ArticlePDF AvailableLiterature Review

Epigenetics in breast cancer therapies - New strategies and future nanomedicine perspectives

MDPI
Cancers
Authors:

Abstract and Figures

Epigenetic dysregulation has been recognized as a critical factor contributing to the development of resistance against standard chemotherapy and to breast cancer progression via epithelial-to-mesenchymal transition. Although the efficacy of the first-generation epigenetic drugs (epi-drugs) in solid tumor management has been disappointing, there is an increasing body of evidence showing that epigenome modulation, in synergy with other therapeutic approaches, could play an important role in cancer treatment, reversing acquired therapy resistance. However, the epigenetic therapy of solid malignancies is not straightforward. The emergence of nanotechnologies applied to medicine has brought new opportunities to advance the targeted delivery of epi-drugs while improving their stability and solubility, and minimizing off-target effects. Furthermore, the omics technologies, as powerful molecular epidemiology screening tools, enable new diagnostic and prognostic epigenetic biomarker identification, allowing for patient stratification and tailored management. In combination with new-generation epi-drugs, nanomedicine can help to overcome low therapeutic efficacy in treatment-resistant tumors. This review provides an overview of ongoing clinical trials focusing on combination therapies employing epi-drugs for breast cancer treatment and summarizes the latest nano-based targeted delivery approaches for epi-drugs. Moreover, it highlights the current limitations and obstacles associated with applying these experimental strategies in the clinics.
This content is subject to copyright.
cancers
Review
Epigenetics in Breast Cancer Therapy—New
Strategies and Future Nanomedicine Perspectives
Verona Buocikova 1, Ivan Rios-Mondragon 2, Eleftherios Pilalis 3,4 ,
Aristotelis Chatziioannou 3,4 , Svetlana Miklikova 1, Michal Mego 5, Karlis Pajuste 6,
Martins Rucins 6, Naouale El Yamani 7, Eleonora Marta Longhin 7, Arkadij Sobolev 6,
Muriel Freixanet 8, Victor Puntes 8,9 ,10 , Aiva Plotniece 6, Maria Dusinska 7,
Mihaela Roxana Cimpan 2, Alena Gabelova 1, and Bozena Smolkova 1, *,
1Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences,
Dubravska Cesta 9, 845 05 Bratislava, Slovakia; verona.buocikova@savba.sk (V.B.);
svetlana.miklikova@savba.sk (S.M.); alena.gabelova@savba.sk (A.G.)
2Department of Clinical Dentistry, University of Bergen, Aarstadveien 19, 5009 Bergen, Norway;
ivan.rios-mondragon@uib.no (I.R.-M.); Mihaela.Cimpan@uib.no (M.R.C.)
3e-NIOS Applications Private Company, Alexandrou Pantou 25, 17671 Kallithea, Greece;
epilalis@e-nios.com (E.P.); achatzi@e-nios.com (A.C.)
4
Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
52nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute,
Klenova 1, 833 10 Bratislava, Slovakia; michal.mego@nou.sk
6Latvian Institute of Organic Synthesis, Aizkraukles str. 21, LV-1006 Riga, Latvia; kpajuste@osi.lv (K.P.);
rucins@osi.lv (M.R.); arkady@osi.lv (A.S.); aiva@osi.lv (A.P.)
7Health Eects Laboratory, NILU-Norwegian Institute for Air Research, 2007 Kjeller, Norway;
ney@nilu.no (N.E.Y.); eml@nilu.no (E.M.L.); mdu@nilu.no (M.D.)
8Vall d Hebron, Institut de Recerca (VHIR), 08035 Barcelona, Spain; muriel.freixanet@vhir.org (M.F.);
victor.puntes@vhir.org (V.P.)
9Institut Catalàde Nanosciència i Nanotecnologia (ICN2), Bellaterra, 08193 Barcelona, Spain
10 InstitucióCatalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
*Correspondence: bozena.smolkova@savba.sk
Alena Gabelova and Bozena Smolkova share the senior authorship.
Received: 31 October 2020; Accepted: 30 November 2020; Published: 3 December 2020


Simple Summary:
Despite advances in cancer treatment, dicult-to-treat tumor subtypes remain
a challenge. New multidisciplinary approaches can help overcome current obstacles posed by
tumor heterogeneity, activation and enrichment of cancer stem cells, and acquired drug resistance
development. Epigenome modulation, currently unsuccessful in solid tumors due to epigenetic drug
instability, toxicity, and o-target eects, might be enabled by implementing nano-based delivery
strategies aiming to improve breast cancer patient outcomes.
Abstract:
Epigenetic dysregulation has been recognized as a critical factor contributing to
the development of resistance against standard chemotherapy and to breast cancer progression via
epithelial-to-mesenchymal transition. Although the ecacy of the first-generation epigenetic drugs
(epi-drugs) in solid tumor management has been disappointing, there is an increasing body of evidence
showing that epigenome modulation, in synergy with other therapeutic approaches, could play
an important role in cancer treatment, reversing acquired therapy resistance.
However, the epigenetic
therapy of solid malignancies is not straightforward. The emergence of nanotechnologies applied to
medicine has brought new opportunities to advance the targeted delivery of epi-drugs while improving
their stability and solubility, and minimizing o-target eects. Furthermore, the omics technologies,
as powerful molecular epidemiology screening tools, enable new diagnostic and prognostic epigenetic
biomarker identification, allowing for patient stratification and tailored management. In combination
with new-generation epi-drugs, nanomedicine can help to overcome low therapeutic ecacy in
Cancers 2020,12, 3622; doi:10.3390/cancers12123622 www.mdpi.com/journal/cancers
Cancers 2020,12, 3622 2 of 32
treatment-resistant tumors. This review provides an overview of ongoing clinical trials focusing on
combination therapies employing epi-drugs for breast cancer treatment and summarizes the latest
nano-based targeted delivery approaches for epi-drugs. Moreover, it highlights the current limitations
and obstacles associated with applying these experimental strategies in the clinics.
Keywords: epigenetics; breast cancer; nanomedicine; epi-drugs; targeted delivery; drug resistance
1. Introduction
The most common cancer diagnosed among women is breast cancer (BC), the second leading cause of
cancer deaths [
1
]. Besides well-studied genetic changes, epigenetic alterations, resulting in aberrant gene
expression, are among the key contributors to breast carcinogenesis. Different mechanisms introduce and
maintain epigenetic modifications, including DNA methylation, post-translational histone modifications,
and non-coding RNA-mediated regulation [
2
]. Epithelial-mesenchymal transition (EMT) is a complex
developmental program, which plays a crucial role in the hematogenous and lymphatic dissemination
of tumors. EMT facilitates phenotypic metamorphosis of epithelial tumor cells into highly motile and
more aggressive mesenchymal cells that can colonize distant organs. Moreover, this multistep process
enables the generation of tumors with stem cell properties, which play a significant role in developing
therapeutic resistance [
3
]. The reversibility of EMT, allowing circulating tumor cells (CTCs) to remain
epithelial in their origin, endowing them with a potential to seed metastasis, supports the hypothesis
about its epigenetic regulation [
4
]. EMT is triggered by extracellular signals, including extracellular matrix
proteins and soluble growth factors, or by intracellular cues. It is mediated by a group of pleiotropic
transcription factors (TFs), which control a heterogeneous network of epigenetic effectors, thus allowing
potent gene expression changes [
4
]. This epigenetic plasticity not only permits dynamic regulation of
expression but also offers numerous therapeutic opportunities.
Currently, BC treatment involves a multidisciplinary approach. Although the effectiveness of various
therapeutic regimens has increased, resulting in reduced mortality [
5
], there are still many obstacles to
overcome. These include serious side effects, hard-to-treat tumor subtypes, intratumoral heterogeneity,
and at present incurable metastatic disease. The success achieved so far in treating hematological
malignancies using epigenetic inhibitors has stimulated interest in their use to treat solid tumors.
Promising preclinical results suggest that epigenetic drugs (epi-drugs) can sensitize resistant cancer cells
to traditional approaches. Unfortunately, these results have not yet been confirmed by clinical studies,
as the early-generation
epi-drugs were basically broad-spectrum reprogrammers, causing large-scale
gene expression changes. This “one size fits all” approach has mostly failed due to off-target effects,
significant toxicities, the risk of large-scale epigenomic repatterning, and the lack of appropriate biomarkers
for patient selection. However, this failure has led to the development of selective new-generation epi-drugs,
which, together with precision medicine design, provide a new chance for epigenetic therapy of solid
tumors [6,7].
Among the options that could contribute to successful clinical applications of epi-drugs are
new technologies for safer and more ecient cancer cell epigenome modulation. Advances in
nanotechnology and material science have provided a broad variety of more precise and safer
nanoscale organic and inorganic nanomaterials for drug delivery (e.g., dendrimers, micelles,
liposomes, gels, metal- and carbon-based nanomaterials). To date, several nanomaterials have
been successfully studied and introduced in cancer treatment, and many others are undergoing clinical
trials. Encapsulation by intelligent nanocarriers of antitumor drugs, conventional chemotherapeutics,
epi-drugs, or both, can improve their solubility and stability by protecting the drugs from fast clearance
and degradation, thus prolonging their half-life in the systemic circulation [
8
]. Nanocarriers can
also be tuned to ensure targeted, controlled, and sustained release, thereby reducing toxicity [
8
10
].
Nanoscale size and unique physicochemical properties (e.g., shape, surface area, and charge) allow
Cancers 2020,12, 3622 3 of 32
the accumulation of nanocarriers in the tumor mass due to the enhanced permeability and retention
(EPR) eect, which is the basis of passive targeting [
11
]. The spatial and temporal heterogeneity of
tumors is one of the limitations of therapeutic ecacy in passive targeting [
12
]. Functional surface
modifications of nanocarriers by specific ligands (antibodies, aptamers, proteins, etc.) with a high
anity for particular receptors overexpressed on the tumor cells allows active targeting ofdrug delivery
to the tumor mass, thereby increasing treatment ecacy and reducing side eects [
13
]. The biggest
challenge to combat BC is to eliminate cancer stem cells (CSCs) that play a crucial role in metastasis and
the development of multidrug resistance to therapy. Nanoscale delivery systems represent a promising
tool for their eradication [14].
In this review, we provide an outline of current achievements in epigenetic therapy of BC,
focusing on the ability of epi-drugs to sensitize resistant cancer cells to standard therapeutic approaches.
Furthermore, we highlight the promise of nanomedicine with regard to overcoming obstacles associated
with the successful use of epi-drugs for the treatment of solid tumors.
2. Molecular Pathology of Breast Cancer
The BC incidence rate varies from 27.9 per 100,000 people in Middle Africa to 92.6 per 100,000 in
Western Europe [
1
]. The dierences in incidence are attributed to dierent risk factors and the availability
of improved imaging techniques for screening and diagnosis [
15
]. The five-year relative survival rate
for women diagnosed with the regional disease was recently estimated at 86%, whereas it was 27% for
those with metastatic disease [
16
]. Therapeutic resistance and metastatic potential are influenced by
the heterogeneity of phenotypic and molecular characteristics [
17
20
]. While the luminal A subtype is
considered a low-grade disease with a good prognosis and likely to benefit from endocrine therapy alone,
luminal B tumors have a higher proliferation rate, worse prognosis, and patients require additional
chemotherapy treatment. The human epidermal receptor 2 (HER2)-overexpressing tumors tend to
grow faster and can have higher histological grade than luminal-like tumors, but generally, they are
successfully treated by targeted anti-HER2 therapies. Triple-negative/basal-like BC is a histologically
high-grade disease associated with a poor prognosis. Patients with this subtype do not benefit from
targeted therapies, and the standard chemotherapy regimen is the only suitable therapeutic approach
at present.
2.1. Role of Epigenetics in BC Pathogenesis
Approximately 10% of BCs are considered to be hereditary. Most of them are associated with
mutations in tumor suppressor genes BRCA1 and BRCA2 or other high or moderate-penetrance
genes, such as CHEK2, ATM, PALB2, PTEN, STK11, and TP53 [
21
]. In general, BC initiation,
followed by histological progression from premalignant stages to invasive carcinoma, develops from
the accumulation of genetic and epigenetic changes. These aberrations involve the inactivation of
tumor suppressor genes and/or oncogene activation, enabling the continuous malignant transformation
of cells [
22
]. The main genetic changes comprise point mutations (single nucleotide substitutions,
small insertions, and deletions), structural rearrangements, and large scale copy number changes [
23
].
The most frequently altered genes reported in early BCs are TP53,PIK3CA,MYC,PTEN, CCDN1,
ERBB2,FGFR, and GATA3 [24].
Apart from genomic alterations, cancer initiation and progression are driven by the combined
action of multiple epigenetic changes [
25
]. The term epigenetics describes heritable DNA modifications
that do not change the DNA sequence but can aect gene expression. Epigenetic regulation is involved
in many normal cellular processes, including cell growth and dierentiation [
26
]. The major epigenetic
modifications encompass changes in DNA methylation, post-translational histone modifications,
and non-coding RNA expression. These modifications are thought to participate in early BC
carcinogenesis events and can be useful as biomarkers for early detection and the determination of
prognosis and response to treatment [27].
Cancers 2020,12, 3622 4 of 32
2.1.1. DNA Methylation
DNA methylation is the chemical modification caused by covalent attachment of a methyl group at
cytosine within CpG dinucleotides, resulting in the formation of 5-methyl-cytosine (5mC). The methyl
group transfer from the S-adenosyl-L-methionine donor is catalyzed by three DNA methyltransferases
(DNMTs), DNMT1, DNMT3A, and DNMT3B. DNMT1 maintains existing methylation patterns
during replication, while DNMT3A and DNMT3B are responsible for de novo methylation [
28
].
DNA methylation occurring in the promoter regions inhibits gene expression by preventing
the binding of the transcriptional machinery to their recognition sequences or by binding proteins
with a methyl-binding domain (MBD) that have a higher anity to promoters [
29
]. MBD proteins
interlink DNA methylation and histone modifications by engaging histone deacetylase complexes
and chromatin remodeling factors, leading to chromatin condensation and thus to transcriptional
repression [30].
Aberrant DNA methylation is a hallmark of cancer. The methylation of normally unmethylated
promoter CpGs, known as hypermethylation, can lead to tumor suppressor genes’ inactivation, thus acting as
a potential biomarker for early cancer detection and prognosis [
31
].
Furthermore, DNA hypomethylation
on
a genome-wide scale, reported in many cancers, can induce genomic instability [
32
].
De Almeida et al.
identified 368 dierentially methylated individual CpG sites in BC tumors compared to healthy breast
tissues [
33
]. Hypermethylated CpG sites were mostly present in upstream promoter regions (56%),
while hypomethylated CpG sites were localized mostly in the gene bodies (66%).
Nevertheless, in many
cancers, CpG island shores, the regions of low CpG density flanking traditional CpG islands (up to 2 kb
distant) show distinct subtype-specific methylation signatures [
34
]. Lately, more than 100 genes
have been found to be hypermethylated in BC [
35
]. They play essential roles in various cell
mechanisms, including DNA repair (e.g., MGMT,BRCA1,MLH1), cell-cycle regulation (e.g., CCND2,
AK5,FOXA2), apoptosis (e.g., BCL2,APC) [
36
], cell adhesion (e.g., CDH1), tissue invasion and
metastasis (e.g., RASSF1A,RAR
β
,TWIST,HIN1), and hormone-mediated cell signaling (ESR1,ESR2,
and THRB) [37].
For a long time, DNA methylation was referred to as an irreversible epigenetic event, which could
only be passively depleted through DNA replication. However, this consideration changed with
the discovery of ten-eleven translocation (TET) proteins [
38
]. The TET family, comprising TET1, TET2,
and TET3 proteins, oxidize 5mC to 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine
and accordingly facilitates active DNA demethylation [
32
]. TET proteins bind preferentially to
unmethylated CpGs within CpG islands, thus maintaining CpG islands in a hypomethylated
state, associated with active transcription [
39
]. Besides, decreased 5-hydroxymethylcytosine and
loss-of-function mutations of TET proteins can potentially aect malignant transformation [40].
2.1.2. Histone Modifications
Chromatin is a dynamic structure composed of nucleosomes. The main components of the nucleosomal
subunit are the histones. These alkaline proteins form an octamer consisting of two identical subunits,
each containing four histones: H2A, H2B, H3, and H4. The nucleosome core particle includes 147 bp
of DNA sequence, wrapped in nearly two superhelical turns around a histone octamer [
41
]. In general,
histones are highly conserved proteins that can be post-translationally modified at the amino acid
residues located on their C- and N-terminal tails [
42
]. Post-translational histone modifications (PTMs)
do not aect the DNA sequence but can influence gene expression by changing chromatin structure
from the non-condensed transcriptionally active state (euchromatin) to the condensed inactive state
(heterochromatin). Histone tail residues can undergo covalent PTMs, including methylation, acetylation,
phosphorylation, sumoylation, glycosylation, ubiquitination, ADP-ribosylation,
and carbonylation [43,44].
For instance, acetylated histone H3 and di- or tri-methylated histone H3 lysine 4 (H3K4me2, H3K4me3)
in the promoter region result in the activation of gene expression. Conversely, repression of
the promoters is usually caused by histone deacetylation and by tri-methylated histone H3 lysine 27
(H3K27me3) and tri-methylated histone H3 lysine 9 (H3K9me3) [
45
,
46
]. It is not surprising that many
Cancers 2020,12, 3622 5 of 32
aberrations in histone modifications are found in cancer due to the essential roles of these modifications
in DNA-mediated cellular processes. Furthermore, it has been shown that global loss of acetylation on
lysine K16 and tri-methylation on lysine K20 of histone H4 is a hallmark of various cancers [
47
]. In BC,
reduced levels of lysine acetylation (H3K9ac, H3K18ac, and H4K12ac), lysine methylation (H3K4me2,
H4K20me3), and arginine methylation (H4R3me2) have been observed in poor prognostic tumors.
Additionally, down-regulation of H4K16ac and the corresponding enzyme histone acetyltransferase
(HAT) hMOF, in most primary breast tumors, represent an early sign of BC [48,49].
Dierent enzymes catalyze histone PTMs, including histone deacetylases (HDACs), which remove
acetyl groups, histone methyltransferases (HMTs), demethylases (HDMs), HATs and others. At least
18 HDACs have been identified in humans and classified into four groups [
50
]. Histone acetylation
marks are read by bromodomain protein modules, which are evolutionarily conserved domains of
110 amino acids within many chromatin-associated proteins (including HATs) and other eector
enzymes [
51
]. HATs are categorized into five families: GNAT, MYST, p300/CBP, SRC, and TAFII250.
Members of the bromodomain family, bromodomain and extra-terminal domain (BET) proteins can
increase proliferation and may potentiate the overexpression of several oncogenes such as MYC [
52
].
HMTs, along with HDMs, introduce or remove methyl groups and modify other proteins in addition
to histones [53].
Molecular changes that aect the expression of histone-modifying enzymes could also
contribute to cancer development and progression. In BC, various alterations have been identified,
including overexpression of p300, HBO1, HDAC1, HDAC2, HDAC3, HDAC6 [
54
], amplification and
overexpression of enhancer of zeste homolog 2 (EZH2) [
55
], depletion of H3K9 trimethyl-demethylase
(JMJD2B) [
56
], downregulation of lysine-specific histone demethylase 1A (LSD1) [
57
], and others.
Due to more demanding methodological approaches, the role of histone modification in cancer is less
studied than DNA methylation or changes in non-coding RNA (ncRNAs).
2.1.3. Epigenetic Regulation by Non-Coding RNA
The ncRNAs are molecules of RNA that are not translated into proteins. However, they play
an important role in epigenetic modifications. RNAs with regulatory functions are divided into two
major classes based on their size: short-chain ncRNAs (including siRNAs, miRNAs, and piRNAs) less
than 200 nucleotides in length, and long ncRNAs (lncRNAs) [
58
]. The ncRNAs can directly influence
gene expression or be epigenetically regulated themselves [
59
]. Alterations in their expression
contribute to the pathogenesis of most human cancers [6062].
MicroRNAs (miRNAs) are endogenous, short, single-stranded RNAs, 19–25 nucleotides in
length, which regulate gene expression by post-transcriptional silencing or the degradation of target
mRNAs [
61
]. In general, they can be categorized into tumor-promoting (oncomiRs) and tumor-suppressing
miRNAs. During carcinogenesis, oncomiRs are usually amplified, and tumor-suppressing miRNAs are
down-regulated [
63
]. miRNAs can be readily identified in tumor samples (non-circulating miRNAs) and
different body fluids (circulating miRNAs). Deregulation of miRNAs is involved in each cancer hallmark,
such as proliferation, invasion and metastasis, apoptosis, etc. [64,65].
In BC, the expression profiles of miRNAs have been determined as promising biomarkers for
diagnosis, more precise subtyping, prognosis, and therapy response [
66
]. It was observed that luminal
A tumors have higher expression of the miR-99a/let-7c/miR-125b-2 miRNA cluster compared to luminal
B tissues [
67
]. The miR-1, miR-92a, miR-133a, and miR-133b were determined as essential diagnostic
markers by analyzing miRNA profiles between tumor and serum samples [
68
]. Furthermore, it was
shown that patients with up-regulated expression of miR-1307-3p, miR-940, and miR-340-3p had
a worse overall survival [
69
], while miR-21 and miR-205 were associated with disease-free survival
interval and miR-205 with overall survival [70]. In addition, miRNA-449a overexpression was found
to have propitious prognostic significance in BC patients [71].
Cancers 2020,12, 3622 6 of 32
2.2. Epigenetics in BC Progression
EMT is a fundamental process having a crucial role during embryogenesis and tissue regeneration.
