound 19 is the best
LSD1 inhibitor with the in vitro IC
50
of 2.98 µM and in
cellulo IC
50
of 17.94 µM against MDA-MB-231 cells.
Isoquercitrin (Figure 6,20), a avonoid compound with anti-
BC activity extracted from Bidens bipinnata L,isan
LSD1 inhibitor that inhibited proliferation of TNBC cell line
MDA-MB-231 via activating mitochondrial-mediated apoptosis
(Xu et al., 2019). Biochanin A (21), a dietary avonoid from Cicer
arietinum L, could inhibit the proliferation and metastasis of BC
in cellulo and in vivo (Moon et al., 2008;Sehdev et al., 2009;Ren
et al., 2018). Compound 21 was found to be effective and
reversible with IC
50
of 2.95 μM and it preferably suppressed
LSD1 over MAO-A/B (>32 μM) (Wang et al., 2020). In gastric
MGC-803 cells, Biochanin A induced the accumulation of
H3K4me1/2 and inhibited cell growth moderately (IC
50
=
6.77 µM) (Wang et al., 2020). Oleacein (Figure 5,22), a
dihydroxy-phenol found in extra virgin olive oil, is a FAD
competitive LSD1 inhibitor with IC
50
of 2.5 μMin vitro
(Cuyàs et al., 2019). Compound 22 reduced the stemness of
BC stem cells via blocking the interaction between LSD1 and the
methylated histone H3, disintegrating the assembled co-
Frontiers in Pharmacology frontiersin.org07
Yang et al. 10.3389/fphar.2022.989575
FIGURE 4
Chemical structures of PCPA-based LSD1 inhibitors in BC therapy.
FIGURE 5
Chemical structures of polyamine analogue-based LSD1 inhibitors.
Frontiers in Pharmacology frontiersin.org08
Yang et al. 10.3389/fphar.2022.989575
repressor complex LSD1/RCOR1/CoREST, disturbing the
occupation of LSD1 to the SOX2 promoter and nally
reducing the SOX2 level. Capsaicin (Figure 6,23), a bioactive
compound from chili peppers with the broad spectrum of
anticancer activity in various subtype of BC cells (Chou et al.,
2009;Thoennissen et al., 2010;Wu et al., 2020;Chen et al., 2021),
was found to act as reversible LSD1 inhibitor with an IC
50
value
of 0.6 μM(Jia et al., 2020). Compound 23 competitively occupied
with FAD-binding sites within the catalytic pocket of LSD1,
raised H3K4me1/2 levels and suppressed the proliferation and
migration of BC cells.
The dried root of Salvia miltiorrhiza is a traditional Chinese
medicine used for over 1,000 years to treat cardiovascular and
cerebrovascular diseases, gynecological diseases, diabetes, and
insomnia (Su et al., 2015;Jia et al., 2019;Shi et al., 2019). S.
miltiorrhiza has also been reported to improve the survival rate of
BC patients and several bioactive components (Figure 6,2429)
mediated its pharmacological actions via multiple anticancer
FIGURE 6
Structure of natural LSD1 inhibitors.
Frontiers in Pharmacology frontiersin.org09
Yang et al. 10.3389/fphar.2022.989575
pathways (Jin et al., 2021;Mahmoud et al., 2021). For example,
rosmarinic acid (24) could suppress proliferation, metastasis and
angiogenesis, and sensitize BC cells to paclitaxel via NF-κB-
p53 pathways (Mahmoud et al., 2021). Dihydroisotanshinone I
(25) inhibited the proliferation of BC cells via inducing their
ferroptosis and apoptosis (Lin et al., 2019). Cryptotanshinone
(26) inhibited migration through inactivating PKM2/β-Catenin
signaling, and mediated drug resistance via reducing the
oligomer formation of breast cancer resistance protein on the
cell membrane, and thus blocking its efux function (Zhou et al.,
2020;Ni et al., 2021). Tanshinone I (27) inhibited the
proliferation of MDA-MB-231 cells via activating the AMP-
activated protein kinase mediating autophagic signaling (Zheng
et al., 2020). Tanshinone IIA (28) sensitized BC cells to
adriamycin via attenuates the stemness of BC cells by
targeting the miR-125b/STARD13 signaling (Li et al., 2022).
Recently, the six extracted monomeric compounds from roots
of S. miltiorrhiza have been identied as LSD1 inhibitors with
their IC
50
values within the range 0.1120.93 μM(Lin et al.,
2020). Among them, salvianolic acid B (29) showed the best
inhibitory activity against LSD1 (IC
50
= 0.11 μM) and it
demethylase-dependently inhibited the proliferation and
migration of MDA-MB-231 cells with an IC
50
value of
54.98 μM at 24 h against MDA-MB-231 cells.
Coptis chinensis, another traditional Chinese medicine widely
used over 2,000 years for treatment of atherosclerosis, diabetes,
and inammation (Alami et al., 2020) has also been used to treat
a varieties of cancers including BC (Wu et al., 2019), and
isoquinoline alkaloids (Figure 6,3034) have been identied
as the main anticancer active components of C. chinensis with
inhibitory activity against LSD1 (Yi et al., 2016). Epierberine (30)
inhibited the proliferation and metastasis of BC cells via induced
cell cycle arrest and induced apoptosis by regulating Wnt/β-
catenin pathway (Dian et al., 2022). Jatrorrhizine (32) exhibited
anti-proliferative activity via attenuating TNIK/Wnt/β-catenin
signaling in BC cells with IC
50
values of 11.08, 17.11, and
22.14 μM against MCF-7, MDA-MB-231, and 4T1 cells,
respectively (Sun et al., 2019). Berberine (27) sensitized the
BC cells to chemotherapeutic agents via reducing XRCC1-
mediated base excision repair (Gao et al., 2019). Palmatine
(34) reduced the lung metastasis of TNBC via downregulating
metastasis-associated protein 1 (MTA1) and increasing p53 level
(Ativui et al., 2022). Recently, Li Z. R. et al. (2020) found that ve
protoberberine alkaloids (3034) also showed the inhibitory
activities against LSD1. All the IC
50
values of them were as
low as micromoles and highly selective to LSD1 over MAOs.
Recently, Ren et al. (2021) also identied four
sesquiterpene-based LSD1 inhibitors (compounds 3538)
with their IC
50
values within the range 3.9722.22 μMfrom
zedoary turmeric oil using CCC strategy. Compound 36 had the
best inhibitory activity (IC
50
=3.95μM) of them and also
exhibited the anti-metastasis activity against MDA-MB-
231 cells.
4.4 Propargylamine derivatives
Due to the similar structure between MAOs and LSD1, two
MAOs inhibitors (Figure 7,39 and 40) with active
propargylamine group also showed a weak inhibitory activity
against LSD1 in the millimolar range (Lee et al., 2006;Matos
et al., 2009). To improve the potency of LSD1 inhibitor, a
covalent and irreversible LSD1 inhibitor (41) was designed
through combining the active propargylamine group with
N-terminal 21 amino acids of LSD1 substrate H3, compound
41 selectively inhibited LSD1 demethylase activity with a Ki value
of 0.107 μM(Culhane et al., 2006). Based on the structure of 41,
Schmitt et al. (2013) designed and synthesized a set of with
propargyl warhead (Figure 7,4245). Among them, compound
44 was the best LSD1 irreversible inhibitor with IC
50
of 44.0 μM
in vitro, while 43 has the best anti-proliferative activity against
MCF7 cells (IC
50
= 91.5 μM) for 24 h (Schmitt et al., 2013). The
docking analysis of the binding mode between 42 and
LSD1 showed that residues Y761 and D555 formed hydrogen
bonds with the amine of N-propargylamine warhead and the
amide nitrogen atom of the benzamide moiety, respectively. The
aromatic substituents extensively form hydrophobic and
T-shaped aromatic interactions with F558, F560, Y807,
and H812.
4.5 Benzohydrazide derivatives
Sorna et al. (2013) identied six benzohydrazides (Figure 8,
4651)IC
50
values within 0.190.333 μM range based on virtual
screening from a compound library containing
2,000,000 compounds and biochemical assay. Then, three
compounds 5355, with IC
50
of 0.128, 0.013, and 0.014 μM,
respectively, were gotten though structure-based optimization.
Compound 55, the best potent LSD1 inhibitor of them, is
reversible and specic for LSD1 and inhibits the proliferation
and survival of seven BC cell lines with IC
50
from 0.468 to
2.730 μM range, which is more potent than a known MAO
inhibitor 2-PCPA. Further study showed that 55 suppressed
the proliferation of BC cells via reducing Sox2 expression,
promoting G1 cell cycle arrest, and inducing the expression of
differentiation-related genes in demethylase-dependent manner
(Zhang et al., 2013). The binding mode analysis using ICM
software showed that 55 could form three H-bonding
interactions with the residues G314, R350, and Y510 via its
benzohydrazide scaffold.
4.6 Phenyl oxazole derivatives
The polyamine/guanidine and methionine were often key
pharmacophores for designing MAOs inhibitors. Dulla et al.
designed a series of compounds (Figure 9,5659) with related
Frontiers in Pharmacology frontiersin.org10
Yang et al. 10.3389/fphar.2022.989575
FIGURE 7
Structure of propargylamine-based LSD1 inhibitors.
FIGURE 8
Structure of benzohydrazide-based LSD1 inhibitors.
Frontiers in Pharmacology frontiersin.org11
Yang et al. 10.3389/fphar.2022.989575
pharmacophores using oxazole as a linker (Dulla et al., 2013).
The results showed that compounds 5658 exhibited good
in vitro inhibitory activity against LSD1 with IC
50
of 16.1,
10.1, and 9.5 μM, respectively. Interestingly, all the synthesized
compounds including compound 59 showed an excellent
cytotoxicity activity against MDA-MB-231 cells (IC
50
=
1.0351.328 nM) in cellulo, which suggested these compounds
have other targets contributing their anticancer activity. Docking
analysis showed that the free -NH2 and the oxazole nitrogen of
compound 56 formed H-bonding with residues T624 and
R316 of LSD1, respectively, and its oxazole ring is involved in
aπ-cation interaction with residue R316. In case of 57, its free
-NH2 formed a H-bond with S760, the sulfur atom formed
H-bond with M332 and V333, and the phenyl ring interacted
with W751 and Y761 residues via π-πstacking. In case of 57, its
replaced guanidine group could form two H-bonds with residues
E308 and R310 whereas its phenyl ring interacted with R316 via
π-cation interaction.
4.7 Dual-target inhibitors and combined
therapy
In BC, LSD1 and KDM6A have been found to co-express and
co-localize with ER, and regulate hormone receptor signaling
(Benedetti et al., 2019), suggesting that developing dual-targeting
agents against the two proteins are potent strategy to improve the
potency of LSD1 inhibitors. Based on this hypothesis, Altuccis
group designed four dual-KDM inhibitors (Figure 10,6063)
targeting LSD1 and KDM6A. Among them, compound 61
exhibited the best anti-BC activity with no signicant toxicity
and good oral potency. In the mechanism, 61 induced cell arrest
and apoptosis of hormone-positive BC in cellulo and in vivo and
exhibited lower toxicity against non-cancerous cells (HaCaT)
compared with clinical HDAC inhibitor Vorinostat.
Additionally, it also reduced the resistance to endocrine
therapies, suggesting dual-target is a feasible and potent
strategy to overcome drug resistance for BC therapy.
Compound 64 was identied as a non-covalent dual LSD1/
G9a inhibitor with anti-leukemia activity (Speranzini et al., 2016;
Menna et al., 2022). Mais group optimized the structure of 64 via
modifying its quinazoline core and got two non-covalent, and
more potent LSD1/G9a inhibitors (Figure 10,65 and 66).
