You are on page 1of 12

Pharmacological Research 163 (2021) 105320

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Breast cancer stem cells, heterogeneity, targeting therapies and


therapeutic implications
Xiaobin Zeng a, b, 1, Chengxiao Liu a, 1, Jie Yao a, Haoqiang Wan a, b, c, Guoqing Wan d,
Yingpeng Li c, **, Nianhong Chen a, e, f, *
a
Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First
Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, PR China
b
Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Medicine School of Shenzhen University, Shenzhen, Guangdong Province, 518037, PR China
c
Department of Gastroenterology, (Longhua Branch), Shenzhen People’s Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen, Guangdong Province,
518120, PR China
d
Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, PR China
e
Department of Cell Biology & University of Pittsburgh Cancer Institute, School of Medicine, University of Pittsburgh, PA, 15261, USA
f
Laboratory of Signal Transduction, Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, 10065, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Both hereditary and sporadic breast cancer are suggested to develop from a stem cell subcomponent retaining
Breast cancer stem cell most key stem cell properties but with dysregulation of self-renewal pathways, which drives tumorigenic dif­
Breast cancer therapy ferentiation and cellular heterogeneity. Cancer stem cells (CSCs), characterized by their self-renewal and dif­
Cancer heterogeneity
ferentiation potential, have been reported to contribute to chemo-/radio-resistance and tumor initiation and to
Ubiquitination
be the main reason for the failure of current therapies in breast cancer and other CSC-bearing cancers. Thus, CSC-
targeted therapies, such as those inducing CSC apoptosis and differentiation, inhibiting CSC self-renewal and
division, and targeting the CSC niche to combat CSC activity, are needed and may become an important
component of multimodal treatment. To date, the understanding of breast cancer has been extended by advances
in CSC biology, providing more accurate prognostic and predictive information upon diagnosis. Recent im­
provements have enhanced the prospect of targeting breast cancer stem cells (BCSCs), which has shown promise
for increasing the breast cancer remission rate. However, targeted therapy for breast cancer remains challenging
due to tumor heterogeneity. One major challenge is determining the CSC properties that can be exploited as
therapeutic targets. Another challenge is identifying suitable BCSC biomarkers to assess the efficacy of novel
BCSC-targeted therapies. This review focuses mainly on the characteristics of BCSCs and the roles of BCSCs in the
formation, maintenance and recurrence of breast cancer; self-renewal signaling pathways in BCSCs; the BCSC
microenvironment; potential therapeutic targets related to BCSCs; and current therapies and clinical trials tar­
geting BCSCs.

1. Introduction breast cancer has decreased significantly in the United States and else­
where, largely due to improvement in early detection and the develop­
WHO statistics indicate that in 2018, breast cancer caused 627,000 ment of more effective adjuvant therapies [1]. However, cancer
deaths and 2.1 million new cases were diagnosed. Among the causes of mortality is still high, and there is no curative treatment.
death, many patients die of metastasis, recurrence and drug resistance Accumulating evidence indicates that normal breast cancer cells can
due to failure of current therapies. Over the past decades, mortality from acquire cancer stem cell (CSC) properties, resulting in an increase in

* Lead corresponding author at: Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People’s Hospital, The Second Clinical Medical
College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, PR China.
** Corresponding author at:Department of Gastroenterology, (Longhua Branch), Shenzhen People’s Hospital, 2nd Clinical Medical College of Jinan University,
Shenzhen, Guangdong Province, 518120, PR China.
E-mail addresses: pqr9hs@163.com (Y. Li), nhchen2004@hotmail.com (N. Chen).
1
These authors have contributed equally to this work.

https://doi.org/10.1016/j.phrs.2020.105320
Received 17 October 2020; Received in revised form 27 November 2020; Accepted 27 November 2020
Available online 1 December 2020
1043-6618/© 2020 Elsevier Ltd. All rights reserved.
X. Zeng et al. Pharmacological Research 163 (2021) 105320

their chemo-/radio-resistance ability. In addition, the proportion of tumors. In addition to these findings based on preclinical and experi­
breast cancer stem cells (BCSCs) in tumors is increased after chemo-/ mental data, a competing hypothesis by Campbell et al. suggests that
radio-therapy, which further increases tumor heterogeneity and the CD44+/CD24− and CD44− /CD24+ cells may be competitive sub­
presence of BCSCs, subsequently leading to therapeutic failure or cancer populations of cancer cells, and this hypothesis is supported by evidence
recurrence. Eradicating CSCs, which are proposed to be the root cause of indicating that CD44+/CD24− cells are not associated with the pro­
cancer initiation and recurrence, has been thought to be a promising gression or prognosis of breast cancer, however, CD44− /CD24+ cells are
approach to improve cancer survival or even to cure cancer. In research negatively correlated with prognosis [14–18]. In addition, the
on killing CSCs, many promising methods have been developed, CD44+/CD24– profile is more closely related to basal breast cancer.
including molecular targeted therapy, microRNA (miRNA)-targeted Expression of CD44 may indicate that any feature of CSCs enhances their
therapy, differentiation therapy, tumor microenvironment-targeted implantation [19]. Notably, luminal-type breast cancer-specific CSCs
therapy, and immunotherapy. have not been identified. However, the above stem cell markers are not
In this article, we review the characteristics of BCSCs, discuss the exclusively expressed on BCSCs but are also expressed on other cells.
regulatory pathways of these cells and potential targets for therapy, and Recently, a study reported the identification of a BCSC-like population
consider the implications of CSC models for clinical trial design. among 3 populations in a breast cancer cell line based on differential
gene transcription detected using single-cell RNA sequencing [20].
2. BCSCs Therefore, DNA/RNA sequencing may reveal more information about
genes and allow more accurate identification of BCSCs in the future.
As early as 1937, CSCs were confirmed to be present in human acute
myeloid leukemia. In 2003, the first solid CSCs were identified in breast 3. The roles of BCSCs in models of heterogeneity in breast cancer
tumors. Subsequently, other CSCs were isolated and identified from and the case for the development of BCSC-targeted therapy
many tumors, including brain, melanoma, prostate, ovarian, colon,
pancreatic, and other types of tumors. CSCs can be defined by three Variability among tumors arising in the same organ or patient is
functional characteristics: the capacity for tumor initiation and forma­ often referred to as intertumor heterogeneity. Because of intertumor
tion, long-term self-renewal, and differentiation into non-self-renewing heterogeneity, tumors can be classified into different types according to
cells [2,3]. Self-renewal can occur via either symmetric division or their morphology, molecular expression profiles or genomic copy
asymmetric cell division. Two daughter stem cells are produced via number patterns [21,22]. Two hypothetical models have been proposed
symmetric self-renewal. Alternatively, via asymmetric self-renewal, a to explain the intertumor heterogeneity: 1) different genetic and/or
daughter stem cell and a nonstem differentiated cell are produced [4,5]. epigenetic events occur within the same target cell, leading to different
The serial tumor transplantation assay, which can demonstrate tumor phenotypes, and 2) different tumor subtypes are derived from
self-renewal and differentiation potential, has been used as a gold different cell of origin in tissues [22] (Fig. 1). Breast cancer can be
standard to ascertain whether a specific population of cells comprises classified into three subtypes based on the intertumor heterogeneity in
CSCs [6]. CSCs may originate from normal adult stem cells, progenitor cell surface molecule expression: 1) hormone receptor-positive (HR+)
cells, differentiated cells, fused cells from stem cells, and cancer cells or breast cancer, with expression of the estrogen receptor (ER) and/or
cells that acquire DNA from other cells [7,8]. CSCs are more likely to be progesterone receptor (PR); 2) human epidermal receptor 2 (HER2)-­
transformed from normal stem cells than from other cells due to accu­ amplified (also called ERBB2-amplified) breast cancer; and 3)
mulated mutations during their relatively long lives [9]. Progenitors and triple-negative breast cancer (TNBC), lacking expression of ER, PR, and
other differentiated cells are also thought to give rise to CSCs via mu­ HER2 [23].
tations, especially mutations in self-renewal genes. However, these cells The cancer cells within a given primary tumor show amazing clonal,
must survive for a relatively shorter time and harbor more mutations genetic and morphological diversity (i.e., intratumor heterogeneity).
than stem cells. Tumor cells often have diverse functional properties. Different pop­
One hypothesis proposed that mutations in mammary stem cells can ulations of cancer cells endow the given tumor with multiple charac­
primarily trigger the formation of breast cancer and that mutations in teristics supporting tumorigenesis, such as angiogenesis, invasion and
progenitor cells contribute to transforming events in the mammary gland. metastasis [24]. The diverse expression of markers in cells within a
Mouse BCSCs, characterized as having an EpCAM+/CD44+/CD24− /low tumor also reflects intratumor heterogeneity. For example, the per­
phenotype, were first identified in a tumorigenesis experiment in xeno­ centage of ER-expressing cells can vary widely and range from 1 to 100
transplanted immune-deficient mice [10]. Later, human cells with a % within a single breast tumor.
CD44+/CD24–/low/Lin– expression phenotype were isolated and identi­ The ‘clonal evolution’ and ‘CSC’ hypotheses constitute the two major
fied as BCSCs. Tumors formed when 100 CD44+/CD24–/low/Lin– cells hypothetical models of tumorigenesis [14, 25]. These two models were
were injected into nonobese diabetic/severe combined immunodefi­ developed to explain intratumoral heterogeneity and intrinsic differ­
ciency (NOD/SCID) mice. Conversely, no tumors formed when 20,000 ences in the breast tumor regeneration capacity [26] (Fig. 2A and B).
cells without this phenotype were injected. This experiment demonstrated The clonal evolution model is based largely on the hypothesis that any
that CD44+/CD24–/low/Lin– subtype cells have key characteristics, undifferentiated or differentiated cell can accumulate mutations that
including self-renewal and differentiation abilities, and that tumors can lead to tumor formation and to the generation of clonal populations of
originate from a rare population of BCSCs with specific markers [10]. cells within a tumor. In contrast, the CSC model proposes that exclu­
Molecular classification of breast cancer has been successfully sively normal stem cells or their early progeny can be transformed into
applied for the design of targeted therapies, resulting in significant ef­ CSCs by the accumulation of mutations. These proliferating stem cells
fects and an increase in the survival rate [11,12]. Breast cancers of then drive tumor formation and recurrence by undergoing a carcino­
different subtypes and histological stages have been reported to express genic differentiation process [25,27]. In addition, a third hypothesis for
unique markers, including the stem cell markers CD44 and CD24, as well tumor generation denies the existence of clear subpopulations of cancer
as other markers, such as vimentin, osteonectin, connexin 43, MUC1, cells within tumors (Fig. 2C), stating instead that cancer cells have
and CK18 [13]. Generally, highly heterogeneous expression of these substantial diversity resulting from the accumulation of random and
markers in different tumor subtypes and histological stages have been gradual mutations [28,29]. The CSC model can explain breast cancer
demonstrated in previous studies. CD44 and CD24 are most commonly tumorigenesis [10]. Some evidence supports the CSC model in tumori­
expressed in basal-like tumors. Another stem cell marker, ALDH1, has genesis: 1) the results from the breast tumorigenesis experiment of CSCs
the highest expression in HER2+ and basal-like tumors. In contrast, in xenotransplanted immune-deficient mice, 2) markers for distant BCSC
CK18, GATA3 and MUC1 are more commonly expressed in luminal-type clones, 3) the model showing intratumor heterogeneity of breast cancer

2
X. Zeng et al. Pharmacological Research 163 (2021) 105320

Fig. 1. Two models describing the establishment of intertumor heterogeneity.

