Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Received: 1 February 2023 Revised: 26 July 2023 Accepted: 27 July 2023

DOI: 10.1002/cso2.1051

REVIEW

Unraveling the dangerous duet between cancer cell


plasticity and drug resistance

Namrata Chatterjee1 Bhavana Pulipaka2 Ayalur Raghu Subbalakshmi3


Mohit Kumar Jolly3 Radhika Nair4,5

1 Department of Biotechnology and


Medical Engineering, NIT Rourkela, Abstract
Odisha, India Cancer cell plasticity is the ability of tumor cells to switch phenotypes and is
2 Department of Biology, Ashoka
one of the predominant requisites of cancer cells capable of undergoing metas-
University, Haryana, India
3 Centre
tasis. Cancer cell plasticity is also recognized as one of the major contributors to
for BioSystems Science and
Engineering, Indian Institute of Science, intratumoral heterogeneity, a critical factor underlying the progression of malig-
Bangalore, India nant tumors, which is known to modify tumor response and induce resistance
4 Rajiv
Gandhi Centre for Biotechnology, against various modes of therapy, thus posing a barrier to efficient cancer man-
Kerala, India
agement. Cancer cell plasticity is acquired by the subversion of cell signaling
5 Centre for Human Genetics, Bangalore,
India
pathways like mitogen-activated protein kinase pathway, phosphoinositide-3-
kinase, signal transducer and activator of transcription 3, Wnt, Hedgehog and
Correspondence Notch as well as cellular programs such as epithelial to mesenchymal transition
Radhika Nair, Centre for Human
Genetics, Bangalore 560100, India, India. and phenotypic plasticity. This complex phenomenon has been studied in many
Email: radhikanair@cgh.res.in cancer types like pancreatic cancer, colon cancer and breast cancer. This review
will explore the current understanding we have in breast cancer on the intrin-
sic molecular mechanisms of cancer cell plasticity and the resistance to various
types of cancer therapy that arise as a result of plasticity. We conclude by explor-
ing the potential novel therapies that specifically target the pathways leading to
plasticity and can be leveraged to treat patients living with the disease.

KEYWORDS
cancer cell plasticity, cancer stem cells (CSCs), epithelial to mesenchymal transition (EMT),
intratumoral heterogeneity, targeted therapy, therapy/drug resistance

1 INTRODUCTION metastasis and resistance to therapy has been recognized.


(Figure 1, 2, Table 1)
Cell plasticity is the phenomenon of specific genotypes Cancer cell plasticity and intratumoral heterogeneity
producing different phenotypes in response to changing are particularly relevant in solid cancers like breast can-
external cues. Plasticity has been widely studied as a cer and are intertwined with the concept of cancer stem
critical process involved in development and evolution, cells (CSCs). CSCs are a subset of cells in the tumor that
but more recently its role in the progression of cancer, inherently have self-renewal and tumorigenic capacity.

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the
original work is properly cited.
© 2023 The Authors. Computational and Systems Oncology published by Wiley Periodicals LLC.

Comput Syst Oncol. 2023;3:e1051. wileyonlinelibrary.com/journal/cso2 1 of 15


https://doi.org/10.1002/cso2.1051
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2 of 15 CHATTERJEE et al.

In addition to driving cancer progression and metasta- pathways as well as extrinsic factors that facilitate cell
sis, CSCs also exhibit plasticity leading to drug resistance, plasticity. Besides covering the mechanisms by which cell
which contributes to the aggressive growth of a tumor. plasticity contributes to resistance, we will also focus on
Furthermore, the CSC paradigm and associated plasticity how we can target these pathways to negate therapeutic
have been proposed as a model to explain the phenomenon resistance with an overarching aim to obliterate cancer
of intratumoral heterogeneity, which refers to the phe- cells.
notypic, genetic, and functional heterogeneity within the
cell population of a single tumor as well as between
primary and metastatic tumors that arise as a result of 2 CELL-INTRINSIC PATHWAYS
intrinsic genetic programs and extracellular triggers. The INVOLVED IN CANCER CELL
phenomenon of cancer cell plasticity [1, 2] allows the PLASTICITY
cells to change their state thus giving rise to a heteroge-
nous cell population. Recent research has also found that Cancer cell plasticity is a consequence of the interplay of
factors such as genetic mutations, external stimuli, or genetic factors and programs, which lead to tumor progres-
signaling from the tumor microenvironment (TME) as sion and metastasis to distant organs and confers thera-
well as different environmental factors [3] can trigger the peutic resistance to breast cancer cells. Major intrinsic cell
transformation of already differentiated cells into CSCs, signaling pathways like MAPK, PI3K/AKT/mammalian
indicating the presence of plasticity even at various stages target of rapamycin (mTOR), Wnt, STAT3, Notch, Hh and
of differentiation in breast cancer [4]. transforming growth factor β (TGFβ) have been found
The molecular mechanisms that are related to the to be dysregulated in many cancer types and harbor the
emergence of plasticity include the activation of intrinsic potential to induce cellular plasticity in cancer cells [13].
mechanisms involved in signaling pathways like Notch, The MAPK pathway or mitogen-activated protein kinase
mitogen-activated protein kinase pathway (MAPK), pathway [14] represents a cascade of protein kinases that
Phosphoinositide-3-Kinase (PI3K), Wingless Related Inte- are considered a key signaling pathway for the regula-
gration Site (Wnt), Hedgehog (Hh) and Signal transducer tion of cellular and molecular functions like proliferation,
and activator of transcription 3 (STAT3) that work in apoptosis, differentiation and stress response [15]. The
tandem to control tumor cell dynamics and cell-to-cell emergence of the MAPK pathway stems back 30 years
communication. Cell-extrinsic factors like TME consisting ago [16] and is grouped into three families in mammals,
of the immune system as well as tumor cells and the namely, extracellular signal regulated kinases (ERKs),
cancer-associated extracellular matrix (ECM) that contain p38/SAKs, and c-Jun N-terminal kinases (JNKs) [17]. The
numerous receptors that have been strongly linked to MAPK pathway in the canonical form gets activated by
providing a niche for CSCs are also required for the a MAPK kinase kinase. In the case of the ERK pathway
development of plasticity. We will not be focussing on of MAPK, rapidly accelerated fibrosarcoma (Raf) is the
the role of the microenvironment in cancer cell plas- Mitogen activated protein kinase kinase (MAPKK), which
ticity, which is an area of intense research [5, 6] with activates MEK 1/2 and then mobilizes ERK 1/2. In JNK and
some excellent review articles on this subject [7–9]. The p38 pathways of the MAPK pathway, Transcription growth
importance of cancer cell plasticity becomes even more factor β activated kinase 1 (TAK1), MEKs, Mixed Linage
relevant in the context of therapeutic intervention in Kinases (MLKs) and Apoptosis Signal Regulating Kinase
the disease. Common treatments that exist for different (ASK1) act as MAPKKs leading to the activation of MKK
stages of cancer include surgery, hormonal therapy, 4/7 and further downstream pathways involving Jun path-
radiation therapy, chemotherapy, targeted therapy and way and MKK3/6, eventually leading to the activation of
immunotherapy [10]. However, the major problem that p38 [18]. Genetic mutation of the MAPK pathway is seen
stands in the way of an effective treatment regime is in 58% of all tumors with different frequencies of muta-
resistance to treatment. Therapy resistance poses a major tions like 42.1% in the JNK pathway, 40.3% in p38, 33.7%
problem in effective cancer treatment with tumor cells in ERK 1/2 pathway, and finally followed by only 6.1% in
that are initially responsive, developing resistance later, or ERK5 pathway [19].
having innate unresponsiveness to different anti-cancer There is evidence that aberrant activation of MAPK
drugs. Resistance in any type of cancer generally stems pathways and their role in cancer cell plasticity may be
from both genetic and epigenetic factors like cell plasticity, due to their role in the maintenance of CSCs. Loss of Dual
epithelial to mesenchymal transition (EMT), heterogene- specificity phosphate-4 (DUSP4), a negative regulator of
ity in cancer cells, or mutations in different genes [11, the MAP kinase superfamily, leads to the development of
12]. The need of the hour is to tackle the phenomenon of a CSC-like phenotype in basal breast cancer, which con-
therapy resistance in cancer cells by targeting the intrinsic fers plasticity to cells [20]. Singh et al. [21] focused on
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CHATTERJEE et al. 3 of 15