Compared to lead compound 63, compounds 65 and 66
exhibited a better inhibitory activity against LSD1 but reduced
the inhibitory activity against G9a. Further study showed that 65
and 66 showed better anticancer activity against MDA-MBA-
231, THP-1, and MV4-11 cells without signicant toxicity to
non-cancer AHH-1 cells compared with epigenetic drug
UNC0638, which suggested that the LSD1 antagonistic activity
of LSD1/G9a inhibitors endorsed with anticancer activity
(Menna et al., 2022).
LSD1 has been found to promote BC proliferation via
interacting with histone deacetylases (HDACs) (Cao et al.,
2017). Huangs group found that HDAC inhibitor
sulforaphane suppressed the proliferation of BC cells via
blocking activity of upstream transcription factor 1 (USF1) and
promoting ubiquitination degradation of LSD1, and LSD1 inhibitor
signicantly sensitized sulforaphane to BC in cellulo and in vivo
(Cao et al., 2018), which inhibited that the combined therapy or
discovery of LSD1/HDAC5 inhibitors is a potential strategy for BC
treatment. Zylla et al. (2022) study found that HDAC/
LSD1 inhibitor 67 (Figure 10) exhibited anti-proliferative and
anti-metastatic activity against TNBC in cellulo and in vivo.
Currently, hormonal drugs and chemotherapy have been
widely combined in the utilization for BC therapy in clinic. But
combined strategies also showed undeniable disadvantages
during clinic use. To overcome this phenomenon, Zhous
group developed a set of dual-target inhibitors based
compound 68. Among these conjugators, compound 69 has
FIGURE 9
Structure of phenyl oxazole-based LSD1 inhibitors.
Frontiers in Pharmacology frontiersin.org12
Yang et al. 10.3389/fphar.2022.989575
the best in vitro and in cellulo activity with IC
50
values of 9.67,
1.36, 1.55, and 8.79 μM against ERα,ERβ, LSD1, and MCF7,
respectively (He et al., 2020). Most notably, 69 exhibited better
anti-BC activity in MCF-7 cells than clinical agent 4-
hydroxytamoxifen.
5 Discussion and future prospects
Mounting evidence supports that LSD1 is overexpressed in
many subtypes of BC and promotes their proliferation (Pollock
et al., 2012;Yang et al., 2018d;Xu et al., 2019;Wang et al., 2022),
differentiation (Wu et al., 2013;Zhang et al., 2013;Ji et al., 2021),
metastasis (Li et al., 2011;Qiu et al., 2018;Zheng et al., 2018;Hu
et al., 2019;Gong et al., 2021), and drug resistance (Bennani-
Baiti, 2012;Verigos et al., 2019;Zhou et al., 2021a;Liu et al.,
2022), which makes LSD1 become a promising target for BC
therapy. But the detailed mechanisms of the LSD1 in BC
progression are unclear and more potential anti-tumor
pathways or downstream genes are yet to clarify due to the
heterogeneity of varieties of BC subtypes. For example, metastasis
and drug resistance are two main factors responsible for BC-
caused death in clinic (Cheng et al., 2021;Cheng et al., 2022a).
Although there are several reported researches on the roles of
LSD1 in BC metastasis and drug resistance, the specic functions
of LSD1 in these two cancer cell events are yet to be investigated.
In addition, many studies showed that LSD1 functioned as an
oncogene or suppressor gene in BC progression dependent on its
transcriptional regulatory activity via assembling into different
complexes with its client proteins. Therefore, mapping the
protein-protein interactome of LSD1 is a potential strategy to
further clarify its function in BC development (Yatim et al., 2012;
Yang et al., 2022). Meanwhile, the exploration on the functions of
LSD1 in homeostasis is also imperative to avoid the potential
health risks during the development and advancement of clinical
trials of LSD1 inhibitors. Moreover, most of the current studies
about LSD1 functions mainly focus on its histone demethylase
activity, and few works are available about the roles of non-
histone substrates, epigenetic modications, and non-enzyme
activity of LSD1 in BC progression (Feng et al., 2016;Liu et al.,
FIGURE 10
Structure of reported dual-target inhibitors.
a
RBA values = ICE2
50 /ICCompound
50 × 100 ± the range (RBA value of E2 as 100%).
Frontiers in Pharmacology frontiersin.org13
Yang et al. 10.3389/fphar.2022.989575
2020a;Gong et al., 2021). Thus, it is imperative to further carry
out research in these areas.
Currently, some LSD1 inhibitors have entered clinical trials
to combat small lung cancer cells and acute myeloid leukemia
and several of them showed encouraging results (Fang et al.,
2019). But there is no LSD1 inhibitor in clinical trials for BC.
Many factors contribute to this phenomenon apart from the
complex etiological factor of BC. First, most of the reported
LSD1 inhibitors have poor selectivity, which adds to the
uncertainty of drug therapy. Second, some LSD1 inhibitors
such as compounds 5659 exhibited intracellular activity
inconsistent with their inhibitory activity against demethylase
activity (Dulla et al., 2013), which suggested that there are
potential off-target effects and unpredictable risks of some
identied LSD1 inhibitors, when they were advanced into
clinical trials for BC therapy. Third, like other enzyme
inhibitors (Yang et al., 2018c;Wu et al., 2018;Yang et al.,
2019;Yang et al., 2021c;Yang et al., 2021d), most of the
reported LSD1 inhibitors were only detected in anti-BC
activity in vitro or in cellulo assays with a dearth of the
studies about in vivo toxicology, pharmacokinetics, and
effectiveness in animals. Finally, due to the existence of
several alternative signaling and isoenzymes in BC cells,
LSD1 inhibitor used alone may be not sufcient to achieve
the desired therapeutic effect sometimes. To solve these
dilemmas, pharmaceutical chemists and pharmacologists have
proposed several strategies. Given allosteric regulation is a
common characteristic for metabolic enzymes (Kremer and
Lyssiotis, 2022), identifying allosteric pockets or sites and
developing corresponding inhibitors is a feasible strategy for
the discovery of LSD1 inhibitors. Targeting the proteinprotein
interaction (PPI) is also an effective method to improve the
selectivity of enzymes (Yang et al., 2018a;Cheng et al., 2020;Yang
et al., 2021a;Yang et al., 2021c;Yang et al., 2021d;Yang et al.,
2022), blocking the interaction between LSD1 and its client
proteins may be also a useful strategy to develop selective
LSD1 inhibitors. Metal complexes have showed promising in
vivo activity for BC treatment (Yang et al., 2018b;Cheng et al.,
2022b), and Yang et al. (2017) have also identied a rhodium-
based LSD1 inhibitor with in cellulo anticancer activity against
prostate cancer, which suggested this kind of compound is a
unique source for the discovery of potent and selective
LSD1 inhibitors against BC treatment. Several drug design
strategy such as computer aided drug optimization and
proteolysis targeting chimera (PROTAC)-strategy have been
introduced to discover lead compounds against LSD1 with
better biocompatibility and in vivo potency (Liu et al., 2020b;
Martín-Acosta and Xiao, 2021). Moreover, combined therapy
and developing dual-target inhibitors have also been used to
improve the anticancer potency and overcome drug resistance of
LSD1 inhibitors (Ota et al., 2016;Cao et al., 2018;Benedetti et al.,
2019;Qin et al., 2019;Verigos et al., 2019;Liu et al., 2022). Drug
delivery by nanocarriers is a potent strategy to increase drug
utilization rate, improve potency, and reduce toxicity and drug
resistance of single agent or combined therapy (Singh and
Mitragotri, 2020;Vallet-Regí et al., 2022). LSD1 has been
found to exhibit its activity by promoting gastric cancer cell
stemness via delivered by endogenous small extracellular vesicles
in vivo (Zhao et al., 2021), and the drug delivery of LSD1 siRNA
using nanocarriers has exhibited potent anticancer activities in
several studies (Suzuki, 2019;Li B. et al., 2020;Jiang et al., 2021).
In a word, LSD1 plays a crucial role in BC progression and
drug resistance, and its inhibition is a potential anti-BC strategy
due to its efcacy in current preclinical studies. Therefore, the
discovery of more specic LSD1 inhibitors is imperative to
deepen our understanding of the role of LSD1 in BC
tumorigenesis and verify the feasibility of targeting LSD1 as
an anti-BC therapy in clinic.
Author contributions
G-JY and JC: supervision, funding acquisition, and
writingoriginal draft preparation. Y-JL, L-JD, FT, M-HZ,
and Z-YS: preparation of gures and tables. J-MW, M-YN,
XL, Z-SL, W-JQ, and C-JF: investigation and validation,
writing, reviewing, and editing. G-JY and JC: conception,
writing, reviewing ,and editing. The authors contributed to the
data preparation and drafted and revised the manuscript.
Funding
This work was supported by the National Natural Science
Foundation of China (31972821), the General ScienticResearch
Project of Education of Zhejiang Province (Y202147351), the Starting
Research Fund of Ningbo University (421912073), and the State Key
Laboratory for Managing Biotic and Chemical Threats to the Quality
and Safety of Agro-products (2010DS700124-ZZ2008).
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could
be construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the
authors and do not necessarily represent those of their afliated
organizations, or those of the publisher, the editors, and the
reviewers. Any product that may be evaluated in this article, or
claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Frontiers in Pharmacology frontiersin.org14
Yang et al. 10.3389/fphar.2022.989575
References
Alami, M. M., Xue, J., Ma, Y., Zhu, D., Abbas, A., Gong, Z., et al. (2020). Structure,
function, diversity, and composition of fungal communities in rhizospheric soil of
Coptis chinensis Franch under a successive cropping system. Plants 9 (2), 244.
doi:10.3390/plants9020244
Andresen, M. S., Ali, H. O., Myklebust, C. F., Sandset, P. M., Stavik, B., Iversen, N.,
et al. (2017). Estrogen induced expression of tissue factor pathway inhibitor-2 in
MCF7 cells involves lysine-specic demethylase 1. Mol. Cell. Endocrinol. 443,
8088. doi:10.1016/j.mce.2017.01.016
Ativui, S., Danquah, C. A., Ossei, P. P. S., and Ofori, M. (2022). Palmatine
attenuates metastatic lung colonization of triple negative breast cancer cells. Front.
Pharmacol. 13, 853230. doi:10.3389/fphar.2022.853230
Ayoub, N. M., Jaradat, S. K., Al-Shami, K. M., and Alkhalifa, A. E. (2022).
Targeting angiogenesis in breast cancer: Current evidence and future perspectives of
novel anti-angiogenic approaches. Front. Pharmacol. 13, 838133. doi:10.3389/fphar.
2022.838133
Bai, J. W., Chen, M. N., Wei, X. L., Li, Y. C., Lin, H. Y., Chen, M., et al. (2017). The
zinc-nger transcriptional factor Slug transcriptionally downregulates ERαby
recruiting lysine-specic demethylase 1 in human breast cancer. Oncogenesis 6
(5), e330. doi:10.1038/oncsis.2017.38
Benedetti, R., DellAversana, C., De Marchi, T., Rotili, D., Liu, N. Q., Novakovic,
B., et al. (2019). Inhibition of histone demethylases LSD1 and UTX regulates ERα
signaling in breast cancer. Cancers 11 (12), 2027. doi:10.3390/cancers11122027
Bennani-Baiti, I. M. (2012). Integration of ERα-PELP1-HER2 signaling by LSD1
(KDM1A/AOF2) offers combinatorial therapeutic opportunities to circumventing
hormone resistance in breast cancer. Breast Cancer Res. 14 (5), 112. doi:10.1186/
bcr3249
Bennesch, M. A., Segala, G., Wider, D., and Picard, D. (2016). LSD1 engages a
corepressor complex for the activation of the estrogen receptor αby estrogen and
cAMP. Nucleic Acids Res. 44 (18), 86558670. doi:10.1093/nar/gkw522
Boulding, T., McCuaig, R., Tan, A., Hardy, K., Wu, F., Dunn, J., et al. (2018).