Fig. 2. Proposed models explaining intratumor heterogeneity in breast cancer. Different models of tumor progression can explain distinct types of intratumor
heterogeneity (A–C), exemplified here by the clonal evolution (A), cancer stem cell (B), and mutator phenotype (C) models. The different models can result in distinct
spatial distributions of subpopulations.

3
X. Zeng et al. Pharmacological Research 163 (2021) 105320

tumors, and 4) the model showing breast cancer tumor relapse and is still unclear, but the resistance is related to antiapoptotic effects and
metastasis. However, the CSC model is controversial in breast cancer induction of quiescence [35]. Most CSCs are in the G0 phase of the cell
principally because the xenotransplanted immune-deficient mouse cycle and divide infrequently, which can lead to resistance to cell
model cannot mimic the normal mouse and human environment, and cycle-specific chemotherapeutic agents. Some ATP-binding cassette
the major difference in BCSCs make presence of BCSCs questionable. A proteins (ABCG2, ABCB1 and ABCC1) highly expressed in the plasma
CSC dynamic concept that emphases evolution and adaptation might be membrane of CSCs have been indicated to export chemotherapeutic
a supplement to explain tumorigenesis, tumor relapse and metastasis agents [36–38]. Furthermore, the BCSC marker ALDH plays important
[30]. In general, CSCs in one tumor are genetically unstable, resulting in roles in the reduction of oxidative stress; resistance to some chemo­
the development of multiple distinct CSC clones (intratumor CSCs clone therapeutic agents, such as oxazolidine, taxanes, and platinum; and
hierarchy) in advanced cancer. After environmental selection including removal of free radicals resulting from radiotherapy [39,40]. In addi­
the niche and niche changes due to metastasis, targeted therapy and tion, the active DNA damage response and its repair pathways in BCSCs
chemotherapy, adapted CSCs survive from the CSC clone pool in the lead to a reduction in apoptosis or other forms of cell necrosis [19]. In
intratumor and intertumor hierarchy and develop to evolved tumors. addition to the intrinsic genetic and epigenetic aberrations involved in
Competition among the CSC clones is another factor in the evolution of resistance, the extrinsic factor, the tumor microenvironment, also reg­
cancer leading to a reduction in the diversity of CSC clones and ulates BCSC heterogeneity and plasticity and further affects the thera­
inter-CSC clone dominance. Distinct BCSC clones can be found in one peutic resistance.
tumor [31]. However, it should be noted that BCSCs can be classified Thus, several treatment options may be efficient for targeting BCSCs:
into clear subtypes by biomarkers so the BCSCs may be more stable 1) therapies targeting self-renewal signaling pathways, resulting in the
genetically. In breast cancer, the evolution and adaptation of tumors death of BCSCs; 2) therapies targeting the microenvironment of breast
promoted by intratumor heterogeneity lead to underestimation by CSCs; 3) therapies targeting drug transporters; 4) differentiation ther­
tumor genomic profiling via single tumor biopsy sampling analysis, apy; and 5) immunotherapy. In addition, therapies targeting miRNAs
which poses a major challenge to personalized medicine and identifi­ and the ubiquitin–proteasome system (UPS) may be options. Interest is
cation of biomarkers, especially those of BCSCs. Intratumor and inter­ increasing in combining such treatments with conventional anticancer
tumor heterogeneity may foster tumor adaptation and therapeutic treatments (chemo-/radiotherapy) targeting the renewal and differen­
failure through Darwinian selection in breast cancer, even though some tiation signaling pathways [such as the Notch and Hedgehog (Hh)
therapies may increase the fraction of CSCs. Killing all types of BCSCs is pathways] and/or with immunomodulatory components (such as PD-1
still a potential basic strategy for curing breast cancer. However, the and PD-L1) exploited by CSCs [41].
strategy only targeting BCSCs may be ineffective as mentioned above, as
new CSCs may re-emerge by dedifferentiation from progenitor cells, 5. BCSC self-renewal pathways and targeted therapies
differentiated cells, fused cells, and cancer cells or cells that acquire
stemness genes from other cells. Thus, eradicating the most types of The current understanding of the pathways that regulate the self-
breast cancer cells may be the most effective basic strategy for curing renewal of breast stem cells indicates that dysregulation of these path­
breast cancer. ways may lead to carcinogenesis. In the mammary gland, the signaling
pathways regulating the self-renewal of breast stem cells, such as the Hh,
4. BCSC-targeted therapies and their implication Wnt/β-catenin, and Notch pathways, play an important role both in
embryogenesis and organogenesis and in the maintenance of adult tis­
Most currently used therapies aim to eradicate as many cancer cells sues via regulation of the balance between self-renewal and differenti­
as possible to reduce the tumor size, but CSCs often survive under these ation of stem cells (Fig. 3). In the regulation of organs, these signaling
therapies, leading to treatment failure or recurrence. CSCs show rela­ pathways promote mammary gland organogenesis, development, and
tively high resistance to chemo-/radiotherapies [32,33]. As mentioned maintenance by regulating the balance between the differentiation and
above, BCSCs have high levels of heterogeneity and plasticity. Thus, self-renewal of stem cells [37]. Studies have demonstrated that dysre­
BCSCs express various proteins that can block drugs and promote the gulation of these pathways may cause malignant transformation [42].
transition between epithelial and mesenchymal states to contribute to Thus, these pathways constitute potential targets for elimination of
therapeutic resistance [34]. How BCSCs acquire the resistance from EMT BCSCs [43].

Fig. 3. Signal transduction networks in the BCSC microenvironment. Schematic illustration of important signal transduction networks, therapeutic targets, and
targeting agents.

4
X. Zeng et al. Pharmacological Research 163 (2021) 105320

5.1. Notch signaling eliminate BCSCs [64]. In addition, the combination of the Hh inhibitor
GDC-0449 (vismodegib) with paclitaxel, epirubicin, and cyclophos­
The Notch signaling pathway helps to determine stem cell fate [44] phamide has been evaluated for the treatment of triple-negative breast
and helps both normal and malignant stem cells to maintain their sur­ cancer in a phase II clinical trial.
vival and activity [45,46]. A Notch transmembrane receptor is activated
by a ligand [Delta-like (DLS) 1, 3, and 4; Jagged 1 and 2] presented by a 5.4. Her2 signaling
neighboring cell and undergoes serial cleavage events mediated by
gamma secretase. The Notch intracellular domain is separated from the Overexpression of the HER-2 gene, a member of the epidermal
Notch protein due to this cleavage and translocates to the nucleus, growth factor receptor family, is an early event in the formation of
leading to activation of downstream transcription factors such as Hes sporadic breast cancer [65]. Approximately 25 %–30 % of breast cancer
and Hay [44]. Aberrant activation of the Notch signaling pathway, patients have tumors of the HER-2-amplified subtype, which is corre­
which promotes chemo-/radioresistance, has been reported in breast lated with a low survival rate and a high risk of relapse [66]. Emerging
cancer cells and BCSCs [46,47]. Moreover, the levels of Notch and the studies have suggested that HER2 regulates BCSC activities, including
p53 inhibitor NUMB were found to be downregulated in approximately self-renewal and radioresistance [67–69]. HER2-targeted therapies are
50 % of human breast tumor samples [48]. Gamma secretase inhibitors very important and successful breast cancer therapeutics, especially in
(GSIs) efficiently block the Notch signaling pathway. Indeed, a phase I/II the treatment of HER-2-overexpressing and HER-2+ breast cancers.
clinical trial showed that the GSI MK-0752 in combination with doce­ Clinical data has shown that patients treated with HER2-targeted anti­
taxel significantly reduced the numbers of CD44+/CD24− stem cells and bodies and agents such as lapatinib, Kadcyla and trastuzumab had
ALDH+ stem cells and decreased the mammosphere formation efficiency increased progression-free survival and overall survival rates among
in breast tumors [49]. Approximately three other GSIs have completed patients with advanced -stage ER+ cancer that was resistant to hormone
clinical trials or are currently in phase II clinical trials in breast cancer, therapy [70]. Moreover, trastuzumab contributes to reducing cancer
although none of these has been approved for treatment [19]. recurrence [71]. If BCSCs are the root cause of breast cancer recurrence,
the significant clinical efficacy of HER-2 inhibitors may result from
5.2. Wnt signaling direct targeting of BCSCs by these drugs. Emerging studies have con­
nected the efficacy of HER-2-targeting agents to their ability to eliminate
Members of the Wnt family, a family of secreted signaling proteins, BCSCs [72]. For example, trastuzumab has been reported to reduce the
stimulate target cells by binding receptors on the cell surface. Canonical CSC population in HER2-overexpressing breast cancer cell lines [73]. In
Wnt signaling is triggered after Wnt proteins bind to receptors of the addition, lapatinib, an EGFR and HER-2/neu (ErbB-2) dual tyrosine ki­
Frizzled family and the receptors low-density lipoprotein-related pro­ nase inhibitor, reduced the percentage of the CD44+/CD24low popula­
teins LRP5 and LRP6. This Wnt/Frizzled/LRP5/6 complex transduces tion and the ability for self-renewal, as assessed by mammosphere
signals to the β-catenin and WNT/stabilization of proteins (STOP) formation, in ALDH1+/HER2+ BCSCs.
signaling cascades. β-Catenin translocates into the nucleus and activates
LEF1/TCF family transcription factors. Noncanonical Wnt signaling 5.5. PTEN and AKT signaling
activates Wnt/planar cell polarity (PCP), Wnt/receptor tyrosine kinase
(RTK) and WNT/Ca2+ signaling cascades via FZD receptors and/or The PTEN tumor suppressor gene encodes a lipid and protein phos­
ROR1/ROR2/RYK coreceptors [50,51]. The Wnt pathway has been phatase (PTEN). Downstream of PTEN is protein kinase B (Akt), which is
shown to be very important for the biological functions and mainte­ a key regulator of Wnt and PI3K signaling pathways and plays a crucial
nance of CSCs [50]. Additionally, activated Wnt/β-Catenin signaling role in energy homeostasis [74]. PTEN regulates PI3K signaling by
plays a key role in epithelial-mesenchymal transition (EMT), in which dephosphorylating PIP3, a product of PI3K. Mutations in PTEN are
cellular plasticity induces the generation of CSCs [52]. Specifically, in usually found in human breast cancers, and these mutations usually lead
BCSCs, aberrant activation of the Wnt signaling pathway has been re­ to accumulation of PIP3, subsequently activating a series of signaling
ported [53,54] and is related to dysregulation of noncoding RNAs cascades including AKT and other kinases [75]. Activated PTEN down­
[55–57]. Treatment with inhibitors of Wnt signaling has demonstrated regulates p27, leading to cell cycle progression and downregulation of
inhibition of BCSC growth in vitro and in a xenograft model [58]. proapoptotic factors such as caspase-9 and BAD. Loss of PTEN was found
Encouragingly, inhibitors targeting BCSCs are entering phase I clinical in 40 % of human breast cancers, and dysregulation of PI3K/AKT
trials [50]. signaling was found in up to 70 % of cancers [76,77]. PTEN has previ­
ously been reported to regulate the self-renewal of neuronal stem cells
5.3. Hh signaling and hematopoietic stem cells [78]. Later, evidence emerged indicating
that repressing PTEN contributes to BCSC self-renewal and survival via
The Hh signaling pathway contributes widely to the regulation of cell the AKT signaling pathway [79]. These findings support the develop­
proliferation, migration, and differentiation in space-, time- and ment of inhibitors of PTEN/AKT signaling for targeting BCSCs and breast
concentration-dependent manners [59–61]. Three lipid-modified cancer. Korkaya et al. reported that perifosine, an AKT inhibitor, inhibits
secreted ligands—Sonic Hedgehog (SHH), Indian Hedgehog (IHH), the formation of mammospheres in vitro and eradicates breast tumor
and Desert Hedgehog (DHH)—transduce signals through Patched xenografts [80]. However, a phase II clinical trial evaluating targeting of
(Ptch1), a 12-pass transmembrane spanning receptor, to the trans­ the AKT protein in advanced breast cancer patients with dysregulation
membrane protein Smoothened (SMO). Activated SMO activates a series of PTEN/AKT/PI3K has been completed, but the effect of this mono­
of downstream intracellular proteins and eventually results in upregu­ therapy was not significant [81].
lation of Hh target genes. Studies have indicated that Hh signaling helps
to regulate CSCs in many cancers, including breast cancer. Hh signaling 6. DNA damage response in BCSCs and therapeutic strategies
can act on the same tumor through various signaling pathways that
mediate interactions between tumor cell populations and their micro­ Cells must repair various genotoxic injuries, such as single and strand
environment [62]. The expression of Ptch1 and the downstream proteins breaks, intrastrand and interstrand crosslinks with bulky adducts, base
Gli1 and Gli2 is upregulated in normal breast stem cells and BCSCs but is mismatches, insertions and deletions and base alkylation, due to
downregulated during cell differentiation [63]. Hh signaling proteins endogenous and exogenous causes via multiple DNA damage responses
are potential therapeutic targets in many types of CSCs, including (DDRs). Normal stem cells have a strong DDR to prevent the accumu­
BCSCs. Inhibitors of these signaling proteins have been reported to lation of genetic damage and prevent mutations from passing to