MAPK pathway inhibitors against different DUSPs in rela- tion of breast CSC phenotype. In breast ductal carcinoma,
tion to their contribution to plasticity and resistance and the MAPK-ERK pathway was involved in the cytoskeletal
found that the downregulation of DUSP1 specifically leads reorganization and transition to a mesenchymal pheno-
to plasticity in melanoma cells. DUSPs are negative regula- type [32] via Rap1Gap (a GTPase-activating protein [33])
tors of the MAPK pathway and act in tandem to maintain suppression, which increased the levels of MAPK/ERK.
cell proliferation and survival. However in cancer, down- MCF10ADCIS cells, which are a model of breast ductal car-
regulation of the DUSP leads to aberrant expression of cinoma, show higher levels of Rap1Gap when compared to
the pathway, but the exact mechanism of downregulation the invasive MCF10ACa1D cells, which represent invasive
still remains unknown. It has also been reported that Six ductal cancer. Rap1Gap silencing in MCF10ADCIS cells
Homeobox (SIX1), which is a developmental homeopro- resulted in elevated levels of ERK/MAPK, which promoted
tein that activates the MAPK/ERK pathway, leads to the their invasive capabilities accompanied by skeletal struc-
development of CSC phenotype in luminal B breast cancer ture re-organization and gain of mesenchymal capabilities
type [22]. It was observed that overexpression of SIX1 in the further consolidating the role of the pathway in cancer cell
MCF7 cell line leads to an increase in tumor-initiating cells plasticity. MAPK pathway can thus be considered as one
through phosphorylation of ERK of the MAPK pathway. of the major pathways that orchestrate plasticity in can-
Atypical MAPK pathway activation could also contribute cer cells and could be an attractive target for therapeutic
to promoting EMT leading to cancer cell plasticity [23, intervention.
24]. ERK3 overexpression is associated with an increase The second pathway that has a major role in can-
in cell migration and morphological changes in breast cer cell plasticity is the PI3K/AKT/mTOR pathway. The
cancer cells [25]. Mutations in the BRAF gene lead to alter- PI3K/AKT/mTOR pathway [34] also known as the phos-
ations in the RAS(Rat Sarcoma)-RAF(Rapidly activated phatidylinositol 3 kinase/protein kinase B pathway, is
fibrosarcoma)-MEK-ERK pathway [13], which has a major a highly conserved pathway affecting cellular processes
role in developing drug resistance. The pivotal transcrip- such as proliferation, survival, growth, metabolism, and
tional factor, Forkhead box C1 (FOXC1) mediates cancer immune response [35]. The signaling pathway came
cell plasticity and metastasis via partial EMT, which in turn into prominence in 1977 with the discovery linking PI
leads to drug resistance. The expression of FOXC1 is medi- kinase activity with viral oncogene-encoded Tyrosine
ated by the p38 MAPK pathway. FOXC1 is a pro-metastatic (Tyr)-kinase [36]. It also has an important role in various
gene, which aids cellular plasticity in cancer cells through oncogenic regulations and thus therapeutic targeting of
transcriptional activation of genes like MMP10, SOX4 and the PI3K pathway has been a key research area in cancer
SOX13. MAPK mediates FOXC1 expression by regulating its biology.
stability by binding and phosphorylation of p38 to Ser241 In physiological conditions, activation of the
and Ser272 sites of FOXC1 [26]. It has also been observed PI3K/AKT/mTOR pathway happens by either recep-
in the majority of cutaneous melanomas that mutations in tor tyrosine kinases or G protein-coupled receptors [37].
the MAPK pathway along with BRAF are the major driv- Various factors like the cancer-testis antigen A-kinase
ing factor for the development of cell plasticity [27]. BRAF anchor protein 3 (AKAP3) [38] are known to aberrantly
gene transcription is regulated by the MAPKKs of the activate the pathway leading to the development of cancer.
MAPK pathway. Thus, when the BRAF gene is mutated, Zhan et al. observed that AKAP3 was upregulated in
it is expressed by the MAPK pathway in its mutated form, cancerous cells of the breast leading to activation of the
leading to aberrant expression of the pathway. PI3K/AKT/mTOR pathway to facilitate cell migration,
The MAPK pathway plays a pivotal role in cellular pro- progression, and migration in breast cancer cells [39].
liferation in tumors like melanomas [28]. In oral squamous AKAP3 is an inhibitor of PTEN, which hinders the pro-
cell carcinoma (OSCC) [29], the MAPK pathway is an inter- gression of cancer cells by binding and inhibiting PI3K.
mediary between the epithelial factor Grainyhead Like Thus, in cancer cells, upregulation of AKAP3 leads to
Transcription Factor 2 (GRHL2) and the EMT-inducing Phosphatase and Tensin Homology (PTEN) inhibition
signal TGFβ [30]. In the OSCC cell line SCC9, inhibition and activation of the PI3K pathway leading to cancer cell
of MAPK signaling activated TGFβ signaling, which in growth and proliferation. This pathway mediates cellular
turn promotes the process of EMT. In the triple-negative plasticity, which results in different cellular populations
breast cancer (TNBC) subtype, MAPK signaling leads to within the tumor. It has been documented in several
the enrichment of breast CSCs [31]. In SUM-159 and MDA- studies that the pathway leads to the repression of the
MB-231 cells, depletion of pluripotency factors SRY-box tight junction proteins, which mediates cell plasticity.
Transcription factor 2 (SOX2), Krϋppel-like factor 4 (KLF4) Resistance of TNBC cells toward chemotherapeutic drugs
and NANOG was seen upon the inhibition of the p38- has been observed due to mutation in the PI3K3CA path-
MAPK signaling pathway indicating its role in the induc- way that encodes a subunit of PI3K leading to inhibition
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4 of 15 CHATTERJEE et al.