LSD1 activation promotes inducible EMT programs and modulates the tumour
microenvironment in breast cancer. Sci. Rep. 8 (1), 73. doi:10.1038/s41598-017-17913-x
Bradley, C., van der Meer, R., Roodi, N., Yan, H. P., Chandrasekharan, M., Sun, Z.
W., et al. (2007). Carcinogen-induced histone alteration in normal human
mammary epithelial cells. Carcinogenesis 28 (10), 21842192. doi:10.1093/
carcin/bgm100
Cao, C. Y., Vasilatos, S. N., Bhargava, R., Fine, J. L., Oesterreich, S., Davidson, N.
E., et al. (2017). Functional interaction of histone deacetylase 5 (HDAC5) and
lysine-specic demethylase 1 (LSD1) promotes breast cancer progression. Oncogene
36 (1), 133145. doi:10.1038/onc.2016.186
Cao, C. Y., Wu, H., Vasilatos, S. N., Chandran, U., Qin, Y., Wan, Y., et al. (2018).
HDAC5-LSD1 axis regulates antineoplastic effect of natural HDAC inhibitor
sulforaphane in human breast cancer cells. Int. J. Cancer 143 (6), 13881401.
doi:10.1002/ijc.31419
Chen, M. J., Xiao, C. C., Jiang, W., Yang, W. P., Qin, Q. H., Tan, Q., et al. (2021).
Capsaicin inhibits proliferation and inducesapoptosis in breast cancer by down-
regulating FBI-1-mediated NF-κB pathway. Drug Des. devel. Ther. 15, 125140.
doi:10.2147/DDDT.S269901
Cheng, S. S., Qu, Y. Q., Wu, J., Yang, G. J., Liu, H., Wang, W. H., et al. (2022a).
Inhibition of the CDK9cyclin T1 proteinprotein interaction as a new approach
against triple-negative breast cancer. Acta Pharm. Sin. B 12 (3), 13901405. doi:10.
1016/j.apsb.2021.10.024
Cheng, S. S., Yang, G. J., Wang, W. H., Leung, C. H., and Ma, D. L. (2020). The
design and development of covalent protein-protein interaction inhibitors for
cancer treatment. J. Hematol. Oncol. 13, 26. doi:10.1186/s13045-020-00850-0
Cheng, S. S., Yang, G. J., Wang, W. H., Ma, D. L., and Leung, C. H. (2021).
Discovery of a tetrahydroisoquinoline-based CDK9-cyclin T1 proteinprotein
interaction inhibitor as an anti-proliferative and anti-migration agent against
triple-negative breast cancer cells. Genes Dis.. doi:10.1016/j.gendis.2021.06.005
Cheng, S. S., Yang, G. J., Wang, W. H., Song, Y. Q., Ko, C. N., Han, Q. B., et al.
(2022b). Identication of a cytisine-based EED-EZH2 protein-protein interaction
inhibitor preventing metastasis in triple-negative breast cancer cells. Acta Mat. Med.
1 (2), 197211. doi:10.15212/AMM-2022-0006
Chou, C. C., Wu, Y. C., Wang, Y. F., Chou, M. J., Kuo, S. J., and Chen, D. R.
(2009). Capsaicin-induced apoptosis in human breast cancer MCF-7 cells through
caspase-independent pathway. Oncol. Rep. 21 (3), 665671. PMID: 19212624.
doi:10.3892/or_00000269
Cortez, V., Mann, M., Tekmal, S., Suzuki, T., Miyata, N., Rodriguez-Aguayo, C.,
et al. (2012). Targeting the PELP1-KDM1 axis as a potential therapeutic strategy for
breast cancer. Breast Cancer Res. 14 (4), R108. doi:10.1186/bcr3229
Culhane, J. C., Szewczuk, L. M., Liu, X., Da, G., Marmorstein, R., and Cole, P. A.
(2006). A mechanism-based inactivator for histone demethylase LSD1. J. Am.
Chem. Soc. 128 (14), 45364537. doi:10.1021/ja0602748
Cuyàs, E., Gumuzio, J., Lozano-Sánchez, J., Carreras, D., Verdura, S., Llorach-
Parés, L., et al. (2019). Extra virgin olive oil contains a phenolic inhibitor of the
histone demethylase LSD1/KDM1A. Nutrients 11 (7), 1656. doi:10.3390/
nu11071656
Cuyàs, E., Gumuzio, J., Verdura, S., Brunet, J., Bosch-Barrera, J., Martin-Castillo,
B., et al. (2020). The LSD1 inhibitor iadademstat (ORY-1001) targets SOX2-driven
breast cancer stem cells: A potential epigenetic therapy in luminal-B and HER2-
positive breast cancer subtypes. Aging 12 (6), 47944814. doi:10.18632/aging.
102887
Dian, L. L., Xu, Z. Z., Sun, Y. F., Li, J. H., Lu, H. F., Zheng, M., et al. (2022).
Berberine alkaloids inhibit the proliferation and metastasis of breast carcinoma cells
involving Wnt/β-catenin signaling and EMT. Phytochemistry 200, 113217. doi:10.
1016/j.phytochem.2022.113217
Disis, M. (2010). Immune regulation of cancer. J. Clin. Oncol. 28, 45314538.
doi:10.1200/JCO.2009.27.2146
Dulla, B., Kirla, K. T., Rathore, V., Deora, G. S., Kavela, S., Maddika, S., et al.
(2013). Synthesis and evaluation of 3-amino/guanidine substituted phenyl oxazoles
as a novel class of LSD1 inhibitors with anti-proliferative properties. Org. Biomol.
Chem. 11 (19), 31033107. doi:10.1039/c3ob40217g
Fang, Y., Liao, G. C., and Yu, B. (2019). LSD1/KDM1A inhibitors in clinical trials:
Advances and prospects. J. Hematol. Oncol. 12 (1), 129. doi:10.1186/s13045-019-
0811-9
Fang, Y., Yang, C., Yu, Z. Q., Li, X., Mu, Q. C., Liao, G. C., et al. (2020). Natural
products as LSD1 inhibitors for cancer therapy. Acta Pharm. Sin. B 11 (3), 621631.
doi:10.1016/j.apsb.2020.06.007
Feng, J., Li, L., Zhang, N., Liu, J., Zhang, L., Gao, H., et al. (2017). Androgen and
AR contribute to breast cancer development and metastasis: An insight of
mechanisms. Oncogene 36 (20), 27752790. doi:10.1038/onc.2016.432
Feng, J. X., Xu, G. Y., Liu, J. W., Zhang, N., Li, L. L., Ji, J. F., et al. (2016).
Phosphorylation of LSD1 at Ser112 is crucial for its function in induction of EMT
and metastasis in breast cancer. Breast Cancer Res. Treat. 159 (3), 443456. doi:10.
1007/s10549-016-3959-9
Ferrari-Amorotti, G., Chiodoni, C., Shen, F., Cattelani, S., Soliera, A., Manzotti,
G., et al. (2014). Suppression of invasion and metastasis of triple-negative breast
cancer lines by pharmacological or genetic inhibition of slug activity. Neoplasia 16
(12), 10471058. doi:10.1016/j.neo.2014.10.006
Forneris, F., Binda, C., Battaglioli, E., and Mattevi, A. (2008). LSD1: Oxidative
chemistry for multifaceted functions in chromatin regulation. Trends biochem. Sci.
33 (4), 181189. doi:10.1016/j.tibs.2008.01.003
Gao, X. J., Wang, J., Li, M. Q., Wang, J., Lv, J., Zhang, L., et al. (2019). Berberine
attenuates XRCC1-mediated base excision repair and sensitizes breast cancer cells
to the chemotherapeutic drugs. J. Cell. Mol. Med. 23 (10), 67976804. doi:10.1111/
jcmm.14560
Gong, Z., Li, A., Ding, J., Li, Q., Zhang, L., Li, Y., et al. (2021). OTUD7B
deubiquitinates LSD1 to govern its binding partner specicity, homeostasis,
and breast cancer metastasis. Adv. Sci. 8 (15), e2004504. doi:10.1002/advs.
202004504
Gu, F. Y., Lin, Y. X., Wang, Z., Wu, X. X., Ye, Z. Y., Wang, Y. Z., et al. (2020).
Biological roles of LSD1 beyond its demethylase activity. Cell. Mol. Life Sci. 77 (17),
33413350. doi:10.1007/s00018-020-03489-9
Han, C., Wang, S. S., Li, Z. R., Chen, C., Hou, J. Q., Xu, D. Q., et al. (2018).
Bioactivity-guided cut countercurrent chromatography for isolation of lysine-
specic demethylase 1 inhibitors from Scutellaria baicalensis Georgi. Anal.
Chim. Acta 1016, 5968. doi:10.1016/j.aca.2018.01.014
He, M., Ning, W. T., Hu, Z. Y., Huang, J., Dong, C. N., and Zhou, H. B. (2020).
Design, synthesis and biological evaluation of novel dual-acting modulators
targeting both estrogen receptor α(ERα) and lysine-specic demethylase 1
(LSD1) for treatment of breast cancer. Eur. J. Med. Chem. 195, 112281. doi:10.
1016/j.ejmech.2020.112281
Hu, X., Xiang, D. X., Xie, Y., Tao, L. W., Zhang, Y., Jin, Y., et al. (2019).
LSD1 suppresses invasion, migration and metastasis of luminal breast cancer cells
via activation of GATA3 and repression of TRIM37 expression. Oncogene 38 (44),
70177034. doi:10.1038/s41388-019-0923-2
Huang, Y., Greene, E., Murray Stewart, T., Goodwin, A. C., Baylin, S. B., Woster,
P. M., et al. (2007). Inhibition of lysine-specic demethylase 1 by polyamine
analogues results in reexpression of aberrantly silenced genes. Proc. Natl. Acad.
Sci. U. S. A. 104, 80238028. doi:10.1073/pnas.0700720104
Frontiers in Pharmacology frontiersin.org15
Yang et al. 10.3389/fphar.2022.989575
Huang, Y., Vasilatos, S. N., Boric, L., Shaw, P. G., and Davidson, N. E. (2012).
Inhibitors of histone demethylation and histone deacetylation cooperate in
regulating gene expression and inhibiting growth in human breast cancer cells.
Breast Cancer Res. Treat. 131 (3), 777789. doi:10.1007/s10549-011-1480-8
Ji, X. Y., Guo, D. X., Ma, J., Yin, M., Yu, Y., Liu, C., et al. (2021). Epigenetic
remodeling hydrogel patches for multidrug-resistant triple-negative breast cancer.
Adv. Mat. 33 (18), e2100949. doi:10.1002/adma.202100949
Ji, Y. Y., Lin, S. D., Wang, Y. J., Su, M., Zhang, W., Gunosewoyo, H., et al. (2017).
Tying up tranylcypromine: Novel selective histone lysine specic demethylase 1
(LSD1) inhibitors. Eur. J. Med. Chem. 141, 101112. doi:10.1016/j.ejmech.2017.
09.073
Jia, G., Cang, S. D., Ma, P. Z., and Song, Z. Y. (2020). Capsaicin: A hotKDM1A/
LSD1 inhibitor from peppers. Bioorg. Chem. 103, 104161. doi:10.1016/j.bioorg.
2020.104161
Jia, Q. Q., Zhu, R. Y., Tian, Y. M., Chen, B. B., Li, R., Li, L., et al. (2019). Salvia
miltiorrhiza in diabetes: A review of its pharmacology, phytochemistry, and safety.