5
X. Zeng et al. Pharmacological Research 163 (2021) 105320

daughter cells, which is particularly harmful to the whole organism CSC biological activities, such as self-renewal. An anti-CD44 antibody
[82]. CSCs originate from normal stem cells (as mentioned above, the was found to significantly reduce the growth of human breast tumors in
CSCs are more likely to originate from normal stem cells than from a xenograft model [104]. Integrins, a family of membrane receptors,
differentiated cancer cells) and have a strong DDR, which is stronger mediate cell-extracellular matrix and cell-cell interactions in BCSCs
than that in normal breast cancer cells [83,84]. Accumulating research [105]. In a mouse model, the number of BCSCs was reduced by ablation
demonstrates that CSCs and cancer cells resist chemo-/radiotherapies [105] or inhibition [106] of focal adhesion kinase (FAK), a key mediator
via the DDR [85]. As a result, theoretically, DDR inhibitors can enhance of integrin signaling, suggesting that FAK is an oncogenic protein [105].
efficiency of the therapies that aim to induce DNA damage in both of Although several FAK inhibitors have been developed, none have been
cancer cells and CSCs. DDR inhibitors have been shown to inhibit DDRs used to treat breast cancer.
of breast cancer cells due to defects in DNA damage repair pathways In addition, reports indicate that antibodies targeting the IL-8 receptor
[82] and to inhibit DDRs in some CSCs such as glioblastoma [86–88] and CXCR1 and the small molecule CXCR1/CXCR2 inhibitor repertaxin can
colon cancer [89,90]. A PARP1 inhibitor combined with the target BCSCs in xenograft models, inhibiting tumor growth and metastasis
cyclin-dependent kinase inhibitor dinaciclib limited the growth of [80]. The effect of these inflammatory cytokines on BCSCs might contribute
BCSCs [91]. BCSCs in TNB were resistant to a PARP1 inhibitor mediated to these clinical effects. Recently, overexpression of Snail or Twist, two
by RAD51 [92]. In addition to PARPs, BCSCs respond to DNA damage by inducible EMT transcription factors, was found to increase the
ATM kinase activation [93,94]. In summary, new DDR inhibitors for CD44+/CD24− subpopulation in immortalized human breast epithelial cells.
BCSCs need to be screened. The cells that underwent EMT exhibited increased mammosphere-forming
capacity and tumor initiation potential in NOD/SCID mice. These results
7. The BCSC microenvironment and related therapies suggested a relationship between EMT and the CSC phenotype. Subse­
quently, specific screened small molecules targeting EMT cells, such as sali­
The microenvironment surrounding stem cells has been termed the nomycin, etoposide, abamectin and nigericin, were confirmed to have the
stem cell niche. BCSCs are regulated by extrinsic factors within their greatest effect in vitro in follow-up studies. Collectively, these findings suggest
niche. The BCSC niche contains various cells, including immune cells, that the cytokine network plays an important role in regulating CSCs in their
adipocytes, mesenchymal stem cells, endothelial cells and fibroblasts niche and that developing strategies to interfere with these loops may provide
[95]. These other cells interact with CSCs through the cytokine network a novel approach to target CSC populations.
via paracrine pathways [96]. Several cytokines secreted mainly from
immune cells, such as IL-6, IL-8, and the receptor CXCR1, have been 8. The UPS in BCSCs and UPS-targeted therapies
demonstrated to regulate BCSC activity in vitro and in xenograft models
[97,98]. CXCR1 expression is higher in ALDH+ cells than in ALDH– cells The UPS plays an important role in cell proliferation, transcription,
of various breast cancer cell lines, and IL-8 increases the number of CSCs the immune response, DNA damage repair, apoptosis, neoplastic growth
[99]. Furthermore, reports indicate that chemotherapy-induced cyto­ and metastasis. It is also crucial for the regulation of cell cycle protein
toxicity may increase the local IL-8 level. The most likely outcome of this degradation [107]. Ubiquitination modifies target proteins through a
increase may be an increase in the proportion of CSCs. The levels of IL-6 series of enzymes, including ubiquitin-activating (E1) enzymes,
and IL-8 in the serum of patients have been correlated with the devel­ ubiquitin-conjugating (E2) enzymes and ubiquitin ligase (E3) enzymes
opment of metastasis and low therapeutic efficacy in advanced breast [108,109]. E3s, which function as key enzymes in this process by
cancers [97,100]. In summary, cytokines and their receptors play recruiting specific target proteins, are classified as RING finger, HECT,
important roles in the regulation of CSC biological characteristics by Ubox, PHD finger and RBR E3s based on their structures [110] (Fig. 4).
changing the cell niche, and inhibitor treatments have been designed to A zinc finger transcription factor, kruppel-like factor 4 (KLF4, GKLF),
block inflammatory cytokines and/or their receptors in BCSCs. In regulates cellular responses to various cellular stresses and signals,
addition, the complex BCSC niche could be targeted for treatment. including DNA damage, oxidative stress, an inflammatory environment
and stem cell renewal-related signals [111]. KLF4 is a fast-turnover
7.1. Transforming growth factor-β (TGF-β) protein, and its turnover half-life is governed by the UPS [112,113].
In breast cancer, the oncogenic effect of KLF4 has been extensively re­
The TGF-β signaling pathway plays an essential role in the regulation ported [113–117]. Notably, other reports have shown the tumor sup­
of stem cell activity. On the one hand, TGF-β induces epithelial cells to pressor function of KLF4 [118,119]. As KLF4 is essential for induced
undergo EMT, leading to the expression of specific proteins in stem cells pluripotent stem (iPS) cell generation, it is believed to play a critical role
and CSCs, which was found to lead in turn to increases in mammosphere in mammary stem cell and BCSC maintenance and differentiation. For
formation in vitro and tumorigenicity in a mouse model [34]. TGF-β example, Ai et al. reported that KLF4 is essential for the maintenance,
signaling is activated in CD44+/CD24–/low BCSCs, leading to upregula­ migration and invasion of BCSCs [120]. Estrogen signals can lead to
tion of vimentin, CTGF, SERPINE1, SPARC and TGFBR2 expression in KLF4 protein accumulation by inhibiting its CUL2/VHL-mediated pro­
these cells [101]. Notably, inhibition of TGF-β can suppress the occur­ tein degradation to further trigger estrogen-induced signaling [121].
rence of breast cancer by affecting CSCs or early progenitor cells in Furthermore, KLF4 is a key node for crosstalk between TGF-β signaling
xenotransplantation models [102]. In addition, the crosstalk between and ER signaling. These results further reveal KLF4 as a critical factor
ER signaling activity and TGF-β signaling has been reported in the that controls normal and CSC maintenance and differentiation and plays
regulation of breast stem cell maintenance as well as cell proliferation an essential role in breast cancer tumor occurrence and progression,
and epithelial cell apoptosis. The anti-TGF-β/PD-1 bifunctional fusion suggesting that KLF4 may serve as a biomarker or therapeutic target for
protein M7824 is in a phase II clinical trial for the treatment of small cell BCSCs. F-box and WD repeat domain-containing 7 (FBW7) is an
lung cancer. However, to date, no phase II-IV clinical trials have been important substrate adaptor for ubiquitin E3 ligases, which act mainly
conducted and no drugs targeting TGF-β in breast cancer have been on many important oncogenic proteins, including Notch, c-Myc, KLF5,
approved. Mcl1, mTOR, presenilin, cyclin E and c-Jun, for ubiquitin-mediated
Currently, therapies specifically targeting the CSC niche are being proteolysis. In addition, FBW7 was confirmed to act as a tumor sup­
developed. As a receptor for hyaluronic acid and other extracellular pressor in a gene knockout mouse model. In various stem cells, such as
matrix proteins, as well as a cofactor for growth factors and cytokines, hematopoietic stem cells, hepatic stem cells and neural stem cells, FBW7
CD44 regulates cell migration, proliferation, differentiation and survival also regulates self-renewal, differentiation and maintenance. Therefore,
by transducing signals from the extracellular matrix to the intracellular FBW7 could be targeted in human diseases and for BCSC-targeted
environment [103]. In BCSCs, CD44 is a marker and regulates various therapy [122].

6
X. Zeng et al. Pharmacological Research 163 (2021) 105320

Fig. 4. Pathway of UPS-mediated degradation in CSCs. Proteins are targeted for degradation by the UPS through enzymatic cascades.