of cell apoptosis and increased cell proliferation [40]. The levels and VIM with increased levels of ECAD [48]. Thus,
mutation in the PI3K3CA gene leads to the upregulation all these studies demonstrate the pivotal role PI3K plays in
of different proteins of the pathway like p110α, Akt and conferring plasticity to the cancer cells by dysregulation of
mTOR and the downregulation of inhibitory protein various factors of the pathway.
PTEN resulting in cell apoptosis inhibition and chemore- STAT3 is another important intrinsic cellular signaling
sistance in the TNBC cell line MDA-MB-231. Another pathway in cancer cells. The signal transducers and acti-
study by Ghebeh et al., which focused on Fascin-mediated vation of transcription (STAT) 3 is a part of the STAT tran-
chemoresistance in breast cancer, demonstrated that the scription factor family, which has seven highly conserved
PI3K/AKT pathway mediates resistance [41]. The group and structurally similar members. Since its discovery in
used xenograft tumor models to evaluate the effect of 1994, STAT3 has been linked with cell growth, apoptosis
Fascin in mediating chemoresistance in breast cancer and disorders including cancer [49]. The STAT3 signaling
cells. The patient data analysis showed a direct correla- pathway is activated by growth factors and cytokines lead-
tion of survival rate of patients who were treated with ing to phosphorylation of STAT3. This causes homodimer-
chemotherapy and Fascin expression. The increase in ization and translocation to the nucleus where the complex
chemoresistance of the cancer cells was also attributed to binds to DNA response elements and regulation of gene
the PI3K pathway in parallel to increased Focal Adhesion transcription [50]. STAT3 is one of the main intrinsic sig-
Kinase (FAK) phosphorylation, expression of X-linked naling pathways that gets aberrantly expressed in cancer
inhibitor of apoptosis protein (XIAP) and Livin, which resulting in cancer cell inflammation. Different studies
are inhibitors of apoptosis proteins and also proapop- have also linked the pathway with cancer cell plasticity
totic markers (Caspase 9, Caspase3, Poly (ADP-ribose) [51, 52]. Another cancer type where STAT3 hyperactiva-
polymerase 1(PARP)) suppression. tion leads to enhanced cancer cell proliferation, tumor
The PI3K pathway can induce EMT directly or through angiogenesis, invasion, and metastasis in colorectal cancer
its cooperation with other signaling pathways [42, 43]. [53]. STAT3 mediates key transcription factors responsi-
Mitochondrial ribosomal protein L13 (MRPL13) was found ble for inducing cellular plasticity and is hyper-activated
to be a pro-EMT factor as its depletion resulted in a in colorectal cancer cells. In Estrogen Receptor-positive
decrease in mesenchymal markers like Snail Family Tran- (ER+) breast cancer cells, the STAT3/progesterone recep-
scriptional Repressor 1 (SNAI1) and Vimentin (VIM) and tor (PR) pathway mediates cell plasticity by inducing
an increase in the levels of E-Cadherin (ECAD). Fur- Aurora-A kinase (AURKA) protein. In a study by Antonino
ther, it was observed that MRPL13 mediated EMT through et al., AURKA activation was mediated by phosphorylation
PI3K/AKT/mTOR signaling pathway. Interestingly the of serine727-STAT3/serine294-PR transcriptional complex.
PI3K/AKT pathway was involved in conferring resistance This led to the regulation of a stemness reprogramming
to tamoxifen in addition to their role as promoters of EMT gene KLF4 by the transcriptional complex, thus promoting
in T47D cells [44] when Connexin43, an EMT inhibitor, plasticity and therapy resistance in the cells [52].
was silenced leading to a reduction in the levels of ECAD The involvement of the Wnt pathway in cancer plas-
as well as an increase of NCAD levels. The addition of ticity in its canonical form is well-established. However,
LY294002, a PI3K/Akt specific signaling inhibitor [45], more recent research has uncovered the effect of the
suppressed EMT, and FAK/PI3K/Akt upregulation con- aberrant functioning of the pathway through genetic and
ferred the cells with resistance to cisplatin in addition epigenetic mechanisms in the initiation, progression, and
to promoting EMT [46], thus corroborating the role of development of drug-resistant characteristics in various
PI3K signaling in EMT progression. In this study, when cancers, which includes the modulation of cancer cell
MDA-M-231 and MCF-7 cells were treated with LY294002, plasticity. The role of the Wnt pathway in cancer was dis-
mesenchymal factors like SNAI1, SNAI2, VIM and N- covered when mammary tumors were observed in mice
Cadherin (NCAD) significantly decreased along with a after infection with the murine mammary tumor virus
concomitant increase in the level of ECAD and made (MMTV) leading to the activation of the murine Int-1 gene,
cells sensitive to cisplatin treatment. Analysis using T47D, which was later named Wnt-1. Later studies found that
MDA-MB-231 cell lines demonstrated that EMT induc- the cytosolic concentrations of β-catenin, a homolog of
tion by inorganic pyrophosphatase 1 was mediated by the Drosophila Armadillo protein with significant roles as
the PI3K/AKT signaling pathway [47]. When the relation- the regulator of intracellular adhesion and as a transcrip-
ship between Ki-Ras2 Kirsten rat sarcoma viral oncology tional activator controlled by the signaling pathway, was
homolog (KRAS), PI3K/AKT and EMT was analyzed in downregulated by a tumor-suppressor called adenomatous
T47D cells, it was seen that KRAS depletion resulted in polyposis coli (APC), while the Wnt1 protein upregulated
the silencing of the signaling factors of PI3K/AKT, along- it, providing a basis for the Wnt β-catenin pathway-cancer
side EMT inhibition as indicated by the reduced NCAD connection. The aberrant functioning of the pathway can
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CHATTERJEE et al. 5 of 15

be observed in different cancers like colorectal, gastric, way as a key modulator for the plasticity of the CSCs
and breast cancers with studies on genetically engineered in TNBC. Hh pathway acts by the reprogramming of the
mouse models revealing that the induction of mammary cancer-associated fibroblasts to a CSC supporting pheno-
tumorigenesis occurs as a consequence of the dysregula- type by activation of the fibroblast growth factor 5 and
tion of the Wnt/β-catenin pathway via the overexpression fibrillary collagen secretion [61]. The Notch pathway is a
of its activators such as Wnt and the targeted disrup- tightly controlled signaling pathway responsible for vari-
tion or silencing of its inhibitors such as APC [54]. In ous aspects of development in metazoans as well as the
most cancers (most notably colorectal and gastrointestinal renewal of tissue. As a conserved signaling pathway, it
cancers), mutations that result in loss-of-function in the plays a critical role in short-range communication between
genes coding for the proteins involved in the Wnt path- cells by establishing physical contact between the target
way that are responsible for the degradation of β-catenin cells. The pathway is responsible for enabling or suppress-
such as APC and Axin have been recognized. As a result, ing, depending on context and molecular cues, cellular
the downregulation of the amount of free cytoplasmic β- functions like cell differentiation, proliferation, cell death
catenin is inhibited, resulting in the transcription of Wnt and controlling cell fates throughout the development of
target genes. A critical role of the Wnt pathway is also the organism as well as in self-renewing adult tissue. Given
observed in the maintenance of cancer cell plasticity. Dif- the vital role it plays, the misregulated loss- or gain-of-
ferent transcription factors like Snal1(Snail1), Snal2 (Slug), function of the Notch pathway can give rise to various
TWIST1, ZEB1, and ZEB2 are the driving factors for the human diseases including cancer. Dysregulation of Notch
transition between the epithelial and mesenchymal cells has also been linked to the maintenance of CSCs. In breast
through the signaling pathways like Wnt, Hh, Notch [55]. cancer specifically, studies have shown that Notch acts as
Genetic alterations in APC and RNF43 result in the upreg- an oncogene. The overexpression of the Notch proteins 1, 3,
ulation of Snail, thus leading to a transition between the or 4 has been shown to transform normal breast epithelial
two phenotypes [56]. Inhibition of GSK3β by the Wnt lig- cells into cancer cells. An ECM glycoprotein, tenascin-C
ands leads to an increase in stability of the transcription (TNC) is observed to promote cancer cell plasticity via acti-
factor Snail, thus leading to plasticity in the cancer cells vation of the Notch pathway in glioblastoma multiforme.
[57]. Li et al. linked the plasticity of the prostate CSCs It was observed using PCR (Polymerase Chain Reaction)
with a circadian rhythm gene PER3. Low levels of the gene and microarray technology that TNC elevates the signal-
lead to activation of the Wnt pathway through phospho- ing components of the pathway—JAG1, ADAMTS15 and
rylation of β-catenin pathway [58]. Upregulation of the NICD 1/2 [62, 63]. Notch pathway is also observed to pro-
Wnt pathway is mediated by inhibitors of other differ- mote CSCs in colon cancer leading to cellular plasticity and
ent pathways as well. For example, MEK inhibitors are chemoresistance [64]. The cascade of events involves the
considered to activate the pathway through AIX1, which induction of another receptor of the pathway—Jagged 1,
is the point of interjection between the Ras-MAPK and which in turn activates CD44, Slug, and SMAD expression,
Wnt pathway. These inhibitors are associated with rapid progressing to an EMT phenotype in the cancer cells [65].
reduction of the AXIN1 protein, which in turn dissoci- In another study by Ishida et al., a hypoxic environment
ates from GSKβ, thus activating the pathway [59]. Under is an inducer of EMT via activation of Notch signaling
physiological conditions, the Hh pathway plays a vital role pathway; however, the exact mechanism of how they are
in embryonic development by patterning the developing linked is still unknown [66].We tried to summarize the role
neural tube, axial skeleton, limbs, skin, lungs, hair and of different pathways in mediating the cellular plasticity
teeth. The first clue connecting the Hh signaling pathway in tumors and the potential role as therapeutic targets in
to cancer came as a result of the discovery that a mutation curbing the same. However, more in-depth knowledge has
in the tumor suppressor gene that encodes for a receptor to be developed to completely understand the role of differ-
molecule found within the pathway called patched was ent pathways and their link with plasticity in cancer cells.
an underlying factor in the development of both famil- (Figure 1)
ial and sporadic forms of basal cell carcinoma. The Hh
pathway is involved in EMT transition in different types
of cancer cells [8]. A study by Zhu et al. found that CSC- 3 DRUG RESISTANCE MEDIATED BY
regulated expression of tetraspanin 8 (TSPAN8) leads to CANCER CELL PLASTICITY
activation of the Sonic-Hh pathway. The cascade of reac-
tions that follow the interaction of TSPAN8 with PTCH1 While chemotherapy is the mainstay in our battle against
in turn inhibits degradation of the SH/PTCH1 complex cancer, intrinsic or extrinsic factors can unfortunately lead
[60] resulting in CSC-like phenotypes and resistance to to cancer cells acquiring the ability to evade the effect
chemotherapy. A study by Cazet et al. linked the Hh path- of drugs [67]. One factor for the development of drug
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
6 of 15 CHATTERJEE et al.