Phytomedicine 58, 152871. doi:10.1016/j.phymed.2019.152871
Jiang, L. X., Gong, X. M., Liao, W. D., Lv, N. H., and Yan, R. W. (2021). Molecular
targeted treatment and drug delivery system for gastric cancer. J. Cancer Res. Clin.
Oncol. 147, 973986. doi:10.1007/s00432-021-03520-x
Jin, Z., Yu, C. H., and Peng, C. (2021). Anticancer effect of tansh inones on female
breast cancer and gynecological cancer. Front. Pharmacol. 12, 824531. doi:10.3389/
fphar.2021.824531
Kim, D. H., Kim, K., and Baek, S. H. (2021). Roles of lysine-spe cic demethylase 1
(LSD1) in homeostasis and diseases. J. Biomed. Sci. 28 (1), 41. doi:10.1186/s12929-
021-00737-3
Kim, J., Park, U. H., Moon, M., Um, S. J., and Kim, E. J. (2013). Negative
regulation of ERαby a novel protein CAC1 through association with histone
demethylase LSD1. FEBS Lett. 587 (1), 1722. doi:10.1016/j.febslet.2012.10.054
Kremer, D. M., and Lyssiotis, C. A. (2022). Targeting allosteric regulation of
cancer metabolism. Nat. Chem. Biol. 18 (5), 441450. doi:10.1038/s41589-022-
00997-6
Lee, J. Y., Park, J. H., Choi, H. J., Won, H. Y., Joo, H. D., Shin, D. H., et al. (2017).
LSD1 demethylates HIF1αto inhibit hydroxylation and ubiquitin-mediated
degradation in tumor angiogenesis. Oncogene 36 (39), 55125521. doi:10.1038/
onc.2017.158
Lee, M. G., Wynder, C., Schmidt, D. M., McCafferty, D. G., and Shiekhattar, R.
(2006). Histone H3 lysine 4 demethylation is a target of nonselective antidepressive
medications. Chem. Biol. 13 (6), 563567. doi:10.1016/j.chembiol.2006.05.004
Li, B. L., Li, R., Zou, H. L., Ariga, K., Li, N. B., and Leong, D. T. (2020). Engineered
functionalized 2D nanoarchitectures for stimuli-responsive drug delivery. Mat.
Horiz. 7 (2), 455469. doi:10.1039/C9MH01300H
Li, L., Liu, X. H., He, L., Yang, J. G., Pei, F., Li, W. M., et al. (2017).
ZNF516 suppresses EGFR by targeting the CtBP/LSD1/CoREST complex to
chromatin. Nat. Commun. 8 (1), 691. doi:10.1038/s41467-017-00702-5
Li, Q., Shi, L., Gui, B., Yu, W. H., Wang, J. M., Zhang, D., et al. (2011). Binding of
the JmjC demethylase JARID1B to LSD1/NuRD suppresses angiogenesis and
metastasis in breast cancer cells by repressing chemokine CCL14. Cancer Res.
71 (21), 68996908. doi:10.1158/0008-5472.Can-11-1523
Li, X. M., Jia, Q., Zhou, Y. Y., Jiang, X., Song, L., Wu, Y. Y., et al. (2022).
Tanshinone IIA attenuates the stemness of breast cancer cells via targeting the miR-
125b/STARD13 axis. Exp. Hematol. Oncol. 11 (1), 2. doi:10.1186/s40164-022-
00255-4
Li, Z. R., Suo, F. Z., Guo, Y. J., Cheng, H. F., Niu, S. H., Shen, D. D., et al. (2020).
Natural protoberberine alkaloids, identied as potent selective LSD1 inhibitors,
induce AML cell differentiation. Bioorg. Chem. 97, 103648. doi:10.1016/j.bioorg.
2020.103648
Lim, S., Janzer, A., Becker, A., Zimmer, A., Schüle, R., Buettner, R., et al. (2010).
Lysine-specic demethylase 1 (LSD1) is highly expressed in ER-negative breast
cancers and a biomarker predicting aggressive biology. Carcinogenesis 31 (3),
512520. doi:10.1093/carcin/bgp324
Lin, T., Ponn, A., Hu, X., Law, B. K., and Lu, J. (2010). Requirement of the
histone demethylase LSD1 in Snai1-mediated transcriptional repression during
epithelial-mesenchymal transition. Oncogene 29 (35), 48964904. doi:10.1038/
onc.2010.234
Lin, Y. L., Han, C., Xu, Q. Q., Wang, W. L., Li, L. N., Zhu, D. R., et al. (2020).
Integrative countercurrent chromatography for the target isolation of lys ine-specic
demethylase 1 inhibitors from the roots of Salvia miltiorrhiza. Talanta 206, 120195.
doi:10.1016/j.talanta.2019.120195
Lin, Y. S., Shen, Y. C., Wu, C. Y., Tsai, Y. Y., Yang, Y. H., Lin, Y. Y., et al. (2019).
Danshen improves survival of patients with breast cancer and
dihydroisotanshinone I induces ferroptosis and apoptosis of breast cancer cells.
Front. Pharmacol. 10, 1226. doi:10.3389/fphar.2019.01226
Lin, Y. W., Kang, T. B., and Zhou, B. P. (2014). Doxorubicin enhances Snail/
LSD1-mediated PTEN suppression in a PARP1-dependent manner. Cell Cycle 13
(11), 17081716. doi:10.4161/cc.28619
Liu, B. B., Liu, X. H., Han, L. L., Chen, X., Wu, X. D., Wu, J. J., et al. (2022). BRD4-
directed super-enhancer organization of transcription repression programs links to
chemotherapeutic efcacy in breast cancer. Proc. Natl. Acad. Sci. U. S. A. 119 (6),
e2109133119. doi:10.1073/pnas.2109133119
Liu, J., Ma, J., Liu, Y., Xia, J., Li, Y. Y., Wang, Z. P., et al. (2020b). PROTACs: A
novel strategy for cancer therapy. Semin. Cancer Biol. 67, 171179. doi:10.1016/j.
semcancer.2020.02.006
Liu, J. W., Feng, J. X., Li, L. L., Lin, L. Y., Ji, J. F., Lin, C., et al. (2020a). Arginine
methylation-dependent LSD1 stability promotes invasion and metastasis of breast
cancer. EMBO Rep. 21 (2), e48597. doi:10.15252/embr.201948597
Luo, H. C., Shenoy, A. K., Li, X. H., Jin, Y., Jin, L. H., Cai, Q. S., et al. (2016). MOF
acetylates the histone demethylase LSD1 to suppress epithelial-to-mesenchymal
transition. Cell Rep. 15 (12), 26652678. doi:10.1016/j.celrep.2016.05.050
Ma, L., Ma, S., Zhao, G. M., Yang, L. Q., Zhang, P., Yi, Q. T., et al. (2016). miR-
708/LSD1 axis regulates the proliferation and invasion of breast cancer cells. Cancer
Med. 5 (4), 684692. doi:10.1002/cam4.623
Mahmoud, M. A., Okda, T. M., Omran, G. A., and Abd-Alhaseeb, M. M. (2021).
Rosmarinic acid suppresses inammation, angiogenesis, and improves paclitaxel
induced apoptosis in a breast cancer model via NF3 κB-p53-caspase-3 pathways
modulation. J. Appl. Biomed. 19 (4), 202209. doi:10.32725/jab.2021.024
Majello, B., Gorini, F., Saccà, C. D., and Amente, S. (2019). Expanding the role of
the histone lysine-specic demethylase LSD1 in cancer. Cancers 11 (3), 324. doi:10.
3390/cancers11030324
Malagraba, G., Yarmohammadi, M., Javed, A., Barceló, C., and Rubio-Tomás, T.
(2022). The role of LSD1 and LSD2 in cancers of the gastrointestinal system: An
update. Biomolecules 12 (3), 462. doi:10.3390/biom12030462
Marabelli, C., Marrocco, B., and Mattevi, A. (2016). The growing structural and
functional complexity of the LSD1/KDM1A histone demethylase. Curr. Opin.
Struct. Biol. 41, 135144. doi:10.1016/j.sbi.2016.07.011
Martín-Acosta, P., and Xiao, X. S. (2021). PROTACs to address the challenges
facing small molecule inhibitors. Eur. J. Med. Chem. 210, 112993. doi:10.1016/j.
ejmech.2020.112993
Matos, M. J., Viña, D., Quezada, E., Picciau, C., Delogu, G., Orallo, F., et al. (2009).
A new series of 3-phenylcoumarins as potent and selective MAO-B inhibitors.
Bioorg. Med. Chem. Lett. 19 (12), 32683270. doi:10.1016/j.bmcl.2009.04.085
Menna, M., Fiorentino, F., Marrocco, B., Lucidi, A., Tomassi, S., Cilli, D., et al.
(2022). Novel non-covalent LSD1 inhibitors endowed with anticancer effects in
leukemia and solid tumor cellular models. Eur. J. Med. Chem. 237, 114410. doi:10.
1016/j.ejmech.2022.114410
Metzger, E., Wissmann, M., Yin, N., Müller, J. M., Schneider, R., Peters, A. H. F.
M., et al. (2005). LSD1 demethylates repressive histone marks to promote
androgen-receptor-dependent transcription. Nature 437 (7057), 436439. doi:10.
1038/nature04020
Moon, Y. J., Shin, B. S., An, G., and Morris, M. E. (2008). Biochanin A inhibits
breast cancer tumor growth in a murine xenograft model. Pharm. Res. 25,
21582163. doi:10.1007/s11095-008-9583-6
Mortier, A., Van Damme, J., and Proost, P. (2008). Regulation of chemokine
activity by posttranslational modication. Pharmacol. Ther. 120 (2), 197217.
doi:10.1016/j.pharmthera.2008.08.006
Ni,W.,Fan,H.,Zheng,X.,Xu,F.,Wu,Y.,Li,X.,etal.(2021).Cryptotanshinoneinhibits
ERα-dependent and-independent BCRP oligomer formation to reverse multidrug
resistance in breast cancer. Front. Oncol. 11, 624811. doi:10.3389/fonc.2021.624811
Nowotarski, S. L., Pachaiyappan, B., Holshouser, S. L., Kutz, C., Li, Y., Huang, Y.,
et al. (2015). Structure-activity study for (bis)ureidopropyl- and (bis)
thioureidopropyldiamine LSD1 inhibitors with 3-5-3 and 3-6-3 carbon
backbone architectures. Bioorg. Med. Chem. 23 (7), 16011612. doi:10.1016/j.
bmc.2015.01.049
Ota, Y., Itoh, Y., Kaise, A., Ohta, K., Endo, Y., Masuda, M., et al. (2016). Targeting
cancer with PCPA-drug conjugates: LSD1 inhibition-triggered release of 4-
hydroxytamoxifen. Angew. Chem. Int. Ed. Engl. 55 (52), 1611516118. doi:10.
1002/anie.201608711
Park, U. H., Kang, M. R., Kim, E. J., Kwon, Y. S., Hur, W., Yoon, S. K., et al. (2016).
ASXL2 promotes proliferation of breast cancer cells by linking ERαto histone
methylation. Oncogene 35 (28), 37423752. doi:10.1038/onc.2015.443
Peng, B., Wang, J., Hu, Y., Zhao, H. L., Hou, W. Y., Zhao, H. L., et al. (2015).
Modulation of LSD1 phosphorylation by CK2/WIP1 regulates RNF168-dependent
Frontiers in Pharmacology frontiersin.org16
Yang et al. 10.3389/fphar.2022.989575
53BP1 recruitment in response to DNA damage. Nucleic Acids Res. 43 (12),
59365947. doi:10.1093/nar/gkv528
Phillips, S., and Kuperwasser, C. (2014). Slug: Critical regulator of epithelial cell
identity in breast development and cancer. Cell adh. Migr. 8 (6), 578587. doi:10.