Accumulating findings have shown that inhibition of the proteasome of EMT [130]. Additionally, lncRNA-Hh enhances EMT via the Hh
is another potential therapeutic approach for cancer. The proteasome signaling pathway [131]. In addition, 16 other lncRNAs have been found
inhibitor PS-341 is an example of a novel class of potent and effective to be involved in the formation of BCSCs and/or maintenance of these
antitumor agents [107]. Chemotherapeutic agents, such as ifosfamide cells [132].
and cisplatin, downregulate ubiquitin–proteasome-mediated proteolysis Accumulating evidence indicates that circular RNAs (circRNAs),
by inhibiting the expression of proteasomal subunits. Given its ability to characterized by their covalently closed circular structure containing
control stem cell differentiation, regulation of the UPS by novel ap­ approximately 1000 nucleotides, play oncogenic roles in breast cancer
proaches is likely to significantly impact the design of new development. CircRNAs have been reported to be associated with EMT
disease-specific therapies for cancer, including therapies targeting in breast cancer [133–137]. Moreover, novel EMT-related circRNAs, for
BCSCs. example, SCYL2 [138] and ANKS1B [139], have been identified in
breast cancer tissue. Recently, a profile of a set of circRNAs in BCSCs was
9. Noncoding RNAs and BCSCs defined; a total of 19 differentially expressed circRNAs were identified,
among which VRK1 was identified as a suppressor of BCSCs [140].
MiRNAs are small noncoding regulatory RNAs that control mRNA Two therapeutic methods—viral vector or nanoparticle (liposome,
translation and posttranslational modification of the encoded proteins polyethylenimine, or dendrimer) delivery systems and antagonists to
via inhibition of ribosomal function, dealkylation of the poly (A) tail, eliminate oncogenic miRNAs or miRNA mimics to restore tumor sup­
uncapping of the 5′ -cap structure and degradation of the target mRNA. pressor miRNAs—have been demonstrated to prevent tumor occurrence
Many miRNAs play essential roles in a wide range of tissues and tumors, and progression in vitro and in mouse tumor models [141]. In BCSCs,
while other miRNAs may have functions only in specific tissues, specific miRNA-34a mimics were reported to be able to eliminate BCSCs directly
tumors or specific tumor progression processes [123]. The miRNA 200 by inhibiting the accumulation of C22ORF28 [142]. In addition,
family (miR-200a, 200b, 200c, 141, 182, 183, and 96), the let-7 family miRNA-200c mimics improved the targeting effect of paclitaxel to
(let-7a, b, c, d, e, f, g, and i) and miR-30e suppress the self-renewal of BCSCs in vitro [143]. Based on these findings, the development of
BCSCs and suppress their chemoresistance and differentiation BCSC-targeted noncoding RNA therapy combined with other therapies is
[124–126]. Furthermore, the miR-200 family suppresses the expression a future direction for breast cancer eradication.
of ZEB1 and ZEB2, resulting in inhibition of EMT [127]. However,
miR-181a, miRNA181 and miR-155 were found to enhance cell 10. Differentiation therapy in BCSCs
self-renewal, sphere formation, or tumor progression in breast cancer. In
addition to the tumor suppressor and oncogenic miRNAs discusses Cell cycle- or DNA damage-dependent chemotherapeutic agents are
above, at least 19 other miRNAs or miRNA families, such as miRNA 600 toxic to rapidly dividing cells, but long-lived and relatively quiescent
and 760, have been identified to be tumorigenesis-related factors in CSCs are more resistant to these cytotoxicities. Therefore, via a method
BCSCs [128]. Recently, a study reported that miRNA-140 inhibits the inducing cell differentiation, CSCs can transform to an active state to
proliferation of BCSCs by downregulating the expression of Wnt (Fig. 5) divide, differentiate and lose their self-renewal potential, an effect that
[129]. can increase their sensitivity to chemo-/radiotherapy. Differentiation
Long noncoding RNAs (lncRNAs) are a population of noncoding therapy has been successfully used to treat specific hematological can­
RNAs with a length of more than 200 nucleotides compared with the cers. However, the application of differentiation therapy specifically
20–24 nucleotides in miRNAs. LncRNAs regulate diverse cellular pro­ targeting BCSCs needs further definition. One differentiation agent, all-
cesses, as well as tumorigenesis, usually as mediators in signaling trans retinoic acid (RA), can promote the differentiation of BCSCs or
pathways, and have been demonstrated to play important roles in CSC suppress the stemness of breast cancer cells, leading to a reduction in
plasticity and EMT programs. Upregulation of the lncRNA HOTAIR in their invasiveness and metastasis and an increase in their sensitivity to
BCSCs contributes to downregulation of miRNA-7, which is a suppressor chemotherapy. A recent report indicated that biological fluids, culture

7
X. Zeng et al. Pharmacological Research 163 (2021) 105320

Fig. 5. A summary of miRNA regulators and biomarkers in the processes of normal mammary gland maintenance and breast cancer initiation, metastasis, and
therapeutic resistance. In these four processes, mammary stem cells (MaSCs), breast cancer stem cells (BCSCs), metastatic CSCs (MCSCs), and therapy-resistant CSCs
(TRCSCs) are pivotal originators and/or critical cellular players. The miRNA regulators are listed as suppressor miRs (in green), oncomiRs (in red), and their targeted
genes (in blue). The miRNA biomarkers are listed as upregulated miRNAs (in red), downregulated miRNAs (in green), and related target genes (in blue).

supernatant from replicative senescent fibroblasts, human ovarian be considered in breast cancer.
follicular fluid and supernatant from fibroblasts subjected to serum Many vaccines for solid tumors have achieved different success rates
starvation for 16 h, regulate Oct4, Sox2, Nanog, and C-myc expression, in preclinical or clinical trials, but only the HPV vaccine for cervical
resulting in increased differentiation of BCSCs [144]. carcinoma has been approved [148,149]. Tumors impair the immune
system, which contributes to cancer surveillance during breast tumor
11. Therapies targeting drug transporters progression and can suppress or eradicate tumors. Dendritic cell (DC)
vaccines are common vaccines in breast cancer treatment, and specific
Overexpression of ABC transporters is not only a property of stem antigens for BCSCs have been identified and targeted to elicit long-term
cells but also a feature of CSCs, including BCSCs [145]. ABC transporters immune responses targeting CSCs. Ideal antigens should be exclusively
display strong drug efflux capacity; thus, these transporters provide a expressed on BCSCs and breast cancer cells but not on normal stem cells or
protective mechanism against xenobiotic toxins and are partially normal cells, although no ideal antigen has been found. The second-best
responsible for the resistance of CSCs to traditional therapies [145]. antigens are the BCSC markers that have been identified, such as CD44,
Inhibition of drug transporters may constitute a novel therapeutic means ALDH, Her-2 and CD49f. A HER-2/neu DC vaccine successfully induced
of resensitizing CSCs. Pheophorbide, a chlorophyll catabolite, is a spe­ the production of anti-HER-2/neu antibodies, stimulated interferon-γ
cific probe for ABCG2 that inhibits the efflux properties of ABCG2 [146]. expression and reduced the size of a spontaneous breast tumor in a mouse
Combined treatment with ABC transporter inhibitors and chemotherapy model [150,151]. More potential onco-antigens in BSCs and breast cancer
could be used to increase the efficiency of chemotherapeutic drugs to kill cells as targets, including some cancer/testis antigens [152], TENM 4
BCSCs [147]. Therefore, effective targeting of these molecules could be [153], TLR2 and xCT, have been identified [154]. In addition to the DC
vital since they play an important role in the resistance of BCSCs to vaccine, adoptive HER-2-specific T cells have been reported to eliminate
treatment. specific breast tumor cells [155]. Determining the number of BCSCs in the
tumor is important; the extent of tumor regression depends on the escape
12. Immunotherapy targeting BCSCs of BCSCs, which itself depends on the number of BCSCs in the tumor and
the ability to eradicate these cells after treatment.
The purpose of immunotherapy is to stimulate the immune response In fact, each type of immunotherapy requires additional treatment,
or help the immune system to recognize and eliminate various patho­ such as targeting cytotoxic T lymphocyte antigen (CTLA)-4, PD-1 or PD-
genic factors, including cancer cells. Current potential immunothera­ L1, to block T cell regulation [156]. In recent years, immune checkpoint
peutic strategies targeting solid tumors, including breast cancer, are inhibitors (ICIs) have shown positive effects in breast cancer treatment
common cancer vaccines and monoclonal antibodies. Adoptive immu­ alone or combination of other therapies. Many clinical trials on immu­
notherapy has been successfully applied as chimeric antigen receptor T notherapy of the ICIs have been performed based on the data from
(CAR-T) or TCR-T cell therapy in leukemia; thus, both treatments should clinicaltrials.gov and PubMed. Combined therapy with atezolizumab, a