F I G U R E 1 Cell intrinsic pathways involved in cancer cell plasticity. The major cellular signaling pathways involving multiple pathways
like MAPK, PI3K-AKT-mTOR, STAT3, Wnt, Notch and Hedgehog are key players in promoting cellular plasticity and therapy resistance in
cancerous cells. Targeted therapeutic approach toward these pathways can lead to avenues leading to curing this deadly phenomenon. JNK:
c-Jun N-terminal kinases, MAPK: mitogen activated protein kinases, Raf: rapidly accelerated fibrosacroma, MEK 1: Mitogen-activated protein
kinase kinase, ERK: extracellular signal regulated kinase, STAT3: signal transducer and activator of transcription 3, PI3K:
phosphoinositide-3-kinase, AkT: Ak strain transforming, mTOR: mammalian target of rapamycin, NCID: Notch intracellular domain, MAM:
mitochondria-associated ER membrane, GLI1/2: human glioma associated oncogene homolog 1/2, TCF/LEF: T-cell factor/lymphoid
enhancer factor.

resistance is plasticity. Different pathways as discussed an indirect connection with the activation of the MAPK
above have been shown to be involved in the phenomenon pathway. Targeting these factors can yield a therapeu-
of cancer cell plasticity leading to resistance toward drugs. tic intervention point to check the treatment resistance
A recent study has linked the translocation of CD24 to phe- mediated by cellular plasticity. Radiotherapy is the use of
notypic switching through cellular plasticity induction via radiation to curb the growth and proliferation of cancer
the p38 MAPK pathway. In breast cancer, CD44 and CD24 cells. The effectiveness of radiotherapy is compromised
are the predominant CSC markers to identify the cells by adaptive radioresistance or the acquisition of resis-
at different stages of differentiation. CD44−/low CD24+ cell tance by the cells over a period of time. In radioresistant
population is predominant in luminal breast cancer, and breast cancer cell lines, a major factor for acquired resis-
CD44+ CD24−/low in basal and mesenchymal breast cancer tance is an increase in CSCs alongside elevated Epithelial
[68–70]. Many studies have linked the presence of different Cell-Adhesion Moelcule (EpCAM) expression. Elevated
breast CSCs (BCSCs) markers to the development of cellu- EpCAM expression conferred resistance toward radiother-
lar plasticity in cancer cells. CD44+ CD24− is observed to apy in breast cancer cell line ZR-75-1 by enhancing Akt
have a correlation with increased metastatic and plastic- expression, which in turn increases the stemness of the
ity potential [71]. A study by Hugh et al. focused on the cells and induces cellular plasticity through inducing EMT
localization pattern of CD24 indicating the development phenotype in the cancer cells. Thus, this pathway act as a
of plasticity [72]. The cellular localization of CD24 is medi- driving factor for EpCAM-mediated cellular plasticity [75].
ated by p38 MAPK pathway activation with phosphoryla- A study by Arnold et al. found radiation to be respon-
tion leading to Bcl-2 overexpression in the cell membrane sible for conferring radiation-mediated cellular plasticity
[73] and treatment resistance. Non-genomic factors like in breast cancer stem-like cells resulting in radioresis-
estrogen mediate cellular plasticity in the tumor cells that tance [76]. The findings of these researchers confirmed
have a propensity toward undergoing EMT. Estrogen bind- that resistance and changes in cellular plasticity in the
ing leads to the activation of c-Src, which in turn mediates TNBC cell line were mediated by STAT3 via an inflam-
cell signaling pathways like MAPK [74]. Thus, estrogen has matory response. The inflammatory response is mediated
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CHATTERJEE et al. 7 of 15