EMT,together with the mesenchymal-epithelial transition (MET), is required to form organs and cellular
structures [
72
]. Moreover, they are involved in malignant progression, where EMT is responsible for
the loss of cellular adhesion, cytoskeleton remodeling, acquisition of cancer cells’ migratory capacity
as well as their ability to intravasate and survive in circulation [
73
]. Typical phenotypic EMT stages
range from fully dierentiated epithelial to dedierentiated mesenchymal cells. EMT is triggered
by extracellular agents such as growth factors (e.g., TGF-
β
[
74
]) and by non-growth factor stimuli
(e.g., hypoxia, oxalate, Galectin-8 [
72
]). Stromal constituents of the tumor microenvironment can
activate EMT via secretion of various cytokines and chemokines acting in a paracrine fashion on nearby
carcinoma cells. Tumor-associated stromal cells include cancer-associated fibroblasts,
regulatory T cells
,
CD4+helper T and CD8+cytotoxic T cells, tumor-associated macrophages, and myeloid-derived
suppressor cells. These cells can promote tumor progression and metastasis. Moreover, activation of
EMT can contribute to the immunosuppressive tumor microenvironment, which has been observed in
BC [
75
,
76
]. In mammary epithelial cells, the up-regulation of TWIST1 or SNAI1-induced formation of
neoplastic cells with CSC properties, which are associated with a more aggressive basal-like BC subtype.
During EMT, epithelial cells down-regulate the expression of several cell adhesion molecules,
among them E-cadherin and
β
-catenin. EMT-inducing TFs play a critical role in this process.
While several of them are well known, notably SNAI1/2, TWIST1/2, ZEB1/2, FOXC1/2, and TCF3,
others may yet be discovered [
77
]. Until recently, the down-regulation of E-cadherin has been
considered a hallmark of EMT; however, this assumption seems to be too simple in light of recent
discoveries. For instance, Padmanaban et al. showed that E-cadherin is required for invasive ductal
carcinoma cell survival and metastasis [
78
]. Although E-cadherin loss was associated with increased
invasion into local tissue, in parallel, it was connected with a reduction in cell growth and survival,
as well as a decreased amount of CTCs in the peripheral blood of patients. Notably, cell clusters
rather than individual invading cells are involved in the initiating steps of malignant progression.
E-cadherin is expressed in the cells that form these clusters [
79
]. Thus, the notion that E-cadherin
is repressed during EMT contradicts recent findings, which have helped to explain why E-cadherin
expression is often observed in the BC tumors and metastases. Although several reports describe
the occurrence of metastases formed by cells that never underwent EMT [
80
], these studies have been
criticized for insucient experimental evidence [81].
While EMT is driving the dissemination of cancer cells, MET is crucial for metastatic colonization [
82
].
MET can occur as a passive consequence of the down-regulation of EMT-TFs or their active repression [
83
].
In BC, MET can be promoted by TFs such as Prrx1 or ZEB1/2 or the expression of ID1 and ID3
genes [
84
].
In the past
few years, evidence has accumulated to demonstrate that the phenotypic plasticity
of the metastatic process is mediated via epigenetic modifications driven by these TFs. After their
binding to promoters and enhancers, they interact with epigenetic regulators and chromatin remodeling
machinery to up-regulate the expression of pro-mesenchymal genes and down-regulate epithelial
genes [
85
]. These DNA-binding proteins act as intermediaries between cellular signaling and chromatin
remodeling, thus guiding the epigenetic machinery to their target sites.
It has been demonstrated that during EMT, HMT G9a, which can also methylate non-histone
proteins, interacts with Snail to recruit it to the CDH1 promoter [
86
,
87
]. A number of these TFs
contain methyl-lysine binding motifs, crucial for correct chromatin recognition, supporting their role
in EMT
´
s epigenetic regulation. It was shown that Zeb2 inhibits E-cadherin via DNA methylation [
88
].
Hypoxia-induced EMT is regulated via HDACs, namely HDAC3, coupled with H3K4Ac, present
at promoter regions of traditional EMT genes such as CDH1 and VIM. Chromatin changes are also
responsible for TGF-
β
mediated EMT, where the new chromatin modifiers were identified, e.g., UTX,
PRMT5, Rad21, or RbBP5 [
89
]. Cells that have undergone EMT can also acquire drug resistance and
stem cell-like properties [
90
]. Traditional EMT-associated TFs were shown to confer resistance to
Cancers 2020,12, 3622 7 of 32
oxaliplatin-based and cisplatin-based chemotherapeutics, while the loss of E-cadherin was associated
with resistance to several growth factors and kinase inhibitors [91,92].
Given their mode of action, epigenetic inhibitors can restore aberrant DNA methylation
or histone deacetylation or impede the recognition of acetylated lysine residues and, therefore,
potentially re-establish epithelial phenotypes during EMT. Comprehensive bioinformatic analysis
has identified and validated various drug combinations together with HDAC inhibition that hamper
EMT [
93
,
94
]. Thus, EMT, the crucial step in cancer progression and metastasis, can be potentially
modulated by epi-drugs.
3. Breast Cancer Therapeutic Opportunities
3.1. The Biomarker-Directed Approach in BC Treatment
To overcome the limitations incurred by resistance mechanisms in the clinical management of
advanced cancers, an increasing effort is being made towards biomarker-driven cancer treatments.
This aims to identify important biomarkers capable of addressing tumor heterogeneity and effectively
predicting a favorable clinical outcome as a response to a particular treatment. BC, in particular,
is considered a family of distinct diseases with a varying molecular basis. Initial gene expression
studies using cDNA microarrays have resulted in a classification of BC into five subtypes, establishing
as major BC biomarkers estrogen receptor (ER), progesterone receptor (PR), and HER2 [
95
,
96
].
Therefore, currently, there is
a mandatory need to define their expression status, lymph node
involvement, and tumor size for all patients with invasive BC for therapy decision making.
These markers are established in international guidelines as essential factors for the clinical management
of primary BC patients [
15
]. However, the stratification of patients based solely on ER, PR, and HER2
expression has proven inecient, unable to capture the substantial phenotypic complexity and
heterogeneity of BC, thus stressing the need to integrate additional biomarkers for a more refined
characterization.
Ki-67, a non-histone
nuclear protein, is used as a marker of cell proliferation.
Ki-67 expression
is significantly higher in malignant tissues with poorly dierentiated tumor cells than
in normal tissue and is thus used to assess tumor aggressiveness [97].
Traditional therapy of non-metastatic BC involves multidisciplinary strategies combining surgery,
radiotherapy, neo-/adjuvant, endocrine, and targeted therapy [
98
]. For non-metastatic BC, the primary
therapy approach consists of eradicating tumor and regional lymph nodes and preventing metastatic
relapse. The first two goals are usually achieved by locoregional therapy that involves surgery
and radiotherapy and/or neoadjuvant therapy in case of locally advanced disease. Prevention of
metastatic relapse is achieved with systemic therapies that comprise anthracycline and taxane-based
chemotherapy, anti-estrogen hormonal therapy, and anti-HER treatment, depending on receptor
status [99].
In contrast to early BC, metastatic disease (stage IV), with common sites of spread in bones, brain,
lung, and liver, is considered incurable, and the therapy aims to prolong life while minimizing symptoms
or side eects. The combinations of endocrine, targeted therapy, chemotherapy, and immunotherapy
can be administered to the metastatic patients, taking into account the tumor subtype, extent,
and localization of the disease and the presence of specific molecular alterations. Beyond HER2 and
ER/PR, new predictive biomarkers for targeted therapy in metastatic BC include BRCA1/2and PI3KCA
mutations for PARP and PI3KCA inhibitors, respectively, and PD-L1 expression and/or MSI status
for immunotherapy. The eective new biological therapies like CDK4/6 or mTOR inhibitors are now
emerging. However, we still lack predictive biomarkers for these treatments [100].
3.2. Precision Medicine Concept
The maturation of omic technologies as powerful molecular epidemiological screening tools
has empowered the emergence of manifold predictive biomarker signatures. The integration of
genomic and transcriptomic profiles of 2000 breast tumors from the METABRIC (Molecular Taxonomy
Cancers 2020,12, 3622 8 of 32
of Breast Cancer International Consortium) cohort revealed ten BC subtypes, termed integrative
clusters (IntClust/s) and characterized by distinct genomic drivers [
101
]. Currently, there are five main
standardized genetic prognostic platforms for BC, aiming to assist decision on therapeutic options,
mainly including hormone therapy, chemotherapy, and anti-HER2 treatment:
Oncotype DX provides prognostic information in terms of 10-year distant recurrence. It predicts
the likelihood of adjuvant chemotherapy benefit in ER+BC patients, based on the expression of
a panel of 21 genes (16 cancer-related and five reference genes) [102].
Breast Cancer Index assesses the expression of 7 genes to predict the benefit from extended,
adjuvant, endocrine therapy (Tamoxifen) in HR+patients. It is a gene expression signature
comprising two functional biomarker panels, the molecular grade index (MGI) and the two-
gene ratio HOXB13/IL17BR (H/I), that evaluate tumor proliferation and estrogen signaling,
respectively [
103
]. MGI is a gene expression assay, measuring the expression of five genes
(BUB1B, CENPA, NEK2, RACGAP1, RRM2) related to histological grade and tumor progression,
which recapitulates tumor grade and can predict the clinical outcome with high performance [
104
].
EndoPredict (Myriad Genetics, Inc., Salt Lake City, UT, USA) is a genomic test for people newly
diagnosed with early-stage, ER+, HER2-negative BC (node-negative). It assesses the expression
of 12 genes (8 target genes, 3 normalization genes, and 1 control gene) to predict response to
chemotherapy [105].
MammaPrint (Agendia, Irvine, CA, USA) is a 70-gene signature test that predicts the clinical
outcome/response to chemotherapy in ER+early-stage BC [106].
Prosigna Breast Cancer Prognostic Gene Signature Assay (Nanostring, Seattle, WA, USA),
formerly PAM50, assesses Tamoxifen response for HR+BC patients based on the expression of
58 genes after 5 years of hormonal therapy treatment in postmenopausal women [107].
Besides Prosigna, Nanostring has developed a more extensive assay, the human nCounter Breast
Cancer 360 panel, which comprises 776 genes across 23 key BC pathways and processes. Results are
grouped in 48 signatures across 13 categories, measuring biological variables crucial to BC tumor
biology. This panel has been developed for the evaluation of diverse BC aspects, including BC
subtyping (luminal A/B, HER2-enriched, basal-like, triple-negative), expression of BC receptors and
signaling (ESR1, PGR, ERBB2, AR, PTEN, CDK4, CDK6), mutational content (HRD, BRCA, P53),
markers for tumor proliferation, apoptosis and dierentiation (FOXA1, SOX2), cell adhesion (claudin),
and immunity (chemokines, TGF-
β
, PD-1). Although a detailed description of the nCounterBC
360 panel exceeds this review’s scope, it contains a vast number of gene expression markers and
indicates the intense molecular heterogeneity characterizing the diversity of BC phenotypes. Therefore,
investigating the epigenetic landscape of BC may provide an additional layer of information that could
improve our fundamental understanding of BC
´
s molecular complexity and the putative rational
development of more eective and precise treatments.
Finally, various studies have identified and suggested epigenetic modifications and regulators
as prognostic biomarkers for BC [
108
110
]. In this context, miRNAs have been considered a pool
of highly potent biomarkers, as they have been linked to the identification of distinct molecular
subtypes and tumor-related processes. miRNA expression profiling was successfully employed to
classify the breast tumors as luminal A, luminal B, basal-like, HER2+, and normal-like BC [
111
].
Aberrant expression of miRNAs has also been correlated with clinical features, such as angiogenesis,
metastasis, and EMT [
112
]. Besides, several lncRNAs have been reported as promising biomarkers
for prognosis, diagnosis, and therapy [
113
]. Overall, there is cumulative evidence focusing on
the systematic screening of epigenetic signals as a promising area for the discovery of novel molecular
BC biomarkers, combining sensitivity, specificity, and robustness, with a potentially decisive impact on
improving the quality of BC treatments in the context of precision medicine.
Cancers 2020,12, 3622 9 of 32
3.3. Potential of Epigenetic Therapy
The role of epigenetics in cancer initiation and progression, including its contribution to
the development of innate and acquired resistance to several therapeutic regimens, has led to
the scientific eort to reverse the aberrant cancer epigenome [
32
]. The lack of knowledge about
subtype-specific epigenome signaling pathways, and missing patient-specific epigenetic biomarker
profiles, are currently the main challenges hampering the wider clinical application of epigenetic agents
in the treatment of solid cancers [
114
]. Over the last decade, several epi-drugs have received US Food
and Drug Administration (FDA) approval for the treatment of blood-borne cancers: 5-azacytidine (AZA,
Vidaza
®
), 5-aza-2
´
-deoxycytidine (decitabine, DAC, Dacogen
®
), vorinostat (VOR, SAHA, Zolinza
®
),
romidepsin (FK228, Istodax
®
), belinostat (Beleodaq
®
), panobinostat (Farydak
®
), and chidamide
(Epidaza®) (Figure 1) [115117]. However, except for tazemetostat (Tazverik®), approved by FDA in
January 2020 for metastatic or locally advanced epithelioid sarcoma, there is no epigenetic therapy
approved for solid tumors, which are considered more epigenetically complex. Moreover, they exhibit
abnormal vascularization, a specific tumor microenvironment, and more dierentiated cells with
decreased epigenetic reprogramming [7,118].
Cancers 2020, 12, x FOR PEER REVIEW 9 of 33
scientific effort to reverse the aberrant cancer epigenome [32]. The lack of knowledge about subtype-
specific epigenome signaling pathways, and missing patient-specific epigenetic biomarker profiles,
are currently the main challenges hampering the wider clinical application of epigenetic agents in the
treatment of solid cancers [114]. Over the last decade, several epi-drugs have received US Food and
Drug Administration (FDA) approval for the treatment of blood-borne cancers: 5-azacytidine (AZA,
Vidaza®), 5-aza-2´-deoxycytidine (decitabine, DAC, Dacogen®), vorinostat (VOR, SAHA, Zolinza®),
romidepsin (FK228, Istodax®), belinostat (Beleodaq®), panobinostat (Farydak®), and chidamide
(Epidaza®) (Figure 1) [115–117]. However, except for tazemetostat (Tazverik®), approved by FDA in
January 2020 for metastatic or locally advanced epithelioid sarcoma, there is no epigenetic therapy
approved for solid tumors, which are considered more epigenetically complex. Moreover, they
exhibit abnormal vascularization, a specific tumor microenvironment, and more differentiated cells
with decreased epigenetic reprogramming [7,118].
Rational epi-drug discovery using validated targets is a recent phenomenon. During early
efforts, epi-drug development has been based on the demonstration of efficacy and phenotypic
observations rather than on knowledge of their molecular targets. The timeline and key events
influencing epi-drug development, including challenges and opportunities associated with their
implementation in clinics, have recently been reviewed in-depth by Ganesan and colleagues [119].
As a detailed characterization of epi-drugs is beyond this paper’s scope, we provide only their basic
characteristics and classification (Figure 1).
Figure 1. Different categories of epi-drugs, assessed in preclinical studies and clinical trials. Eight of
them (indicated by asterisks) were approved to treat several human malignancies (modified from
7,119,120). Abbreviations: DNMTIs-DNA methyltransferase inhibitors; HDACIs-histone deacetylase
inhibitors; HMTIs-histone methyltransferase inhibitors; HDMIs-histone demethylase inhibitors;
BETIs-bromodomain and extra-terminal domain inhibitors; HATIs-histone acetyltransferase
inhibitors; ncRNAs-non-coding RNAs; DOT1LIs-DOT1-like histone lysine methyltransferase
Figure 1.
Dierent categories of epi-drugs, assessed in preclinical studies and clinical trials.
Eight of them (indicated by asterisks) were approved to treat several human malignancies (modified
from 7,119,120). Abbreviations: DNMTIs-DNA methyltransferase inhibitors; HDACIs-histone
deacetylase inhibitors; HMTIs-histone methyltransferase inhibitors; HDMIs-histone demethylase
inhibitors; BETIs-bromodomain and extra-terminal domain inhibitors; HATIs-histone acetyltransferase
inhibitors; ncRNAs-non-coding RNAs; DOT1LIs-DOT1-like histone lysine methyltransferase inhibitors;
EZH2Is-enhancer of zeste homolog 2 inhibitors; PRMTIs-protein arginine methyltransferase inhibitor;
LSD1Is-lysine-specific histone demethylase 1A inhibitors.
Cancers 2020,12, 3622 10 of 32
Rational epi-drug discovery using validated targets is a recent phenomenon. During early eorts,
epi-drug development has been based on the demonstration of ecacy and phenotypic observations
rather than on knowledge of their molecular targets. The timeline and key events influencing epi-drug
development, including challenges and opportunities associated with their implementation in clinics,
have recently been reviewed in-depth by Ganesan and colleagues [
119
]. As a detailed characterization
of epi-drugs is beyond this paper’s scope, we provide only their basic characteristics and classification
(Figure 1).
Given that overexpression of DNMTs and HDACs are considered the critical factors in
carcinogenesis, demethylating agents and HDAC inhibitors (HDACIs) seem to be promising anticancer
drugs [
120
122
]. Cytidine analogs, AZA and DAC, were the first DNMT inhibitors (DNMTIs) approved
by the FDA in 2004 for the treatment of myelodysplastic syndrome and acute myeloid leukemia [
123
].
Although they were initially defined as cytotoxic agents, their therapeutic properties were achieved
at lower doses and with prolonged exposure [
124
]. In general, HDACIs block histone deacetylation,
causing reactivation of tumor suppressor genes that can inhibit cancer cell proliferation. Moreover,
they have been shown to induce cancer cell death at concentrations to which normal cells are relatively
resistant [
125
,
126
]. Chemically, HDACIs are classified into dierent subgroups: carboxylic acids,
benzamides, cyclic peptides, and hydroxamic acids [
127
]. Similar to DNMTs, the first-generation
HDACIs were characterized by poor bioavailability, low stability, and short half-life.
The development of second-generation epi-drugs has, therefore, been aimed to circumvent
these shortcomings. Guadecitabine, a second-generation DNMTI, has a novel molecular structure,
which prolongs its
in vivo
half-life, and increases efficacy [
128
]. As nucleoside analogs require active DNA
synthesis to incorporate them into the DNA, their use is limited in hypoproliferative cancers and could
be a major obstacle in the therapy of solid tumors [
129
]. Their common side effects could be avoided
using non-nucleoside analogs, such as hydralazine, procainamide, RG108, and MG98 [
130
]. Recently,
more efficient bi-substrate analogs have become potent DNMTIs [
119
].
Although the second-generation
HDACIs, such as hydroxamic acid, belinostat, panobinostat, chidamide, or valproic acid, possess improved
pharmacological properties, they achieved limited efficacy as single agents. However, their combination
with other therapeutic approaches has allowed new avenues of their clinical investigation [7].
The principle of precision medicine is now being applied to the development and the use
of third-generation epi-drugs, defined by a high degree of selectivity. This family includes HMT
inhibitors (e.g., EZH2, DOT1-like histone-lysine methyltransferase (DOT1L), G9a and PRMT inhibitors),
HDM inhibitors (e.g., LSD1 or Jumonji C domain inhibitors), BET inhibitors (BETIs) and HAT inhibitors
(HATIs) [
7
,
131
]. HMTIs are emerging therapies targeting specific modifications. For example, it has
been found that mutations in lymphomas activate the H3K27 histone methyltransferase EZH2,
leading to disease progression. Therefore, the EZH2I can selectively target and induce cell death
in cell lines with these mutations [
132
]. DOT1L is the only histone methyltransferase that targets
the histone H3 lysine 79 (H3K79) residue [
133
]. Aberrant H3K79 methylation is associated with
aggressive mixed-lineage leukemia and poor patient prognosis in lung, colorectal, and BCs [
134
].
This suggests that pharmacological inhibition of DOT1L can have therapeutic potential in several
cancer types. The H3K4 and H3K9 demethylase enzyme LSD1 has an essential regulatory role
in cell proliferation [
135
]. Its overexpression in several tumors has been correlated with a worse
prognosis [
136
]. LSD1 inhibition may slow down cell growth in LSD1-overexpressing tumor cells. BETIs,
JQ1, and I-BET762, are cell-permeable agents that reversibly and specifically bind the bromodomain
proteins, thus impeding their interaction with acetylated histone lysine residues. It has been shown that
they inhibit proliferation and induce apoptosis in various cancer cells [
137
]. HATIs include peptides,
small molecules derived from natural products (e.g., curcumin), and synthetic molecules [
138
].
Peptide-CoA bisubstrate inhibitors mimic the formation of the substrate and cofactor complex binding
to the HAT enzyme [
131
]. Well-conceived computational strategies and new screening platforms will
be needed to predict loci specific epi-drugs sensitivities.
Cancers 2020,12, 3622 11 of 32
The ncRNAs, like miRNAs or siRNAs, with their power to selectively “switch-o specific
cancer genes, are attractive targets for the development of personalized cancer therapy. The main
hindrance to the implementation of ncRNA-based therapy in clinical practice is the absence of
eective delivery systems that can protect the RNA molecules from fast nuclease degradation before
delivering them into the target cells
´
cytoplasm [
139
]. There are two dierent possibilities to use
miRNAs as therapeutic agents; substitution of depleted miRNAs (MRX34, miR34a replacement)
and inhibition of overexpressed miRNAs by antagonistic oligonucleotides [
140
]. The inhibitor of
miR-155, MRG-106, has been successfully investigated in phase I clinical trials for the treatment of
hematological malignancies [
141
]. The miRNA-based therapeutic strategy also has great potential to
regulate lncRNAs. The siRNA-mediated silencing by oligonucleotide inhibitors results in the inhibition
of lncRNA-protein interactions and secondary structure changes, thus competing for their binding
partners [
142
]. Down-regulation of cancer-related genes by siRNAs, e.g., CALAA-01 (targeting RRM2),
Atu-027 (targeting PKN3), has been assessed in phase I/II clinical trials [139].
Although several ongoing clinical trials, including epi-drugs, exist on a wide range of diseases,
many obstacles remain to be resolved. Among them are enzyme isoform selectivity, dual substrates,
multimeric enzyme complexes involved in epigenetic regulations, high-order chromatin structure,
functional effects of inhibition, and off-target effects. Other challenges are the pharmacology of
the compounds, doses to be used, therapeutic regimens or duration of the treatment, and patient selection.