4161/19336918.2014.972740
Pilotto, S., Speranzini, V., Tortorici, M., Durand, D., Fish, A., Valente, S., et al.
(2015). Interplay among nucleosomal DNA, histone tails, and corepressor CoREST
underlies LSD1-mediated H3 demethylation. Proc. Natl. Acad. Sci. U. S. A. 112 (9),
27522757. doi:10.1073/pnas.1419468112
Pollock, J. A., Larrea, M. D., Jasper, J. S., McDonnell, D. P., and McCafferty, D. G.
(2012). Lysine-specic histone demethylase 1 inhibitors control breast cancer
proliferation in ERα-dependent and -independent manners. ACS Chem. Biol. 7
(7), 12211231. doi:10.1021/cb300108c
Qin, Y., Vasilatos, S. N., Chen, L., Wu, H., Cao, Z. S., Fu, Y. M., et al. (2019).
Inhibition of histone lysine-specic demethylase 1 elicits breast tumor immunity
and enhances antitumor efcacy of immune checkpoint blockade. Oncogene 38 (3),
390405. doi:10.1038/s41388-018-0451-5
Qiu, R. F., Shi, H., Wang, S., Leng, S., Liu, R. Q., Zheng, Y., et al. (2018).
BRMS1 coordinates with LSD1 and suppresses breast cancer cell metastasis. Am.
J. Cancer Res. 8 (10), 20302045. PMCID: PMC6220148.
Ren, C. L., Lin, Y. L., Liu, X. Q., Yan, D., Xu, X., Zhu, D. R., et al. (2021). Target
separation and antitumor metastasis activity of sesquiterpene-based lysine-specic
demethylase 1 inhibitors from zedoary turmeric oil. Bioorg. Chem. 108, 104666.
doi:10.1016/j.bioorg.2021.104666
Ren, G. X., Shi, Z. X., Teng, C., and Yao, Y. (2018). Antiproliferative activity of
combined Biochanin A and ginsenoside Rhon MDA-MB-231 and MCF-7 human
breast cancer cells.. Molecules 23 (11), 2908. doi:10.3390/molecules23112908
Schmitt, M. L., Hauser, A. T., Carlino, L., Pippel, M., Schulz-Fincke, J., Metzger,
E., et al. (2013). Nonpeptidic propargylamines as inhibitors of lysine specic
demethylase 1 (LSD1) with cellular activity. J. Med. Chem. 56 (18), 73347342.
doi:10.1021/jm400792m
Sehdev, V., Lai, J. C., and Bhushan, A. (2009). Biochanin A modulates cell
viability, invasion, and growth promoting signaling pathways in HER-2-positive
breast cancer cells. J. Oncol. 2009, 121458. doi:10.1155/2009/121458
Shen, D. D., Pang, J. R., Bi, Y. P., Zhao, L. F., Li, Y. R., Zhao, L. J., et al. (2022).
LSD1 deletion decreases exosomal PD-L1 and restores T-cell response in gastric
cancer. Mol. Cancer 21 (1), 75. doi:10.1186/s12943-022-01557-1
Shi, M., Huang, F. F., Deng, C. P., Wang, Y., and Kai, G. Y. (2019). Bioactivities,
biosynthesis and biotechnological production of phenolic acids in Salvia
miltiorrhiza. Crit. Rev. Food Sci. Nutr. 59 (6), 953964. doi:10.1080/10408398.
2018.1474170
Shi, Y. J., Lan, F., Matson, C., Mulligan, P., Whetstine, J. R., Cole, P. A., et al.
(2004). Histone demethylation mediated by the nuclear amine oxidase homolog
LSD1. Cell 119 (7), 941953. doi:10.1016/j.cell.2004.12.012
Siegel, R. L., Miller, K. D., Fuchs, H. E., and Jemal, A. (2021). Cancer statistics,
2021. Ca. Cancer J. Clin. 72 (1), 733. doi:10.3322/caac.21654
Siegel, R. L., Miller, K. D., Fuchs, H. E., and Jemal, A. (2022). Cancer statistics,
2022. Ca. Cancer J. Clin. 72, 733. doi:10.3322/caac.21708
Singh, B., and Mitragotri, S. (2020). Harnessing cells to deliver nanoparticle drugs
to treat cancer. Biotechnol. Adv. 42, 107339. doi:10.1016/j.biotechadv.2019.01.006
Sobczak, M., Strachowska, M., Gronkowska, K., and Robaszkiewicz, A. (2022).
Activation of ABCC genes by Cisplatin depends on the CoREST occurrence at their
promoters in A549 and MDA-MB-231 cell lines. Cancers 14 (4), 894. doi:10.3390/
cancers14040894
Song, Y. H., Zhang, H. Q., Yang, X. K., Shi, Y. T., and Yu, B. (2022). Annual review
of lysine-specic demethylase 1 (LSD1/KDM1A) inhibitors in 2021. Eur. J. Med.
Chem. 228, 114042. doi:10.1016/j.ejmech.2021.114042
Sorna, V., Theisen, E. R., Stephens, B., Warner, S. L., Bearss, D. J., Vankayalapati,
H., et al. (2013). High-throughput virtual screening identies novel N-(1-
phenylethylidene)-benzohydrazides as potent, specic, and reversible
LSD1 inhibitors. J. Med. Chem. 56 (23), 94969508. doi:10.1021/jm400870h
Speranzini,V.,Rotili,D.,Ciossani,G.,Pilotto,S.,Marrocco,B.,Forgione,M.,
et al. (2016). Polymyxins and quinazolines are LSD1/KDM1A inhibitors with
unusual structural features. Sci. Adv. 2 (9), e1601017. doi:10.1126/sciadv.
1601017
Strachowska, M., Gronkowska, K., Michlewska, S., and Robaszkiewicz, A. (2021).
CBP/p300 bromodomain inhibitor-I-CBP112 declines transcription of the key ABC
transporters and sensitizes cancer cells to chemotherapy drugs. Cancers 13 (18),
4614. doi:10.3390/cancers13184614
Su, C. Y., Ming, Q. L., Rahman, K., Han, T., and Qin, L. P. (2015). Salvia
miltiorrhiza: Traditional medicinal uses, chemistry, and pharmacology. Chin.
J. Nat. Med. 13 (3), 163182. doi:10.1016/S1875-5364(15)30002-9
Sugino, N., Kawahara, M., Tatsumi, G., Kanai, A., Matsui, H., Yamamoto, R., et al.
(2017). A novel LSD1 inhibitor NCD38 ameliorates MDS-related leukemia with
complex karyotype by attenuating leukemia programs via activating super-
enhancers. Leukemia 31 (11), 23032314. doi:10.1038/leu.2017.59
Sukocheva, O. A., Lukina, E., Friedemann, M., Menschikowski, M., Hagelgans, A.,
and Aliev, G. (2020). The crucial role of epigenetic regulation in breast cancer anti-
estrogen resistance: Current ndings and future perspectives. Semin. Cancer Biol.
82, 3559. doi:10.1016/j.semcancer.2020.12.004
Sun, Y. F., Gao, X. Y., Wu, P. P., Wink, M., Li, J. H., Dian, L. L., et al. (2019).
Jatrorrhizine inhibits mammary carcinoma cells by targeting TNIK mediated Wnt/
β-catenin signalling and epithelial-mesenchymal transition (EMT). Phytomedicine
63, 153015. doi:10.1016/j.phymed.2019.153015
Suzuki, T. (2019). Lysine-specic histone demethylases 1/2 (LSD1/2) and their
inhibitors. Chem. Epigenetics 2019, 197219. doi:10.1007/7355_2019_74
Tan, A. H. W., Tu, W. J., McCuaig, R., Hardy, K., Donovan, T., Tsimbalyuk, S.,
et al. (2019). Lysine-specic histone demethylase 1A regulates macrophage
polarization and checkpoint molecules in the tumor microenvironment of
triple-negative breast cancer. Front. Immunol. 10, 1351. doi:10.3389/mmu.2019.
01351
Thoennissen, N. H., Okelly, J., Lu, D., Iwanski, G. B., La, D. T., Abbassi, S., et al.
(2010). Capsaicin causes cell-cycle arrest and apoptosis in ER-positive and-negative
breast cancer cells by modulating the EGFR/HER-2 pathway. Oncogene 29 (2),
285296. doi:10.1038/onc.2009.335
Tu, W. J., McCuaig, R. D., Tan, A. H. Y., Hardy, K., Seddiki, N., Ali, S., et al.
(2020). Targeting nuclear LSD1 to reprogram cancer cells and reinvigorate
exhausted T cells via a novel LSD1-EOMES switch. Front. Immunol. 11, 1228.
doi:10.3389/mmu.2020.01228
Vallet-Regí, M., Schüth, F., Lozano, D., Colilla, M., and Manzano, M. (2022).
Engineering mesoporous silica nanoparticles for drug delivery: Where are we after
two decades? Chem. Soc. Rev. 51 (13), 53655451. doi:10.1039/d1cs00659b
Vasilatos, S. N., Katz, T. A., Oesterreich, S., Wan, Y., Davidson, N. E., and Huang,
Y. (2013). Crosstalk between lysine-specic demethylase 1 (LSD1) and histone
deacetylases mediates antineoplastic efcacy of HDAC inhibitors in human breast
cancer cells. Carcinogenesis 34 (6), 11961207. doi:10.1093/carcin/bgt033
Verigos, J., Karakaidos, P., Kordias, D., Papoudou-Bai, A., Evangelou, Z., Harissis,
H. V., et al. (2019). The histone demethylase LSD1/ΚDM1A mediates
chemoresistance in breast cancer via regulation of a stem cell program. Cancers
11 (10), 1585. doi:10.3390/cancers11101585
Wang, L., Li, L. Z., Han, Q. X., Wang, X. F., Zhao, D., and Liu, J. Q. (2020).
Identication and biological evaluation of natural product Biochanin A. Bioorg.
Chem. 97, 103674. doi:10.1016/j.bioorg.2020.103674
Wang, T., Zhang, F. L., and Sun, F. L. (2022). ORY-1001, a KDM1A inhibitor,
inhibits proliferation, and promotes apoptosis of triple negative breast cancer cells
by inactivating androgen receptor. Drug Dev. Res. 83 (1), 208216. doi:10.1002/ddr.
21860
Wang, Y., Zhang, H., Chen, Y. P., Sun, Y. M., Yang, F., Yu, W. H., et al. (2009).
LSD1 is a subunit of the NuRD complex and targets the metastasis programs in
breast cancer. Cell 138 (4), 660672. doi:10.1016/j.cell.2009.05.050
Wen, N., Lv, Q., and Du, Z. G. (2020). MicroRNAs involved in drug resistance of
breast cancer by regulating autophagy. J. Zhejiang Univ. Sci. B 21 (9), 690702.
doi:10.1631/jzus.B2000076
Wu, D., Jia, H. Y., Zhang, Z. R., and Li, S. J. (2020). Capsaicin suppresses breast
cancer cell viability by regulating the CDK8/PI3K/Akt/Wnt/β-catenin signaling
pathway. Mol. Med. Rep. 22 (6), 48684876. doi:10.3892/mmr.2020.11585
Wu, J. S., Luo, Y., Deng, D. H., Su, S. Y., Li, S., Xiang, L., et al. (2019). Coptisine
from Coptis chinensis exerts diverse benecial properties: A concise review. J. Cell.
Mol. Med. 23 (12), 79467960. doi:10.1111/jcmm.14725
Wu, K. J., Zhong, H. J., Yang, G. J., Wu, C., Huang, J. M., Li, G. D., et al. (2018).