by Interleukin-6 (IL6) leading to the activation of the the plasticity of cancer cells and provide newer therapeutic
Janus family kinase and the STAT3 pathway. Resistance avenues.
toward the drug lapatinib in locally advanced Human One of the initial theories to explain the phenomenon of
Epidermal Growth factor Receptor-2 positive (HER2+) drug resistance in breast cancer involves BCSCs, which are
breast cancer is mainly mediated by Interleukin-8 (IL-8) thought to be inherently resistant to cytotoxic and targeted
that is secreted from tumor macrophages via activation therapies. Recent research has found evidence that links
of Src/STAT3/ERK1/2 pathway [77] and confers radio- Notch1 and Notch4 in the development of drug-resistant
resistance to breast cancer cell lines. Activation of the behavior exhibited by BCSCs. A study using polyoma
STAT3 pathway occurs by phosphorylation of Tyr705 middle T (MMTV-PyMT) mammary tumor mouse model
residue of the signal transducer [78] and underscores the also showed that cells showed a temporal increase in
importance of targeting STAT3 in breast cancer. expression of the Notch ligand, DII1, as the tumor pro-
Hyperactivated Wnt pathway and the resultant dysreg- gressed from early to late stages. Moreover, these DII1+
ulation of the proteolytic degradation of β-catenin is also cells also exhibit chemotherapeutic-resistant gene signa-
strongly linked to the development of drug resistance tures, and the consequent blocking of the ligand sensitized
in many invasive cancers [79]. Conventional chemother- the cells to chemotherapy as well as inhibiting tumor
apy affects cancer cells by exposing them to cytotoxic growth and metastasis [86]. The Dll1+ cells were enriched
agents that inhibit cell proliferation and induce cell death with genes that aid the process of plasticity and are thus
in rapidly proliferating cells, while radiotherapy involves associated with increased cellular plasticity. Moreover, the
the utilization of ionizing radiation, damaging their DNA small interfering RNA- mediated knockdown of Notch1 in
and resulting in cell death. CSCs are generally known ALDH+ cells, resulted in an inhibition of their growth and
to be more resistant to chemo and radiation therapies as increased apoptosis and tumor growth in mice models [87].
compared to the non-stem cell populations in the tumor, Jagged-1 receptor of the Notch pathway is overexpressed
causing a relapse even after treatment [80]. Given that the through SLUG upregulation and in turn, represses E-
Wnt signaling pathway plays an important role in main- cadherin thus allowing EMT and resistance in the cancer
taining CSCs, it could be a major mechanism underlying cells [88].
the development of therapeutic resistance. A study using
carboplatin-resistant in vivo patient-derived xenografts as
well as isogenic TNBC cell-line models found that there 4 POTENTIAL TREATMENTS
was an increased activation of the Wnt/β-catenin path- TARGETING CELL-INTRINSIC
way, which was correlated with the expression of stem PATHWAYS MEDIATING CANCER CELL
cell markers in the models studied. Moreover, they also PLASTICITY
demonstrated that the inhibition of the Wnt signaling
pathway in these models resulted in their resensitization to The molecular mechanisms in cancer cell plasticity that
carboplatin from their resistant states—thus highlighting contribute to resistance remain to be elucidated com-
a potential mechanism for overcoming chemotherapeutic pletely. Research has paved the way for targeting various
resistance, especially in breast cancer [81]. More evidence crucial factors that play a role in conferring cellular
comes from studies where ST8SIA1 and its correspond- plasticity and therapy resistance in breast cancer cells.
ing mRNA were upregulated in chemotherapy-resistant These factors can thus act as potential biomarkers for tar-
TNBC patients. Inhibition of ST8SIA1 led to increased geted therapy and for the development of new treatment
efficacy of chemotherapy through the suppression of the modalities.
Wnt/β-catenin pathway [82]. One of the proteins in the MAPK pathway that is linked
Studies have also revealed the role of plasticity-mediated to plasticity and resistance is the MEK protein. Drugs tar-
drug resistance in cancer cells [83] via the Hh pathway geting the MEK protein have been observed to lead to
through the upregulation of different proteins like OCT4, improvement in the prognosis of breast cancer patients.
BMI 1 and SOX 2, which increased stemness and drug resis- One such targeted therapy involved a combinational ther-
tance [84]. In studies by Magistri et al. and Nguyen et al., apy of RAF/MEK inhibitor CH5126766 with eribulin in
it was observed that Smoothened (SMO) inhibition leads TNBC that successfully in inhibiting apoptosis and cell
to the overexpression of two Hh target genes, GL1 and migration, thus contributing to tumor growth [89]. In
GL2, which are the transcription factors responsible for another study, dual inhibition of MEK1/2 and MEK 5 using
the upregulation of the master gene Snail that is known inhibitors cobimetinib and trametinib resulted in the sup-
for promoting plasticity in the cancer cells [85]. Targeting pression of EMT in TNBC [90]. Mohan et al. have also
these factors of the pathway could lead to a reduction in shown that brucein D, which is an inhibitor targeting the
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
8 of 15 CHATTERJEE et al.

p38 pathway leads to apoptosis and decreases the viability dependent cancers and as a mechanism to inhibit the
of breast cancer cells by inhibiting the MAPK pathway [91]. therapy resistance conferred by this oncogenic signaling
PI3K/AKT/mTOR pathway is another altered pathway pathway. For example, the introduction of a truncated
in breast cancer cell plasticity. Inhibition of the pathway version of the APC protein was shown to have mod-
using small molecule inhibitors targeting different compo- erate success in mediating the degradation of β-catenin
nents of the pathway has emerged as an area of research in colorectal cancers where the APC gene has a loss-of-
interest. In vitro studies using inhibitors like buparlisib, function mutation [100]. Another potential mechanism
pictilisib, alpelisib, and taselisib have demonstrated pro- to increase β-catenin degradation involves the modula-
apoptotic and anti-proliferative activity on breast cancer tion of Axin production through the active inhibition
cell lines [92]. Natural compounds have also started to of Tankyrases, which are proteins that degrade Axin.
gain popularity for potential targeting of the pathway. One Tankyrase inhibitors such as XAV939 strongly suppress
such bibenzyl compound is gigantol extracted from orchids the Wnt/beta-catenin pathway, resulting in an anti-cancer
belonging to the Dendrobium genus. It has been observed effect in Wnt-mediated cancers [101]. PAF-Wnt signaling
to downregulate the PI3K/Akt/mTOR pathway in cancer is involved in inducing cellular plasticity in breast cancer
cells thus in turn inhibiting proliferation and inducing cells and can be considered as a potential therapeutic tar-
apoptosis. The study involved in silico determination of get. In a study by Wang et al., PAF was evaluated as a target
the potential pathway by which gigantol acts followed by of the Wnt pathway, and drugs were screened against the
in vitro assays to elucidate their effect on breast cancer cell pathway. NVP-AUY922, an Hsp90 inhibitor, leads to sig-
lines [93]. Possible target identification for gigantol identi- nificant downregulation of β-catenin, thus reducing the
fied PI3K, mTOR, and Akt as potential targets. Molecular stemness of the cancer cells.
docking studies provided the exact binding sites of the The Hh pathway is a strong contender for therapeutic
drug, which were then validated using in vitro methods. targeting in breast cancer given its important role in the
Another natural compound that has emerged as a potential progression of the disease by using Hh inhibitor molecules.
candidate for therapy in TNBC is a novel anilide shikonin For example, SMO inhibitors that target the SMO recep-
ester, M9. Studies on the breast cancer cell line MDA-MB- tor have proved to be fairly successful. Two different
231 showed that M9 has the ability to induce apoptosis and SMO inhibitors, vismodegib and sonidegib, have received
arrest the cell cycle by affecting both the PI3K/Akt/mTOR approval from the Food and Drug Administration (FDA)
and Wnt/β-catenin pathways by inhibiting Akt and down- for the treatment of breast cancer after it was found that
regulating the PI3K pathway [94]. Inhibitors of Akt are these molecules were able to successfully bind to SMO and
at the forefront of therapeutic intervention strategies that inhibit the downstream activation of the Hh target genes.
promote anti-cancer effects in breast cancer. Zhu et al. Novel SMO inhibitors such as saridegib and taladegib are
observed that AZD5363, an Akt inhibitor, was successful now under development, demonstrating the scope in the
in reducing stemness in the breast cancer cell line MCF-7. area of SMO inhibitor-based treatment mechanisms for
It was used in combination with 3,3′-diindoylmethane to several cancer types [102]. In addition, cyclin-dependent
induce apoptosis in the cells [95]. kinase inhibitors such as dinaciclib targeting the Hh sig-
The dysregulation of the signal transducer 3 in cell plas- naling pathway are also currently being tested in clinical
ticity and therapy resistance in breast cancer makes it an trials as shown in a recent study leading to the reduction
attractive target for small molecule inhibitors and drugs. of stemness and other malignant properties in TNBC cells
Resveratrol is a natural phytoalexin that regulates the cas- [103].
cade of STAT signaling and has the potential to mitigate Targeted therapies involving the inhibition of the Notch
breast cancer progression [96]. Studies have been con- pathway in breast cancer are chiefly based on the use of
ducted in vitro that showed the target of Resveratrol is gamma-secretase inhibitors, transcriptional inhibitors,
STAT3 protein [97]. Resveratrol triggers a stepwise inhibi- and monoclonal antibodies, which target the ligand and
tion cascade involving Src tyrosine kinase and STAT3 pro- receptor components of the Notch pathway. In vitro
teins. In order to circumvent the problem of trastuzumab studies with ER+ breast cancer cells treated with the
resistance in Her2+ patients, scientists have targeted the gamma-secretase inhibitor Z-Leu-Leu-Nle-CHO [104] led
proto-oncogene MUC1, which is a downstream target of to the strong inhibition of cells and reduction of plasticity
STAT3 [98]. 6Br-6a is a STAT3 inhibitor that decreased cell of the cancer cells. Furthermore, a study using a TNBC
proliferation, increased apoptosis and reduced plasticity murine model showed that the inhibition of a deubiquiti-
making it a promising target in breast cancer [99]. nase USP9x using the small molecule inhibitor G9 reduces
The use of molecules that inhibit the various con- Notch activity [105]. ϒ-secretase inhibitor MRK-003
stituents of the Wnt signaling cascade is currently being reversed the role of Jagged-1-mediated bone metastasis
studied as a potential therapeutic strategy in Wnt- in cancer and consequently decreases tumor growth.
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CHATTERJEE et al. 9 of 15