Epigenetic Therapy in BC
Several clinical studies have investigated the ecacy of DNMTIs (DAC, AZA) and HDACIs (VOR,
phenyl butyrate) administration as BC monotherapy. However, these epi-drugs have shown limited
antitumor ecacy at the maximum tolerated dose, suggesting the unsuitability of this approach [
143
].
In BC, the epi-drugs have been investigated in combination with cytotoxic agents, radiotherapy,
targeted and hormone therapy, immunotherapy, as well as the combination of epi-drugs themselves
(Table 1).
Table 1. Epigenetic drugs in clinical trials focused on breast cancer.
Epi-Drug Other Interventions Status Phase Trial No.
Decitabine
LBH589, Tamoxifen Terminated 1, 2 NCT01194908
Paclitaxel Unknown 1b NCT03282825
Carboplatin Recruiting 2 NCT03295552
Doxorubicin and 4 more Recruiting 2 NCT02957968
Azacitidine
Nab-paclitaxel Completed 1, 2 NCT00748553
Entinostat Active, not recruiting 2 NCT01349959
Definitive breast Withdrawn NA NCT01292083
Fulvestrant Terminated 2 NCT02374099
Durvalumab Active, not recruiting 2 NCT02811497
Valproic acid
FEC100 Terminated 2 NCT01010854
Hydralazine Terminated 2 NCT00395655
Bevacizumab, Cetuximab Recruiting 1 NCT01552434
Entinostat
Capecitabine Recruiting 1 NCT03473639
Exemestane Recruiting
Completed
Active, not recruiting
Active, not recruiting
3
2
3
2
NCT03538171,
NCT00676663,
NCT02115282,
NCT03291886
Fulvestrant Withdrawn 2 NCT02115594
Lapatinib Ditosylate, Trastuzumab Completed 1 NCT01434303
Anastrozole Terminated 2 NCT01234532
Nivolumab, Ipilimumab Active, not recruiting 1 NCT02453620
Atezolizumab and 6 more Recruiting 1, 2 NCT03280563
Romidepsin
Cisplatin, Nivolumab Suspended 1, 2 NCT02393794
Abraxane Terminated 1, 2 NCT01938833
Alone Completed 2 NCT00098397
Cancers 2020,12, 3622 12 of 32
Table 1. Cont.
Epi-Drug Other Interventions Status Phase Trial No.
Vorinostat
Olaparib Not yet recruiting 1 NCT03742245
Paclitaxel and 3 more Completed 1, 2 NCT00574587
Alone Completed
Terminated
Completed
Withdrawn
Completed
Terminated
1
2
2
NA
1
2
NCT00719875,
NCT00132002,
NCT00262834,
NCT01695057,
NCT00788112,
NCT00126451
Tamoxifen Completed
Terminated
2
2
NCT00365599,
NCT01194427
Tamoxifen, Pembrolizumab Terminated
Not yet recruiting
2
2
NCT02395627,
NCT04190056
Carboplatin, Nab-paclitaxel Active, not recruiting 2 NCT00616967
Ixabepilone Completed 1 NCT01084057
Lapatinib Terminated 1, 2 NCT01118975
Paclitaxel, Bevacizumab Completed 1, 2 NCT00368875
Anastrozole, Letrozole, Exemestane Completed
Completed
NA
NA
NCT01720602,
NCT01153672
Trastuzumab Completed 1, 2 NCT00258349
Belinostat Ribociclib Not yet recruiting 1 NCT04315233
Panobinostat
Alone Completed
Terminated
Withdrawn
2
2
1
NCT00777049,
NCT00777335,
NCT00993642
Trastuzumab Terminated 1, 2 NCT00567879
Letrozole Completed 1, 2 NCT01105312
Trastuzumab, Paclitaxel Completed 1 NCT00788931
Capecitabine, Lapatinib Completed 1 NCT00632489
The DAC-chemotherapy combination has been widely studied in BC preclinical studies,
and the results
suggest enhanced sensitivity compared to cytotoxic therapy alone [
144
146
].
Despite these findings, clinical trials have been disappointing due to systemic toxicities and limited
ecacy. Among the reasons leading to unsatisfactory results were the design of clinical trials using
unselected patient populations and high-dose administration. This has revealed a need to identify
appropriate epigenetic biomarkers for allowing a personalized approach and targeted delivery of
epi-drugs [7].
The preclinical data with HDACIs on HR+BC cell lines indicate an increase in antiproliferative
endocrine therapy action [
147
]. In ER- BCs, an AZA-entinostat combination, and HDACI therapy
(entinostat, valproic acid, TSA) alone, have shown a high ER re-expression and ecient restoration
of the sensitivity to antioestrogen treatment [
148
151
]. Furthermore, the BET inhibitor JQ1 alone
or in combination with the selective ER down-regulation by fulvestrant can eectively suppress
the growth of tamoxifen-resistant cells [
152
]. The synergy of combination therapy has been preclinically
demonstrated on triple-negative BC cells with mTOR-BETIs [
153
] and with PARP-HDACIs [
154
].
Based on promising preclinical results, the joint ecacy of HDACIs and anti-HER2 therapy with
trastuzumab has been assessed in a clinical study (NCT00258349). Unfortunately, the hypothesis
of reversing trastuzumab resistance by adding VOR in the therapeutic regimen has not been
confirmed [
155
].
However, with the appropriate
drug combination, it could be possible to target
several oncogenic pathways [156].
Cancers 2020,12, 3622 13 of 32
All in all, for the successful utilization of epi-drugs in clinics, there is an urgent need for
identification of new epigenetic therapeutic targets and mechanisms ensuring prolonged stability of
epi-drugs, decreasing undesirable side eects while allowing increased ecacy via targeted delivery.
4. Nanomedicine as a Tool to Overcome the Current Limitations of Epigenetic Therapy
Nanomedicine applies the knowledge from material science and nanobiotechnologies to healthcare
in order to improve the diagnosis, imaging, monitoring, prevention and regeneration, and to increase
the ecacy and safety of the treatment [
157
]. According to the European Commission definition,
a nanomaterial is a natural, incidental, or manufactured material with one or more external dimensions
in the nanoscale (size range 1 nm–100 nm), and having a volume-specific surface area larger than
60 m
2
/cm
3
[
158
]. The term nanomaterial in nanomedicine is broader and includes particles with
dimensions up to 1000 nm [
159
]. Besides, the International Organization for Standardization has defined
a nanoparticle (NP) as a nanomaterial, with the size in all three dimensions in the nanoscale [
160
].
However, these varied definitions and classifications allow a wide interpretation. Notably, the beneficial
or detrimental potential of nanomaterials for the environment and human health depends on their
physical and chemical properties, such as shape, composition, size, surface charge, and electron
transfer [161].
4.1. Smart Nanoformulations for Drug Delivery Applications
Nowadays, a central area for the use of nanomedical products, considering the number of scientific
publications and product approvals by regulatory agencies, is cancer treatment [
162
]. The conventional
cytotoxic anticancer therapy is often associated with low therapeutic ecacy and increased systemic
toxicity. The chemotherapeutic agents also aect rapidly dividing non-cancerous cells, causing common
(nausea, vomiting, fatigue, hair loss) and drug-specific(e.g., cardiotoxicity, neurotoxicity, nephrotoxicity)
side eects. Besides that, tumor cells have several defense mechanisms against the cytotoxic eect of
chemotherapy. The most significant one is the multidrug resistance resulting from the overexpression
of ATP-dependent eux pumps with broad drug specificity [163].
Innovative materials, including NPs, with their unique properties, have several advantages as
potential transport vehicles for standard and experimental anticancer agents. They allow encapsulation
of hydrophobic and lipophilic molecules, enhancing their pharmacokinetics and circulation time in
the body, leading to increased drug accumulation in the tumor site via passive targeting by the EPR
eect. Moreover, they can be conjugated with specific ligands to bypass biological barriers and deliver
a high drug concentration into the target tissue via active targeting. They also allow the drug
´
s
release in a stable and controlled manner, triggered by pH, temperature, redox potential, etc. [
164
,
165
].
However, the ecient
penetration of anticancer agents encapsulated in NPs remains an important
issue in solid tumor therapy [
166
]. Systemically administered NPs should range from 10 to 200 nm
in size to avoid early elimination by kidneys or subsequent entrapment by spleen and liver [
167
].
Biocompatibility, low toxicity, and low immunotoxicity are the prerequisite attributes of all biomedical
nanomaterials. Before being approved for clinical use by the FDA, they have to undergo complex
preclinical and clinical testing to confirm their biosafety. FDA-approved materials are, therefore,
preferentially employed for the development of innovative nanocarriers. There is an increasing body of
evidence from preclinical studies that the encapsulation of existing epi-drugs into dierent formulations
has resulted in their improved stability and enhanced targeted delivery while minimizing o-target
eects [168,169].
Nowadays, the range of nano-based delivery systems is growing [
170
]. Liposomes, solid lipid
NPs (SLNs), polymer NPs, polymer micelles, dendrimers, nanoemulsions, and polymer-lipid hybrid
NPs have been studied as promising nanocarriers for targeted drug delivery (Figure 2) [168].
Cancers 2020,12, 3622 14 of 32
Cancers 2020, 12, x FOR PEER REVIEW 14 of 33
Figure 2. Main types of soft nanocarriers for drug delivery. Schematic examples of surface
modifications and functionalization.
With the large body of evidence accumulated over the past decades, three important elements
in the development of controlled drug delivery can be defined: (1) utilization of the EPR effect for
passive targeting, (2) surface modification of NPs/nanocarriers to prolong their circulation time, (3)
their effective design [171]. Coating the surface of NPs with PEG (PEGylation), an FDA approved
polymer, is a commonly used approach for improving the efficiency of drug and gene delivery to
target cells and tissues. PEGylation protects the surface from aggregation, opsonization, and
phagocytosis and prolongs systemic circulation time [172]. PEG is a hydrophilic, biocompatible, and
biologically inert polymer, which is used to enhance the stability of NPs [173]. Moreover, the coating
allows loading of specific ligands, antibodies, peptides, drugs, folic acid, aptamers, tumor markers,
transferrin, vitamins, etc., on the surface of particles offering an exciting tool to make NPs target-
specific and increase their therapeutic benefit [174–176]. These ligands can be easily conjugated onto
PEG´s distal end via various chemical coupling strategies [177]. Cancer cells overexpress various
kinds of receptors and antigens that may be used as potential drug targets in cancer therapy [178].
Entrapment of the fluorescent compound in the NPs platform allows tracking the fate of NPs in vitro
and monitoring of the accumulation of the drug in the tumor and the treatment efficacy [179]. The
Quantum Dots (QDs) are fluorescent semiconductor nanocrystals of about 10 nm, composed of a core
coated with an envelope. QD-containing NPs have found applications in cancer medical imaging,
cancer cell tracking, cancer photodynamic therapy, and cancer diagnosis [180].
4.1.1. Liposomes
Liposomes (Figure 2A) are the most utilized nanomaterial for drug delivery, with many clinical
products currently available [181]. Liposomal vesicles consist of amphiphilic poly-molecular lipid
compounds that assemble into bi-layered self-closed spherical NPs [182]. Different structural
modifications of polar and non-polar parts of phospholipids have allowed the development of
various synthetic lipids with improved properties [183]. The advantage of liposomes is their ability
to deliver both hydrophilic and hydrophobic drugs. Surface functionalization of liposomes enhances
Figure 2.
Main types of soft nanocarriers for drug delivery. Schematic examples of surface modifications
and functionalization.
With the large body of evidence accumulated over the past decades, three important elements in
the development of controlled drug delivery can be defined: (1) utilization of the EPR eect for passive
targeting, (2) surface modification of NPs/nanocarriers to prolong their circulation time, (3) their
eective design [
171
]. Coating the surface of NPs with PEG (PEGylation), an FDA approved polymer,
is a commonly used approach for improving the eciency of drug and gene delivery to target cells
and tissues. PEGylation protects the surface from aggregation, opsonization, and phagocytosis and
prolongs systemic circulation time [
172
]. PEG is a hydrophilic, biocompatible, and biologically inert
polymer, which is used to enhance the stability of NPs [
173
]. Moreover, the coating allows loading of
specific ligands, antibodies, peptides, drugs, folic acid, aptamers, tumor markers, transferrin, vitamins,
etc., on the surface of particles oering an exciting tool to make NPs target-specific and increase
their therapeutic benefit [
174
176
]. These ligands can be easily conjugated onto PEG
´
s distal end
via various chemical coupling strategies [
177
]. Cancer cells overexpress various kinds of receptors
and antigens that may be used as potential drug targets in cancer therapy [
178
]. Entrapment of
the fluorescent compound in the NPs platform allows tracking the fate of NPs
in vitro
and monitoring
of the accumulation of the drug in the tumor and the treatment ecacy [
179
]. The Quantum Dots
(QDs) are fluorescent semiconductor nanocrystals of about 10 nm, composed of a core coated with
an envelope. QD-containing NPs have found applications in cancer medical imaging, cancer cell
tracking, cancer photodynamic therapy, and cancer diagnosis [180].
4.1.1. Liposomes
Liposomes (Figure 2A) are the most utilized nanomaterial for drug delivery, with many clinical
products currently available [
181
]. Liposomal vesicles consist of amphiphilic poly-molecular lipid
compounds that assemble into bi-layered self-closed spherical NPs [
182
]. Dierent structural
modifications of polar and non-polar parts of phospholipids have allowed the development of
various synthetic lipids with improved properties [
183
]. The advantage of liposomes is their ability to
Cancers 2020,12, 3622 15 of 32
deliver both hydrophilic and hydrophobic drugs. Surface functionalization of liposomes enhances
their stability and facilitates targeted drug, gene, or imaging agent delivery, even across the biological
and physiological barriers [
172
,
175
,
176
,
184
186
]. They are biocompatible, of low immunogenicity,
and they increase the solubility of a wide range of chemotherapeutics [
187
]. Several liposomal
formulations, namely Doxil
®
/Caelyx
(pegylated liposomal doxorubicin), Myocet
®
(non-pegylated
liposomal doxorubicin), DaunoXome
®
(liposomal daunorubicin), Marqibo
®
(liposomal vincristine),
Mepact®(liposomal mifamurtide)
, and Onivyde
®
(pegylated liposomal irinotecan) have already been
EMA and/or FDA-approved for the treatment of various cancer types, including BC [164,188].
To improve the ecacy of HDACI in BCs and other solid tumors, PEGylated liposomes were
prepared for encapsulation of trichostatin A (TSA), CG1521 (CG), and PXD101 (PXD) (Table 2) [
189
].
These liposome formulations are promising nanocarriers as they remained stable in size, charge,
and biological activity for one month when stored at 4 C. Wang et al. utilized the iron complexation
technique on VOR and LAQ824 before encapsulating them in PEGylated liposomes [
190
]. This strategy
resulted in an improved aqueous solubility of these drugs as well as liposomal encapsulation eciency.
Table 2. Nanoplatforms for epigenetic drug delivery.
Nanocarrier Loaded Drug Reference
PEGylated liposomes trichostatin A, CG1521, and PXD101 [189]
PEGylated liposomes with Fe complex VOR and LAQ824 [190]
Hybrid lipid-polymer NPs DAC [191,192]
Solid lipid NPs VOR [193]
Solid lipid NPs decorated with hyaluronic acid VOR [194]
Norbornene polyethylene oxide macromonomer CI-994 (tacedinaline) [195]
POEG blocks VOR [196]
PLGA NPs decorated with PGON belinostat and VOR [197,198]
PLGE-PEG nano-micelles AZA [199]
PEG-PLA di-block copolymer DAC [200]
Gelatinases-stimuli di-block copolymers (PEG, PCL) DAC [201,202]
LGE block copolymer VOR [203]
Lipid-polymer (DSPE-PEG-COOH-
PLGA-lecithin-PEG) core-shell NPs VOR and quisinostat [204]
Nanogels DAC [205]
4.1.2. Solid Lipid Nanoparticles
SLNs (Figure 2B) consist of a hydrophobic lipid core composed of lipids (e.g., fatty acids,
steroids, waxes, or triglycerides) that are in the solid state at room temperature. The lipid core is
stabilized by surfactants (emulsifiers) to prevent particle agglomeration [
206
]. The advantage of
SLNs is the easy entrapment of hydrophobic drugs in the lipidic matrix, while the outer hydrophilic
shell allows the conjugation of hydrophilic drugs to lipidic components. The solid state of the lipid
permits better-controlled drug release and improves the stability of the drug. Moreover, SLNs can be
administered by various routes, including systemic injection, oral, transdermal, pulmonary, and ocular
application [
168
]. SLNs can be modified to exhibit various advantages over liposomes and polymeric
NPs; they are non-toxic, biocompatible, biodegradable, and highly stable [207].
Lipid-based nanostructures as carriers of the potential oral delivery of DAC were designed and
synthesized by Neupane et al. [
191
] An ex vivo gut permeation study showed a nearly four-fold
increment in the drug
´
s permeation from the nanostructured lipid carrier compared with the plain
DAC solution and better accumulation of DAC encapsulated in lipid 4 h after oral administration
Cancers 2020,12, 3622 16 of 32
compared with i.v. injection of free DAC into the tumor. Based on these results, the authors suppose
that lipid-based nanocarrier systems represent a promising cancer cell treatment strategy through oral
delivery. Solid lipids also represent promising nanocarriers for HDACI delivery. Encapsulation of VOR
into solid lipid NPs enhanced its bioavailability and reduced the drug clearance rate compared to free
VOR
in vivo
, whether administered orally or i.v. and increased VOR’s toxicity in multidrug-resistant
BC cells
in vitro
[
193
,
194
,
208
,
209
]. Cyclin-dependent kinase 4 (CDK4) siRNA packaged in a lipid NP
(LNP)-based delivery system that consists of an ionizable cationic lipid, helper lipid, and polyethylene
glycol (PEG)-lipid showed a 16-fold increase in intracellular uptake of siRNA by BC cells and
a significant G1 cell cycle arrest due to ecient down-regulation of CDK4 at both mRNA and protein
levels [
210
]. The ncRNA nano-based drug patirisan (Onpattro
®
) has recently been approved by
the FDA (in 2018) to treat polyneuropathy. It is the first clinically approved example of an RNAi
therapy-delivering NP administered intravenously (i.v.) and the first FDA-approved RNAi therapeutic
in general [
211
]. ALN-18328, the active ingredient of patirisan, is formulated as a lipid nanoparticle to
targeted delivery to hepatocytes.
4.1.3. Polymeric Nanoparticles
Polymeric NPs (Figure 2C) are the simplest form of soft-materials for nanobiomedical
applications [
212
]. They can be composed of synthetic biodegradable polymers, some of which
were already approved by the FDA for tissue engineering scaolds and drug delivery vehicles [
213
].
Alternatively, they can be made of natural polymers such as chitosan, gelatin alginate, and albumin.
The advantages of polymeric NPs include the capacity to encapsulate both hydrophilic and hydrophobic
drugs and straightforward activation with specific molecules for targeted delivery to the tumor
mass [
214
]. While synthetic polymers allow sustained drug release, within a period of days to weeks,
natural polymers are more easily and rapidly degraded [
215
]. Polymeric NPs are stable, bioavailable,
have prolonged circulation time in peripheral blood, enable controlled drug release, and, compared to
liposomes, they possess superior drug loading capacities [
216
]. There are several FDA-approved
polymeric nano-drugs, such as Adagen
®
, Cimzia
®
, Macugen
®
, Neulasta
®
, Pegasys
®
, PegIntron
®
,
Renagel®, and Somavert®[212].
An innovative aspect of the nano-based delivery platform is the conjugation of CI-994 (tacedinaline)
through a clickable acid-responsive linker to a macromonomer. Such a prodrug monomer could
be polymerized to create a formulation that releases the HDACI in a pH-dependent manner while
minimizing
in vivo
drug release during blood circulation [
195
]. A pendant SAHA derivatized polymer
able to form spherical micelles is a promising novel prodrug carrier. This nanoformulation retains
the pharmacological activity of SAHA, inhibits the proliferation of tumor cells, and induces histone
acetylation [
196
]. Belinostat-loaded polymer NPs, decorated with a novel cell-penetrating polymer,
poly(guanidinium oxanorbornene), significantly enhanced intracellular uptake,
decreased the tumor
volume, and increased the intratumoral acetylation of histone H4 [
197
]. The safety of orally administered
SAHA-loaded polymer NPs manifested low cytotoxicity in lung cancer cells and favorable
biodistribution patterns
in vivo
[
198
]. A VOR nanofiber-coated stent was shown to be a promising
candidate for cholangiocarcinoma treatment [
217
]. Using the emulsion-solvent diusion technique,
Alp et al
. have developed a biocompatible starch NP formulation for delivery of CG-1521 to
hard-to-treat BCs [
218
]. This formulation improves the bioavailability and half-life of this HDACI
without aecting the drug´s mechanism of action.
A stable nanosized ribophorin II (RPN2) siRNA–atelocollagen complex was prepared to improve
the
in vitro
and
in vivo
resistance of BC cells to DOX and taxanes. RPN2 silencing caused reduced
glycosylation of the P-glycoprotein in vitro and markedly reduced tumor growth in vivo [219].
4.1.4. Polymeric Micelles
Polymeric micelles (Figure 2D) are supramolecular delivery systems composed of multiple
amphiphilic block copolymers, self-assembling in aqueous environments at a defined concentration [
206
].
Cancers 2020,12, 3622 17 of 32
The micelles
´
core-shell structure presents a hydrophobic core and the outer hydrophilic shell. The core
can be tailored for the controlled delivery of hydrophobic drugs, while hydrophilic drugs can be adsorbed
or chemically linked to the shell [
9
]. Polymeric micelles have high stability and exhibit prolonged
circulation time in the blood [
168
]. Poly(ethylene glycol)–poly(lactic-co-glycolide) (PEG–PLGA) di-block
copolymer micelles represent one of the most promising biocompatible platforms for drug delivery [
220
].
Lipids can also be used to form the hydrophobic core of these hybrids for encapsulation of many types of
poorly water-soluble drugs [
221
]. FDA approved polymeric micelles, already used in clinical practice,
include Copaxone
®
, Eligard
®
, Estrasorb
, Oncaspar
®
, Krystexxa
®
, Plegridy
®
, and Adynovate
®
used
for the treatment of sclerosis, prostate cancer, and blood malignities [212].