Small molecule Pin1 inhibitor blocking NF-κB signaling in prostate cancer cells.
Chem. Asian J. 13 (3), 275279. doi:10.1002/asia.201701216
Wu, Y. D., Wang, Y. F., Yang, X. W. H., Kang, T. B., Zhao, Y. X., Wang, C., et al.
(2013). The deubiquitinase USP28 stabilizes LSD1 and confers stem-cell-like traits
to breast cancer cells. Cell Rep. 5 (1), 224236. doi:10.1016/j.celrep.2013.08.030
Wu, Z. Q., Li, X. Y., Hu, C. Y., Ford, M., Kleer, C. G., and Weiss, S. J. (2012).
Canonical Wnt signaling regulates Slug activity and links epithelial-mesenchymal
transition with epigenetic Breast Cancer 1, Early Onset (BRCA1) repression. Proc.
Natl. Acad. Sci. U. S. A. 109 (41), 1665416659. doi:10.1073/pnas.1205822109
Xiao, Y., and Yu, D. H. (2021). Tumor microenvironment as a therapeutic
target in cancer. Pharmacol. Ther. 221, 107753. doi:10.1016/j.pharmthera.2020.
107753
Xu, X., Peng, W. H., Liu, C. Y., Li, S. X., Lei, J. L., Wang, Z., et al. (2019). Flavone-
based natural product agents as new lysine-specic demethylase 1 inhibitors
Frontiers in Pharmacology frontiersin.org17
Yang et al. 10.3389/fphar.2022.989575
exhibiting cytotoxicity against breast cancer cells in vitro.Bioorg. Med. Chem. 27 (2),
370374. doi:10.1016/j.bmc.2018.12.013
Yakulov, T., Raggioli, A., Franz, H., and Kemler, R. (2013). Wnt3a-dependent and
-independent protein interaction networks of chromatin-bound β-catenin in mouse
embryonic stem cells. Mol. Cell. Proteomics 12 (7), 19801994. doi:10.1074/mcp.
M112.026914
Yang, C., Li, D., Zang, S. H., Zhang, L., Zhong, Z. F., and Zhou, Y. T. (2022).
Mechanisms of carcinogenic activity triggered by lysine-specic demethylase 1A.
Front. Pharmacol. 2804, 955218. doi:10.3389/fphar.2022.955218
Yang, C., Wang, W. H., Liang, J. X., Li, G. D., Vellaisamy, K., Wong, C. Y., et al.
(2017). A rhodium (III)-based inhibitor of lysine-specic histone demethylase 1 as
an epigenetic modulator in prostate cancer cells. J. Med. Chem. 60 (6), 25972603.
doi:10.1021/acs.jmedchem.7b00133
Yang, G. J., Wang, W. H., Mok, S. W. F., Wu, C., Law, B. Y. K., Miao, X. M., et al.
(2018b). Selective Inhibition of lysine-specic demethylase 5A (KDM5A) using a
rhodium(III) complex for triple-negative breast cancer therapy. Angew. Chem. Int.
Ed. Engl. 57 (40), 1309113095. doi:10.1002/anie.201807305
Yang, G. J., Wu, J., Leung, C. H., Ma, D. L., and Chen, J. (2021c). A review on the
emerging roles of pyruvate kinase M2 in anti-leukemia therapy. Int. J. Biol.
Macromol. 193, 14991506. doi:10.1016/j.ijbiomac.2021.10.213
Yang, G. J., Ko, C. N., Zhong, H. J., Leung, C. H., and Ma, D. L. (2019). Structure-
based discovery of a Selective KDM5A inhibitor that exhibits anti-cancer activity via
inducing cell cycle arrest and senescence in breast cancer cell lines. Cancers 11 (1),
92. doi:10.3390/cancers11010092
Yang, G. J., Lei, P. M., Wong, S. Y., Ma, D. L., and Leung, C. H. (2018a).
Pharmacological inhibition of LSD1 for cancer treatment. Molecules 23 (12), 3194.
doi:10.3390/molecules23123194
Yang, G. J., Song, Y. Q., Wang, W. H., Han, Q. B., Ma, D. L., and Leung, C. H.
(2021b). An optimized BRD4 inhibitor effectively eliminates NF-κB-driven triple-
negative breast cancer cells. Bioorg. Chem. 114, 105158. doi:10.1016/j.bioorg.2021.
105158
Yang, G. J., Wang, W. H., Lei, P. M., Leung, C. H., and Ma, D. L. (2020). A 7-
methoxybicoumarin derivative selectively inhibits BRD4 BD2 for anti-melanoma
therapy. Int. J. Biol. Macromol. 164, 32043220. doi:10.1016/j.ijbiomac.2020.08.194
Yang, G. J., Wu, J., Miao, L., Zhu, M. H., Zhou, Q. J., Lu, X. J., et al. (2021d).
Pharmacological inhibition of KDM5A for cancer treatment. Eur. J. Med. Chem.
226, 113855. doi:10.1016/j.ejmech.2021.113855
Yang, G. J., Zhong, H. J., Ko, C. N., Wong, S. Y., Vellaisamy, K., Ye, M., et al.
(2018c). Identication of a rhodium(iii) complex as a Wee1 inhibitor against TP53-
mutated triple-negative breast cancer cells. Chem. Commun. 54 (20), 24632466.
doi:10.1039/c7cc09384e
Yang, G. J., Zhu, M. H., Lu, X. J., Liu, Y. J., Lu, J. F., Leung, C. H., et al. (2021a). The
emerging role of KDM5A in human cancer. J. Hematol. Oncol. 14 (1), 30. doi:10.
1186/s13045-021-01041-1
Yang, M. J., Culhane, J. C., Szewczuk, L. M., Jalili, P., Ball, H. L., Machius, M., et al.
(2007). Structural basis for the inhibition of the LSD1 histone demethylase by the
antidepressant trans-2-phenylcyclopropylamine. Biochemistry 46, 80588065.
doi:10.1021/bi700664y
Yang, Y., Huang, W., Qiu, R. F., Liu, R. Q., Zeng, Y., Gao, J., et al. (2018d).
LSD1 coordinates with the SIN3A/HDAC complex and maintains sensitivity to
chemotherapy in breast cancer. J. Mol. Cell Biol. 10 (4), 285301. doi:10.1093/jmcb/
mjy021
Yatim, A., Benne, C., Sobhian, B., Laurent-Chabalier, S., Deas, O., Judde, J. D.,
et al. (2012). NOTCH1 nuclear interactome reveals key regulators of its
transcriptional activity and oncogenic function. Mol. Cell 48 (3), 445458.
doi:10.1016/j.molcel.2012.08.022
Yi, L., Cui, Y., Xu, Q. F., and Jiang, Y. J. (2016). Stabilization of LSD1 by
deubiquitinating enzyme USP7 promotes glioblastoma cell tumorigenesis and
metastasis through suppression of the p53 signaling pathway. Oncol. Rep. 36
(5), 29352945. doi:10.3892/or.2016.5099
Yoneyama, M., Tochio, N., Umehara, T., Koshiba, S., Inoue, M., Yabuki, T., et al.
(2007). St, ructural and functional differences of SWIRM domain subtypes. J. Mol.
Biol. 369 (1), 222238. doi:10.1016/j.jmb.2007.03.027
Zhang, X. M., Lu, F., Wang, J., Yin, F., Xu, Z. S., Qi, D. H., et al. (2013). Pluripotent
stem cell protein Sox2 confers sensitivity to LSD1 inhibition in cancer cells. Cell Rep.
5 (2), 445457. doi:10.1016/j.celrep.2013.09.018
Zhao, L. J., Li, Y. Y., Zhang, Y. T., Fan, Q. Q., Ren, H. M., Zhang, C., et al. (2021).
Lysine demethylase LSD1 delivered via small extracellular vesicles promotes gastric
cancer cell stemness. EMBO Rep. 22, e50922. doi:10.15252/embr.202050922
Zheng, L. H., Zhang, Y., Liu, G. J., Cheng, S., Zhang, G., An, C., et al. (2020).
Tanshinone I regulates autophagic signaling via the activation of AMP-activated
protein kinase in cancer cells. Anticancer. Drugs 31 (6), 601608. doi:10.1097/CAD.
0000000000000908
Zheng,Y.,Zeng,Y.,Qiu,R.F.,Liu,R.Q.,Huang,W.,Hou,Y.Q.,etal.(2018).The
homeotic protein SIX3 suppresses carcinogenesis and metastasis through recruiting the
LSD1/NuRD(MTA3) complex. Theranostics 8(4),972989. doi:10.7150/thno.22328
Zhou, A. D., Lin, K. Y., Zhang, S. C., Chen, Y. H., Zhang, N., Xue, J. F., et al.
(2016). Nuclear GSK3βpromotes tumorigenesis by phosphorylating KDM1A and
inducing its deubiquitylation by USP22. Nat. Cell Biol. 18 (9), 954966. doi:10.1038/
ncb3396
Zhou, C., Kang, D., Xu, Y. G., Zhang, L. Y., and Zha, X. M. (2015). Identication
of novel selective lysine-specic demethylase 1 (LSD1) inhibitors using a
pharmacophore-based virtual screening combined with docking. Chem. Biol.
Drug Des. 85 (6), 659671. doi:10.1111/cbdd.12461
Zhou, J., Su, C. M., Chen, H. A., Du, S., Li, C. W., Wu, H., et al. (2020).
Cryptanshinone inhibits the glycolysis and inhibits cell migration through PKM2/
β-catenin axis in breast cancer. Onco. Targets. Ther. 13, 86298639. doi:10.2147/
OTT.S239134
Zhou, M., Venkata, P. P., Viswanadhapalli, S., Palacios, B., Alejo, S., Chen, Y. H.,
et al. (2021a). KDM1A inhibition is effective in reducing stemness and treating
triple negative breast cancer. Breast Cancer Res. Treat. 185 (2), 343357. doi:10.
1007/s10549-020-05963-1
Zhou, Z. L., Van der Jeught, K., Fang, Y. Z., Yu, T., Li, Y. J., Ao, Z., et al. (2021b).
An organoid-based screen for epigenetic inhibitors that stimulate antigen
presentation and potentiate T-cell-mediated cytotoxicity. Nat. Biomed. Eng. 5
(11), 13201335. doi:10.1038/s41551-021-00805-x
Zhu, Q. S., Huang, Y., Marton, L. J., Woster, P. M., Davidson, N. E., and Jr Casero,
R. A. (2012). Polyamine analogs modulate gene expression by inhibiting lysine-
specic demethylase 1 (LSD1) and altering chromatin structure in human breast
cancer cells. Amino Acids 42, 887898. doi:10.1007/s00726-011-1004-1
Zylla, J. L., Hoffman, M. M., Plesselova, S., Bhattacharya, S., Calar, K., Afeworki,
Y., et al. (2022). Reduction of metastasis via epigenetic modulation in a murine
model of metastatic triple negative breast cancer (TNBC). Cancers 14 (7), 1753.
doi:10.3390/cancers14071753
Frontiers in Pharmacology frontiersin.org18
Yang et al. 10.3389/fphar.2022.989575
... 26,27) Therefore, LSD1 inhibitors are of interest both as tools for studying LSD1 functions and as potential therapeutic agents. [28][29][30][31][32][33][34] Thus far, many LSD1 inhibitors have been reported. [28][29][30][31][32][33][34] However, most of these inhibitors are small molecules, and peptide-based inhibitors are relatively scarce. ...
... [28][29][30][31][32][33][34] Thus far, many LSD1 inhibitors have been reported. [28][29][30][31][32][33][34] However, most of these inhibitors are small molecules, and peptide-based inhibitors are relatively scarce. 12,[35][36][37][38][39][40][41][42][43][44][45][46] A series of the reported peptide-based LSD1 inhibitors were designed based on SNAIL1, 17,[43][44][45][46][47] a member of the SNAIL/SCRATCH family of transcription factors. ...