TA B L E 1 Molecular pathways involved in cancer plasticity, which impact drug resistance and possible therapeutic strategies.
Pathway Related drug resistance Potential treatment Inhibitor targeted
MAPK pathway 1. Chemoresistance 1. CH5126766 along with eribulin 1. RAF/MEK inhibitor [89]
2. Radiation resistance 2. Dual inhibitors cobimetinib and 2. MEK1/2 and MEK 5 [90]
3. Enzyme therapy resistance trametinib 3. p38 pathway [91]
3. Brucein D
PI3K/AKT/mTOR 1. Chemotherapy Resistance 1. Gigantol 1. Akt [93]–[95]
pathway 2. Radiotherapy resistance 2. Anilide Shikonin ester- M9. 2. Luminal A breast cancer
3. AZD5363 alongside cells [119]
3,3′-Diindoylmethane
4. Dietary phytochemicals
STAT3 pathway 1. Radiotherapy resistance 1. Resveratrol 1. STAT3 [96, 98, 99]
2. Chemotherapy resistance 2. Trastuzumab
3. 6Br-6a
Wnt/β-catenin 1. Chemoresistance 1. Tankyrase inhibitors such as XAV939 1. Axin degradation [101]
pathway 2. Radioresistance 2. Introduction of truncated APC 2. Lack of APC [101]
3. Immunotherapy resistance 3. Targeting β-catenin 3. NVP-AUY922
Hedgehog pathway 1. Chemoresistance 1. SMO inhibitors like vismodegib, 1. SMO receptor [102, 103]
2. Immunotherapy resistance sonidegib, saridegib, and taladegib
Notch pathway 1. Chemoresistance 1. Gamma-secretase inhibitors such as 1. Gamma-secretase protein
2. Immunotherapy resistance Z-Leu-Leu-Nle-CHO [104, 105]
2. Monoclonal antibodies like OMP-59R5 2. Notch 2/ Notch 3 receptors
[120]
Abbreviations: AKT, Ak strain transforming; APC, adenomatous polyposis coli; MAPK, mitogen activated protein kinases; MEK, mitogen-activated protein kinase
kinase; mTOR, mammalian target of rapamycin; PI3K, phosphoinositide-3-kinase; RAF, rapidly accelerated fibrosacroma; STAT3, signal transducer and activator
of transcription 3.

Different treatment avenues have been developed over Unfortunately, even with the advancement of therapy
the years to target the deadly phenomenon of cellular targeting different pathways that facilitate cellular plas-
plasticity. Targeting the complex network of cell signaling ticity, the prognosis has not improved much for patients.
pathways to curb the phenomenon of resistance aris- Various hurdles in curbing the problem of cancer cell
ing from cellular plasticity has led to different efficient plasticity and associated drug resistance remain. In depth
therapies. However, the complexity involved in cancer understanding of CSCs and their crosstalk with the TME
cell plasticity is still to be uncovered completely and as well as role of the TME in pathway dysregulation and
newer targets explored to curb the scourge of therapeutic its role in cancer cell plasticity is of foremost importance.
resistance. (Table 1) (Figure 2) The TME [107] is a complex network of different cells,
which along with the cancer cells provide an avenue for
the induction of cellular plasticity as well as therapy resis-
5 CONCLUSION AND FUTURE tance. Targeting the microenvironment will also lead to
PERSPECTIVES potential therapeutic avenues for curbing this deadly phe-
nomenon of plasticity and therapy resistance. The exact
Interest in understanding cancer cell plasticity as well as its mechanisms, both intrinsic and extrinsic mechanisms
crucial role in therapy resistance has led to the uncovering leading to cancer cell plasticity and therapy resistance in
of several mechanisms behind this process. The main focus cancer, are yet to be untangled. This will be of immense
of this review is the cell-intrinsic pathways that modulate value to researchers who are continuously looking for
the phenomenon of plasticity with the aim of targeting designing new therapeutic approaches that aim at perturb-
major cell-intrinsic as well as extrinsic drivers of the pro- ing the interplay of cellular factors resulting in plasticity
cess. Cell intrinsic mechanisms include major signaling [108] and therapy resistance.
pathways, which are mostly dysregulated in cancer lead- Another area of development concerns the major set-
ing to the ability of the cells to transition between different back that occurs with treatment strategies is clonal evo-
phenotypes [13, 106]. These dysregulated pathways also lution leading to acquired resistance against these tar-
are a major driving factor for the development of resis- geted therapies and inhibitors. One strategy to overcome
tance toward treatment and targeting the same has been acquired resistance is to make next-generation inhibitors
the major focus of researchers for a long time. that are able to target the resistance arising due to clonal
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
10 of 15 CHATTERJEE et al.

F I G U R E 2 Targeting molecular pathways contributing to cancer cell plasticity. Different therapeutic molecules and treatment
modalities that target pathways have been developed with the inhibitors having the potential to effect its target molecule and reversing
plasticity. MAPK: mitogen-activated protein kinases, Raf: Rapidly accelerated fibrosacroma, MEK 1: mitogen-activated protein kinase kinase,
STAT3: Signal transducer and activator of transcription 3, Akt: Ak strain transforming.

evolution. In a study by Hayashi et al., inhibitors of Epi- progression [116]. This multi-scale model included a
dermal Growth Factor Receptors (EGFR) and anaplastic combination of partial differential equations, deep rein-
lymphoma kinase were developed targeting the altered forcement learning and agent-based modeling. It helped
phenotypes of the gene [109]. Various genetic mutations recreate patterns of tumor growth and the evolution of
in therapy-resistant cancer cells like BRAF, PTEN, MET, microvascular network morphology by estimating the
and TP53 have been observed in colorectal cancer, which gradient of factors like oxygen and Vascular Endothelial
are considered as clonal markers [110]. Studies to deter- Growth Factor (VEGF) along with the temporal changes
mine the reasons for these markers that lead to clonal observed in the cellular phenotype. In a study involving
evolution of the tumor are in PTEN, ROBO1, CDH1, high-grade serous ovarian carcinoma, a mathemati-
PIK3CG, DMD, and LRP1B [111]. Targeting the geneti- cal model was used to determine the effectiveness of
cally altered factors leading to clonal evolution through treatment strategies such as initial surgery followed by
precision medicine and personalized therapy can be an chemotherapy and multiple chemotherapy cycles [117].
alternative to overcome the barrier. Mathematical models have also been developed to study
Computational approaches such as modeling the therapeutic resistance during cancer treatment [118], such
dynamics of gene regulatory networks, metabolic model- as quantifying the role of point mutations, phenotypic
ing and machine learning are being increasingly used to switching, gene amplification and the amount of drug
study tumor development and suggest new cancer treat- dosage accessible to cells in enabling drug resistance.
ment strategies [112]. Metastatic castrate-resistant prostate To conclude, we strongly believe that unraveling the
cancer progression involves a complex circuit involving molecular circuitry involved in cancer cell plasticity lead-
TGFβ and receptor activator of nuclear kappa β (κβ) ing to drug resistance will open new avenues of therapeutic
ligand, which have shown to possess multiple potential targeting, which will ultimately benefit patients.
therapeutic targets [113, 114]. To address the complexity
and multi-scale nature of the disease, initial predictions AC K N OW L E D G M E N T S
were generated using discrete-continuum hybrid cellular The authors would like to acknowledge Ms Mubeena S for
automata agent-based model; these predictions were later her help with editing the paper. The authors would also
validated using in vitro and in vivo studies [115]. In a like to thank RGCB for funding.
study involving breast-cancer-bearing mice, microscale
computed tomography images were compounded with a D A T A AVA I L A B I L I T Y S T A T E M E N T
multi-scale computational model to predict the growth Data sharing is not applicable to this article as no datasets
patterns of the microvascular networks and cancer cell were generated or analyzed during the current study.
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CHATTERJEE et al. 11 of 15