The micelles are predominantly utilized as versatile nanocarriers for the delivery of DNMT
inhibitors. Encapsulation of AZA or DAC into micelles has increased their stability under physiological
conditions, markedly enhanced their therapeutic ecacy, provided controlled pH-dependent drug
release, significantly down-regulated DNMT1 and DNMT3b expression, and increased expression
of caspase-9 in murine xenograft models of BC [
199
,
200
]. Intelligent DAC-loaded micelles have
been prepared by inserting cancer-specific gelatinase-cleavable peptide between two polymers.
These micelles manifested superior cellular uptake in the tumor-bearing xenografts due to an active
(gelatinase-stimuli) targeting strategy and more ecient gene demethylation compared with their
counterparts lacking gelatinase features [
201
,
202
]. DAC-loaded hybrid lipid-polymer micelles were
eciently internalized by BC cells and rescued the expression of silenced tumor suppressor genes
in cancer cells [
192
]. Several micelle formulations have been proposed and tested as a promising
nanocarrier for HDAI delivery. Kwak et al. revealed higher ecacy of tumor growth inhibition,
enhanced drug accumulation, and higher inhibition rate of HDAC expression in a xenograft mice
model by VOR-encapsulated micelles compared with the free drug [
203
]. Similarly, higher therapeutic
ecacy was confirmed by VOR- or quisinostat-loaded hybrid lipid-polymer micelles compared with
free HDACIs [
204
]. These micelles suppressed DNA double-strand break repair in tumor cells and
showed a synergistic eect in mouse xenograft
in vivo
models. Micelles loaded with antisense-miR-21
and antisense-miR-10b were prepared to block the functions of endogenous miRNAs, regulating genes
involved in cell proliferation, dierentiation, and apoptosis. Treatment of mice bearing subcutaneous
tumor xenografts with this formulation resulted in a substantial tumor growth reduction, thus indicating
a potential new therapeutic approach for triple-negative BC [222].
4.1.5. Dendrimers
Dendrimers (Figure 2E) are three-dimensional, hyper-branched, or tree-like polymers having
a central core with repeated branches of interior layers and exterior terminal functionality. These structures
offer high availability and a wide range of molecular moieties for the chemical conjugations and
internal cavities to encapsulate drugs and nucleic acids [
223
]. Dendrimers can be used to deliver
various molecules (therapeutics, imaging, and targeting agents) in a single particle due to the presence
of different functional monomers [
220
]. Dendrimers are suitable drug-delivery systems because of
their nanometric size, ease of fabrication, monodispersity, lipophilicity, multidrug loading capacity,
controlled drug release, low toxicity, biocompatibility, and ability to easily penetrate cell membranes [
9
].
Among different commercially available dendrimers, diaminobutyric polypropylenimine (DAB),
polyamidoamine (PAMAM), poly (amidoamine-organosilicon) (PAMAMOS), poly (Lysine), and poly
(propylene imine) (PPI) are the most promising platforms for delivering cargos to target sites [223].
Finlay et al. used a modified poly (amidoamine) (PAMAM) dendrimer for delivery of siRNA,
targeting the TWIST1 transcription factor [
224
]. TWIST1 is often overexpressed in aggressive BCs and
is involved in regulating cell migration through EMT. These PAMAM-siRNA complexes significantly
down-regulated TWIST1 and EMT-related target genes
in vitro
as well as in xenograft orthotopic
tumors. PAMAM dendrimer was also used to deliver antisense oligonucleotides targeting the vascular
endothelial growth factor VEGF-ASODN to inhibit the tumor vascularization of breast tumor tissue
using a human breast tumor xenograft mice model [225].
Cancers 2020,12, 3622 18 of 32
4.1.6. Nanogels
Nanogels (Figure 2F) are three-dimensional polymeric networks with a high capacity for
water uptake. They are mostly made of synthetic polymers or biopolymers, which contain
hydrophilic and hydrophobic monomers, cross-linked by either physical or chemical bonds, which
influence many network properties, like swelling, elastic modulus, and transport properties [
226
,
227
].
In aqueous media, nanogels form semi-solid states (hydrogels). The porous network allows
high-drug entrapment eciency. Nanogels can carry various drugs with low or high molecular
weight and either hydrophilic or hydrophobic. The advantages of nanogels include high drug
encapsulation capacity, enhanced drug stability, minimal toxicity, biocompatibility, prolonged blood
circulation time, and stimulus responsiveness to drug release (pH, magnetic field, light, ionic content,
and temperature) [206].
Nanogels (NGs) have also been explored for treating solid tumors. DAC-loaded nanogels
decorated with PEG manifested sustained DNMT1 depletion, prolonged cancer cell arrest in the G2/M
cell-cycle phase, and significantly enhanced the antiproliferative eect of DAC [205].
4.2. Nanoplatforms for Combination Therapy
A promising strategy to achieve more significant therapeutic benefits appears to be the combination
therapy involving epi-drug-loaded NPs and either a chemotherapeutic-encapsulated nanocarrier or
free standard anticancer drugs, or co-packaging of epi-drugs and anticancer drugs in one nanoplatform.
Li et al. have recently shown that combination therapy with low-dose DAC-loaded NPs and
DOX-loaded NPs is more eective and opens up new possibilities for the management of BC [
200
].
In line with these results, Vijayaraghavalu et al. have revealed that a combination of epigenetic drugs
(DAC +SAHA) encapsulated in biodegradable NGs more eectively overcomes drug resistance than the
same drugs in solution [
228
]. Moreover, pretreatment with epigenetic drugs in nanogels, then with DOX
in nanogels, was most eective in overcoming resistance even at low doses of DOX. Co-packaging DAC
and arsenic trioxide (ATO) into alendronate-conjugated bone-targeting hybrid lipid-polymer micelles
should enhance the synergistic eect of DAC and ATO in the treatment of MDS and reduce systemic
toxicity of chemotherapeutics [
229
]. Using co-axial and multi-needle electrohydrodynamic atomization
(EHDA) technology, Parhizkaz et al. have co-encapsulated DAC and cisplatin in a single PLGA-based
nanocarrier [
230
]. The EHDA technique enables fine-tuning of individual drug release characteristics,
with rapid DAC and slower cisplatin release, thus achieving the maximum synergistic therapeutic eect
and overcoming the chemoresistance to cisplatin. Lysophosphatidic acid receptor 1 (LPAR1)-targeted
lipid nanoemulsions (nanoscale oil-in-water emulsions) were developed for encapsulation of two
drugs with dierent chemistries, DAC (hydrophilic) and panobinostat (PAN–HDACI, hydrophobic)
and targeted co-delivery in triple-negative BC tissues [231].
Pendant SAHA derivatized polymer (POEG-b-PSAHA) micelles were used as nanocarriers for
DOX delivery. A DOX/POEG-b-PSAHA formulation resulted in an improved therapeutic eect
in vivo
compared to free DOX, Doxil, or POEG-b-POM-loaded DOX micelles, indicating that SAHA-based
prodrug micelles may serve as a dual functional carrier for combination strategies in epigenetic-oriented
anticancer therapy [
196
]. Ruttala et al. developed a transferrin-anchored albumin nanoplatform
with PEGylated lipid bilayers (Tf-L-APVN) for the targeted co-delivery of paclitaxel and VOR in
solid tumors [
232
]. At
in vitro
conditions, Tf-L-APVN significantly enhanced the synergistic eects of
paclitaxel and VOR on the proliferation of breast and liver cancer cells. In HepG2 tumor-bearing mice,
the co-delivery of paclitaxel and VOR in one nano-based platform significantly inhibited the tumor
growth, thus oering great potential in the chemotherapy of solid tumors.
Using a layer-by-layer approach, Deng et al. generated a siRNA-loaded film superimposed on
a simple DOX-loaded liposome [
233
]. They showed that combination therapy with siRNA targeting
multidrug resistance protein 1 (MRP1) significantly enhanced DOX ecacy
in vitro
, and
in vivo
resulted in a substantial reduction in the tumor of tumors, which otherwise are nonresponsive to
treatment with DOX.
Cancers 2020,12, 3622 19 of 32
5. Biosafety of Soft Nanocarriers
Despite the many advantages of using nano-based delivery systems, there are also significant
drawbacks that must be considered, as they might present a real challenge for clinical use [
186
].
Currently, there is very little regulatory guidance in the field of biomedical applications of NPs [
234
].
Although the EPR eect has been described as a basis for successful passive targeting of tumors,
the complexity of this process must also be highlighted. The EPR eect can be influenced by
the interactions of NPs with the biological systems, including proteins, blood flow, and the tumor
microenvironment. Nanoparticle properties, including size, shape, surface properties, porosity,
and structure, can aect the EPR eect [
235
]. For example, it has been reported that the high interstitial
fluid pressure in tumors can hinder the accumulation of nanocarriers since the high density of
the extracellular matrix can reduce the chances of NP penetration [236].
It is also dicult to predict the behavior and responses of nanocarrier interactions with biological
systems during the drug delivery process [
220
]. For instance, the interaction of NPs with the renal
system and its role in blood clearance is a crucial aspect to be taken into account. An ecient
bloodstream clearance is important to avoid NPs accumulating, which might lead to adverse long-term
eects. The eciency of kidney clearance has been reported to be aected by small changes in the size
of the NPs. At the same time, unspecific clearance, possibly leading to drug release far from the tumor
site, must also be avoided [237,238].
Thus, the nanocarriers
´
possible toxicity must be taken into account and thoroughly
investigated [
239
]. Nanomaterials pose particular challenges in the evaluation of their toxicity
in comparison with chemical substances. Besides the chemical composition, the particles physical
properties (e.g., their size, shape, agglomeration/aggregation, solubility/dispersibility, surface charge,
redox potential) play an important role in eliciting biological responses [
240
]. In this sense, a proper
hazard and risk assessment needs to be applied. This means that external and internal exposures, as
well as cellular uptake of the NPs, must be defined. In order to investigate all the possible pathways
through which toxicity might arise, a battery of relevant toxicity assays should be applied. As NPs
have been reported to de-regulate gene expression and aect epigenetic mechanisms involved in
several biological processes, including cancer development [
44
,
165
], these eects have to be taken into
consideration in addition to cytotoxicity and genotoxicity.
Attention must also be given to the choice of exposure models. Although the use of
in vivo
models cannot yet be completely excluded, alternative
in vitro
tests can be valid substitutes for initial
screenings of cyto- and genotoxicity and for investigating the underlying molecular mechanisms
of toxicity of nanomaterials. More advanced and relevant
in vitro
models to assess ecacy and
safety have been developed, moving beyond the traditional 2D monocultural models, towards 3D,
e.g., spherical tumor
models, and more complex multicellular models, which better resemble and
mimic real-life conditions [
241
]. Several nanomaterials have not yet had their potential adverse
biological eects fully assessed due to costs and time constraints associated with the experimental
assessment, frequently involving animals [242].
There are OECD test guidelines for
in vitro
assays adapted to the testing of NPs.
In vitro
assays
are usually robust, fast, and cost-eective compared to
in vivo
methods, and several high-throughput
screening methods for relevant cells from humans and other mammals are available to study the eects
of NPs [
243
,
244
]. Nowadays,
in vitro
assays adapted for testing nanomaterials are performed in
a controlled manner, taking physicochemical characterization and cellular uptake into consideration.
However, such tests need further validation because of putative NM-induced interferences [245].
To fully understand the advantages and disadvantages of NP-based therapeutics, more clinical
data are needed to identify the best applications for nano-chemotherapeutics [
13
]. In the field of cancer
nanomedicine research, it is important to take into account the complex heterogeneity within and
among human tumors and to focus on the design of precision nanomedicines to achieve personalized
cancer treatments [
246
]. In this sense, understanding which physicochemical properties are coupled
with adverse eects is critical for designing safer and more eective nanomaterials for cancer treatment.
Cancers 2020,12, 3622 20 of 32
This so-called “Safe(r)-by-Design” approach requires the implementation of safety evaluation early
in the development of nanomaterials [
247
], together with understanding their toxicokinetics and
modulation of the immune system [248250].
6. Conclusions
In BC, there are tumor subtypes, which are considered to be hard to cure, and do not respond to
standard therapeutic agents, and have a high recurrence probability. In recent decades, the intense
study of epigenetic deregulation in many cancers has uncovered the potential of new therapeutic
approaches, targeting reversible changes in the epigenome. The limited tolerability, low ecacy,
and o-target eects of most epi-drugs remain significant challenges in this experimental approach as
applied to solid tumors. The exploration of lower doses, sequential scheduling, and their targeted
delivery might considerably improve the therapeutic index. In this regard, nanotechnology has
revolutionized the field of drug delivery for epigenetic therapy of solid tumors. Dierent formulations
of soft nanocarriers have been employed to increase the stability, solubility, and specificity of DNMT
and histone inhibitors as well as non-coding RNAs. In addition, conventional chemotherapeutic agents
have been successfully co-packed with epi-drugs in nanocarriers for combination therapy. Data from
preclinical and clinical trials of nano-based epi-drugs for BC treatment demonstrate reduced systemic
toxicity and improved ecacy compared to conventional free-drug formulations.
Furthermore, nanomedicine offers the possibility to refine the detection, diagnosis, and conventional
therapy of many cancers by combining different agents in a single multi-component nano-drug.
Although promising, nano-based delivery systems should follow the Safe(r)-by-Design approach and be
rigorously tested at all phases of development to prevent adverse health effects and environmental hazards.
The use of nano-based delivery systems, together with the benefits of a new generation of epi-drugs
and the emergence of robust biomarker data, will help to advance personalized-targeted therapy and
the ecacy of treatment for breast and other cancers.
Author Contributions:
Conceptualization, B.S., A.G., and V.B.; Writing—original draft preparation and literature
search, V.B., B.S., A.G., M.M., E.P., A.C., K.P., M.R., A.S., A.P., M.F., E.M.L., N.E.Y., and S.M.; Writing—review and
editing, M.R.C., I.R.-M., V.P., and M.D.; Supervision, V.P.; Visualization, B.S. and V.B. All authors have read and
agreed to the published version of the manuscript.
Funding:
This research was funded by ERA-NET EuroNanoMed II INNOCENT, by the Slovak Research and
Development Agency (APVV), grant No. APVV-16-0010, APVV-16-0178, by the European Union’s Horizon 2020
research and innovation program under grant agreement No. 857381, project VISION (Strategies to strengthen
scientific excellence and innovation capacity for early diagnosis of gastrointestinal cancers), by ERA-NET
EuroNanoMed III CELLUX, H2020-NMBP-TO-IND-2019 SABYDOMA, contract No. 862296 and by Scientific
Grant Agency (VEGA), contracts No. 2/0271/17, 2/0138/20 and 2/0052/18.
Acknowledgments:
We are grateful to Andrew Collins for his critical reading and English corrections.
Graphical abstract and Figure 2were created with BioRender.com.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018:
GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer
J. Clin. 2018,68, 394–424. [CrossRef] [PubMed]
2.
Dworkin, A.M.; Huang, T.H.M.; Toland, A.E. Epigenetic alterations in the breast: Implications for breast
cancer detection, prognosis and treatment. Semin. Cancer Biol. 2009,19, 165–171. [CrossRef] [PubMed]
3. Roche, J. The Epithelial-to-Mesenchymal Transition in Cancer. Cancers 2018,10, 52. [CrossRef] [PubMed]
4.
Sun, L.; Fang, J. Epigenetic regulation of epithelial-mesenchymal transition. Cell. Mol. Life Sci.
2016
,
73, 4493–4515. [CrossRef] [PubMed]
5.
Moo, T.A.; Sanford, R.; Dang, C.; Morrow, M. Overview of Breast Cancer Therapy. PET Clin.
2018
,13, 339–354.
[CrossRef] [PubMed]
Cancers 2020,12, 3622 21 of 32
6.
Lu, Y.; Chan, Y.T.; Tan, H.Y.; Li, S.; Wang, N.; Feng, Y. Epigenetic regulation in human cancer: The potential
role of epi-drug in cancer therapy. Mol. Cancer 2020,19, 79. [CrossRef] [PubMed]
7.
Morel, D.; Jeery, D.; Aspeslagh, S.; Almouzni, G.; Postel-Vinay, S. Combining epigenetic drugs with other
therapies for solid tumours—Past lessons and future promise. Nat. Rev. Clin. Oncol.
2020
,17, 91–107.
[CrossRef] [PubMed]
8.
Magro, M.; Venerando, A.; Macone, A.; Canettieri, G.; Agostinelli, E.; Vianello, F. Nanotechnology-Based
Strategies to Develop New Anticancer Therapies. Biomolecules 2020,10, 735. [CrossRef] [PubMed]
9.
Jain, V.; Kumar, H.; Anod, H.V.; Chand, P.; Gupta, N.V.; Dey, S.; Kesharwani, S.S. A review of
nanotechnology-based approaches for breast cancer and triple-negative breast cancer. J. Control. Release
2020
,
326, 628–647. [CrossRef] [PubMed]
10.
Thakur, V.; Kutty, R.V. Recent advances in nanotheranostics for triple negative breast cancer treatment. J. Exp.
Clin. Cancer Res. 2019,38, 430. [CrossRef] [PubMed]
11.
Hobbs, S.K.; Monsky, W.L.; Yuan, F.; Roberts, W.G.; Grith, L.; Torchilin, V.P.; Jain, R.K. Regulation of
transport pathways in tumor vessels: Role of tumor type and microenvironment. Proc. Natl. Acad. Sci. USA
1998,95, 4607–4612. [CrossRef] [PubMed]
12.
Ernsting, M.J.; Murakami, M.; Roy, A.; Li, S.D. Factors controlling the pharmacokinetics, biodistribution and
intratumoral penetration of nanoparticles. J. Control. Release 2013,172, 782–794. [CrossRef]
13.
Navya, P.N.; Kaphle, A.; Srinivas, S.P.; Bhargava, S.K.; Rotello, V.M.; Daima, H.K. Current trends and
challenges in cancer management and therapy using designer nanomaterials. Nano Converg.
2019
,6, 23.
[CrossRef] [PubMed]
14.
Gao, Y.; Tang, M.; Leung, E.; Svirskis, D.; Shelling, A.; Wu, Z. Dual or multiple drug loaded nanoparticles to
target breast cancer stem cells. RSC Adv. 2020,10, 19089–19105. [CrossRef]
15.
Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.;
Cardoso, F. Breast cancer. Nat. Rev. Dis. Primers 2019,5, 66. [CrossRef] [PubMed]
16.
Howlader, N.; Noone, A.; Krapcho, M. SEER Cancer Statistics Review, 1975–2014, Based on November 2016
SEER Data Submission; National Cancer Institute: Bethesda, MD, USA, 2017.
17.
Goncalves, H., Jr.; Guerra, M.R.; Duarte Cintra, J.R.; Fayer, V.A.; Brum, I.V.; Bustamante Teixeira, M.T.
Survival Study of Triple-Negative and Non-Triple-Negative Breast Cancer in a Brazilian Cohort. Clin. Med.
Insights Oncol. 2018,12. [CrossRef]
18.
Hinohara, K.; Polyak, K. Intratumoral heterogeneity: More than just mutations. Trends Cell Biol.
2019
,
29, 569–579. [CrossRef] [PubMed]
19.
Koren, S.; Bentires-Alj, M. Breast Tumor Heterogeneity: Source of Fitness, Hurdle for Therapy. Mol. Cell
2015,60, 537–546. [CrossRef] [PubMed]
20. Turashvili, G.; Brogi, E. Tumor Heterogeneity in Breast Cancer. Front. Med. 2017,4, 227. [CrossRef]
21.
Taylor, A.; Brady, A.F.; Frayling, I.M.; Hanson, H.; Tischkowitz, M.; Turnbull, C.; Side, L. Consensus for genes
to be included on cancer panel tests oered by UK genetics services: Guidelines of the UK Cancer Genetics
Group. J. Med. Genet. 2018,55, 372–377. [CrossRef]
22.
Vogelstein, B.; Kinzler, K.W. Cancer genes and the pathways they control. Nat. Med.
2004
,10, 789–799.
[CrossRef]
23.
Desmedt, C.; Yates, L.; Kulka, J. Catalog of genetic progression of human cancers: Breast cancer.
Cancer Metastasis Rev. 2016,35, 49–62. [CrossRef] [PubMed]
24.
Nik-Zainal, S.; Davies, H.; Staaf, J.; Ramakrishna, M.; Glodzik, D.; Zou, X.; Martincorena, I.; Alexandrov, L.B.;
Martin, S.; Wedge, D.C.; et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences.
Nature 2016,534, 47–54. [CrossRef] [PubMed]
25.
Dawson, M.A.; Kouzarides, T. Cancer epigenetics: From mechanism to therapy. Cell
2012
,150, 12–27.
[CrossRef]
26.
Sharma, S.; Kelly, T.K.; Jones, P.A. Epigenetics in cancer. Carcinogenesis
2010
,31, 27–36. [CrossRef] [PubMed]
27. Kanwal, R.; Gupta, S. Epigenetic modifications in cancer. Clin. Genet. 2012,81, 303–311. [CrossRef]
28.
Veeck, J.; Esteller, M. Breast cancer epigenetics: From DNA methylation to microRNAs. J. Mammary Gland
Biol. Neoplasia 2010,15, 5–17. [CrossRef]
29.
Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology
2013
,38, 23–38.
[CrossRef]
Cancers 2020,12, 3622 22 of 32
30.
Esteller, M. Epigenetic gene silencing in cancer: The DNA hypermethylome. Hum. Mol. Genet.
2007
,
16, R50–R59. [CrossRef]
31.
Laird, P.W. The power and the promise of DNA methylation markers. Nat. Rev. Cancer
2003
,3, 253–266.
[CrossRef]
32.
Guo, M.; Peng, Y.; Gao, A.; Du, C.; Herman, J.G. Epigenetic heterogeneity in cancer. Biomark Res.
2019
,7, 23.
[CrossRef] [PubMed]
33.