8
X. Zeng et al. Pharmacological Research 163 (2021) 105320

PD-L1 inhibitor, and albumin-bound paclitaxel is approved by the FDA [3] S.P. McDermott, M.S. Wicha, Targeting breast cancer stem cells, Mol. Oncol. 4 (5)
(2010) 404–419.
for the treatment of metastatic triple negative breast cancer (TNBC)
[4] M. Al-Hajj, M.F. Clarke, Self-renewal and solid tumor stem cells, Oncog 23 (43)
[153]. It has been reported that PD-L1 upregulates in BCSCs in vitro, but (2004) 7274–7282.
further studies are still needed [157]. [5] M.F. Clarke, Self-renewal and solid-tumor stem cells, Biol. Blood Marrow
Transplant. 11 (2 Suppl 2) (2005) 14–16.
[6] K. Rycaj, D.G. Tang, Cell-of-origin of cancer versus cancer stem cells: assays and
13. Perspective interpretations, Canc Res 75 (19) (2015) 4003–4011.
[7] J.E. Visvader, G.J. Lindeman, Cancer stem cells in solid tumours: accumulating
evidence and unresolved questions, Nat Rev Canc 8 (10) (2008) 755–768.
Current cancer treatment methods have shown effectiveness in [8] R.K. Nimmakayala, S.K. Batra, M.P. Ponnusamy, Unraveling the journey of cancer
removing a large number of differentiated cancer cells but fail to elimi­ stem cells from origin to metastasis, Biochim Biophys Acta Rev Canc 1871 (1)
nate CSCs that cause tumor recurrence. CSCs are resistant to the current (2019) 50–63.
[9] L.V. Nalla, K. Kalia, A. Khairnar, Self-renewal signaling pathways in breast cancer
drugs, a property constituting an important obstacle for cancer treatment.
stem cells, Int. J. Biochem. Cell Biol. 107 (2019) 140–153.
Targeting BCSCs is an extremely attractive approach for reducing breast [10] M. Al-Hajj, M.S. Wicha, A. Benito-Hernandez, S.J. Morrison, M.F. Clarke,
cancer metastasis and recurrence to markedly improve the remission rate Prospective identification of tumorigenic breast cancer cells, Proc Natl Acad Sci U
and quality of life of patients. Although many drugs and experimental S A 100 (7) (2003) 3983–3988.
[11] K. Polyak, Heterogeneity in breast cancer, J. Clin. Invest. 121 (10) (2011)
therapies have been developed for targeting CSCs, a main challenge is 3786–3788.
distinguishing CSCs from normal stem cells. Breast cancer is a very het­ [12] E.A. Perez, Breast cancer management: opportunities and barriers to an
erogeneous disease, and no universal molecular, cellular or genetic sets of individualized approach, Oncologist 16 (Suppl 1) (2011) 20–22.
[13] S.Y. Park, H.E. Lee, H. Li, M. Shipitsin, R. Gelman, K. Polyak, Heterogeneity for
markers for BCSCs have been identified. Therefore, the target on BCSCs stem cell-related markers according to tumor subtype and histologic stage in
needs further clarification to effectively target the BCSC compartment, breast cancer, Clin Canc Res 16 (3) (2010) 876–887.
thus avoiding the normal stem cell niche but destroying the CSC niche. [14] L.L. Campbell, K. Polyak, Breast tumor heterogeneity: cancer stem cells or clonal
evolution? Cell Cycle 6 (19) (2007) 2332–2338.
We must improve our understanding of both normal stem cell biology and [15] B.K. Abraham, P. Fritz, M. McClellan, P. Hauptvogel, M. Athelogou, H. Brauch,
CSC biology in order to identify such targets. In particular, a deeper un­ Prevalence of CD44+/CD24-/low cells in breast cancer may not be associated
derstanding of the control of stem cell self-renewal may lead to the with clinical outcome but may favor distant metastasis, Clin Canc Res 11 (3)
(2005) 1154–1159.
development of new therapies in the near future. In addition, approaches [16] E. Mylona, I. Giannopoulou, E. Fasomytakis, A. Nomikos, C. Magkou,
for targeting BCSCs that would be fully optimal for combination therapies P. Bakarakos, L. Nakopoulou, The clinicopathologic and prognostic significance
with existing treatments need further development. Moreover, mini­ of CD44+/CD24(-/low) and CD44-/CD24+ tumor cells in invasive breast
carcinomas, Hum. Pathol. 39 (7) (2008) 1096–1102.
mizing or avoiding long-term and/or side effects should remain the ulti­
[17] H.J. Kim, M.J. Kim, S.H. Ahn, B.H. Son, S.B. Kim, J.H. Ahn, W.C. Noh, G. Gong,
mate goal of targeting BCSCs. Different prognostic significance of CD24 and CD44 expression in breast cancer
according to hormone receptor status, Breast 20 (1) (2011) 78–85.
[18] C. Sheridan, H. Kishimoto, R.K. Fuchs, S. Mehrotra, P. Bhat-Nakshatri, C.
Authors’ contributions H. Turner, R. Goulet Jr, S. Badve, H. Nakshatri, CD44+/CD24- breast cancer cells
exhibit enhanced invasive properties: an early step necessary for metastasis,
Xiaobin Zeng and Chenxiao Liu had performed and updated the Breast Canc Res 8 (5) (2006) R59.
[19] S. Palomeras, S. Ruiz-Martinez, T. Puig, Targeting breast cancer stem cells to
literature search, and drafted for the article. Nianhong Chen had the
overcome treatment resistance, Mol 23 (9) (2018) 2193.
idea, performed the literature search, data analysis, and drafted for the [20] E. Jonasson, S. Ghannoum, E. Persson, J. Karlsson, T. Kroneis, E. Larsson,
article. Nianhong Chen and Yingpeng Li contributed to interpretation of G. Landberg, A. Stahlberg, Identification of breast cancer stem cell related genes
using functional cellular assays combined with single-cell RNA sequencing in
results and made critical revisions. Jie Yao, Haoqiang Wan and Guoqing
MDA-MB-231 cells, Front. Genet. 10 (2019) 500.
Wan gave some important suggestions, also made critical revisions. All [21] H.G. Russnes, N. Navin, J. Hicks, A.L. Borresen-Dale, Insight into the
authors have reviewed the final version of the manuscript and approved heterogeneity of breast cancer through next-generation sequencing, J. Clin.
it for publication. Invest. 121 (10) (2011) 3810–3818.
[22] J.E. Visvader, Cells of origin in cancer, Nat 469 (7330) (2011) 314–322.
[23] M.J. Higgins, J. Baselga, Targeted therapies for breast cancer, J. Clin. Invest. 121
Funding (10) (2011) 3797–3803.
[24] A. Marusyk, K. Polyak, Tumor heterogeneity: causes and consequences, Biochim.
Biophys. Acta 1805 (1) (2010) 105–117.
This work was supported by grants from the Natural Science Foun­ [25] S.M. Pontier, W.J. Muller, Integrins in mammary-stem-cell biology and breast-
dation of China (81503221, 81703939), the Science and Technology cancer progression–a role in cancer stem cells? J. Cell. Sci. 122 (Pt 2) (2009)
207–214.
Planning Project of Shenzhen Municipality (JCYJ20170307095556333,
[26] K. Kai, Y. Arima, T. Kamiya, H. Saya, Breast cancer stem cells, Breast Canc 17 (2)
JCYJ20170413093108233, JCYJ20170307100726777), and the Natu­ (2010) 80–85.
ral Science Foundation of Guangdong Province (2017A030313659, [27] T. Reya, S.J. Morrison, M.F. Clarke, I.L. Weissman, Stem cells, cancer, and cancer
2014A030310365). stem cells, Nat 414 (6859) (2001) 105–111.
[28] C. Odoux, H. Fohrer, T. Hoppo, L. Guzik, D.B. Stolz, D.W. Lewis, S.M. Gollin, T.
C. Gamblin, D.A. Geller, E. Lagasse, A stochastic model for cancer stem cell origin
in metastatic colon cancer, Canc Res 68 (17) (2008) 6932–6941.
Declaration of Competing Interest [29] L.A. Loeb, C.F. Springgate, N. Battula, Errors in DNA replication as a basis of
malignant changes, Canc Res 34 (9) (1974) 2311–2321.
The authors reported no declarations of interest. [30] I. Baccelli, A. Trumpp, The evolving concept of cancer and metastasis stem cells,
J. Cell Biol. 198 (3) (2012) 281–293.
[31] S. Liu, Y. Cong, D. Wang, Y. Sun, L. Deng, Y. Liu, R. Martin-Trevino, L. Shang, S.
Acknowledgements P. McDermott, M.D. Landis, S. Hong, A. Adams, R. D’Angelo, C. Ginestier,
E. Charafe-Jauffret, S.G. Clouthier, D. Birnbaum, S.T. Wong, M. Zhan, J.C. Chang,
M.S. Wicha, Breast cancer stem cells transition between epithelial and
We thank Mr. Joe X. Qiao for his efforts in figure draft. We apologize mesenchymal states reflective of their normal counterparts, Stem Cell Rep 2 (1)
to colleagues in the field whose work was not included due to space (2014) 78–91.
[32] T.M. Savarese, H.P. Low, I. Baik, W.C. Strohsnitter, C.C. Hsieh, Normal breast
limitation.
stem cells, malignant breast stem cells, and the perinatal origin of breast cancer,
Stem Cell Rev. 2 (2) (2006) 103–110.
References [33] H. Yin, J. Glass, The phenotypic radiation resistance of CD44+/CD24(-or low)
breast cancer cells is mediated through the enhanced activation of ATM signaling,
PLoS One 6 (9) (2011), e24080.
[1] J. Birnbaum, V.K. Gadi, E. Markowitz, R. Etzioni, The effect of treatment
[34] B. Sousa, A.S. Ribeiro, J. Paredes, Heterogeneity and plasticity of breast cancer
advances on the mortality results of breast cancer screening trials: a
stem cells, Adv. Exp. Med. Biol. 1139 (2019) 83–103.
microsimulation model, Ann. Intern. Med. 164 (4) (2016) 236–243.
[35] J.A. Brown, Y. Yonekubo, N. Hanson, A. Sastre-Perona, A. Basin, J.A. Rytlewski,
[2] D. Nassar, C. Blanpain, Cancer stem cells: basic concepts and therapeutic
I. Dolgalev, S. Meehan, A. Tsirigos, S. Beronja, M. Schober, TGF-beta-Induced
implications, Annu. Rev. Pathol. Mech. Dis. 11 (2016) 47–76.