Article
Peptides have recently garnered attention as middle-molecular-weight drugs with the characteristics of small molecules and macromolecules. Lysine-specific demethylase 1 (LSD1) is a potential therapeutic target for lung cancer, neuroblastoma, and leukemia, and some peptide-based LSD1 inhibitors designed based on the N-terminus of SNAIL1, a member of the SNAIL/SCRATCH family of transcription factors, have been reported. The N-terminus of SNAIL1 peptide acts as a cap of the catalytic site of LSD1, inhibiting interactions with LSD1. However, the structure–activity relationship (SAR) of these inhibitors is not yet fully understood. Therefore, in the present study, we aimed to uncover the SAR and to identify novel SNAIL1 peptide-based LSD1 inhibitors. We synthesized peptide inhibitor candidates based on truncating the N-terminus of SNAIL1 or substituting its amino acid residues. In the truncation study, we found that SNAIL1 1–16 (2), which was composed of 16 residues, strongly inhibited LSD1. Furthermore, we investigated the SAR at residues-3 and -5 from the N-terminus and found that peptides 2j and 2k, in which leucine 5 of the parent peptide is substituted with unnatural amino acids, cyclohexylalanine and norleucine, respectively, strongly inhibited LSD1. This result suggests that the hydrophobic interaction between the inhibitor peptides and LSD1 affects the LSD1-inhibitory activity. We believe that this SAR information provides a basis for the development of more potent LSD1 inhibitors. Fullsize Image
... KDMs consist of two classes of proteins of over 30 members, including the (i) flavin-dependent monoamine oxidases (LSD), and (ii) the Fe(II)-and α-ketoglutarate (2OG)-dependent oxygenases that possess a conserved catalytic Jumonji C domain (JmjC) [16]. Many KDMs members, such as LSD1, KDM4A/B/C/D, KDM6A, and KDM7A, are associated with BC progress in a subtype/ content-dependent manner, and inhibitors targeting them have been extensively developed [16][17][18][19][20][21]. KDM5, also known as JARID1 (jumonji domain ARID-containing protein), belongs to the JmjC family and comprises the four members KDM5A/B/C/D, which are encoded in the human genome at loci 12p13.33, ...
Article
Full-text available
Breast cancer (BC) is the most frequent malignant cancer diagnosis and is a primary factor for cancer deaths in women. The clinical subtypes of BC include estrogen receptor (ER) positive, progesterone receptor (PR) positive, human epidermal growth factor receptor 2 (HER2) positive, and triple-negative BC (TNBC). Based on the stages and subtypes of BC, various treatment methods are available with variations in the rates of progression-free disease and overall survival of patients. However, the treatment of BC still faces challenges, particularly in terms of drug resistance and recurrence. The study of epigenetics has provided new ideas for treating BC. Targeting aberrant epigenetic factors with inhibitors represents a promising anticancer strategy. The KDM5 family includes four members, KDM5A, KDM5B, KDM5C, and KDMD, all of which are Jumonji C domain-containing histone H3K4me2/3 demethylases. KDM5 proteins have been extensively studied in BC, where they are involved in suppressing or promoting BC depending on their specific upstream and downstream pathways. Several KDM5 inhibitors have shown potent BC inhibitory activity in vitro and in vivo, but challenges still exist in developing KDM5 inhibitors. In this review, we introduce the subtypes of BC and their current therapeutic options, summarize KDM5 family context-specific functions in the pathobiology of BC, and discuss the outlook and pitfalls of KDM5 inhibitors in this disease.
... Breast cancer, characterized by the uncontrolled proliferation of mammary epithelial cells, has a high incidence rate among women [82,83]. The oncogenic role of the insulin receptor (IR) has been observed in diverse cancer types. ...
Article
Full-text available
RNA-binding proteins (RBPs) are kinds of proteins with either singular or multiple RNA-binding domains (RBDs), and they can assembly into ribonucleic acid-protein complexes, which mediate transportation, editing, splicing, stabilization, translational efficiency, or epigenetic modifications of their binding RNA partners, and thereby modulate various physiological and pathological processes. CUG-BP, Elav-like family 1 (CELF1) is a member of the CELF family of RBPs with high affinity to the GU-rich elements in mRNA, and thus exerting control over critical processes including mRNA splicing, translation, and decay. Mounting studies support that CELF1 is correlated with occurrence, genesis and development and represents a potential therapeutical target for these malignant diseases. Herein, we present the structure and function of CELF1, outline its role and regulatory mechanisms in varieties of homeostasis and diseases, summarize the identified CELF1 regulators and their structure-activity relationships, and prospect the current challenges and their solutions during studies on CELF1 functions and corresponding drug discovery, which will facilitate the establishment of a targeted regulatory network for CELF1 in diseases and advance CELF1 as a potential drug target for disease therapy.
... Breast cancer, characterized by the uncontrolled proliferation of mammary epithelial cells, has a high incidence rate among women [82,83]. The oncogenic role of the insulin receptor (IR) has been observed in diverse cancer types. ...
Article
Full-text available
RNA-binding proteins (RBPs) are kinds of proteins with either singular or multiple RNA-binding domains (RBDs), and they can assembly into ribonucleic acid–protein complexes, which mediate transportation, editing, splicing, stabilization, translational efficiency, or epigenetic modifications of their binding RNA partners, and thereby modulate various physiological and pathological processes. CUG-BP, Elav-like family 1 (CELF1) is a member of the CELF family of RBPs with high affinity to the GU-rich elements in mRNA, and thus exerting control over critical processes including mRNA splicing, translation, and decay. Mounting studies support that CELF1 is correlated with occurrence, genesis and development and represents a potential therapeutical target for these malignant diseases. Herein, we present the structure and function of CELF1, outline its role and regulatory mechanisms in varieties of homeostasis and diseases, summarize the identified CELF1 regulators and their structure–activity relationships, and prospect the current challenges and their solutions during studies on CELF1 functions and corresponding drug discovery, which will facilitate the establishment of a targeted regulatory network for CELF1 in diseases and advance CELF1 as a potential drug target for disease therapy. Graphical Abstract
... 297 Moreover, Chen et al. reported the competence of LSD1 to reduce the expression of tumor suppressor genes by interacting with β-catenin. 298,299 In addition, the down-regulation of KDM5B in TNBC inhibits long non-coding RNA MALAT1, thereby inhibiting cancer cell invasion. 300,301 One thing we should pay attention to in the above research is that dimethylation and trimethylation at the H3K4 site have opposite clinical significance for the prognosis of breast cancer. ...
Article
Full-text available
Histones are DNA-binding basic proteins found in chromosomes. After the histone translation, its amino tail undergoes various modifications, such as methylation, acetylation, phosphorylation, ubiquitination, malonylation, propionylation, butyrylation, crotonylation, and lactylation, which together constitute the "histone code." The relationship between their combination and biological function can be used as an important epigenetic marker. Methylation and demethylation of the same histone residue, acetylation and deacetylation, phosphorylation and dephosphorylation, and even methylation and acetylation between different histone residues cooperate or antagonize with each other, forming a complex network. Histone-modifying enzymes, which cause numerous histone codes, have become a hot topic in the research on cancer therapeutic targets. Therefore, a thorough understanding of the role of histone post-translational modifications (PTMs) in cell life activities is very important for preventing and treating human diseases. In this review, several most thoroughly studied and newly discovered histone PTMs are introduced. Furthermore, we focus on the histone-modifying enzymes with carcinogenic potential, their abnormal modification sites in various tumors, and multiple essential molecular regulation mechanism. Finally, we summarize the missing areas of the current research and point out the direction of future research. We hope to provide a comprehensive understanding and promote further research in this field.
Article
Although certain members of the Ubiquitin-specific peptidases (USPs) have been recognized as promising therapeutic targets for various diseases, research progress regarding USP21 has been relatively sluggish in its early stages. USP21 is a crucial member of the USPs subfamily, involved in diverse cellular processes such as apoptosis, DNA repair, and signal transduction. Research findings from the past decade demonstrate that USP21 mediates the deubiquitination of multiple well-known target proteins associated with critical cellular processes relevant to both disease and homeostasis, particularly in various cancers. This review comprehensively summarizes the structure and biological functions of USP21 with an emphasis on its role in tumorigenesis, and elucidates the advances on the discovery of tens of small-molecule inhibitors targeting USP21, which suggests that targeting USP21 may represent a potential strategy for cancer therapy.
Article
Full-text available
Emerging role of Jumonji domain-containing protein D3 (JMJD3) in inflammatory diseases, Journal of Pharmaceutical Analysis, https://doi. Abstract 3 Jumonji domain-containing protein D3 (JMJD3) is a 2-oxoglutarate-dependent 4 dioxygenase that specifically removes transcriptional repression marks di-and tri-5 methylated groups from lysine 27 on histone 3 (H3K27me2/3). The erasure of these 6 marks leads to the activation of some associated genes, thereby influencing various 7 biological processes, such as development, differentiation, and immune response. 8 However, comprehensive descriptions regarding the relationship between JMJD3 and 9 inflammation are lacking. Here, we provide a comprehensive overview of JMJD3, 10 including its structure, functions, and involvement in inflammatory pathways. In addition, 11 we summarize the evidence supporting JMJD3's role in several inflammatory diseases, as 12 well as the potential therapeutic applications of JMJD3 inhibitors. Additionally, we also 13 discuss the challenges and opportunities associated with investigating the functions of 14 JMJD3 and developing targeted inhibitors and propose feasible solutions to provide 15 valuable insights into the functional exploration and discovery of potential drugs 16 targeting JMJD3 for inflammatory diseases.
Article
Histone demethylation is a kind of epigenetic modification mediated by a variety of enzymes and participates in regulating multiple physiological and pathological events. Lysine-specific demethylase 7A is a kind of α-ketoglutarate- and Fe(II)-dependent demethylase belonging to the PHF2/8 subfamily of the JmjC demethylases. KDM7A is mainly localized in the nucleus and contributes to transcriptional activation via removing mono- and di-methyl groups from the lysine residues 9 and 27 of Histone H3. Mounting studies support that KDM7A is not only necessary for normal embryonic, neural, and skeletal development, and but also associated with cancer, inflammation, osteoporosis, and other diseases. Herein, the structure of KDM7A is described by comparing the similarities and differences of its amino acid sequences of KDM7A and other Histone demethylases; the functions of KDM7A in homeostasis and dyshomeostasis are summarized via documenting its content and related signaling; the currently known KDM7A-specific inhibitors and their structural relationship are listed based on their structure optimization and pharmacological activities; and the challenges and opportunities in exploring functions and developing targeted agents of KDM7A are also prospected via presenting encountered problems and potential solutions, which will provide an insight in functional exploration and drug discovery for KDM7A-related diseases.
Article
Full-text available
Epigenetics has emerged as a prime focus area in the field of cancer research. Lysine-specific demethylase 1A (LSD1), the first discovered histone demethylase, is mainly responsible for catalysing demethylation of histone 3 lysine 4 (H3K4) and H3K9 to activate or inhibit gene transcription. LSD1 is abnormally expressed in various cancers and participates in cancer proliferation, apoptosis, metastasis, invasion, drug resistance and other processes by interacting with regulatory factors. Therefore, it may serve as a potential therapeutic target for cancer. This review summarises the major oncogenic mechanisms mediated by LSD1 and provides a reference for developing novel and efficient anticancer strategies targeting LSD1.