De Almeida, B.P.; Apol
ó
nio, J.D.; Binnie, A.; Castelo-Branco, P. Roadmap of DNA methylation in breast
cancer identifies novel prognostic biomarkers. BMC Cancer 2019,19, 219. [CrossRef] [PubMed]
34.
Irizarry, R.A.; Ladd-Acosta, C.; Wen, B.; Wu, Z.; Montano, C.; Onyango, P.; Cui, H.; Gabo, K.; Rongione, M.;
Webster, M.; et al. The human colon cancer methylome shows similar hypo- and hypermethylation at
conserved tissue-specific CpG island shores. Nat. Genet. 2009,41, 178–186. [CrossRef]
35.
Hinshelwood, R.A.; Clark, S.J. Breast cancer epigenetics: Normal human mammary epithelial cells as a model
system. J. Mol. Med. 2008,86, 1315–1328. [CrossRef] [PubMed]
36.
Jovanovic, J.; Rønneberg, J.A.; Tost, J.; Kristensen, V. The epigenetics of breast cancer. Mol. Oncol.
2010
,
4, 242–254. [CrossRef] [PubMed]
37.
Pasculli, B.; Barbano, R.; Parrella, P. Epigenetics of breast cancer: Biology and clinical implication in the era
of precision medicine. Semin. Cancer Biol. 2018,51, 22–35. [CrossRef] [PubMed]
38.
Tahiliani, M.; Koh, K.P.; Shen, Y.; Pastor, W.A.; Bandukwala, H.; Brudno, Y.; Agarwal, S.; Iyer, L.M.; Liu, D.R.;
Aravind, L.; et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL
partner TET1. Science 2009,324, 930–935. [CrossRef]
39.
Zhang, H.; Zhang, X.; Clark, E.; Mulcahey, M.; Huang, S.; Shi, Y.G. TET1 is a DNA-binding protein that
modulates DNA methylation and gene transcription via hydroxylation of 5-methylcytosine. Cell Res.
2010
,
20, 1390–1393. [CrossRef]
40.
Huang, Y.; Rao, A. Connections between TET proteins and aberrant DNA modification in cancer. Trends
Genet. 2014,30, 464–474. [CrossRef]
41.
Audia, J.E.; Campbell, R.M. Histone Modifications and Cancer. Cold Spring Harb. Perspect. Biol.
2016
,
8, a019521. [CrossRef]
42.
Chervona, Y.; Costa, M. Histone modifications and cancer: Biomarkers of prognosis? Am. J. Cancer Res.
2012
,
2, 589–597. [PubMed]
43.
Kurdistani, S.K. Histone modifications as markers of cancer prognosis: A cellular view. Br. J. Cancer
2007
,
97, 1–5. [CrossRef] [PubMed]
44.
Stoccoro, A.; Karlsson, H.L.; Copped
è
, F.; Migliore, L. Epigenetic eects of nano-sized materials. Toxicology
2013,313, 3–14. [CrossRef]
45.
Horn, P.J.; Peterson, C.L. Heterochromatin assembly: A new twist on an old model. Chromosome Res.
2006
,
14, 83–94. [CrossRef] [PubMed]
46.
Yan, W.; Herman, J.G.; Guo, M. Epigenome-based personalized medicine in human cancer. Epigenomics
2016
,
8, 119–133. [CrossRef] [PubMed]
47.
Fraga, M.F.; Ballestar, E.; Villar-Garea, A.; Boix-Chornet, M.; Espada, J.; Schotta, G.; Bonaldi, T.; Haydon, C.;
Ropero, S.; Petrie, K.; et al. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common
hallmark of human cancer. Nat. Genet. 2005,37, 391–400. [CrossRef] [PubMed]
48.
Elsheikh, S.E.; Green, A.R.; Rakha, E.A.; Powe, D.G.; Ahmed, R.A.; Collins, H.M.; Soria, D.; Garibaldi, J.M.;
Paish, C.E.; Ammar, A.A.; et al. Global histone modifications in breast cancer correlate with tumor phenotypes,
prognostic factors, and patient outcome. Cancer Res. 2009,69, 3802–3809. [CrossRef]
49.
Kapoor-Vazirani, P.; Kagey, J.D.; Powell, D.R.; Vertino, P.M. Role of hMOF-dependent histone H4 lysine
16 acetylation in the maintenance of TMS1/ASC gene activity. Cancer Res. 2008,68, 6810–6821. [CrossRef]
50.
Karsli-Ceppioglu, S.; Dagdemir, A.; Judes, G.; Ngollo, M.; Penault-Llorca, F.; Pajon, A.; Bignon, Y.J.;
Bernard-Gallon, D. Epigenetic mechanisms of breast cancer: An update of the current knowledge. Epigenomics
2014,6, 651–664. [CrossRef]
51.
Marmorstein, R.; Zhou, M.M. Writers and readers of histone acetylation: Structure, mechanism, and inhibition.
Cold Spring Harb. Perspect. Biol. 2014,6, a018762. [CrossRef]
52.
Yang, Z.; He, N.; Zhou, Q. Brd4 recruits P-TEFb to chromosomes at late mitosis to promote G1 gene expression
and cell cycle progression. Mol. Cell. Biol. 2008,28, 967–976. [CrossRef] [PubMed]
Cancers 2020,12, 3622 23 of 32
53.
You, J.S.; Jones, P.A. Cancer genetics and epigenetics: Two sides of the same coin? Cancer Cell
2012
,22, 9–20.
[CrossRef] [PubMed]
54.
Hosseini, A.; Minucci, S. Chapter 6—Alterations of histone modifications in cancer. In Epigenetics in Human
Disease, 2nd ed.; Tollefsbol, T.O., Ed.; Academic Press: Cambridge, MA, USA, 2018; Volume 6, pp. 141–217.
55.
Yang, X.; Karuturi, R.K.; Sun, F.; Aau, M.; Yu, K.; Shao, R.; Miller, L.D.; Tan, P.B.; Yu, Q. CDKN1C (p57) is
a direct target of EZH2 and suppressed by multiple epigenetic mechanisms in breast cancer cells. PLoS ONE
2009,4, e5011. [CrossRef]
56.
Shi, L.; Sun, L.; Li, Q.; Liang, J.; Yu, W.; Yi, X.; Yang, X.; Li, Y.; Han, X.; Zhang, Y.; et al. Histone demethylase
JMJD2B coordinates H3K4/H3K9 methylation and promotes hormonally responsive breast carcinogenesis.
Proc. Natl. Acad. Sci. USA 2011,108, 7541–7546. [CrossRef] [PubMed]
57.
Wang, Y.; Zhang, H.; Chen, Y.; Sun, Y.; Yang, F.; Yu, W.; Liang, J.; Sun, L.; Yang, X.; Shi, L.; et al. LSD1 is
a subunit of the NuRD complex and targets the metastasis programs in breast cancer. Cell
2009
,138, 660–672.
[CrossRef] [PubMed]
58.
Wei, J.W.; Huang, K.; Yang, C.; Kang, C.S. Non-coding RNAs as regulators in epigenetics (Review). Oncol. Rep.
2017,37, 3–9. [CrossRef]
59.
Peschansky, V.J.; Wahlestedt, C. Non-coding RNAs as direct and indirect modulators of epigenetic regulation.
Epigenetics 2014,9, 3–12. [CrossRef]
60.
Ferreira, H.J.; Esteller, M. Non-coding RNAs, epigenetics, and cancer: Tying it all together.
Cancer Metastasis Rev. 2018,37, 55–73. [CrossRef]
61.
Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther.
2016
,1, 15004.
[CrossRef]
62.
Zhang, W.; Liu, J.; Wang, G. The role of microRNAs in human breast cancer progression. Tumor Biol.
2014
,
35, 6235–6244. [CrossRef]
63.
Kasinski, A.L.; Slack, F.J. Epigenetics and genetics. MicroRNAs en route to the clinic: Progress in validating
and targeting microRNAs for cancer therapy. Nat. Rev. Cancer 2011,11, 849–864. [CrossRef] [PubMed]
64.
Bertoli, G.; Cava, C.; Castiglioni, I. MicroRNAs: New Biomarkers for Diagnosis, Prognosis, Therapy Prediction
and Therapeutic Tools for Breast Cancer. Theranostics 2015,5, 1122–1143. [CrossRef] [PubMed]
65.
Van Roosbroeck, K.; Calin, G.A. Cancer Hallmarks and MicroRNAs: The Therapeutic Connection.
Adv. Cancer Res. 2017,135, 119–149. [CrossRef] [PubMed]
66.
Rahman, M.M.; Brane, A.C.; Tollefsbol, T.O. MicroRNAs and Epigenetics Strategies to Reverse Breast Cancer.
Cells 2019,8, 1214. [CrossRef]
67.
Søkilde, R.; Persson, H.; Ehinger, A.; Pirona, A.C.; Fernö, M.; Hegardt, C.; Larsson, C.; Loman, N.;
Malmberg, M.; Ryd
é
n, L.; et al. Refinement of breast cancer molecular classification by miRNA expression
profiles. BMC Genom. 2019,20, 503. [CrossRef] [PubMed]
68.
Chan, M.; Liaw, C.S.; Ji, S.M.; Tan, H.H.; Wong, C.Y.; Thike, A.A.; Tan, P.H.; Ho, G.H.; Lee, A.S. Identification
of circulating microRNA signatures for breast cancer detection. Clin. Cancer Res.
2013
,19, 4477–4487.
[CrossRef]
69.
Martinez-Gutierrez, A.D.; Cant
ú
de Le
ó
n, D.; Millan-Catalan, O.; Coronel-Hernandez, J.; Campos-Parra, A.D.;
Porras-Reyes, F.; Exayana-Alderete, A.; L
ó
pez-Camarillo, C.; Jacobo-Herrera, N.J.; Ramos-Payan, R.
Identification of miRNA Master Regulators in Breast Cancer. Cells 2020,9, 1610. [CrossRef]
70.
Markou, A.; Yousef, G.M.; Stathopoulos, E.; Georgoulias, V.; Lianidou, E. Prognostic significance of
metastasis-related microRNAs in early breast cancer patients with a long follow-up. Clin. Chem.
2014
,
60, 197–205. [CrossRef]
71.
Tormo, E.; Ballester, S.; Adam-Artigues, A.; Burgu
é
s, O.; Alonso, E.; Bermejo, B.; Men
é
ndez, S.; Zazo, S.;
Madoz-G
ú
rpide, J.; Rovira, A.; et al. The miRNA-449 family mediates doxorubicin resistance in triple-negative
breast cancer by regulating cell cycle factors. Sci. Rep. 2019,9, 5316. [CrossRef]
72.
Lavin, D.P.; Tiwari, V.K. Unresolved Complexity in the Gene Regulatory Network Underlying EMT.
Front. Oncol. 2020,10, 554. [CrossRef]
73.
Felipe Lima, J.; Nofech-Mozes, S.; Bayani, J.; Bartlett, J.M. EMT in Breast Carcinoma—A Review. J. Clin. Med.
2016,5, 65. [CrossRef] [PubMed]
74.
Miettinen, P.J.; Ebner, R.; Lopez, A.R.; Derynck, R. TGF-beta induced transdierentiation of mammary
epithelial cells to mesenchymal cells: Involvement of type I receptors. J. Cell Biol.
1994
,127, 2021–2036.
[CrossRef] [PubMed]
Cancers 2020,12, 3622 24 of 32
75.
Akalay, I.; Janji, B.; Hasmim, M.; Noman, M.Z.; Andr
é
, F.; De Cremoux, P.; Bertheau, P.; Badoual, C.; Vielh, P.;
Larsen, A.K.; et al. Epithelial-to-mesenchymal transition and autophagy induction in breast carcinoma
promote escape from T-cell-mediated lysis. Cancer Res. 2013,73, 2418–2427. [CrossRef] [PubMed]
76.
Dongre, A.; Rashidian, M.; Reinhardt, F.; Bagnato, A.; Keckesova, Z.; Ploegh, H.L.; Weinberg, R.A.
Epithelial-to-Mesenchymal Transition Contributes to Immunosuppression in Breast Carcinomas. Cancer Res.
2017,77, 3982–3989. [CrossRef] [PubMed]
77.
Moreno-Bueno, G.; Portillo, F.; Cano, A. Transcriptional regulation of cell polarity in EMT and cancer.
Oncogene 2008,27, 6958–6969. [CrossRef]
78.
Padmanaban, V.; Krol, I.; Suhail, Y.; Szczerba, B.M.; Aceto, N.; Bader, J.S.; Ewald, A.J. E-cadherin is required
for metastasis in multiple models of breast cancer. Nature 2019,573, 439–444. [CrossRef]
79.
Cheung, K.J.; Padmanaban, V.; Silvestri, V.; Schipper, K.; Cohen, J.D.; Fairchild, A.N.; Gorin, M.A.; Verdone,J.E.;
Pienta, K.J.; Bader, J.S.; et al. Polyclonal breast cancer metastases arise from collective dissemination of
keratin 14-expressing tumor cell clusters. Proc. Natl. Acad. Sci. USA 2016,113, E854–E863. [CrossRef]
80.
Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.; Choi, H.; El Rayes, T.; Ryu, S.; Troeger, J.; et al.
Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance.
Nature 2015,527, 472–476. [CrossRef]
81.
Ye, X.; Brabletz, T.; Kang, Y.; Longmore, G.D.; Nieto, M.A.; Stanger, B.Z.; Yang, J.; Weinberg, R.A.
Upholding a role for EMT in breast cancer metastasis. Nature 2017,547, E1–E3. [CrossRef]
82.
Brabletz, T. To dierentiate or not—Routes towards metastasis. Nat. Rev. Cancer
2012
,12, 425–436. [CrossRef]
83.
Beerling, E.; Seinstra, D.; de Wit, E.; Kester, L.; van der Velden, D.; Maynard, C.; Schäfer, R.; van Diest, P.;
Voest, E.; van Oudenaarden, A.; et al. Plasticity between Epithelial and Mesenchymal States Unlinks EMT
from Metastasis-Enhancing Stem Cell Capacity. Cell Rep. 2016,14, 2281–2288. [CrossRef] [PubMed]
84.
Stankic, M.; Pavlovic, S.; Chin, Y.; Brogi, E.; Padua, D.; Norton, L.; Massagu
é
, J.; Benezra, R.
TGF-
β
-Id1 signaling opposes Twist1 and promotes metastatic colonization via a mesenchymal-to-epithelial
transition. Cell Rep. 2013,5, 1228–1242. [CrossRef] [PubMed]
85.
Bedi, U.; Mishra, V.K.; Wasilewski, D.; Scheel, C.; Johnsen, S.A. Epigenetic plasticity: A central regulator of
epithelial-to-mesenchymal transition in cancer. Oncotarget 2014,5, 2016–2029. [CrossRef] [PubMed]
86.
Casciello, F.; Windloch, K.; Gannon, F.; Lee, J.S. Functional Role of G9a Histone Methyltransferase in Cancer.
Front. Immunol. 2015,6, 487. [CrossRef] [PubMed]
87.
Dong, C.; Wu, Y.; Yao, J.; Wang, Y.; Yu, Y.; Rychahou, P.G.; Evers, B.M.; Zhou, B.P. G9a interacts with Snail
and is critical for Snail-mediated E-cadherin repression in human breast cancer. J. Clin. Investig.
2012
,
122, 1469–1486. [CrossRef] [PubMed]
88.
Skrypek, N.; Bruneel, K.; Vandewalle, C.; De Smedt, E.; Soen, B.; Loret, N.; Taminau, J.; Goossens, S.;
Vandamme, N.; Berx, G. ZEB2 stably represses RAB25 expression through epigenetic regulation by SIRT1
and DNMTs during epithelial-to-mesenchymal transition. Epigenetics Chromatin 2018,11, 70. [CrossRef]
89.
Lin, Y.T.; Wu, K.J. Epigenetic regulation of epithelial-mesenchymal transition: Focusing on hypoxia and
TGF-βsignaling. J. Biomed. Sci. 2020,27, 39. [CrossRef]
90.
Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on
cancer. Oncogene 2010,29, 4741–4751. [CrossRef]
91.
Guo, W.; Keckesova, Z.; Donaher, J.L.; Shibue, T.; Tischler, V.; Reinhardt, F.; Itzkovitz, S.; Noske, A.;
Zürrer-Härdi, U.; Bell, G.; et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell
2012,148, 1015–1028. [CrossRef]
92.
Lim, S.; Becker, A.; Zimmer, A.; Lu, J.; Buettner, R.; Kirfel, J. SNAI1-mediated epithelial-mesenchymal
transition confers chemoresistance and cellular plasticity by regulating genes involved in cell death and
stem cell maintenance. PLoS ONE 2013,8, e66558. [CrossRef]
93.
Barneh, F.; Mirzaie, M.; Nickchi, P.; Tan, T.Z.; Thiery, J.P.; Piran, M.; Salimi, M.; Goshadrou, F.; Aref, A.R.;
Jafari, M. Integrated use of bioinformatic resources reveals that co-targeting of histone deacetylases, IKBK and
SRC inhibits epithelial-mesenchymal transition in cancer. Brief. Bioinform. 2019,20, 717–731. [CrossRef]
94.
Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.;
Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell
2008,133, 704–715. [CrossRef]
95.
Perou, C.M.; Sorlie, T.; Eisen, M.B.; van de Rijn, M.; Jerey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.;
Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000,406, 747–752. [CrossRef]
Cancers 2020,12, 3622 25 of 32
96.
Sørlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.;
Van De Rijn, M.;
Jerey, S.S. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical
implications. Proc. Natl. Acad. Sci. USA 2001,98, 10869–10874. [CrossRef] [PubMed]
97.
Li, L.T.; Jiang, G.; Chen, Q.; Zheng, J.N. Ki67 is a promising molecular target in the diagnosis of cancer.
Mol. Med. Rep. 2015,11, 1566–1572. [CrossRef] [PubMed]
98.
Chew, H.K. Adjuvant therapy for breast cancer: Who should get what? West. J. Med.
2001
,174, 284–287.
[CrossRef]
99.
Cardoso, F.; Kyriakides, S.; Ohno, S.; Penault-Llorca, F.; Poortmans, P.; Rubio, I.T.; Zackrisson, S.; Senkus, E.
Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol.
2019,30, 1674. [CrossRef]
100.
Cardoso, F.; Senkus, E.; Costa, A.; Papadopoulos, E.; Aapro, M.; Andr
é
, F.; Harbeck, N.; Aguilar Lopez, B.;
Barrios, C.H.; Bergh, J.; et al. 4th ESO-ESMO International Consensus Guidelines for Advanced Breast
Cancer (ABC 4).Ann. Oncol. 2018,29, 1634–1657. [CrossRef] [PubMed]
101.
Curtis, C.; Shah, S.P.; Chin, S.-F.; Turashvili, G.; Rueda, O.M.; Dunning, M.J.; Speed, D.; Lynch, A.G.;
Samarajiwa, S.; Yuan, Y. The genomic and transcriptomic architecture of 2000 breast tumours reveals novel
subgroups. Nature 2012,486, 346–352. [CrossRef] [PubMed]
102.
Paik, S.; Shak, S.; Tang, G.; Kim, C.; Baker, J.; Cronin, M.; Baehner, F.L.; Walker, M.G.; Watson, D.; Park, T. A
multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. New Engl. J. Med.
2004,351, 2817–2826. [CrossRef]
103.
Bartlett, J.; Sgroi, D.; Treuner, K.; Zhang, Y.; Ahmed, I.; Piper, T.; Salunga, R.; Brachtel, E.; Pirrie, S.; Schnabel, C.
Breast Cancer Index and prediction of benefit from extended endocrine therapy in breast cancer patients
treated in the Adjuvant Tamoxifen—To Oer More?(aTTom) trial. Ann. Oncol.
2019
,30, 1776–1783. [CrossRef]
[PubMed]
104.
Ma, X.-J.; Salunga, R.; Dahiya, S.; Wang, W.; Carney, E.; Durbecq, V.; Harris, A.; Goss, P.; Sotiriou, C.;
Erlander, M. A five-gene molecular grade index and HOXB13: IL17BR are complementary prognostic factors
in early stage breast cancer. Clin. Cancer Res. 2008,14, 2601–2608. [CrossRef] [PubMed]
105.
Simon, R.M.; Paik, S.; Hayes, D.F. Use of archived specimens in evaluation of prognostic and predictive
biomarkers. J. Natl. Cancer Inst. 2009,101, 1446–1452. [CrossRef]
106.
Cardoso, F.; van’t Veer, L.J.; Bogaerts, J.; Slaets, L.; Viale, G.; Delaloge, S.; Pierga, J.-Y.; Brain, E.; Causeret, S.;
DeLorenzi, M. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. New Engl.
J. Med. 2016,375, 717–729. [CrossRef] [PubMed]
107.
Jensen, M.-B.; Lænkholm, A.-V.; Nielsen, T.O.; Eriksen, J.O.; Wehn, P.; Hood, T.; Ram, N.;
Buckingham, W.; Ferree, S.; Ejlertsen, B. The Prosigna gene expression assay and responsiveness to
adjuvant cyclophosphamide-based chemotherapy in premenopausal high-risk patients with breast cancer.
Breast Cancer Res. 2018,20, 79. [CrossRef]
108.
Bae, Y.K.; Brown, A.; Garrett, E.; Bornman, D.; Fackler, M.J.; Sukumar, S.; Herman, J.G.; Gabrielson, E.
Hypermethylation in histologically distinct classes of breast cancer. Clin. Cancer Res.
2004
,10, 5998–6005.
[CrossRef]
109.
De Oca, R.M.; Gurard-Levin, Z.A.; Berger, F.; Rehman, H.; Martel, E.; Corpet, A.; de Koning, L.; Vassias, I.;
Wilson, L.O.; Meseure, D. The histone chaperone HJURP is a new independent prognostic marker for luminal
A breast carcinoma. Mol. Oncol. 2015,9, 657–674. [CrossRef]
110.