9
X. Zeng et al. Pharmacological Research 163 (2021) 105320

quiescence mediates chemoresistance of tumor-propagating cells in squamous cell [60] J.J. Lee, D.P. von Kessler, S. Parks, P.A. Beachy, Secretion and localized
carcinoma, Cell Stem Cell 21 (5) (2017) 650–664, e8. transcription suggest a role in positional signaling for products of the
[36] D.A. Berry, K.A. Cronin, S.K. Plevritis, D.G. Fryback, L. Clarke, M. Zelen, J. segmentation gene hedgehog, Cell 71 (1) (1992) 33–50.
S. Mandelblatt, A.Y. Yakovlev, J.D. Habbema, E.J. Feuer, I. Cancer, [61] H. Hahn, C. Wicking, P.G. Zaphiropoulous, M.R. Gailani, S. Shanley,
C. Surveillance Modeling Network, Effect of screening and adjuvant therapy on A. Chidambaram, I. Vorechovsky, E. Holmberg, A.B. Unden, S. Gillies, K. Negus,
mortality from breast cancer, N. Engl. J. Med. 353 (17) (2005) 1784–1792. I. Smyth, C. Pressman, D.J. Leffell, B. Gerrard, A.M. Goldstein, M. Dean,
[37] B.J. Morrison, C.W. Schmidt, S.R. Lakhani, B.A. Reynolds, J.A. Lopez, Breast R. Toftgard, G. Chenevix-Trench, B. Wainwright, A.E. Bale, Mutations of the
cancer stem cells: implications for therapy of breast cancer, Breast Canc Res 10 human homolog of Drosophila patched in the nevoid basal cell carcinoma
(4) (2008) 210. syndrome, Cell 85 (6) (1996) 841–851.
[38] J. Dittmer, Breast cancer stem cells: features, key drivers and treatment options, [62] C.D. Peacock, Q. Wang, G.S. Gesell, I.M. Corcoran-Schwartz, E. Jones, J. Kim, W.
Semin Canc Biol 53 (2018) 59–74. L. Devereux, J.T. Rhodes, C.A. Huff, P.A. Beachy, D.N. Watkins, W. Matsui,
[39] L. Yang, P. Shi, G. Zhao, J. Xu, W. Peng, J. Zhang, G. Zhang, X. Wang, Z. Dong, Hedgehog signaling maintains a tumor stem cell compartment in multiple
F. Chen, H. Cui, Targeting cancer stem cell pathways for cancer therapy, Signal myeloma, Proc Natl Acad Sci U S A 104 (10) (2007) 4048–4053.
Transduct. Target. Ther. 5 (1) (2020) 8. [63] S. Liu, G. Dontu, I.D. Mantle, S. Patel, N.S. Ahn, K.W. Jackson, P. Suri, M.
[40] S. Singh, C. Brocker, V. Koppaka, Y. Chen, B.C. Jackson, A. Matsumoto, D. S. Wicha, Hedgehog signaling and Bmi-1 regulate self-renewal of normal and
C. Thompson, V. Vasiliou, Aldehyde dehydrogenases in cellular responses to malignant human mammary stem cells, Canc Res 66 (12) (2006) 6063–6071.
oxidative/electrophilic stress, Free Radic. Biol. Med. 56 (2013) 89–101. [64] M. Liu, W. Zhang, W. Tang, Y. Wang, X. Zhao, X. Wang, X. Qi, J. Li,
[41] J. Xin Yu, J.P. Hodge, C. Oliva, S.T. Neftelinov, V.M. Hubbard-Lucey, J. Tang, Isocyclopamine, a novel synthetic derivative of cyclopamine, reverts doxorubicin
Trends in clinical development for PD-1/PD-L1 inhibitors, Nat. Rev. Drug Discov. resistance in MCF-7/ADR cells by increasing intracellular doxorubicin
19 (3) (2020) 163–164. accumulation and downregulating breast cancer stem-like cells, Tumour Biol. 37
[42] J.L. McKenzie, O.I. Gan, M. Doedens, J.C. Wang, J.E. Dick, Individual stem cells (2) (2016) 1919–1931.
with highly variable proliferation and self-renewal properties comprise the [65] M.A. Olayioye, Update on HER-2 as a target for cancer therapy: intracellular
human hematopoietic stem cell compartment, Nat. Immunol. 7 (11) (2006) signaling pathways of ErbB2/HER-2 and family members, Breast Canc Res 3 (6)
1225–1233. (2001) 385–389.
[43] G. Dontu, S. Liu, M.S. Wicha, Stem cells in mammary development and [66] T. Holbro, R.R. Beerli, F. Maurer, M. Koziczak, C.F. Barbas 3rd, N.E. Hynes, The
carcinogenesis: implications for prevention and treatment, Stem Cell Rev. 1 (3) ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3
(2005) 207–213. to drive breast tumor cell proliferation, Proc Natl Acad Sci U S A 100 (15) (2003)
[44] E.N. Kontomanolis, S. Kalagasidou, S. Pouliliou, X. Anthoulaki, N. Georgiou, 8933–8938.
V. Papamanolis, Z.N. Fasoulakis, The Notch pathway in breast cancer [67] S. Ithimakin, K.C. Day, F. Malik, Q. Zen, S.J. Dawsey, T.F. Bersano-Begey, A.
progression, Transfus. Apher. Sci. 2018 (2018), 2415489. A. Quraishi, K.W. Ignatoski, S. Daignault, A. Davis, C.L. Hall, N. Palanisamy, A.
[45] A. Androutsellis-Theotokis, R.R. Leker, F. Soldner, D.J. Hoeppner, R. Ravin, S. N. Heath, N. Tawakkol, T.K. Luther, S.G. Clouthier, W.A. Chadwick, M.L. Day, C.
W. Poser, M.A. Rueger, S.K. Bae, R. Kittappa, R.D. McKay, Notch signalling G. Kleer, D.G. Thomas, D.F. Hayes, H. Korkaya, M.S. Wicha, HER2 drives luminal
regulates stem cell numbers in vitro and in vivo, Nat 442 (7104) (2006) 823–826. breast cancer stem cells in the absence of HER2 amplification: implications for
[46] G. Farnie, R.B. Clarke, K. Spence, N. Pinnock, K. Brennan, N.G. Anderson, N. efficacy of adjuvant trastuzumab, Canc Res 73 (5) (2013) 1635–1646.
J. Bundred, Novel cell culture technique for primary ductal carcinoma in situ: role [68] N. Duru, M. Fan, D. Candas, C. Menaa, H.C. Liu, D. Nantajit, Y. Wen, K. Xiao,
of Notch and epidermal growth factor receptor signaling pathways, J. Natl. A. Eldridge, B.A. Chromy, S. Li, D.R. Spitz, K.S. Lam, M.S. Wicha, J.J. Li, HER2-
Cancer Inst. Monographs 99 (8) (2007) 616–627. associated radioresistance of breast cancer stem cells isolated from HER2-
[47] P.B. Gupta, T.T. Onder, G. Jiang, K. Tao, C. Kuperwasser, R.A. Weinberg, E. negative breast cancer cells, Clin Canc Res 18 (24) (2012) 6634–6647.
S. Lander, Identification of selective inhibitors of cancer stem cells by high- [69] J. Diessner, V. Bruttel, R.G. Stein, E. Horn, S.F. Hausler, J. Dietl, A. Honig,
throughput screening, Cell 138 (4) (2009) 645–659. J. Wischhusen, Targeting of preexisting and induced breast cancer stem cells with
[48] S. Pece, M. Serresi, E. Santolini, M. Capra, E. Hulleman, V. Galimberti, S. Zurrida, trastuzumab and trastuzumab emtansine (T-DM1), Cell Death Dis. 5 (2014)
P. Maisonneuve, G. Viale, P.P. Di Fiore, Loss of negative regulation by Numb over e1149.
Notch is relevant to human breast carcinogenesis, J. Cell Biol. 167 (2) (2004) [70] S. Massarweh, C.K. Osborne, C.J. Creighton, L. Qin, A. Tsimelzon, S. Huang,
215–221. H. Weiss, M. Rimawi, R. Schiff, Tamoxifen resistance in breast tumors is driven by
[49] A.F. Schott, M.D. Landis, G. Dontu, K.A. Griffith, R.M. Layman, I. Krop, L. growth factor receptor signaling with repression of classic estrogen receptor
A. Paskett, H. Wong, L.E. Dobrolecki, M.T. Lewis, A.M. Froehlich, J. Paranilam, D. genomic function, Canc Res 68 (3) (2008) 826–833.
F. Hayes, M.S. Wicha, J.C. Chang, Preclinical and clinical studies of gamma [71] G. Mariani, A. Fasolo, E. De Benedictis, L. Gianni, Trastuzumab as adjuvant
secretase inhibitors with docetaxel on human breast tumors, Clin Canc Res 19 (6) systemic therapy for HER2-positive breast cancer, Nat. Clin. Pract. Oncol. 6 (2)
(2013) 1512–1524. (2009) 93–104.
[50] M. Katoh, Canonical and non-canonical WNT signaling in cancer stem cells and [72] H. Korkaya, M.S. Wicha, HER2 and breast cancer stem cells: more than meets the
their niches: cellular heterogeneity, omics reprogramming, targeted therapy and eye, Canc Res 73 (12) (2013) 3489–3493.
tumor plasticity (Review), Int. J. Oncol. 51 (5) (2017) 1357–1369. [73] H. Korkaya, A. Paulson, F. Iovino, M.S. Wicha, HER2 regulates the mammary
[51] Y. Li, G. Bu, LRP5/6 in Wnt signaling and tumorigenesis, Future Oncol. 1 (5) stem/progenitor cell population driving tumorigenesis and invasion, Oncog 27
(2005) 673–681. (47) (2008) 6120–6130.
[52] C. Zhao, J. Blum, A. Chen, H.Y. Kwon, S.H. Jung, J.M. Cook, A. Lagoo, T. Reya, [74] M. Keniry, R. Parsons, The role of PTEN signaling perturbations in cancer and in
Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo, targeted therapy, Oncogene 27 (41) (2008) 5477–5485.
Canc Cell 12 (6) (2007) 528–541. [75] R. Hill, H. Wu, PTEN, stem cells, and cancer stem cells, J. Biol. Chem. 284 (18)
[53] M. Majumder, X. Xin, L. Liu, E. Tutunea-Fatan, M. Rodriguez-Torres, K. Vincent, (2009) 11755–11759.
L.M. Postovit, D. Hess, P.K. Lala, COX-2 induces breast cancer stem cells via EP4/ [76] N. Hinz, M. Jucker, Distinct functions of AKT isoforms in breast cancer: a
PI3K/AKT/NOTCH/WNT Axis, Stem Cells 34 (9) (2016) 2290–2305. comprehensive review, Cell Commun. Signal 17 (1) (2019) 154.
[54] G. Domenici, I. Aurrekoetxea-Rodriguez, B.M. Simoes, M. Rabano, S.Y. Lee, J. [77] A.R. Panigrahi, S.E. Pinder, S.Y. Chan, E.C. Paish, J.F. Robertson, I.O. Ellis, The
S. Millan, V. Comaills, E. Oliemuller, J.A. Lopez-Ruiz, I. Zabalza, B.A. Howard, R. role of PTEN and its signalling pathways, including AKT, in breast cancer; an
M. Kypta, M.D. Vivanco, A Sox2-Sox9 signalling axis maintains human breast assessment of relationships with other prognostic factors and with outcome,
luminal progenitor and breast cancer stem cells, Oncog 38 (17) (2019) J. Pathol. 204 (1) (2004) 93–100.
3151–3169. [78] D.J. Rossi, I.L. Weissman, Pten, tumorigenesis, and stem cell self-renewal, Cell
[55] T. Tang, C. Guo, T. Xia, R. Zhang, K. Zen, Y. Pan, L. Jin, LncCCAT1 promotes 125 (2) (2006) 229–231.
breast Cancer stem cell function through activating WNT/beta-catenin signaling, [79] A. Baker, D. Wyatt, M. Bocchetta, J. Li, A. Filipovic, A. Green, D.S. Peiffer,
Theranostics 9 (24) (2019) 7384–7402. S. Fuqua, L. Miele, K.S. Albain, C. Osipo, Notch-1-PTEN-ERK1/2 signaling axis
[56] X. Sun, C. Xu, S.C. Tang, J. Wang, H. Wang, P. Wang, N. Du, S. Qin, G. Li, S. Xu, promotes HER2+ breast cancer cell proliferation and stem cell survival, Oncog 37
Z. Tao, D. Liu, H. Ren, Let-7c blocks estrogen-activated Wnt signaling in induction (33) (2018) 4489–4504.
of self-renewal of breast cancer stem cells, Canc Gene Ther 23 (4) (2016) 83–89. [80] H. Korkaya, A. Paulson, E. Charafe-Jauffret, C. Ginestier, M. Brown, J. Dutcher, S.
[57] R. El Helou, G. Pinna, O. Cabaud, J. Wicinski, R. Bhajun, L. Guyon, C. Rioualen, G. Clouthier, M.S. Wicha, Regulation of mammary stem/progenitor cells by
P. Finetti, A. Gros, B. Mari, P. Barbry, F. Bertucci, G. Bidaut, A. Harel-Bellan, PTEN/Akt/beta-catenin signaling, PLoS Biol. 7 (6) (2009), e1000121.
D. Birnbaum, E. Charafe-Jauffret, C. Ginestier, miR-600 acts as a bimodal switch [81] Y. Xing, N.U. Lin, M.A. Maurer, H. Chen, A. Mahvash, A. Sahin, A. Akcakanat,
that regulates breast cancer stem cell fate through WNT signaling, Cell Rep. 18 (9) Y. Li, V. Abramson, J. Litton, M. Chavez-MacGregor, V. Valero, S.A. Piha-Paul,
(2017) 2256–2268. D. Hong, K.A. Do, E. Tarco, D. Riall, A.K. Eterovic, G.M. Wulf, L.C. Cantley, G.
[58] A. Gurney, F. Axelrod, C.J. Bond, J. Cain, C. Chartier, L. Donigan, M. Fischer, B. Mills, L.A. Doyle, E. Winer, G.N. Hortobagyi, A.M. Gonzalez-Angulo, F. Meric-
A. Chaudhari, M. Ji, A.M. Kapoun, A. Lam, S. Lazetic, S. Ma, S. Mitra, I.K. Park, Bernstam, Phase II trial of AKT inhibitor MK-2206 in patients with advanced
K. Pickell, A. Sato, S. Satyal, M. Stroud, H. Tran, W.C. Yen, J. Lewicki, T. Hoey, breast cancer who have tumors with PIK3CA or AKT mutations, and/or PTEN
Wnt pathway inhibition via the targeting of Frizzled receptors results in loss/PTEN mutation, Breast Canc Res 21 (1) (2019) 78.
decreased growth and tumorigenicity of human tumors, Proc Natl Acad Sci U S A [82] R. Ali, E.A. Rakha, S. Madhusudan, H.E. Bryant, DNA damage repair in breast
109 (29) (2012) 11717–11722. cancer and its therapeutic implications, Pathology 49 (2) (2017) 156–165.
[59] C. Nusslein-Volhard, E. Wieschaus, Mutations affecting segment number and [83] F. Karimi-Busheri, A. Rasouli-Nia, J.R. Mackey, M. Weinfeld, Senescence evasion
polarity in Drosophila, Nat 287 (5785) (1980) 795–801. by MCF-7 human breast tumor-initiating cells, Breast Canc Res 12 (3) (2010) R31.
[84] M. Diehn, R.W. Cho, N.A. Lobo, T. Kalisky, M.J. Dorie, A.N. Kulp, D. Qian, J.
S. Lam, L.E. Ailles, M. Wong, B. Joshua, M.J. Kaplan, I. Wapnir, F.M. Dirbas,