Article
Full-text available
The present review details a chronological description of the events that took place during the development of mesoporous materials, their different synthetic routes and their use as drug delivery systems. The outstanding textural properties of these materials quickly inspired their translation to the nanoscale dimension leading to mesoporous silica nanoparticles (MSNs). The different aspects of introducing pharmaceutical agents into the pores of these nanocarriers, together with their possible biodistribution and clearance routes, would be described here. The development of smart nanocarriers that are able to release a high local concentration of the therapeutic cargo on-demand after the application of certain stimuli would be reviewed here, together with their ability to deliver the therapeutic cargo to precise locations in the body. The huge progress in the design and development of MSNs for biomedical applications, including the potential treatment of different diseases, during the last 20 years will be collated here, together with the required work that still needs to be done to achieve the clinical translation of these materials. This review was conceived to stand out from past reports since it aims to tell the story of the development of mesoporous materials and their use as drug delivery systems by some of the story makers, who could be considered to be among the pioneers in this area.
Article
Full-text available
Enhancer of zeste homolog 2 (EZH2) is activated in breast cancer, particularly in triple-negative breast cancer (TNBC), and is critical for cell invasion. It interacts with embryonic ectoderm development (EED) in maintaining cancer stem cells (CSC) and epithelial-mesenchymal transition (EMT) properties, hence promoting CSC metastasis. Because the association of EZH2 with EED promotes the catalytic activity of EZH2, inhibiting the EED-EZH2 interaction is a potential therapeutic strategy for treating EZH2-dependent cancer. Although several EED-EZH2 protein-protein interaction (PPI) inhibitors have been developed, few target EED. Here, we identified that a cytisine derivative compound ( 1 ) potently binds EED, thus blocking the EED-EZH2 PPI. Compound 1 was found to inhibit cell proliferation and suppress the growth of 3D tumor spheres of TNBC cells. Moreover, by reversing EMT and decreasing the ratio of CSCs, the compound inhibited TNBC metastasis and invasion ability. Therefore, targeting EED to disrupt the EED-EZH2 PPI may provide a new approach for treating TNBC metastasis. To our knowledge, compound 1 is the first cytisine-based EED-EZH2 PPI inhibitor preventing metastasis in TNBC cells. This study may provide a new avenue for the development of more efficacious EED-EZH2 PPI inhibitors in TNBC treatment.
Article
Full-text available
Background: Metastatic breast cancer to the lungs is a serious, life-threatening complication that is difficult to cure. Circulating tumor cells play a key role in the metastatic spread of breast cancer to the lungs via the lymphatic or circulatory system. Palmatine is a protoberberine alkaloid, identified as an active component of traditional African herbal preparations. Palmatine has antimetastatic and antiproliferative effects. The inhibitory activity of palmatine on the metastatic colonization of triple negative breast cancer cells in the lungs was investigated in this study. Methods: 4T1 triple breast cancer cells were transplanted synergically to the thoracic duct of the female balb/c mice via the lymphatic system. Palmatine 1, 5 and 10 mg/kg were administered for 28 days. The lungs were analyzed for levels of arterial blood gas, histological damage, immunohistochemical expression of the metastasis-associated protein 1 (MTA1) and tumor suppressor p53 (p53). Results: Administering palmatine 1–10 mg/kg dose dependently improved hypoxemia, ameliorated metastasis associated lung injury; histology score of 3.33 ± 0.33, 1.67 ± 0.33, 1.33 ± 0.33, decreased lung MTA1 (2.19 ± 0.12, 1.83 ± 0.04, 1.84 ± 0.05) and increased p53 expression (1.99 ± 0.06, 2.27 ± 0.12, 2.34 ± 0.12) respectively. Conclusion: Palmatine preserved lung morphology and demonstrated therapeutic potential in aiding the treatment of lung metastasis.
Article
Full-text available
Simple Summary We investigate the use of the small molecule epigenetic modulator 4SC-202 as a potential cancer therapeutic or adjuvant for Triple Negative Breast Cancer (TNBC). Epigenetic modulation involves alteration of the cellular phenotype without altering the genotype. Here, we investigate how 4SC-202 affects tumor growth, the ability of tumor cells to migrate, and the growth of tumors in vivo. This study demonstrates that 4SC-202 kills tumor cells, reduces their ability to migrate, and decreases metastasis of the tumor cells and tumor burden in a highly metastatic murine model of TNBC. These preclinical studies provide critical information on the potential efficacy of 4SC-202 as a potential therapeutic or adjuvant for TNBC. Abstract This study investigates the effects of a dual selective Class I histone deacetylase (HDAC)/lysine-specific histone demethylase 1A (LSD1) inhibitor known as 4SC-202 (Domatinostat) on tumor growth and metastasis in a highly metastatic murine model of Triple Negative Breast Cancer (TNBC). 4SC-202 is cytotoxic and cytostatic to the TNBC murine cell line 4T1 and the human TNBC cell line MDA-MB-231; the drug does not kill the normal breast epithelial cell line MCF10A. Furthermore, 4SC-202 reduces cancer cell migration. In vivo studies conducted in the syngeneic 4T1 model, which closely mimics human TNBC in terms of sites of metastasis, reveal reduced tumor burden and lung metastasis. The mechanism of action of 4SC-202 may involve effects on cancer stem cells (CSC) which can self-renew and form metastatic lesions. Approximately 5% of the total 4T1 cell population grown in three-dimensional scaffolds had a distinct CD44high/CD24low CSC profile which decreased after treatment. Bulk transcriptome (RNA) sequencing analyses of 4T1 tumors reveal changes in metastasis-related pathways in 4SC-202-treated tumors, including changes to expression levels of genes implicated in cell migration and cell motility. In summary, 4SC-202 treatment of tumors from a highly metastatic murine model of TNBC reduces metastasis and warrants further preclinical studies.
Article
Full-text available
Epigenetic mechanisms are known to play a key role in cancer progression. Specifically, histone methylation involves reversible post-translational modification of histones that govern chromatin structure remodelling, genomic imprinting, gene expression, DNA damage repair, and meiotic crossover recombination, among other chromatin-based activities. Demethylases are enzymes that catalyse the demethylation of their substrate using a flavin adenine dinucleotide-dependent amine oxidation process. Lysine-specific demethylase 1 (LSD1) and its homolog, lysine-specific demethylase 2 (LSD2), are overexpressed in a variety of human cancer types and, thus, regulate tumour progression. In this review, we focus on the literature from the last 5 years concerning the role of LSD1 and LSD2 in the main gastrointestinal cancers (i.e., gastric cancer, liver cancer, pancreatic cancer, and colorectal cancer).
Article
Full-text available
Background Histone lysine-specific demethylase 1 (LSD1) expression has been shown to be significantly elevated in gastric cancer (GC) and may be associated with the proliferation and metastasis of GC. It has been reported that LSD1 repressed tumor immunity through programmed cell death 1 ligand 1 (PD-L1) in melanoma and breast cancer. The role of LSD1 in the immune microenvironment of GC is unknown. Methods Expression LSD1 and PD-L1 in GC patients was analyzed by immunohistochemical (IHC) and Western blotting. Exosomes were isolated from the culture medium of GC cells using an ultracentrifugation method and characterized by transmission electronic microscopy (TEM), nanoparticle tracking analysis (NTA), sucrose gradient centrifugation, and Western blotting. The role of exosomal PD-L1 in T-cell dysfunction was assessed by flow cytometry, T-cell killing and enzyme-linked immunosorbent assay (ELISA). Results Through in vivo exploration, mouse forestomach carcinoma (MFC) cells with LSD1 knockout (KO) showed significantly slow growth in 615 mice than T-cell-deficient BALB/c nude mice. Meanwhile, in GC specimens, expression of LSD1 was negatively correlated with that of CD8 and positively correlated with that of PD-L1. Further study showed that LSD1 inhibited the response of T cells in the microenvironment of GC by inducing the accumulation of PD-L1 in exosomes, while the membrane PD-L1 stayed constant in GC cells. Using exosomes as vehicles, LSD1 also obstructed T-cell response of other cancer cells while LSD1 deletion rescued T-cell function. It was found that while relying on the existence of LSD1 in donor cells, exosomes can regulate MFC cells proliferation with distinct roles depending on exosomal PD-L1-mediated T-cell immunity in vivo. Conclusion LSD1 deletion decreases exosomal PD-L1 and restores T-cell response in GC; this finding indicates a new mechanism with which LSD1 may regulate cancer immunity in GC and provides a new target for immunotherapy against GC.
Article
Berberine alkaloids belong to the class of isoquinoline alkaloids that have been shown to possess anticancer potential, berberine exhibits inhibitory effects on breast cancer development. However, the exact mechanisms of action for anti-breast carcinoma of the alkaloids, including epiberberine, berberrubine and dihydroberberine are still unclear. MTT assay, colony formation, wound healing and transwell invasion assays detected these alkaloids suppressed proliferation, migration and invasion of breast cancer cells. Hoechst and Annexin V-FITC/propidium iodide staining were used to analyze the apoptosis of breast cancer cells. Western blotting investigated the changes noted in the expression levels of the key proteins involved in the Wnt signaling pathway and epithelial to mesenchymal transition (EMT). The results showed that inhibited the proliferation of breast cancer cells. Berberine alkaloids inhibited the cell cycle at G2/M phase in MCF-7 cells, but in MDA-MB-231 cells berberine alkaloids arrested the cell cycle in G0/G1 and G2/M phases. By decreasing β-catenin expression, increasing GSK-3β expression and decreasing N-cadherin expression, increasing E-cadherin expression, which proved that epiberberine, berberrubine and dihydroberberine inhibited of metastasis of breast cancer cells through Wnt signaling pathway and reversed EMT except berberine. Furthermore, berberine alkaloids exert their anti-breast cancer effects through the synergistic action of intrinsic and extrinsic pathways of apoptosis. These findings highlight the different effects of different berberine alkaloids on breast cancer cells and confirm that berberine alkaloids may be potentially used in the treatment of breast cancer.
Article
Metabolic reprogramming is observed across all cancer types. Indeed, the success of many classic chemotherapies stems from their targeting of cancer metabolism. Contemporary research in this area has refined our understanding of tumor-specific metabolic mechanisms and has revealed strategies for exploiting these vulnerabilities selectively. Based on this growing understanding, new small-molecule tools and drugs have been developed to study and target tumor metabolism. Here, we highlight allosteric modulation of metabolic enzymes as an attractive mechanism of action for small molecules that target metabolic enzymes. We then discuss the mechanistic insights garnered from their application in cancer studies and highlight the achievements of this approach in targeting cancer metabolism. Finally, we discuss technological advances in drug discovery for allosteric modulators of enzyme activity.
Article
LSD1 is a histone lysine demethylase proposed as therapeutic target in cancer. Chemical modifications applied at C2, C4 and/or C7 positions of the quinazoline core of the previously reported dual LSD1/G9a inhibitor 1 led to a series of non-covalent, highly active, and selective LSD1 inhibitors (2–4 and 6–30) and to the dual LSD1/G9a inhibitor 5 that was more potent than 1 against LSD1. In THP-1 and MV4-11 leukemic cells, the most potent compounds (7, 8, and 29) showed antiproliferative effects at sub-micromolar level without significant toxicity at 1 μM in non-cancer AHH-1 cells. In MV4-11 cells, the new derivatives increased the levels of the LSD1 histone mark H3K4me2 and induced the re-expression of the CD86 gene silenced by LSD1, thereby confirming the inhibition of LSD1 at cellular level. In breast MDA-MB-231 as well as in rhabdomyosarcoma RD and RH30 cells, taken as examples of solid tumors, the same compounds displayed cell growth arrest in the same IC50 range, highlighting a crucial anticancer role for LSD1 inhibition and suggesting no added value for the simultaneous G9a inhibition in these tumor cell lines.