Roessler, J.; Ammerpohl, O.; Gutwein, J.; Steinemann, D.; Schlegelberger, B.; Weyer, V.; Sariyar, M.; Geers, R.;
Arnold, N.; Schmutzler, R. The CpG island methylator phenotype in breast cancer is associated with
the lobular subtype. Epigenomics 2015,7, 187–199. [CrossRef]
111.
Blenkiron, C.; Goldstein, L.D.; Thorne, N.P.; Spiteri, I.; Chin, S.-F.; Dunning, M.J.; Barbosa-Morais, N.L.;
Teschendor, A.E.; Green, A.R.; Ellis, I.O. MicroRNA expression profiling of human breast cancer identifies
new markers of tumor subtype. Genome Biol. 2007,8, R214. [CrossRef]
112.
Braicu, C.; Raduly, L.; Morar-Bolba, G.; Cojocneanu, R.; Jurj, A.; Pop, L.-A.; Pileczki, V.; Ciocan, C.;
Moldovan, A.; Irimie, A. Aberrant miRNAs expressed in HER-2 negative breast cancers patient. J. Exp. Clin.
Cancer Res. 2018,37, 257. [CrossRef]
113.
Qi, P.; Du, X. The long non-coding RNAs, a new cancer diagnostic and therapeutic gold mine. Mod. Pathol.
2013,26, 155–165. [CrossRef] [PubMed]
Cancers 2020,12, 3622 26 of 32
114.
Valdespino, V.; Valdespino, P.M. Potential of epigenetic therapies in the management of solid tumors.
Cancer Manag. Res. 2015,7, 241–251. [CrossRef]
115.
Jones, P.A.; Issa, J.P.; Baylin, S. Targeting the cancer epigenome for therapy. Nat. Rev. Genet.
2016
,17, 630–641.
[CrossRef] [PubMed]
116.
Ito, Y.; Makita, S.; Tobinai, K. Development of new agents for peripheral T-cell lymphoma. Expert Opin.
Biol. Ther. 2019,19, 197–209. [CrossRef] [PubMed]
117.
Roberti, A.; Valdes, A.F.; Torrecillas, R.; Fraga, M.F.; Fernandez, A.F. Epigenetics in cancer therapy and
nanomedicine. Clin. Epigenetics 2019,11, 81. [CrossRef] [PubMed]
118.
Ramachandran, S.; Ient, J.; Göttgens, E.L.; Krieg, A.J.; Hammond, E.M. Epigenetic Therapy for Solid Tumors:
Highlighting the Impact of Tumor Hypoxia. Genes 2015,6, 935–956. [CrossRef]
119.
Ganesan, A.; Arimondo, P.B.; Rots, M.G.; Jeronimo, C.; Berdasco, M. The timeline of epigenetic drug
discovery: From reality to dreams. Clin. Epigenetics 2019,11, 174. [CrossRef]
120.
Gnyszka, A.; Jastrzebski, Z.; Flis, S. DNA methyltransferase inhibitors and their emerging role in epigenetic
therapy of cancer. Anticancer Res. 2013,33, 2989–2996.
121.
Bohl, S.R.; Bullinger, L.; Rücker, F.G. Epigenetic therapy: Azacytidine and decitabine in acute myeloid
leukemia. Expert Rev. Hematol. 2018,11, 361–371. [CrossRef]
122.
Roche, J.; Bertrand, P. Inside HDACs with more selective HDAC inhibitors. Eur. J. Med. Chem.
2016
,
121, 451–483. [CrossRef]
123.
Ghasemi, S. Cancer’s epigenetic drugs: Where are they in the cancer medicines? Pharm. J.
2020
,20, 367–379.
[CrossRef] [PubMed]
124. Filippova, E.V.; Zemaitaitis, B.; Aung, T.; Wolfe, A.J.; Anderson, W.F. Structural Basis for DNA Recognition
by the Two-Component Response Regulator RcsB. mBio 2018,9, e01993-17. [CrossRef] [PubMed]
125.
Marks, P.A.; Dokmanovic, M. Histone deacetylase inhibitors: Discovery and development as anticancer
agents. Expert Opin. Investig. Drugs 2005,14, 1497–1511. [CrossRef] [PubMed]
126.
Montalvo-Casimiro, M.; Gonz
á
lez-Barrios, R.; Meraz-Rodriguez, M.A.; Ju
á
rez-Gonz
á
lez, V.T.;
Arriaga-Canon, C.; Herrera, L.A. Epidrug Repurposing: Discovering New Faces of Old Acquaintances in
Cancer Therapy. Front. Oncol. 2020,10, 2461. [CrossRef]
127.
West, A.C.; Johnstone, R.W. New and emerging HDAC inhibitors for cancer treatment. J. Clin. Investig.
2014
,
124, 30–39. [CrossRef] [PubMed]
128.
Garcia-Manero, G.; Roboz,G.; Walsh, K.; Kantarjian, H.; Ritchie, E.; Kropf,P.; O’Connell, C.; Tibes,R.; Lunin, S.;
Rosenblat, T.; et al. Guadecitabine (SGI-110) in patients with intermediate or high-risk myelodysplastic
syndromes: Phase 2 results from a multicentre, open-label, randomised, phase 1/2 trial. Lancet Haematol.
2019,6, e317–e327. [CrossRef]
129.
Graça, I.; Pereira-Silva, E.; Henrique, R.; Packham, G.; Crabb, S.J.; Jer
ó
nimo, C. Epigenetic modulators as
therapeutic targets in prostate cancer. Clin. Epigenetics 2016,8, 98. [CrossRef]
130.
Cheng, Y.; He, C.; Wang, M.; Ma, X.; Mo, F.; Yang, S.; Han, J.; Wei, X. Targeting epigenetic regulators for cancer
therapy: Mechanisms and advances in clinical trials. Signal Transduct. Target. Ther. 2019,4, 62. [CrossRef]
131.
Wapenaar, H.; Dekker, F.J. Histone acetyltransferases: Challenges in targeting bi-substrate enzymes.
Clin. Epigenetics 2016,8, 59. [CrossRef]
132.
McCabe, M.T.; Ott, H.M.; Ganji, G.; Korenchuk, S.; Thompson, C.; Van Aller, G.S.; Liu, Y.; Graves, A.P.;
Della Pietra, A., III; Diaz, E.; et al. EZH2 inhibition as a therapeutic strategy for lymphoma with
EZH2-activating mutations. Nature 2012,492, 108–112. [CrossRef]
133.
Stein, E.M.; Garcia-Manero, G.; Rizzieri, D.A.; Tibes, R.; Berdeja, J.G.; Savona, M.R.; Jongen-Lavrenic, M.;
Altman, J.K.; Thomson, B.; Blakemore, S.J.; et al. The DOT1L inhibitor pinometostat reduces H3K79
methylation and has modest clinical activity in adult acute leukemia. Blood
2018
,131, 2661–2669. [CrossRef]
134.
Wong, M.; Polly, P.; Liu, T. The histone methyltransferase DOT1L: Regulatory functions and a cancer therapy
target. Am. J. Cancer Res. 2015,5, 2823–2837.
135.
Wojtala, M.; D ˛abek, A.; Rybaczek, D.; ´
Sliwi´nska, A.; ´
Swiderska, E.; Słapek, K.; El-Osta, A.; Balcerczyk, A.
Silencing Lysine-Specific Histone Demethylase 1 (LSD1) Causes Increased HP1-Positive Chromatin,
Stimulation of DNA Repair Processes, and Dysregulation of Proliferation by Chk1 Phosphorylation in
Human Endothelial Cells. Cells 2019,8, 1212. [CrossRef]
136.
Majello, B.; Gorini, F.; Sacc
à
, C.D.; Amente, S. Expanding the Role of the Histone Lysine-Specific Demethylase
LSD1 in Cancer. Cancers 2019,11, 324. [CrossRef]
Cancers 2020,12, 3622 27 of 32
137.
Alqahtani, A.; Choucair, K.; Ashraf, M.; Hammouda, D.M.; Alloghbi, A.; Khan, T.; Senzer, N.; Nemunaitis, J.
Bromodomain and extra-terminal motif inhibitors: A review of preclinical and clinical advances in cancer
therapy. Future Sci. OA 2019,5, Fso372. [CrossRef]
138.
Luan, Y.; Ngo, L.; Han, Z.; Wang, X.; Qu, M.; Zheng, Y.G. Histone acetyltransferases: Enzymes, assays, and
inhibitors. In Epigenetic Technological Applications; Elsevier: Amsterdam, The Netherlands, 2015; pp. 291–317.
139.
Meseure, D.; Drak Alsibai, K.; Nicolas, A.; Bieche, I.; Morillon, A. Long Noncoding RNAs as New Architects
in Cancer Epigenetics, Prognostic Biomarkers, and Potential Therapeutic Targets. BioMed Res. Int.
2015
,
2015, 320214. [CrossRef]
140.
Baumann, V.; Winkler, J. miRNA-based therapies: Strategies and delivery platforms for oligonucleotide and
non-oligonucleotide agents. Future Med. Chem. 2014,6, 1967–1984. [CrossRef]
141.
Takahashi, R.-U.; Prieto-Vila, M.; Kohama, I.; Ochiya, T. Development of miRNA-based therapeutic
approaches for cancer patients. Cancer Sci. 2019,110, 1140–1147. [CrossRef]
142.
Ma, L.; Chua, M.S.; Andrisani, O.; So, S. Epigenetics in hepatocellular carcinoma: An update and future
therapy perspectives. World J. Gastroenterol. 2014,20, 333–345. [CrossRef]
143.
Falahi, F.; van Kruchten, M.; Martinet, N.; Hospers, G.A.; Rots, M.G. Current and upcoming approaches to
exploit the reversibility of epigenetic mutations in breast cancer. Breast Cancer Res.
2014
,16, 412. [CrossRef]
144.
Ari, F.; Napieralski, R.; Ulukaya, E.; Dere, E.; Colling, C.; Honert, K.; Krüger, A.; Kiechle, M.; Schmitt, M.
Modulation of protein expression levels and DNA methylation status of breast cancer metastasis genes
by anthracycline-based chemotherapy and the demethylating agent decitabine. Cell Biochem. Funct.
2011
,
29, 651–659. [CrossRef] [PubMed]
145.
Hurtubise, A.; Momparler, R.L. Evaluation of antineoplastic action of 5-aza-2
0
-deoxycytidine (Dacogen)
and docetaxel (Taxotere) on human breast, lung and prostate carcinoma cell lines. Anticancer Drugs
2004
,
15, 161–167. [CrossRef]
146.
Mirza, S.; Sharma, G.; Pandya, P.; Ralhan, R. Demethylating agent 5-aza-2-deoxycytidine enhances
susceptibility of breast cancer cells to anticancer agents. Mol. Cell. Biochem. 2010,342, 101–109. [CrossRef]
147.
Hodges-Gallagher, L.; Valentine, C.D.; Bader, S.E.; Kushner, P.J. Inhibition of histone deacetylase enhances
the anti-proliferative action of antiestrogens on breast cancer cells and blocks tamoxifen-induced proliferation
of uterine cells. Breast Cancer Res. Treat. 2007,105, 297–309. [CrossRef]
148.
Basse, C.; Arock, M. The increasing roles of epigenetics in breast cancer: Implications for pathogenicity,
biomarkers, prevention and treatment. Int. J. Cancer 2015,137, 2785–2794. [CrossRef]
149.
Connolly, R.M.; Li, H.; Jankowitz, R.C.; Zhang, Z.; Rudek, M.A.; Jeter, S.C.; Slater, S.A.; Powers, P.; Wol, A.C.;
Fetting, J.H.; et al. Combination Epigenetic Therapy in Advanced Breast Cancer with 5-Azacitidine and
Entinostat: A Phase II National Cancer Institute/Stand Up to Cancer Study. Clin. Cancer Res.
2017
,
23, 2691–2701. [CrossRef]
150.
Fortunati, N.; Bertino, S.; Costantino, L.; De Bortoli, M.; Compagnone, A.; Bandino, A.; Catalano, M.G.;
Boccuzzi, G. Valproic acid restores ER alpha and antiestrogen sensitivity to ER alpha-negative breast cancer
cells. Mol. Cell. Endocrinol. 2010,314, 17–22. [CrossRef]
151.
Jang, E.R.; Lim, S.J.; Lee, E.S.; Jeong, G.; Kim, T.Y.; Bang, Y.J.; Lee, J.S. The histone deacetylase inhibitor
trichostatin A sensitizes estrogen receptor alpha-negative breast cancer cells to tamoxifen. Oncogene
2004
,
23, 1724–1736. [CrossRef]
152.
Feng, Q.; Zhang, Z.; Shea, M.J.; Creighton, C.J.; Coarfa, C.; Hilsenbeck, S.G.; Lanz, R.; He, B.; Wang, L.; Fu, X.;
et al. An epigenomic approach to therapy for tamoxifen-resistant breast cancer. Cell Res.
2014
,24, 809–819.
[CrossRef]
153.
V
á
zquez, R.; Riveiro, M.E.; Astorgues-Xerri, L.; Odore, E.; Rezai, K.; Erba, E.; Panini, N.; Rinaldi, A.;
Kwee, I.; Beltrame, L.; et al. The bromodomain inhibitor OTX015 (MK-8628) exerts anti-tumor activity in
triple-negative breast cancer models as single agent and in combination with everolimus. Oncotarget
2017
,
8, 7598–7613. [CrossRef]
154.
Min, A.; Im, S.A.; Kim, D.K.; Song, S.H.; Kim, H.J.; Lee, K.H.; Kim, T.Y.; Han, S.W.; Oh, D.Y.; Kim, T.Y.;
et al. Histone deacetylase inhibitor, suberoylanilide hydroxamic acid (SAHA), enhances anti-tumor eects
of the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib in triple-negative breast cancer cells.
Breast Cancer Res. 2015,17, 33. [CrossRef]
Cancers 2020,12, 3622 28 of 32
155.
Goldstein, L.J.; Zhao, F.; Wang, M.; Swaby, R.F.; Sparano, J.A.; Meropol, N.J.; Bhalla, K.N.; Pellegrino, C.M.;
Katherine Alpaugh, R.; Falkson, C.I.; et al. A Phase I/II study of suberoylanilide hydroxamic acid (SAHA) in
combination with trastuzumab (Herceptin) in patients with advanced metastatic and/or local chest wall
recurrent HER2-amplified breast cancer: A trial of the ECOG-ACRIN Cancer Research Group (E1104).
Breast Cancer Res. Treat. 2017,165, 375–382. [CrossRef]
156.
Damaskos, C.; Garmpis, N.; Valsami, S.; Kontos, M.; Spartalis, E.; Kalampokas, T.; Kalampokas, E.;
Athanasiou, A.; Moris, D.; Daskalopoulou, A.; et al. Histone Deacetylase Inhibitors: An Attractive
Therapeutic Strategy Against Breast Cancer. Anticancer Res. 2017,37, 35–46. [CrossRef]
157.
Tinkle, S.; McNeil, S.E.; Mühlebach, S.; Bawa, R.; Borchard, G.; Barenholz, Y.C.; Tamarkin, L.; Desai, N.
Nanomedicines: Addressing the scientific and regulatory gap. Ann. N. Y. Acad. Sci.
2014
,1313, 35–56.
[CrossRef]
158.
EUR-Lex: EU. Available online: lawhttps://eur-lex.europa.eu/legal-content/EN/TXT/PDF/?uri=CELEX:
32011H0696&from=EN (accessed on 17 August 2020).
159. Schütz, C.A.; Juillerat-Jeanneret, L.; Mueller, H.; Lynch, I.; Riediker, M. Therapeutic nanoparticles in clinics
and under clinical evaluation. Nanomedicine 2013,8, 449–467. [CrossRef]
160.
Boverhof, D.R.; Bramante, C.M.; Butala, J.H.; Clancy, S.F.; Lafranconi, M.; West, J.; Gordon, S.C.
Comparative assessment of nanomaterial definitions and safety evaluationconsiderations.
Regul. Toxicol. Pharmacol.
2015,73, 137–150. [CrossRef]
161. Nanomaterials definition matters. Nat. Nanotechnol. 2019,14, 193. [CrossRef]
162.
Kim, K.; Khang, D. Past, Present, and Future of Anticancer Nanomedicine. Int. J. Nanomed.
2020
,15, 5719.
[CrossRef]
163.
Wong, H.L.; Bendayan, R.; Rauth, A.M.; Li, Y.; Wu, X.Y. Chemotherapy with anticancer drugs encapsulated
in solid lipid nanoparticles. Adv. Drug Deliv. Rev. 2007,59, 491–504. [CrossRef]
164.
Martinelli, C.; Pucci, C.; Ciofani, G. Nanostructured carriers as innovative tools for cancer diagnosis and
therapy. APL Bioeng. 2019,3, 011502. [CrossRef]
165.
Smolkova, B.; Dusinska, M.; Gabelova, A. Nanomedicine and epigenome. Possible health risks.
Food Chem. Toxicol. 2017,109, 780–796. [CrossRef]
166.
Takechi-Haraya, Y.; Goda, Y.; Sakai-Kato, K. Control of Liposomal Penetration into Three-Dimensional
Multicellular Tumor Spheroids by Modulating Liposomal Membrane Rigidity. Mol. Pharm.
2017
,
14, 2158–2165. [CrossRef]
167.
Davis, M.E.; Chen, Z.; Shin, D.M. Nanoparticle therapeutics: An emerging treatment modality for cancer.
In Nanoscience and Technology: A Collection of Reviews from Nature Journals; World Scientific: Singapore, 2010;
pp. 239–250.
168.
Du, M.; Ouyang, Y.; Meng, F.; Ma, Q.; Liu, H.; Zhuang, Y.; Pang, M.; Cai, T.; Cai, Y. Nanotargeted agents:
An emerging therapeutic strategy for breast cancer. Nanomedicine 2019,14, 1771–1786. [CrossRef]
169.
Caster, J.M.; Patel, A.N.; Zhang, T.; Wang, A. Investigational nanomedicines in 2016: A review of
nanotherapeutics currently undergoing clinical trials. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol.
2017
,
9, e1416. [CrossRef]
170.
Liyanage, P.Y.; Hettiarachchi, S.D.; Zhou, Y.; Ouhtit, A.; Seven, E.S.; Oztan, C.Y.; Celik, E.; Leblanc, R.M.
Nanoparticle-mediated targeted drug delivery for breast cancer treatment. Biochim. Biophys. Acta Rev. Cancer
2019,1871, 419–433. [CrossRef]
171.
Anarjan, F.S. Active targeting drug delivery nanocarriers: Ligands. Nano-Struct. Nano-Objects
2019
,19, 100370.
172.
Bozzuto, G.; Molinari, A. Liposomes as nanomedical devices. Int. J. Nanomed.
2015
,10, 975–999. [CrossRef]
173.
Cole, A.J.; David, A.E.; Wang, J.; Galb
á
n, C.J.; Hill, H.L.; Yang, V.C. Polyethylene glycol modified,
cross-linked starch-coated iron oxide nanoparticles for enhanced magnetic tumor targeting. Biomaterials
2011,32, 2183–2193. [CrossRef]
174.
Saraf, S.; Jain, A.; Tiwari, A.; Verma, A.; Panda, P.K.; Jain, S.K. Advances in liposomal drug delivery to cancer:
An overview. J. Drug Deliv. Sci. Technol. 2020,56, 101549. [CrossRef]
175.
Bottai, G.; Tru, M.; Corsi, F.; Santarpia, L. Progress in nonviral gene therapy for breast cancer and what
comes next? Expert Opin. Biol. Ther. 2017,17, 595–611. [CrossRef]
176.
Aftab, S.; Shah, A.; Nadhman, A.; Kurbanoglu, S.; Ozkan, S.A.; Dionysiou, D.D.; Shukla, S.S.; Aminabhavi, T.M.
Nanomedicine: An eective tool in cancer therapy. Int. J. Pharm. 2018,540, 132–149. [CrossRef] [PubMed]
Cancers 2020,12, 3622 29 of 32
177. Zhang, D.; Zhang, J. Surface engineering of nanomaterials with phospholipid-polyethylene glycol-derived
functional conjugates for molecular imaging and targeted therapy. Biomaterials
2020
,230, 119646. [CrossRef]
[PubMed]
178.
Park, J.W.; Hong, K.; Kirpotin, D.B.; Colbern, G.; Shalaby, R.; Baselga, J.; Shao, Y.; Nielsen, U.B.; Marks, J.D.;
Moore, D. Anti-HER2 immunoliposomes: Enhanced ecacy attributable to targeted delivery.
Clin. Cancer Res.
2002,8, 1172–1181. [PubMed]
179.
Snipstad, S.; Hak, S.; Baghirov, H.; Sulheim, E.; Mørch,
Ý
.; L
é
lu, S.; von Haartman, E.; Bäck, M.; Nilsson, K.P.R.;
Klymchenko, A.S. Labeling nanoparticles: Dye leakage and altered cellular uptake. Cytom. Part A
2017
,
91, 760–766. [CrossRef] [PubMed]
180.
Moumaris, M.; Bretagne, J.-M.; Abuaf, N. Nanomedical Devices and Cancer Theranostics. Open Nanomed.
Nanotechnol. J. 2020,6, 1–11. [CrossRef]
181.
Gurunathan, S.; Kang, M.-H.; Qasim, M.; Kim, J.-H. Nanoparticle-Mediated Combination Therapy:
Two-in-One Approach for Cancer. Int. J. Mol. Sci. 2018,19, 3264. [CrossRef]
182.
Puri, A.; Loomis, K.; Smith, B.; Lee, J.H.; Yavlovich, A.; Heldman, E.; Blumenthal, R. Lipid-based nanoparticles
as pharmaceutical drug carriers: From concepts to clinic. Crit. Rev. Ther. Drug Carr. Syst.
2009
,26, 523–580.
[CrossRef]
183.
Monteiro, N.; Martins, A.; Reis, R.L.; Neves, N.M. Liposomes in tissue engineering and regenerative medicine.
J. R. Soc. Interface 2014,11, 20140459. [CrossRef]
184.
Riaz, M.K.; Riaz, M.A.; Zhang, X.; Lin, C.; Wong, K.H.; Chen, X.; Zhang, G.; Lu, A.; Yang, Z. Surface
Functionalization and Targeting Strategies of Liposomes in Solid Tumor Therapy: A Review. Int. J. Mol. Sci.