10
X. Zeng et al. Pharmacological Research 163 (2021) 105320

G. Somlo, C. Garberoglio, B. Paz, J. Shen, S.K. Lau, S.R. Quake, J.M. Brown, I. [107] J. Adams, V.J. Palombella, E.A. Sausville, J. Johnson, A. Destree, D.D. Lazarus,
L. Weissman, M.F. Clarke, Association of reactive oxygen species levels and J. Maas, C.S. Pien, S. Prakash, P.J. Elliott, Proteasome inhibitors: a novel class of
radioresistance in cancer stem cells, Nat 458 (7239) (2009) 780–783. potent and effective antitumor agents, Canc Res 59 (11) (1999) 2615–2622.
[85] E. Abad, L. Civit, D. Potesil, Z. Zdrahal, A. Lyakhovich, Enhanced DNA damage [108] C.M. Pickart, Mechanisms underlying ubiquitination, Annu. Rev. Biochem. 70
response through RAD50 in triple negative breast cancer resistant and cancer (2001) 503–533.
stem-like cells contributes to chemoresistance, FEBS J. (2020). [109] C. Rathinam, L.E. Matesic, R.A. Flavell, The E3 ligase Itch is a negative regulator
[86] M. Venere, P. Hamerlik, Q. Wu, R.D. Rasmussen, L.A. Song, A. Vasanji, N. Tenley, of the homeostasis and function of hematopoietic stem cells, Nat. Immunol. 12 (5)
W.A. Flavahan, A.B. Hjelmeland, J. Bartek, J.N. Rich, Therapeutic targeting of (2011) 399–407.
constitutive PARP activation compromises stem cell phenotype and survival of [110] D. Rotin, S. Kumar, Physiological functions of the HECT family of ubiquitin
glioblastoma-initiating cells, Cell Death Differ. 21 (2) (2014) 258–269. ligases, Nat. Rev. Mol. Cell Biol. 10 (6) (2009) 398–409.
[87] S.U. Ahmed, R. Carruthers, L. Gilmour, S. Yildirim, C. Watts, A.J. Chalmers, [111] A.M. Gamper, X. Qiao, J. Kim, L. Zhang, M.C. DeSimone, W.K. Rathmell, Y. Wan,
Selective inhibition of parallel DNA damage response pathways optimizes Regulation of KLF4 turnover reveals an unexpected tissue-specific role of pVHL in
radiosensitization of glioblastoma stem-like cells, Canc Res 75 (20) (2015) tumorigenesis, Mol. Cell 45 (2) (2012) 233–243.
4416–4428. [112] Z.Y. Chen, X. Wang, Y. Zhou, G. Offner, C.C. Tseng, Destabilization of Kruppel-
[88] R.D. Carruthers, S.U. Ahmed, S. Ramachandran, K. Strathdee, K.M. Kurian, like factor 4 protein in response to serum stimulation involves the ubiquitin-
A. Hedley, N. Gomez-Roman, G. Kalna, M. Neilson, L. Gilmour, K.H. Stevenson, E. proteasome pathway, Canc Res 65 (22) (2005) 10394–10400.
M. Hammond, A.J. Chalmers, Replication stress drives constitutive activation of [113] M.O. Kim, S.H. Kim, Y.Y. Cho, J. Nadas, C.H. Jeong, K. Yao, D.J. Kim, D.H. Yu, Y.
the DNA damage response and radioresistance in glioblastoma stem-like cells, S. Keum, K.Y. Lee, Z. Huang, A.M. Bode, Z. Dong, ERK1 and ERK2 regulate
Canc Res 78 (17) (2018) 5060–5071. embryonic stem cell self-renewal through phosphorylation of Klf4, Nat. Struct.
[89] A. Jarrar, F. Lotti, J. DeVecchio, S. Ferrandon, G. Gantt, A. Mace, G. Karagkounis, Mol. Biol. 19 (3) (2012) 283–290.
M. Orloff, M. Venere, M. Hitomi, J. Lathia, J.N. Rich, M.F. Kalady, Poly(ADP- [114] K.W. Foster, A.R. Frost, P. McKie-Bell, C.Y. Lin, J.A. Engler, W.E. Grizzle, J.
Ribose) polymerase inhibition sensitizes colorectal cancer-initiating cells to M. Ruppert, Increase of GKLF messenger RNA and protein expression during
chemotherapy, Stem Cells 37 (1) (2019) 42–53. progression of breast cancer, Canc Res 60 (22) (2000) 6488–6495.
[90] A. Lucena-Cacace, D. Otero-Albiol, M.P. Jimenez-Garcia, S. Munoz-Galvan, [115] A.Y. Pandya, L.I. Talley, A.R. Frost, T.J. Fitzgerald, V. Trivedi, M. Chakravarthy,
A. Carnero, NAMPT is a potent oncogene in colon cancer progression that D.C. Chhieng, W.E. Grizzle, J.A. Engler, H. Krontiras, K.I. Bland, A.F. LoBuglio, S.
modulates cancer stem cell properties and resistance to therapy through Sirt1 and M. Lobo-Ruppert, J.M. Ruppert, Nuclear localization of KLF4 is associated with an
PARP, Clin Canc Res 24 (5) (2018) 1202–1215. aggressive phenotype in early-stage breast cancer, Clin Canc Res 10 (8) (2004)
[91] J.P.W. Carey, C. Karakas, T. Bui, X. Chen, S. Vijayaraghavan, Y. Zhao, J. Wang, 2709–2719.
K. Mikule, J.K. Litton, K.K. Hunt, K. Keyomarsi, Synthetic lethality of PARP [116] B.D. Rowland, R. Bernards, D.S. Peeper, The KLF4 tumour suppressor is a
inhibitors in combination with MYC blockade is independent of BRCA status in transcriptional repressor of p53 that acts as a context-dependent oncogene, Nat.
triple-negative breast Cancer, Canc Res 78 (3) (2018) 742–757. Cell Biol. 7 (11) (2005) 1074–1082.
[92] Y. Liu, M.L. Burness, R. Martin-Trevino, J. Guy, S. Bai, R. Harouaka, M.D. Brooks, [117] H. Okuda, F. Xing, P.R. Pandey, S. Sharma, M. Watabe, S.K. Pai, Y.Y. Mo,
L. Shang, A. Fox, T.K. Luther, A. Davis, T.L. Baker, J. Colacino, S.G. Clouthier, Z. M. Iiizumi-Gairani, S. Hirota, Y. Liu, K. Wu, R. Pochampally, K. Watabe, miR-7
M. Shao, M.S. Wicha, S. Liu, RAD51 mediates resistance of Cancer stem cells to suppresses brain metastasis of breast cancer stem-like cells by modulating KLF4,
PARP inhibition in triple-negative breast Cancer, Clin Canc Res 23 (2) (2017) Canc Res 73 (4) (2013) 1434–1444.
514–522. [118] J.L. Yori, D.D. Seachrist, E. Johnson, K.L. Lozada, F.W. Abdul-Karim, L.
[93] D. Yang, M. Peng, Y. Hou, Y. Qin, X. Wan, P. Zhu, S. Liu, L. Yang, H. Zeng, T. Jin, A. Chodosh, W.P. Schiemann, R.A. Keri, Kruppel-like factor 4 inhibits tumorigenic
Y. Qiu, Q. Li, M. Liu, Oxidized ATM promotes breast cancer stem cell enrichment progression and metastasis in a mouse model of breast cancer, Neoplasia 13 (7)
through energy metabolism reprogram-mediated acetyl-CoA accumulation, Cell (2011) 601–610.
Death Dis. 11 (7) (2020) 508. [119] T. Nagata, Y. Shimada, S. Sekine, R. Hori, K. Matsui, T. Okumura, S. Sawada,
[94] S.Y. Kim, J.G. Rhee, X. Song, E.V. Prochownik, D.R. Spitz, Y.J. Lee, Breast cancer J. Fukuoka, K. Tsukada, Prognostic significance of NANOG and KLF4 for breast
stem cell-like cells are more sensitive to ionizing radiation than non-stem cells: cancer, Breast Canc 21 (1) (2014) 96–101.
role of ATM, PLoS One 7 (11) (2012), e50423. [120] F. Yu, J. Li, H. Chen, J. Fu, S. Ray, S. Huang, H. Zheng, W. Ai, Kruppel-like factor
[95] H. Korkaya, S. Liu, M.S. Wicha, Breast cancer stem cells, cytokine networks, and 4 (KLF4) is required for maintenance of breast cancer stem cells and for cell
the tumor microenvironment, J. Clin. Invest. 121 (10) (2011) 3804–3809. migration and invasion, Oncog 30 (18) (2011) 2161–2172.
[96] A. Orimo, P.B. Gupta, D.C. Sgroi, F. Arenzana-Seisdedos, T. Delaunay, R. Naeem, [121] D. Hu, Y. Wan, Regulation of Kruppel-like factor 4 by the anaphase promoting
V.J. Carey, A.L. Richardson, R.A. Weinberg, Stromal fibroblasts present in complex pathway is involved in TGF-beta signaling, J. Biol. Chem. 286 (9) (2011)
invasive human breast carcinomas promote tumor growth and angiogenesis 6890–6901.
through elevated SDF-1/CXCL12 secretion, Cell 121 (3) (2005) 335–348. [122] Z. Wang, H. Inuzuka, H. Fukushima, L. Wan, D. Gao, S. Shaik, F.H. Sarkar,
[97] P. Sansone, G. Storci, S. Tavolari, T. Guarnieri, C. Giovannini, M. Taffurelli, W. Wei, Emerging roles of the FBW7 tumour suppressor in stem cell
C. Ceccarelli, D. Santini, P. Paterini, K.B. Marcu, P. Chieco, M. Bonafe, IL-6 differentiation, EMBO Rep. 13 (1) (2011) 36–43.
triggers malignant features in mammospheres from human ductal breast [123] H. Liu, MicroRNAs in breast cancer initiation and progression, Cell. Mol. Life Sci.
carcinoma and normal mammary gland, J. Clin. Invest. 117 (12) (2007) 69 (21) (2012) 3587–3599.
3988–4002. [124] F. Yu, H. Yao, P. Zhu, X. Zhang, Q. Pan, C. Gong, Y. Huang, X. Hu, F. Su,
[98] K. Inoue, J.W. Slaton, B.Y. Eve, S.J. Kim, P. Perrotte, M.D. Balbay, S. Yano, J. Lieberman, E. Song, let-7 regulates self renewal and tumorigenicity of breast
M. Bar-Eli, R. Radinsky, C.A. Pettaway, C.P. Dinney, Interleukin 8 expression cancer cells, Cell 131 (6) (2007) 1109–1123.
regulates tumorigenicity and metastases in androgen-independent prostate [125] D. Iliopoulos, H.A. Hirsch, K. Struhl, An epigenetic switch involving NF-kappaB,
cancer, Clin Canc Res 6 (5) (2000) 2104–2119. Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation, Cell
[99] E. Charafe-Jauffret, C. Ginestier, F. Iovino, J. Wicinski, N. Cervera, P. Finetti, M. 139 (4) (2009) 693–706.
H. Hur, M.E. Diebel, F. Monville, J. Dutcher, M. Brown, P. Viens, L. Xerri, [126] F. Yu, H. Deng, H. Yao, Q. Liu, F. Su, E. Song, Mir-30 reduction maintains self-
F. Bertucci, G. Stassi, G. Dontu, D. Birnbaum, M.S. Wicha, Breast cancer cell lines renewal and inhibits apoptosis in breast tumor-initiating cells, Oncog 29 (29)
contain functional cancer stem cells with metastatic capacity and a distinct (2010) 4194–4204.
molecular signature, Canc Res 69 (4) (2009) 1302–1313. [127] P.A. Gregory, A.G. Bert, E.L. Paterson, S.C. Barry, A. Tsykin, G. Farshid, M.
[100] A. Albini, M.B. Sporn, The tumour microenvironment as a target for A. Vadas, Y. Khew-Goodall, G.J. Goodall, The miR-200 family and miR-205
chemoprevention, Nat Rev Canc 7 (2) (2007) 139–147. regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1, Nat.
[101] M. Shipitsin, L.L. Campbell, P. Argani, S. Weremowicz, N. Bloushtain-Qimron, Cell Biol. 10 (5) (2008) 593–601.
J. Yao, T. Nikolskaya, T. Serebryiskaya, R. Beroukhim, M. Hu, M.K. Halushka, [128] F. Vahidian, H. Mohammadi, M. Ali-Hasanzadeh, A. Derakhshani, M. Mostaan,
S. Sukumar, L.M. Parker, K.S. Anderson, L.N. Harris, J.E. Garber, A.L. Richardson, M. Hemmatzadeh, B. Baradaran, MicroRNAs and breast cancer stem cells:
S.J. Schnitt, Y. Nikolsky, R.S. Gelman, K. Polyak, Molecular definition of breast potential role in breast cancer therapy, J. Cell. Physiol. 234 (4) (2019)
tumor heterogeneity, Canc Cell 11 (3) (2007) 259–273. 3294–3306.
[102] X. Zu, Q. Zhang, R. Cao, J. Liu, J. Zhong, G. Wen, D. Cao, Transforming growth [129] D. Wu, J. Zhang, Y. Lu, S. Bo, L. Li, L. Wang, Q. Zhang, J. Mao, miR-140-5p
factor-beta signaling in tumor initiation, progression and therapy in breast cancer: inhibits the proliferation and enhances the efficacy of doxorubicin to breast
an update, Cell Tissue Res. 347 (1) (2012) 73–84. cancer stem cells by targeting Wnt1, Canc Gene Ther 26 (3-4) (2019) 74–82.
[103] Y. Yan, X. Zuo, D. Wei, Concise review: emerging role of CD44 in cancer stem [130] H. Zhang, K. Cai, J. Wang, X. Wang, K. Cheng, F. Shi, L. Jiang, Y. Zhang, J. Dou,
cells: a promising biomarker and therapeutic target, Stem Cells Transl. Med. 4 (9) MiR-7, inhibited indirectly by lincRNA HOTAIR, directly inhibits SETDB1 and
(2015) 1033–1043. reverses the EMT of breast cancer stem cells by downregulating the STAT3
[104] E. Marangoni, N. Lecomte, L. Durand, G. de Pinieux, D. Decaudin, C. Chomienne, pathway, Stem Cells 32 (11) (2014) 2858–2868.
F. Smadja-Joffe, M.F. Poupon, CD44 targeting reduces tumour growth and [131] M. Zhou, Y. Hou, G. Yang, H. Zhang, G. Tu, Y.E. Du, S. Wen, L. Xu, X. Tang,
prevents post-chemotherapy relapse of human breast cancers xenografts, Br. J. S. Tang, L. Yang, X. Cui, M. Liu, LncRNA-hh strengthen Cancer stem cells
Cancer Suppl. 100 (6) (2009) 918–922. generation in twist-positive breast Cancer via activation of hedgehog signaling
[105] J.L. Guan, Integrin signaling through FAK in the regulation of mammary stem pathway, Stem Cells 34 (1) (2016) 55–66.
cells and breast cancer, IUBMB Life 62 (4) (2010) 268–276. [132] J.M. Brown, M.D. Wasson, P. Marcato, The missing lnc: the potential of targeting
[106] V.N. Kolev, W.F. Tam, Q.G. Wright, S.P. McDermott, C.M. Vidal, I.M. Shapiro, triple-negative breast Cancer and Cancer stem cells by inhibiting long non-coding
Q. Xu, M.S. Wicha, J.A. Pachter, D.T. Weaver, Inhibition of FAK kinase activity RNAs, Cells 9 (3) (2020) 763.
preferentially targets cancer stem cells, Oncotarg 8 (31) (2017) 51733–51747.