ORCID 13. D. Kong, C. J. Hughes, and H. L. Ford, Cellular plasticity in


Namrata Chatterjee https://orcid.org/0000-0001-9568- breast cancer progression and therapy, Front Mol. Biosci. 7
887X (2020), 72. https://doi.org/10.3389/fmolb.2020.00072
14. T. Mohapatra, and M. Dixit, IQ motif containing GTPase activat-
Mohit Kumar Jolly https://orcid.org/0000-0002-6631-
ing proteins (IQGAPs), a-kinase anchoring proteins (AKAPs) and
2109
kinase suppressor of Ras proteins (KSRs) in scaffolding oncogenic
pathways and their therapeutic potential, ACS Omega. 7 (2022),
PEER REVIEW 45837–45848. https://doi.org/10.1021/acsomega.2c05505
The peer review history for this article is available 15. Y.‑J. Guo, W.‑W. Pan, S.‑B. Liu, Z.‑F. Shen, Y. Xu, and L.‑L. Hu,
at https://publons.com/publon/10.1002/cso2.1051 ERK/MAPK signalling pathway and tumorigenesis (Review),
Exp. Ther. Med. 19 (2020), 1997–2007. https://doi.org/10.3892/
REFERENCES etm.2020.8454
16. N. G. Ahn, The MAP kinase cascade. Discovery of a new signal
1. A. P. Thankamony, K. Saxena, R. Murali, M. K. Jolly, and
transduction pathway, Mol. Cell. Biochem. 127 (1993), 201–209.
R. Nair, Cancer stem cell plasticity–A deadly deal, Front Mol.
https://doi.org/10.1007/BF01076771
Biosci. 7 (2020), 79. https://doi.org/10.3389/fmolb.2020.00079
17. D. K. Morrison, MAP kinase pathways, Cold Spring Harb. Per-
2. G. Lee, and R. R Hall, Cancer stem cells: Cellular plasticity, niche,
spect. Biol. 4 (2012), no. 11, a011254–a011254. https://doi.org/10.
and its clinical relevance, J. Stem Cell Res. Ther. 6 (2016), 363.
1101/cshperspect.a011254
https://doi.org/10.4172/2157-7633.1000363
18. N. A. Turner, and N. M. Blythe, Cardiac fibroblast p38 MAPK: A
3. G. Fusco, and A. Minelli, Phenotypic plasticity in development
critical regulator of myocardial remodeling, J. Cardiovasc. Dev.
and evolution: Facts and concepts. Introduction, Philos. Trans.
Dis. 6 (2019), 27. https://doi.org/10.3390/jcdd6030027
R. Soc. Lond. B Biol. Sci. 365 (2010), 547–556. https://doi.org/
19. M. Sinkala, P. Nkhoma, N. Mulder, and D. P. Martin, Inte-
10.1098/rstb.2009.0267
grated molecular characterisation of the MAPK pathways in
4. J. M. Smigiel, S. E. Taylor, B. L. Bryson, I. Tamagno, K. Polak,
human cancers reveals pharmacologically vulnerable mutations
and M. W. Jackson, Cellular plasticity and metastasis in breast
and gene dependencies, Commun. Biol. 4 (2021), 9. https://doi.
cancer: A pre- and post-malignant problem, J. Cancer Metastasis
org/10.1038/s42003-020-01552-6
Treat. 5 (2019), 47. https://doi.org/10.20517/2394-4722.2019.26
20. J. M. Balko, L. J. Schwarz, N. E. Bhola, R. Kurupi, P. Owens,
5. L. Arora, M. Kalia, S. Dasgupta, N. Singh, A. K. Verma, and
T. W. Miller, H. Gómez, R. S. Cook, and C. L. Arteaga, Activa-
D. Pal, Development of a multicellular 3D tumor model to study
tion of MAPK pathways due to DUSP4 loss promotes cancer stem
cellular heterogeneity and plasticity in NSCLC tumor microen-
cell-like phenotypes in basal-like breast cancer DUSP4 loss pro-
vironment, Front. Oncol. 12 (2022), 881207. https://doi.org/10.
motes CSC phenotypes in basal-like breast cancer, Cancer Res.
3389/fonc.2022.881207
73 (2013), 6346–6358.
6. H. Luo, X. Xia, L.-B. Huang, H. An, M. Cao, G. D. Kim, H.-N.
21. M. K. Singh, S. Altameemi, M. Lares, M. A. Newton, and
Chen, W.-H. Zhang, Y. Shu, X. Kong, Z. Ren, P.-H. Li, Y. Liu, H.
V. Setaluri, Role of dual specificity phosphatases (DUSPs) in
Tang, R. Sun, C. Li, B. Bai, W. Jia, Y. Liu, W. Zhang, L. Yang,
melanoma cellular plasticity and drug resistance, Sci. Rep. 12
Y. Peng, L. Dai, H. Hu, Y. Jiang, Y. Hu, J. Zhu, H. Jiang, Z.
(2022), 14395. https://doi.org/10.1038/s41598-022-18578-x
Li, C. Caulin, J. Park, and H. Xu, Pan-cancer single-cell anal-
22. R. Iwanaga, C.-A. Wang, D. S. Micalizzi, J. C. Harrell, P.
ysis reveals the heterogeneity and plasticity of cancer-associated
Jedlicka, C. A. Sartorius, P. Kabos, S. M. Farabaugh, A. P.
fibroblasts in the tumor microenvironment, Nat. Commun. 13
Bradford, and H. L. Ford, Expression of Six1 in luminal breast
(2022), 6619. https://doi.org/10.1038/s41467-022-34395-2
cancers predicts poor prognosis and promotes increases in tumor
7. E. Faurobert, A.-P. Bouin, and C. Albiges-Rizo, Microenviron-
initiating cells by activation of extracellular signal-regulated
ment, tumor cell plasticity, and cancer, Curr. Opin. Oncol. 27
kinase and transforming growth factor-beta signaling pathways,
(2015), 64–70. https://doi.org/10.1097/cco.0000000000000154
Breast Cancer Res. 14 (2012), 1–14.
8. R. Zhang, J. Tu, and S. Liu, Novel molecular regulators of breast
23. S. Janta, K. Pranweerapaiboon, P. Vivithanaporn, A.
cancer stem cell plasticity and heterogeneity, Semin. Cancer
Plubrukarn, A. Chairoungdua, P. Prasertsuksri, S.
Biol. 82 (2022), 11–25, https://doi.org/10.1016/j.semcancer.2021.
Apisawetakan, and K. Chaithirayanon, Holothurin A inhibits
03.008
RUNX1-enhanced EMT in metastasis prostate cancer via the
9. X. Wang, and J. P. Thiery, Harnessing carcinoma cell plasticity
Akt/JNK and P38 MAPK signaling pathway, Mar. Drugs. 21
mediated by TGF-β signaling, Cancers. 13 (2021), 3397.
(2023), 345.
10. T.-A. Moo, R. Sanford, C. Dang, and M. Morrow, Overview of
24. S. Zhan, T. Wang, J. Li, H. Zhu, W. Ge, and J. Li, Asporin inter-
breast cancer therapy, PET Clin. 13 (2018), 339–354. https://doi.
acts with HER2 to promote thyroid cancer metastasis via the
org/10.1016/j.cpet.2018.02.006
MAPK/EMT signaling pathway, Front. Oncol. 12 (2022), 762180.
11. B. Mansoori, A. Mohammadi, S. Davudian, S. Shirjang, and B.
25. R. Al-Mahdi, N. Babteen, K. Thillai, M. Holt, B. Johansen,
Baradaran, The different mechanisms of cancer drug resistance:
H. L. Wetting, O.-M. Seternes, and C. M. Wells, A novel role
A brief review, Adv. Pharm. Bull. 7 (2017), 339–348. https://doi.
for atypical MAPK kinase ERK3 in regulating breast cancer cell
org/10.15171/apb.2017.041
morphology and migration, Cell Adh. Migr. 9 (2015), 483–494.
12. S. Shen, and J. Clairambault, Cell plasticity in cancer cell pop-
https://doi.org/10.1080/19336918.2015.1112485
ulations, F1000Res. 9 (2020), F1000. https://doi.org/10.12688/
26. T. Ray, T. Ryusaki, and P. S. Ray, Therapeutically targeting
f1000research.24803.1
cancers that overexpress FOXC1: A transcriptional driver of cell
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
12 of 15 CHATTERJEE et al.