2018,19, 195. [CrossRef]
185.
Olusanya, T.O.B.; Haj Ahmad, R.R.; Ibegbu, D.M.; Smith, J.R.; Elkordy, A.A. Liposomal Drug Delivery
Systems and Anticancer Drugs. Molecules 2018,23, 907. [CrossRef]
186.
Inglut, C.T.; Sorrin, A.J.; Kuruppu, T.; Vig, S.; Cicalo, J.; Ahmad, H.; Huang, H.C. Immunological and
Toxicological Considerations for the Design of Liposomes. Nanomaterials
2020
,10, 190. [CrossRef] [PubMed]
187.
Deshpande, P.P.; Biswas, S.; Torchilin, V.P. Current trends in the use of liposomes for tumor targeting.
Nanomedicine 2013,8, 1509–1528. [CrossRef] [PubMed]
188.
Leo, C.P.; Hentschel, B.; Szucs, T.D.; Leo, C. FDA and EMA Approvals of New Breast Cancer
Drugs—A Comparative Regulatory Analysis. Cancers 2020,12, 437. [CrossRef] [PubMed]
189.
Urbinati, G.; Marsaud, V.; Plassat, V.; Fattal, E.; Lesieur, S.; Renoir, J.M. Liposomes loaded with histone
deacetylase inhibitors for breast cancer therapy. Int. J. Pharm. 2010,397, 184–193. [CrossRef]
190.
Wang, Y.; Tu, S.; Steen, D.; Xiong, M. Iron complexation to histone deacetylase inhibitors SAHA and LAQ824
in PEGylated liposomes can considerably improve pharmacokinetics in rats. J. Pharm. Pharm. Sci.
2014
,
17, 583–602. [CrossRef]
191.
Neupane, Y.R.; Srivastava, M.; Ahmad, N.; Kumar, N.; Bhatnagar, A.; Kohli, K. Lipid based nanocarrier
system for the potential oral delivery of decitabine: Formulation design, characterization, ex vivo, and
in vivo
assessment. Int. J. Pharm. 2014,477, 601–612. [CrossRef]
192.
Su, X.; Wang, Z.; Li, L.; Zheng, M.; Zheng, C.; Gong, P.; Zhao, P.; Ma, Y.; Tao, Q.; Cai, L. Lipid-polymer
nanoparticles encapsulating doxorubicin and 2
0
-deoxy-5-azacytidine enhance the sensitivity of cancer cells
to chemical therapeutics. Mol. Pharm. 2013,10, 1901–1909. [CrossRef]
193.
Tran, T.H.; Ramasamy, T.; Truong, D.H.; Shin, B.S.; Choi, H.G.; Yong, C.S.; Kim, J.O. Development of
vorinostat-loaded solid lipid nanoparticles to enhance pharmacokinetics and ecacy against multidrug-
resistant cancer cells. Pharm. Res. 2014,31, 1978–1988. [CrossRef]
194.
Tran, T.H.; Choi, J.Y.; Ramasamy, T.; Truong, D.H.; Nguyen, C.N.; Choi, H.G.; Yong, C.S.; Kim, J.O.
Hyaluronic acid-coated solid lipid nanoparticles for targeted delivery of vorinostat to CD44 overexpressing
cancer cells. Carbohydr. Polym. 2014,114, 407–415. [CrossRef]
195.
Denis, I.; el Bahhaj, F.; Collette, F.; Delatouche, R.; Gueugnon, F.; Pouliquen, D.; Pichavant, L.; H
é
roguez, V.;
Gr
é
goire, M.; Bertrand, P.; et al. Histone deacetylase inhibitor-polymer conjugate nanoparticles for
acid-responsive drug delivery. Eur. J. Med. Chem. 2015,95, 369–376. [CrossRef]
196.
Xu, J.; Sun, J.; Wang, P.; Ma, X.; Li, S. Pendant HDAC inhibitor SAHA derivatised polymer as a novel prodrug
micellar carrier for anticancer drugs. J. Drug Target. 2018,26, 448–457. [CrossRef] [PubMed]
Cancers 2020,12, 3622 30 of 32
197.
Martin, D.T.; Hoimes, C.J.; Kaimakliotis, H.Z.; Cheng, C.J.; Zhang, K.; Liu, J.; Wheeler, M.A.; Kelly, W.K.;
Tew, G.N.; Saltzman, W.M.; et al. Nanoparticles for urothelium penetration and delivery of the histone
deacetylase inhibitor belinostat for treatment of bladder cancer. Nanomedicine
2013
,9, 1124–1134. [CrossRef]
[PubMed]
198.
Sankar, R.; Ravikumar, V. Biocompatibility and biodistribution of suberoylanilide hydroxamic acid loaded
poly (DL-lactide-co-glycolide) nanoparticles for targeted drug delivery in cancer. Biomed. Pharmacother.
2014
,
68, 865–871. [CrossRef] [PubMed]
199.
Naz, A.; Cui, Y.; Collins, C.J.; Thompson, D.H.; Irudayaraj, J. PLGA-PEG nano-delivery system for epigenetic
therapy. Biomed. Pharmacother. 2017,90, 586–597. [CrossRef] [PubMed]
200.
Li, S.Y.; Sun, R.; Wang, H.X.; Shen, S.; Liu, Y.; Du, X.J.; Zhu, Y.H.; Jun, W. Combination therapy with
epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy
response and overcome resistance by breast cancer stem cells. J. Control. Release
2015
,205, 7–14. [CrossRef]
201.
Hong, Y.D.; Zhang, J.; Zhuang, M.; Li, W.; Wu, P.U.; Li, R.T.; Hu, N.; Bian, B.X.; Song, Z.Y.; Wu, F.L. Ecacy of
decitabine-loaded gelatinases-stimuli nanoparticles in overcoming cancer drug resistance is mediated via its
enhanced demethylating activity to transcription factor AP-2 epsilon. Oncotarget
2017
,8, 114495–114505.
[CrossRef]
202.
Wu, F.L.; Li, R.T.; Yang, M.; Yue, G.F.; Wang, H.Y.; Liu, Q.; Cui, F.B.; Wu, P.Y.; Ding, H.; Yu, L.X.;
et al. Gelatinases-stimuli nanoparticles encapsulating 5-fluorouridine and 5-aza-2
0
-deoxycytidine enhance
the sensitivity of gastric cancer cells to chemical therapeutics. Cancer Lett. 2015,363, 7–16. [CrossRef]
203.
Kwak, T.W.; Kim, D.H.; Jeong, Y.I.; Kang, D.H. Antitumor activity of vorinostat-incorporated nanoparticles
against human cholangiocarcinoma cells. J. Nanobiotechnol. 2015,13, 60. [CrossRef]
204.
Wang, E.C.; Min, Y.; Palm, R.C.; Fiordalisi, J.J.; Wagner, K.T.; Hyder, N.; Cox, A.D.; Caster, J.M.; Tian, X.;
Wang, A.Z. Nanoparticle formulations of histone deacetylase inhibitors for eective chemoradiotherapy in
solid tumors. Biomaterials 2015,51, 208–215. [CrossRef]
205.
Vijayaraghavalu, S.; Labhasetwar, V. Ecacy of decitabine-loaded nanogels in overcoming cancer drug
resistance is mediated via sustained DNA methyltransferase 1 (DNMT1) depletion. Cancer Lett.
2013
,
331, 122–129. [CrossRef]
206.
Houshmand, M.; Garello, F.; Circosta, P.; Stefania, R.; Aime, S.; Saglio, G.; Giachino, C. Nanocarriers as Magic
Bullets in the Treatment of Leukemia. Nanomaterials 2020,10, 276. [CrossRef] [PubMed]
207.
Duan, Y.; Dhar, A.; Patel, C.; Khimani, M.; Neogi, S.; Sharma, P.; Kumar, N.S.; Vekariya, R.L. A brief review
on solid lipid nanoparticles: Part and parcel of contemporary drug delivery systems. RSC Adv.
2020
,
10, 26777–26791. [CrossRef]
208.
Tran, T.H.; Chu, D.T.; Truong, D.H.; Tak, J.W.; Jeong, J.H.; Hoang, V.L.; Yong, C.S.; Kim, J.O. Development of
lipid nanoparticles for a histone deacetylases inhibitor as a promising anticancer therapeutic. Drug Deliv.
2016,23, 1335–1343. [CrossRef] [PubMed]
209.
Ishii, Y.; Hattori, Y.; Yamada, T.; Uesato, S.; Maitani, Y.; Nagaoka, Y. Histone deacetylase inhibitor prodrugs in
nanoparticle vector enhanced gene expression in human cancer cells. Eur. J. Med. Chem.
2009
,44, 4603–4610.
[CrossRef] [PubMed]
210.
Wang, X.; Yu, B.; Wu, Y.; Lee, R.J.; Lee, L.J. Ecient down-regulation of CDK4 by novel lipid
nanoparticle-mediated siRNA delivery. Anticancer Res. 2011,31, 1619–1626.
211.
Anselmo, A.C.; Mitragotri, S. Nanoparticles in the clinic: An update. Bioeng. Transl. Med.
2019
,4, e10143.
[CrossRef]
212.
Bobo, D.; Robinson, K.J.; Islam, J.; Thurecht, K.J.; Corrie, S.R. Nanoparticle-Based Medicines: A Review of
FDA-Approved Materials and Clinical Trials to Date. Pharm. Res. 2016,33, 2373–2387. [CrossRef]
213.
Park, J.H.; Lee, B.K.; Park, S.H.; Kim, M.G.; Lee, J.W.; Lee, H.Y.; Lee, H.B.; Kim, J.H.; Kim, M.S. Preparation of
biodegradable and elastic poly (
ε
-caprolactone-co-lactide) copolymers and evaluation as a localized and
sustained drug delivery carrier. Int. J. Mol. Sci. 2017,18, 671. [CrossRef]
214.
Lu, B.; Huang, X.; Mo, J.; Zhao, W. Drug delivery using nanoparticles for cancer stem-like cell targeting.
Front. Pharmacol. 2016,7, 84. [CrossRef]
215.
Nasir, A.; Kausar, A.; Younus, A. A review on preparation, properties and applications of polymeric
nanoparticle-based materials. Polym. Plast. Technol. Eng. 2015,54, 325–341. [CrossRef]
216.
De Souza, C.; Ma, Z.; Lindstrom, A.R.; Chatterji, B.P. Nanomaterials as potential transporters of HDAC
inhibitors. Med. Drug Discov. 2020,6, 100040. [CrossRef]
Cancers 2020,12, 3622 31 of 32
217.
Kwak, T.W.; Lee, H.L.; Song, Y.H.; Kim, C.; Kim, J.; Seo, S.J.; Jeong, Y.I.; Kang, D.H.
Vorinostat-eluting poly(DL-lactide-co-glycolide) nanofiber-coated stent for inhibition of cholangiocarcinoma
cells. Int. J. Nanomed. 2017,12, 7669–7680. [CrossRef] [PubMed]
218.
Alp, E.; Damkaci, F.; Guven, E.; Tenniswood, M. Starch nanoparticles for delivery of the histone deacetylase
inhibitor CG-1521 in breast cancer treatment. Int. J. Nanomed. 2019,14, 1335–1346. [CrossRef] [PubMed]
219.
Honma, K.; Iwao-Koizumi, K.; Takeshita, F.; Yamamoto, Y.; Yoshida, T.; Nishio, K.; Nagahara, S.; Kato, K.;
Ochiya, T. RPN2 gene confers docetaxel resistance in breast cancer. Nat. Med.
2008
,14, 939–948. [CrossRef]
220.
Lombardo, D.; Kiselev, M.A.; Caccamo, M.T. Smart Nanoparticles for Drug Delivery Application:
Development of Versatile Nanocarrier Platforms in Biotechnology and Nanomedicine. J. Nanomater.
2019,2019. [CrossRef]
221.
Hanafy, N.A.; El-Kemary, M.; Leporatti, S. Micelles structure development as a strategy to improve smart
cancer therapy. Cancers 2018,10, 238. [CrossRef]
222.
Devulapally, R.; Sekar, N.M.; Sekar, T.V.; Foygel, K.; Massoud, T.F.; Willmann, J.K.; Paulmurugan, R.
Polymer nanoparticles mediated codelivery of antimiR-10b and antimiR-21 for achieving triple negative
breast cancer therapy. ACS Nano 2015,9, 2290–2302. [CrossRef]
223.
Mandal, A.K. Dendrimers in targeted drug delivery applications: A review of diseases and cancer. Int. J.
Polym. Mater. Polym. Biomater. 2020, 1–11. [CrossRef]
224.
Finlay, J.; Roberts, C.M.; Lowe, G.; Loeza, J.; Rossi, J.J.; Glackin, C.A. RNA-based TWIST1 inhibition via
dendrimer complex to reduce breast cancer cell metastasis. Biomed. Res. Int.
2015
,2015, 382745. [CrossRef]
225.
Wang, P.; Zhao, X.H.; Wang, Z.Y.; Meng, M.; Li, X.; Ning, Q. Generation 4 polyamidoamine dendrimers is
a novel candidate of nano-carrier for gene delivery agents in breast cancer treatment. Cancer Lett.
2010
,
298, 34–49. [CrossRef]
226.
Suhail, M.; Rosenholm, J.M.; Minhas, M.U.; Badshah, S.F.; Naeem, A.; Khan, K.U.; Fahad, M. Nanogels as
drug-delivery systems: A comprehensive overview. Ther. Deliv. 2019,10, 697–717. [CrossRef] [PubMed]
227.
Reddy, N.; Reddy, R.; Jiang, Q. Crosslinking biopolymers for biomedical applications. Trends Biotechnol.
2015
,
33, 362–369. [CrossRef] [PubMed]
228.
Vijayaraghavalu, S.; Labhasetwar, V. Nanogel-mediated delivery of a cocktail of epigenetic drugs plus
doxorubicin overcomes drug resistance in breast cancer cells. Drug Deliv. Transl. Res.
2018
,8, 1289–1299.
[CrossRef] [PubMed]
229. Wu, X.; Hu, Z.; Nizzero, S.; Zhang, G.; Ramirez, M.R.; Shi, C.; Zhou, J.; Ferrari, M.; Shen, H. Bone-targeting
nanoparticle to co-deliver decitabine and arsenic trioxide for eective therapy of myelodysplastic syndrome
with low systemic toxicity. J. Control. Release 2017,268, 92–101. [CrossRef]
230.
Parhizkar, M.; Reardon, P.; Harker, A.; Browning, R.; Stride, E.; Pedley, R.; Knowles, J.; Edirisinghe, M.
Enhanced ecacy in drug-resistant cancer cells through synergistic nanoparticle mediated delivery of
cisplatin and decitabine. Nanoscale Adv. 2020,2, 1177–1186. [CrossRef]
231.
Kim, B.; Pena, C.D.; Auguste, D.T. Targeted Lipid Nanoemulsions Encapsulating Epigenetic Drugs Exhibit
Selective Cytotoxicity on CDH1(-)/FOXM1(+) Triple Negative Breast Cancer Cells. Mol. Pharm.
2019
,
16, 1813–1826. [CrossRef]
232.
Ruttala, H.B.; Ramasamy, T.; Poudal, B.K.; Choi, Y.; Choi, J.Y.; Kim, J.; Kwang Ku, S.; Choi, H.G.; Soon Yong, C.;
Oh Kim, J. Molecularly targeted co-delivery of a histone deacetylase inhibitor and paclitaxel by lipid-protein
hybrid nanoparticles for synergistic combinational chemotherapy. Oncotarget
2017
,8, 14925–14940. [CrossRef]
233.
Deng, Z.J.; Morton, S.W.; Ben-Akiva, E.; Dreaden, E.C.; Shopsowitz, K.E.; Hammond, P.T. Layer-by-layer
nanoparticles for systemic codelivery of an anticancer drug and siRNA for potential triple-negative breast
cancer treatment. ACS Nano 2013,7, 9571–9584. [CrossRef]
234.
Foulkes, R.; Man, E.; Thind, J.; Yeung, S.; Joy, A.; Hoskins, C. The regulation of nanomaterials and
nanomedicines for clinical application: Current and future perspectives. Biomater. Sci.
2020
,8, 4653–4664.
[CrossRef]
235.
Lungu, I.I.; Grumezescu, A.M.; Volceanov, A.; Andronescu, E. Nanobiomaterials used in cancer therapy:
An up-to-date overview. Molecules 2019,24, 3547. [CrossRef]
236.
Gao, H. Shaping tumor microenvironment for improving nanoparticle delivery. Curr. Drug Metab.
2016
,
17, 731–736. [CrossRef] [PubMed]
237.
Choi, H.S.; Liu, W.; Misra, P.; Tanaka, E.; Zimmer, J.P.; Ipe, B.I.; Bawendi, M.G.; Frangioni, J.V. Renal clearance
of quantum dots. Nat. Biotechnol. 2007,25, 1165–1170. [CrossRef] [PubMed]
Cancers 2020,12, 3622 32 of 32
238.
Longmire, M.; Choyke, P.L.; Kobayashi, H. Clearance properties of nano-sized particles and molecules as
imaging agents: Considerations and caveats. Future Med. 2008. [CrossRef] [PubMed]
239.
Hossen, S.; Hossain, M.K.; Basher, M.; Mia, M.; Rahman, M.; Uddin, M.J. Smart nanocarrier-based drug
delivery systems for cancer therapy and toxicity studies: A review. J. Adv. Res.
2019
,15, 1–18. [CrossRef]
[PubMed]
240.
Dusinska, M.; Tulinska, J.; El Yamani, N.; Kuricova, M.; Liskova, A.; Rollerova, E.; Rund
é
n-Pran, E.;
Smolkova, B. Immunotoxicity, genotoxicity and epigenetic toxicity of nanomaterials: New strategies for
toxicity testing? Food Chem. Toxicol. 2017,109, 797–811. [CrossRef]
241.
Weiswald, L.-B.; Bellet, D.; Dangles-Marie, V. Spherical cancer models in tumor biology. Neoplasia
2015
,
17, 1–15. [CrossRef]
242.
Afantitis, A.; Melagraki, G.; Isigonis, P.; Tsoumanis, A.; Varsou, D.D.; Valsami-Jones, E.; Papadiamantis, A.;
Ellis, L.-J.A.; Sarimveis, H.; Doganis, P. NanoSolveIT Project: Driving nanoinformatics research to develop
innovative and integrated tools for in silico nanosafety assessment. Comput. Struct. Biotechnol. J.
2020
,
18, 583–602. [CrossRef]
243.
Collins, A.R.; Annangi, B.; Rubio, L.; Marcos, R.; Dorn, M.; Merker, C.; Estrela-Lopis, I.; Cimpan, M.R.;
Ibrahim, M.; Cimpan, E. High throughput toxicity screening and intracellular detection of nanomaterials.
Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017,9, e1413. [CrossRef]
244.
Ostermann, M.; Sauter, A.; Xue, Y.; Birkeland, E.; Schoelermann, J.; Holst, B.; Cimpan, M.R.
Label-free impedance flow cytometry for nanotoxicity screening. Sci. Rep. 2020,10, 1–14. [CrossRef]
245.
Ruzycka, M.; Cimpan, M.R.; Rios-Mondragon, I.; Grudzinski, I.P. Microfluidics for studying metastatic
patterns of lung cancer. J. Nanobiotechnol. 2019,17, 71. [CrossRef]
246.
Bor, G.; Mat Azmi, I.D.; Yaghmur, A. Nanomedicines for cancer therapy: Current status, challenges and
future prospects. Ther. Deliv. 2019,10, 113–132. [CrossRef] [PubMed]
247.
Dusinska, M.; Mariussen, E.; Rund
é
n-Pran, E.; Hudecova, A.M.; Elje, E.; Kazimirova, A.; El Yamani, N.;
Dommershausen, N.; Tharmann, J.; Fieblinger, D.; et al. In Vitro Approaches for Assessing the Genotoxicity
of Nanomaterials. Methods Mol. Biol. 2019,1894, 83–122. [CrossRef] [PubMed]
248.
Jesus, S.; Schmutz, M.; Som, C.; Borchard, G.; Wick, P.; Borges, O. Hazard assessment of polymeric
nanobiomaterials for drug delivery: What can we learn from literature so far. Front. Bioeng. Biotechnol.
2019
,
7, 261. [CrossRef] [PubMed]
249.
Kanwal, Z.; Raza, M.A.; Manzoor, F.; Riaz, S.; Jabeen, G.; Fatima, S.; Naseem, S. A comparative assessment of
nanotoxicity induced by metal (silver, nickel) and metal oxide (cobalt, chromium) nanoparticles in Labeo
rohita. Nanomaterials 2019,9, 309. [CrossRef] [PubMed]
250. Gatto, F.; Bardi, G. Metallic nanoparticles: General research approaches to immunological characterization.
Nanomaterials 2018,8, 753. [CrossRef] [PubMed]
Publisher’s Note:
MDPI stays neutral with regard to jurisdictional claims in published maps and institutional
aliations.
©
2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).
... Precision epigenomic therapies may fulfill the potential of early epigenetic approaches that, despite potent effects, were plagued by poor pharmacokinetic and safety/tolerability profiles 5,30 . We describe a platform that enables the rapid design and development of programmable mRNA therapeutics enabling precise and durable epigenomic controlachieving the targeted specificity of CRISPR and other gene editing approaches without permanent changes to target cell DNA. ...
Article
Full-text available
Pathogenic gene dysregulation can be attributed to chromatin state change that pre-transcriptionally regulates expression. Recent breakthroughs elucidating the rules governing this DNA control layer, an epigenetic code, unlock a modality in precision medicine to target gene dysregulation across myriad diseases. Here we present a modular platform to design programmable mRNA therapeutics, Epigenomic Controllers (EC), that control gene expression through directed epigenetic change. By leveraging natural mechanisms, ECs tune expression levels of one or multiple genes with durable effect of weeks-to-months in female mice following a single dose. We design and characterize ECs to multiple target genes and identify an EC that effectively inhibits the cancer- and inflammatory-disorder-associated multi-gene cluster CXCL1-8. With precision targeting of NF-kB signaling and identification of homologous murine surrogates, ECs significantly reduce neutrophil migration in vivo during acute lung inflammation in female mice. A platform approach to EC design for epigenomic modulation expands treatment frontiers for diverse gene targets, including those considered “undruggable.”