11
X. Zeng et al. Pharmacological Research 163 (2021) 105320

[133] M. Liu, C. Luo, J. Dong, J. Guo, Q. Luo, C. Ye, Z. Guo, CircRNA_103809 suppresses camptothecin against breast cancer stem cells using TIRF, Integr. Biol. (Camb) 8
the proliferation and metastasis of breast cancer cells by sponging MicroRNA-532- (6) (2016) 704–711.
3p (miR-532-3p), Front. Genet. 11 (2020) 485. [146] R.W. Robey, K. Steadman, O. Polgar, K. Morisaki, M. Blayney, P. Mistry, S.
[134] G. Pan, A. Mao, J. Liu, J. Lu, J. Ding, W. Liu, Circular RNA hsa_circ_0061825 (circ- E. Bates, Pheophorbide a is a specific probe for ABCG2 function and inhibition,
TFF1) contributes to breast cancer progression through targeting miR-326/TFF1 Canc Res 64 (4) (2004) 1242–1246.
signalling, Cell Prolif. 53 (2) (2020), e12720. [147] J.M. Houthuijzen, L.G. Daenen, J.M. Roodhart, E.E. Voest, The role of
[135] X. Song, Y. Liang, Y. Sang, Y. Li, H. Zhang, B. Chen, L. Du, Y. Liu, L. Wang, mesenchymal stem cells in anti-cancer drug resistance and tumour progression,
W. Zhao, T. Ma, C. Wang, Q. Yang, circHMCU promotes proliferation and Br. J. Cancer Suppl. 106 (12) (2012) 1901–1906.
metastasis of breast cancer by sponging the let-7 family, Mol. Ther. Nucleic Acids [148] J.A. Berzofsky, M. Terabe, J.B. Trepel, I. Pastan, D.F. Stroncek, J.C. Morris, L.
20 (2020) 518–533. V. Wood, Cancer vaccine strategies: translation from mice to human clinical trials,
[136] J.H. Xu, Y. Wang, D. Xu, Hsa_circ_001569 is an unfavorable prognostic factor and Cancer Immunol. Immunother. 67 (12) (2018) 1863–1869.
promotes cell proliferation and metastasis by modulating PI3K-AKT pathway in [149] J. Banchereau, R.M. Steinman, Dendritic cells and the control of immunity, Nat
breast cancer, Canc Biomark 25 (2) (2019) 193–201. 392 (6673) (1998) 245–252.
[137] Y.B. Xue, M.Q. Ding, L. Xue, J.H. Luo, CircAGFG1 sponges miR-203 to promote [150] L.X. Wang, G.E. Plautz, T cells sensitized with breast tumor progenitor cell
EMT and metastasis of non-small-cell lung cancer by upregulating ZNF281 vaccine have therapeutic activity against spontaneous HER2/neu tumors, Breast
expression, Thorac Canc 10 (8) (2019) 1692–1701. Canc Res Treat 134 (1) (2012) 61–70.
[138] C. Yuan, X. Luo, X. Zhan, H. Zeng, S. Duan, EMT related circular RNA expression [151] S. Baek, C.S. Kim, S.B. Kim, Y.M. Kim, S.W. Kwon, Y. Kim, H. Kim, H. Lee,
profiles identify circSCYL2 as a novel molecule in breast tumor metastasis, Int. J. Combination therapy of renal cell carcinoma or breast cancer patients with
Mol. Med. 45 (6) (2020) 1697–1710. dendritic cell vaccine and IL-2: results from a phase I/II trial, J. Transl. Med. 9
[139] K. Zeng, B. He, B.B. Yang, T. Xu, X. Chen, M. Xu, X. Liu, H. Sun, Y. Pan, S. Wang, (2011) 178.
The pro-metastasis effect of circANKS1B in breast cancer, Mol Canc 17 (1) (2018) [152] R. Yamada, A. Takahashi, T. Torigoe, R. Morita, Y. Tamura, T. Tsukahara,
160. T. Kanaseki, T. Kubo, K. Watarai, T. Kondo, Y. Hirohashi, N. Sato, Preferential
[140] N. Yan, H. Xu, J. Zhang, L. Xu, Y. Zhang, L. Zhang, Y. Xu, F. Zhang, Circular RNA expression of cancer/testis genes in cancer stem-like cells: proposal of a novel sub-
profile indicates circular RNA VRK1 is negatively related with breast cancer stem category, cancer/testis/stem gene, Tissue Antig 81 (6) (2013) 428–434.
cells, Oncotarg 8 (56) (2017) 95704–95718. [153] E. Quaglino, L. Conti, F. Cavallo, Breast cancer stem cell antigens as targets for
[141] A. Tiwari, B. Mukherjee, M. Dixit, MicroRNA key to angiogenesis regulation: immunotherapy, Semin. Immunol. 47 (2020), 101386.
MiRNA biology and therapy, Curr Canc Drug Targ 18 (3) (2018) 266–277. [154] S. Lanzardo, L. Conti, R. Rooke, R. Ruiu, N. Accart, E. Bolli, M. Arigoni,
[142] X. Lin, W. Chen, F. Wei, B.P. Zhou, M.C. Hung, X. Xie, Nanoparticle delivery of M. Macagno, G. Barrera, S. Pizzimenti, L. Aurisicchio, R.A. Calogero, F. Cavallo,
miR-34a eradicates long-term-cultured breast Cancer stem cells via targeting Immunotargeting of antigen xCT attenuates stem-like cell behavior and
C22ORF28 directly, Theranostics 7 (19) (2017) 4805–4824. metastatic progression in breast Cancer, Canc Res 76 (1) (2016) 62–72.
[143] J. Liu, T. Meng, M. Yuan, L. Wen, B. Cheng, N. Liu, X. Huang, Y. Hong, H. Yuan, [155] H. Bernhard, J. Neudorfer, K. Gebhard, H. Conrad, C. Hermann, J. Nahrig,
F. Hu, MicroRNA-200c delivered by solid lipid nanoparticles enhances the effect F. Fend, W. Weber, D.H. Busch, C. Peschel, Adoptive transfer of autologous,
of paclitaxel on breast cancer stem cell, Int. J. Nanomed. Nanosurg. 11 (2016) HER2-specific, cytotoxic T lymphocytes for the treatment of HER2-overexpressing
6713–6725. breast cancer, Cancer Immunol. Immunother. 57 (2) (2008) 271–280.
[144] R. Pourbagher, H. Akhavan-Niaki, S.G.A. Jorsaraei, S. Fattahi, D. Sabour, [156] W. Nie, J. Chen, Q. Xiu, Cytotoxic T-lymphocyte associated antigen 4
E. Zabihi, Z. Abedian, M. Ghasemi, M. Golpour, A. Mostafazadeh, Targeting LA7 polymorphisms and asthma risk: a meta-analysis, PLoS One 7 (7) (2012), e42062.
breast cancer stem cells of rat through repressing the genes of stemness-related [157] P. Darvin, V. Sasidharan Nair, E. Elkord, PD-L1 expression in human breast
transcription factors using three different biological fluids, Gene 734 (2020), Cancer stem cells is epigenetically regulated through posttranslational histone
144381. modifications, J. Oncol. 2019 (2019), 3958908.
[145] P. Arumugam, J.M. Song, Quantitative evaluation of ABC transporter-mediated
drug resistance based on the determination of the anticancer activity of

12

You might also like