... Epigenetic modifications play a critical role in the evolution of breast cancer, standing apart from the mutational changes traditionally associated with cancer's molecular genesis [24]. These epigenetic changes, characterized by their frequency, reversibility, and specificity to certain genes, significantly contribute to the process of cell transformation, primarily through DNA methylation patterns [25]. ...
Article
Full-text available
Purpose of Review This review aims to explore the multifaceted landscape of breast cancer, focusing on the crucial role of genetic and genomic studies in understanding its heterogeneity and guiding personalized treatment strategies. It seeks to address the genetic underpinnings of breast cancer and the potential for developing targeted therapies. Recent Findings Recent advancements in genome-wide analysis and next-generation sequencing technologies have significantly advanced our understanding of the genetic complexities of breast cancer. Key discoveries include gene copy number alterations, mutational signatures, and intricate patterns of intra-tumoral heterogeneity. Specific genetic aberrations, such as the “kataegis” phenomenon and mutations in the integrin signaling pathway, have been identified, providing new avenues for therapeutic intervention. These findings highlight the diversity of breast cancer subtypes, including triple-negative breast cancer (TNBC), and underscore the importance of personalized medicine. Summary The review concludes that dissecting the genomic landscape of breast cancer reveals significant genetic diversity and potential targets for therapy. Identifying specific genetic alterations holds promise for developing targeted therapies, emphasizing the necessity of personalized medicine in breast cancer care. Despite challenges in integrating these genetic insights into clinical practice, the potential for more effective and tailored treatment approaches is substantial. Future research should focus on overcoming these challenges to fully realize the benefits of personalized medicine for breast cancer patients.
... Several liposomal formulations have received EMA and/or FDA approval for the treatment of various cancer types. 72 To enhance the effectiveness of histone deacetylase inhibitors (HDACIs) in solid tumors, PEGylated liposomes were developed for the encapsulation of trichostatin A (TSA), CG1521 (CG), and PXD101 (PXD). 73 These liposome formulations serve as promising nanocarriers, demonstrating stability in size, charge, and biological activity for one month when stored at 4°C. ...
Article
Full-text available
Epigenetic dysregulation can significantly trigger the onset and progression of various diseases, epigenetic therapy is a new treatment strategy by changing DNA methylation, histone modification, N6-methyladenosine, chromatin modification and other epigenetic modifications to regulate gene expression levels for therapeutic purposes. However, small-molecule epigenetic drugs face challenges in disease treatment, such as lack of selectivity, limited therapeutic efficacy, and insufficient safety. Nanomedicine delivery systems offer significant advantages in addressing these issues by enhancing drug targeting, improving bioavailability, and reducing nonspecific distribution. This help minimize side effects while increasing both therapeutic effectiveness and safety of epigenetic drugs. In this review, we focus on the mechanism and role of epigenetic regulatory factors in diseases, as well as the challenges faced by small molecule inhibitors in treatment strategies, especially the research advancements in epigenetic drug delivery systems, review and discuss the therapeutic potential and challenges of using nanotechnology to develop epigenetic drug delivery systems.
... Nanomedicine has revolutionized cancer treatment by leveraging nanoparticles (NPs), typically within the size range of 1 to 100 nm, for various therapeutic and diagnostic purposes. NPs-based epigenetic drug delivery, offering a precision-oriented strategy to target the underlying molecular mechanisms driving tumorigenesis [115]. One of its key advantages lies in targeted drug delivery, where NPs can be precisely engineered to target cancer cells, sparing healthy tissues and minimizing the often debilitating side effects associated with traditional cancer treatments [40,43]. ...
Article
Full-text available
Cancer has arisen from both genetic mutations and epigenetic changes, making epigenetics a crucial area of research for innovative cancer prevention and treatment strategies. This dual perspective has propelled epigenetics into the forefront of cancer research. This review highlights the important roles of DNA methylation, histone modifications and non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs) and long non-coding RNAs, which are key regulators of cancer-related gene expression. It explores the potential of epigenetic-based therapies to revolutionize patient outcomes by selectively modulating specific epigenetic markers involved in tumorigenesis. The review examines promising epigenetic biomarkers for early cancer detection and prognosis. It also highlights recent progress in oligonucleotide-based therapies, including antisense oligonucleotides (ASOs) and antimiRs, to precisely modulate epigenetic processes. Furthermore, the concept of epigenetic editing is discussed, providing insight into the future role of precision medicine for cancer patients. The integration of nanomedicine into cancer therapy has been explored and offers innovative approaches to improve therapeutic efficacy. This comprehensive review of recent advances in epigenetic-based cancer therapy seeks to advance the field of precision oncology, ultimately culminating in improved patient outcomes in the fight against cancer.
... In addition, processes such as angiogenesis and evasion of the host immune system are regulated by c-Myc. Increased c-Myc levels result in increased expression of vascular endothelial growth factor (VEGF) via activation of the b-catenin pathway [63]. ...
Article
Full-text available
This review shows how mammary stem cells (MaSCs) influence breast development, breastfeeding, and breast cancer risk. MaSCs, which can differentiate into various cell types, are vital for breast tissue health, but also disease development in breast tissue. Research shows that breastfeeding affects MaSCs, offering protection against breast cancer through various mechanisms. Hormonal changes such as increased prolactin concentration, oxytocin secretion, lower progesterone levels, and reduced exposure to estrogen during lactation promote apoptosis in potential cancer cells, boost immune surveillance, and modulate inflammation. Key findings reveal that pregnancy at an earlier age and extended breastfeeding reduce MaSC numbers, lowering cancer risk. Additionally, breastfeeding induces various epigenetic changes, such as DNA methylation and histone modification, which provide long-term protection against the development of cancer. Components of breast milk, like alpha-lactalbumin and lactoferrin, contribute by promoting cancer cell apoptosis and inhibiting tumor growth. The dual benefits of breastfeeding are reduced breast cancer risk for mothers and immunological advantages for infants. Multicenter epidemiology research has focused particular attention on longer breastfeeding duration associated with a reduced risk of triple-negative breast cancer. This review offers comprehensive evidence that breastfeeding protects against breast cancer through various biological, hormonal, and molecular mechanisms, showing the importance of promoting breastfeeding as a natural cancer prevention method. This article reviews the role of mammary stem cells in breast development, lactation, and breast cancer.
... Despite advances in diagnosis and treatment, the molecular mechanisms underlying BC development/progression remain incompletely understood [2]. Identifying novel genetic and epigenetic markers associated with BC risk and outcomes is essential for improving preventive measurements, early detection, and personalized therapeutic strategies [3,4]. ...
Article
Full-text available
MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression and play critical roles in tumorigenesis. Genetic variants in miRNA processing genes, DROSHA and DICER, have been implicated in cancer susceptibility and progression in various populations. However, their role in Egyptian patients with breast cancer (BC) remains unexplored. This study aims to investigate the association of DROSHA rs10719 and DICER rs3742330 polymorphisms with BC risk and clinical outcomes. This case–control study included 209 BC patients and 106 healthy controls. Genotyping was performed using TaqMan assays in blood, tumor tissue, and adjacent non-cancerous tissue samples. Associations were analyzed using logistic regression and Fisher’s exact test. The DROSHA rs10719 AA genotype was associated with a 3.2-fold increased risk (95%CI = 1.23–9.36, p < 0.001), and the DICER rs3742330 GG genotype was associated with a 3.51-fold increased risk (95%CI = 1.5–8.25, p = 0.001) of BC. Minor allele frequencies were 0.42 for rs10719 A and 0.37 for rs3742330 G alleles. The risk alleles were significantly more prevalent in tumor tissue than adjacent normal tissue (rs10719 A: 40.8% vs. 0%; rs3742330 G: 42.7% vs. 0%; p < 0.001). However, no significant associations were observed with clinicopathological features or survival outcomes over a median follow-up of 17 months. In conclusion, DROSHA rs10719 and DICER rs3742330 polymorphisms are associated with increased BC risk and more prevalent in tumor tissue among our cohort, suggesting a potential role in miRNA dysregulation during breast tumorigenesis. These findings highlight the importance of miRNA processing gene variants in BC susceptibility and warrant further validation in larger cohorts and different ethnic populations.
Article
Full-text available
This article provides a comprehensive analysis of the signaling pathways implicated in breast cancer (BC), the most prevalent malignancy among women and a leading cause of cancer-related mortality globally. Special emphasis is placed on the structural dynamics of protein complexes that are integral to the regulation of these signaling cascades. Dysregulation of cellular signaling is a fundamental aspect of BC pathophysiology, with both upstream and downstream signaling cascade activation contributing to cellular process aberrations that not only drive tumor growth, but also contribute to resistance against current treatments. The review explores alterations within these pathways across different BC subtypes and highlights potential therapeutic strategies targeting these pathways. Additionally, the influence of specific mutations on therapeutic decision-making is examined, underscoring their relevance to particular BC subtypes. The article also discusses both approved therapeutic modalities and ongoing clinical trials targeting disrupted signaling pathways. However, further investigation is necessary to fully elucidate the underlying mechanisms and optimize personalized treatment approaches.
Article
Introduction: Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), approved by the US FDA for obesity treatment, are typically administered subcutaneously, an invasive method leading to suboptimal patient adherence and peripheral side effects. Additionally, this route requires the drug to cross the restrictive blood-brain barrier (BBB), limiting its safety and effectiveness in weight management and cognitive addiction disorders. Delivering the drug intranasally could overcome these drawbacks. Areas covered: This review summarizes GLP-1 RAs used as anti-obesity agents, focusing on the intranasal route as a potential pathway to deliver these biomolecules to the brain. It also discusses strategies to overcome challenges associated with nasal delivery. Expert opinion: Nose-to-brain (N2B) pathways can address limitations of the subcutaneous route for GLP-1 RAs. However, peptide delivery to the brain is challenging due to nasal physiological barriers and the drug's physicochemical properties. Innovative approaches, such as cell permeation enhancers, mucoadhesive systems, and nanocarriers in nasal formulations, along with efficient drug delivery devices, show promising preclinical results. Despite this, successful preclinical data does not guarantee clinical effectiveness, highlighting the need for comprehensive clinical investigations to optimize formulations and fully utilize the nose-to-brain interface for peptide delivery.
Article
Full-text available
Gene mutations are strongly associated with tumor progression and are well known in cancer development. However, recently discovered epigenetic alterations have shown the potential to greatly influence tumoral response to therapy regimens. Such epigenetic alterations have proven to be dynamic, and thus could be restored. Due to their reversible nature, the promising opportunity to improve chemotherapy response using epigenetic therapy has arisen. Beyond helping to understand the biology of the disease, the use of modern clinical epigenetics is being incorporated into the management of the cancer patient. Potential epidrug candidates can be found through a process known as drug repositioning or repurposing, a promising strategy for the discovery of novel potential targets in already approved drugs. At present, novel epidrug candidates have been identified in preclinical studies and some others are currently being tested in clinical trials, ready to be repositioned. This epidrug repurposing could circumvent the classic paradigm where the main focus is the development of agents with one indication only, while giving patients lower cost therapies and a novel precision medical approach to optimize treatment efficacy and reduce toxicity. This review focuses on the main approved epidrugs, and their druggable targets, that are currently being used in cancer therapy. Also, we highlight the importance of epidrug repurposing by the rediscovery of known chemical entities that may enhance epigenetic therapy in cancer, contributing to the development of precision medicine in oncology.
Article
Full-text available
This review aims to summarize the methods that have been used till today, highlight methods that are currently being developed, and predict the future roadmap for anticancer therapy. In the beginning of this review, established approaches for anticancer therapy, such as conventional chemotherapy, hormonal therapy, monoclonal antibodies, and tyrosine kinase inhibitors are summarized. To counteract the side effects of conventional chemotherapy and to increase limited anticancer efficacy, nanodrug- and stem cell-based therapies have been introduced. However, current level of understanding and strategies of nanodrug and stem cell-based therapies have limitations that make them inadequate for clinical application. Subsequently, this manuscript reviews methods with fewer side effects compared to those of the methods mentioned above which are currently being investigated and are already being applied in the clinic. The newer strategies that are already being clinically applied include cancer immunotherapy, especially T cell-mediated therapy and immune checkpoint inhibitors, and strategies that are gaining attention include the manipulation of the tumor microenvironment or the activation of dendritic cells. Tumor-associated macrophage repolarization is another potential strategy for cancer immunotherapy, a method which activates macrophages to immunologically attack malignant cells. At the end of this review, we discuss combination therapies, which are the future of cancer treatment. Nanoparticle-based anticancer immunotherapies seem to be effective, in that they effectively use nanodrugs to elicit a greater immune response. The combination of these therapies with others, such as photothermal or tumor vaccine therapy, can result in a greater anticancer effect. Thus, the future of anticancer therapy aims to increase the effectiveness of therapy using various therapies in a synergistic combination rather than individually.
Article
Full-text available
Drug delivery technology has a wide spectrum, which is continuously being upgraded at a stupendous speed. Different fabricated nanoparticles and drugs possessing low solubility and poor pharmacokinetic profiles are the two major substances extensively delivered to target sites. Among the colloidal carriers, nanolipid dispersions (liposomes, deformable liposomes, virosomes, ethosomes, and solid lipid nanoparticles) are ideal delivery systems with the advantages of biodegradation and nontoxicity. Among them, nano-structured lipid carriers and solid lipid nanoparticles (SLNs) are dominant, which can be modified to exhibit various advantages, compared to liposomes and polymeric nanoparticles. Nano-structured lipid carriers and SLNs are non-biotoxic since they are biodegradable. Besides, they are highly stable. Their (nano-structured lipid carriers and SLNs) morphology, structural characteristics, ingredients used for preparation, techniques for their production, and characterization using various methods are discussed in this review. Also, although nano-structured lipid carriers and SLNs are based on lipids and surfactants, the effect of these two matrixes to build excipients is also discussed together with their pharmacological significance with novel theranostic approaches, stability and storage.
Article
Full-text available
The use of nanomaterials in biomedicine has increased over the past 10 years, with many different nanoparticle systems being utilised within the clinical setting. With limited emerging success in clinical trials, polymeric, metallic, and lipid based nanoparticles have all found a place in medicine, with these generally providing enhanced drug efficacy or therapeutic effect compared to the standard drug treatments. Although there is great anticipation surrounding the field of nanomedicine and its influence on the pharmaceutical industry, there is currently very little regulatory guidance in this area, despite repeated calls from the research community, something that is critical to provide legal certainty to manufacturers, policymakers, healthcare providers and the general public. This is reflected in the lack of an international definition of what these materials are, with several bodies, including the National Institute of Health USA, the European Science Foundation and the European Technology Platform, have differing definitions, and the FDA having no clear definition at all. The uncertainty created by the lack of consistency across the board may ultimately impact funding, research and development of such products negatively thus destroying public acceptance and perception of nano-products. This review aims to discuss the use of nanomaterials within the clinical setting, why regulation of these materials is so important, and the challenges faced in regulating these materials generally, as well as the current regulation used in different nations.
Article
Full-text available
Breast cancer is the neoplasm with the highest number of deaths in women. Although the molecular mechanisms associated with the development of this tumor have been widely described, metastatic disease has a high mortality rate. In recent years, several studies show that microRNAs or miRNAs regulate complex processes in different biological systems including cancer. In the present work, we describe a group of 61 miRNAs consistently over-expressed in breast cancer (BC) samples that regulate the breast cancer transcriptome. By means of data mining from TCGA, miRNA and mRNA sequencing data corresponding to 1091 BC patients and 110 normal adjacent tissues were downloaded and a miRNA–mRNA network was inferred. Calculations of their oncogenic activity demonstrated that they were involved in the regulation of classical cancer pathways such as cell cycle, PI3K–AKT, DNA repair, and k-Ras signaling. Using univariate and multivariate analysis, we found that five of these miRNAs could be used as biomarkers for the prognosis of overall survival. Furthermore, we confirmed the over-expression of two of them in 56 locally advanced BC samples obtained from the histopathological archive of the National Cancer Institute of Mexico, showing concordance with our previous bioinformatic analysis.
Article
Full-text available
Breast cancer stem(-like) cells (BCSCs) have been found to be responsible for therapeutic resistance and disease relapse. BCSCs are difficult to eradicate due to their high resistance to conventional treatments and high plasticity. Functionalised nanoparticles have been investigated as smart vehicles to transport across various barriers and increase the interaction of therapeutic agents with cancer cells, as well as BCSCs. In this review, we discuss the different characteristics of BCSCs, and challenges to tackle BCSCs at cellular and molecular levels. The mechanisms of action and physicochemical properties of the current BCSC targeting agents are also covered. We will focus on the rational design and recent advances of “Nano + Nano” or single tumour targeting nanoparticle systems loaded with dual or multiple agents to kill all cancer cells including BCSCs. These cocktail therapies include the combination of a chemotherapy agent with a BCSC-specific inhibitor, a phytochemical agent or RNA based therapy. Given the heterogeneity of breast tumour tissue, targeting both BCSCs and bulk breast cancer cells simultaneously with multiple agents holds great promise in eliminating breast cancer. The future research needs to focus on overcoming various barriers in the ‘clinical translation’ of BCSC-targeting nanomedicines to cure breast cancer, which requires a significant multidisciplinary effort.
Article
Full-text available
Epithelial to mesenchymal transition (EMT) is the process whereby a polarized epithelial cell ceases to maintain cell-cell contacts, loses expression of characteristic epithelial cell markers, and acquires mesenchymal cell markers and properties such as motility, contractile ability, and invasiveness. A complex process that occurs during development and many disease states, EMT involves a plethora of transcription factors (TFs) and signaling pathways. Whilst great advances have been made in both our understanding of the progressive cell-fate changes during EMT and the gene regulatory networks that drive this process, there are still gaps in our knowledge. Epigenetic modifications are dynamic, chromatin modifying enzymes are vast and varied, transcription factors are pleiotropic, and signaling pathways are multifaceted and rarely act alone. Therefore, it is of great importance that we decipher and understand each intricate step of the process and how these players at different levels crosstalk with each other to successfully orchestrate EMT. A delicate balance and fine-tuned cooperation of gene regulatory mechanisms is required for EMT to occur successfully, and until we resolve the unknowns in this network, we cannot hope to develop effective therapies against diseases that involve aberrant EMT such as cancer. In this review, we focus on data that challenge these unknown entities underlying EMT, starting with EMT stimuli followed by intracellular signaling through to epigenetic mechanisms and chromatin remodeling.
Article
Full-text available
The blooming of nanotechnology has made available a limitless landscape of solutions responding to crucial issues in many fields and, nowadays, a wide choice of nanotechnology-based strategies can be adopted to circumvent the limitations of conventional therapies for cancer. Herein, the current stage of nanotechnological applications for cancer management is summarized encompassing the core nanomaterials as well as the available chemical–physical approaches for their surface functionalization and drug ligands as possible therapeutic agents. The use of nanomaterials as vehicles to delivery various therapeutic substances is reported emphasizing advantages, such as the high drug loading, the enhancement of the pay-load half-life and bioavailability. Particular attention was dedicated to highlight the importance of nanomaterial intrinsic features. Indeed, the ability of combining the properties of the transported drug with the ones of the nano-sized carrier can lead to multifunctional theranostic tools. In this view, fluorescence of carbon quantum dots, optical properties of gold nanoparticle and superparamagnetism of iron oxide nanoparticles, are fundamental examples. Furthermore, smart anticancer devices can be developed by conjugating enzymes to nanoparticles, as in the case of bovine serum amine oxidase (BSAO) and gold nanoparticles. The present review is aimed at providing an overall vision on nanotechnological strategies to face the threat of human cancer, comprising opportunities and challenges.
Article
Full-text available
Nanomedicine that combines technology with medical diagnostics and therapeutics is now advancing to greater heights for its significant potential in drug delivery. Epigenetic modifications that alter gene expressions patterns are often associated with disease progression and therapeutic resistance. Histone Deacetylase (HDAC) enzymes that are upregulated in Cancer and Alzheimer's are being highly researched as promising targets for treatment strategies. Even though there have been a spate of HDAC inhibitor drugs currently in clinical trials or in the market, side effects associated with toxicity and non specific targeting warrant the need for a better design for efficient drug delivery. To date there are very few clinically approved nanovectors and even fewer nanoparticles that can safely deliver HDAC inhibitors in vitro and in vivo. This review will examine the different types of nanomaterials that are currently available and enumerate the current studies being carried out with regard to nanomediated HDAC inhibitor delivery. We will discuss the challenges that face combining HDAC inhibitors with different nanoparticles and underscore the potential that exists to develop these strategies further with a specific focus on anti-cancer therapy.
Article
Breast cancer (BC) is one of the most prevalent cancers in women. Triple-negative breast cancer (TNBC) in which the three major receptors i.e. estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2), are absent is known to express the most aggressive phenotype and increased metastasis which results in the development of resistance to chemotherapy. It offers various therapeutic advantages in treating BC and TNBC. Nanotechnology offers various unique characteristics such as small size (nanometric), active and passive targeting, and the ability to attach multiple targeting moieties, controlled release, and site-specific targeting. This review focuses on conventional drug therapies, recent treatment strategies, and unique therapeutic approaches available for BC and TNBC. The role of breast cancer stem cells in the recurrence of BC and TNBC has also been highlighted. Several chemotherapeutic agents delivered using nanocarriers such as polymeric nanoparticles/micelles, metallic/inorganic NPs, and lipid-based NPs (Liposome, solid-lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs)), etc. with excellent responses in the treatment of BC/TNBC along with breast cancer stem cells have been discussed in details. Moreover, the application of nanomedicine including CRISPR nanoparticle, exosomes for the treatment of BC/TNBC and other molecular targets available such as poly (ADP-ribose) polymerase (PARP), epidermal growth factor receptor (EGFR), Vascular endothelial growth factor (VEGF), etc. for further exploration have also been discussed.