plasticity, partial EMT, and cancer metastasis, Front. Oncol. 11 tein 3 facilitates breast cancer progression via activation of the
(2021), 721959. https://doi.org/10.3389/fonc.2021.721959 PTEN/PI3K/AKT/mTOR signaling, Bioengineered. 13 (2022),
27. S. Diazzi, S. Tartare-Deckert, and M. Deckert, The mechanical 8478–8489. https://doi.org/10.1080/21655979.2022.2051687
phenotypic plasticity of melanoma cell: An emerging driver of 40. H. Hu, J. Zhu, Y. Zhong, R. Geng, Y. Ji, Q. Guan, C. Hong, Y.
therapy cross-resistance, Oncogenesis. 12 (2023), 7. https://doi. Wei, N. Min, A. Qi, Y. Zhang, and X. Li, PIK3CA mutation con-
org/10.1038/s41389-023-00452-8 fers resistance to chemotherapy in triple-negative breast cancer by
28. M. Sala, N. Allain, M. Moreau, A. Jabouille, E. Henriet, A. inhibiting apoptosis and activating the PI3K/AKT/mTOR signal-
Abou-Hammoud, A. Uguen, S. Di-Tommaso, C. Dourthe, A.- ing pathway, Ann. Transl. Med. 9 (2021), 410. https://doi.org/10.
A. Raymond, J.-W. Dupuy, E. Gerard, N. Dugot-Senant, B. 21037/atm-21-698
Rousseau, J.-P. Merlio, A. Pham-Ledart, B. Vergier, S. Tartare- 41. H. Ghebeh, S. Al-Khaldi, S. Olabi, A. Al-Dhfyan, F. Al-
Deckert, V. Moreau, and F. Saltel, Discoidin domain receptor 2 Mohanna, R. Barnawi, A. Tulbah, T. Al-Tweigeri, D. Ajarim,
orchestrates melanoma resistance combining phenotype switch- and M. Al-Alwan, Fascin is involved in the chemotherapeutic
ing and proliferation, Oncogene. 41 (2022), 2571–2586. https:// resistance of breast cancer cells predominantly via the PI3K/Akt
doi.org/10.1038/s41388-022-02266-1 pathway, Br. J. Cancer. 111 (2014), 1552–1561. https://doi.org/10.
29. M. Valipour, Recent advances of antitumor shikonin/alkannin 1038/bjc.2014.453
derivatives: A comprehensive overview focusing on structural 42. W. Xu, Z. Yang, and N. Lu, A new role for the PI3K/Akt signal-
classification, synthetic approaches, and mechanisms of action, ing pathway in the epithelial-mesenchymal transition, Cell Adh.
Eur. J. Med. Chem. 235 (2022), 114314. https://doi.org/10.1016/j. Migr. 9 (2015), 317–324. https://doi.org/10.1080/19336918.2015.
ejmech.2022.114314 1016686
30. W. Chen, K. L. Kang, A. Alshaikh, S. Varma, Y.-L. Lin, K.- 43. M. Cai, H. Li, R. Chen, and X. Zhou, MRPL13 promotes tumor
H. Shin, R. Kim, C.-Y. Wang, N.-H. Park, K. Walentin, K. cell proliferation, migration and EMT process in breast cancer
M. Schmidt-Ott, and M. K. Kang, Grainyhead-like 2 (GRHL2) through the PI3K-AKT-mTOR pathway, Cancer Manag. Res. 13
knockout abolishes oral cancer development through reciprocal (2021), 2009–2024. https://doi.org/10.2147/cmar.S296038
regulation of the MAP kinase and TGF-β signaling pathways, 44. D.-P. Wu, Y. Zhou, L.-X. Hou, X.-X. Zhu, W. Yi, S.-M. Yang, T.-
Oncogenesis. 7 (2018), 38. https://doi.org/10.1038/s41389-018- Y. Lin, J.-L. Huang, B. Zhang, and X.-X. Yin, Cx43 deficiency
0047-5 confers EMT-mediated tamoxifen resistance to breast cancer via
31. H. Lu, L. Tran, Y. Park, I. Chen, J. Lan, Y. Xie, and G. L. c-Src/PI3K/Akt pathway, Int. J. Biol. Sci. 17 (2021), 2380–2398.
Semenza, Reciprocal regulation of DUSP9 and DUSP16 expres- https://doi.org/10.7150/ijbs.55453
sion by HIF1 controls ERK and p38 MAP kinase activity and 45. D.-P. Wu, Y. Zhou, L.-X. Hou, X.-X. Zhu, W. Yi, S.-M. Yang, T.-
mediates chemotherapy-induced breast cancer stem cell enrich- Y. Lin, J.-L. Huang, B. Zhang, and X.-X. Yin, Cx43 deficiency
ment, Cancer Res. 78 (2018), 4191–4202. https://doi.org/10.1158/ confers EMT-mediated tamoxifen resistance to breast cancer via
0008-5472.Can-18-0270 c-Src/PI3K/Akt pathway, Int. J. Biol. Sci. 17 (2021), 2380–2398.
32. S. Shah, E. J. Brock, R. M. Jackson, K. Ji, J. L. Boerner, B. https://doi.org/10.7150/ijbs.55453
F. Sloane, and R. R. Mattingly, Downregulation of rap1gap: A 46. J. Luo, J.-F. Yao, X.-F. Deng, X.-D. Zheng, M. Jia, Y.-Q. Wang,
switch from DCIS to invasive breast carcinoma via ERK/MAPK Y. Huang, and J.-H. Zhu, 14, 15-EET induces breast cancer cell
activation, Neoplasia. 20 (2018), 951–963. https://doi.org/10. EMT and cisplatin resistance by up-regulating integrin αvβ3 and
1016/j.neo.2018.07.002 activating FAK/PI3K/AKT signaling, J. Exp. Clin. Cancer Res.
33. M. Wu, and H. Zhao, Analysis of key genes and pathways in 37 (2018), 23. https://doi.org/10.1186/s13046-018-0694-6
breast ductal carcinoma in situ, Oncol. Lett. 20 (2020), 217. 47. C. Guo, S. Li, A. Liang, M. Cui, Y. Lou, and H. Wang, PPA1
https://doi.org/10.3892/ol.2020.12080 promotes breast cancer proliferation and metastasis through
34. M. Below, and C. Osipo, Notch signaling in breast cancer: A role PI3K/AKT/GSK3β signaling pathway, Front. Cell Dev. Biol. 9
in drug resistance, Cells. 9 (2020), 2204. (2021), 730558. https://doi.org/10.3389/fcell.2021.730558
35. X. Shi, J. Wang, Y. Lei, C. Cong, D. Tan, and X. Zhou, Research 48. Y. Zhang, J. L. Liu, and J. Wang, KRAS gene silencing inhibits
progress on the PI3K/AKT signaling pathway in gynecological the activation of PI3K-Akt-mTOR signaling pathway to regulate
cancer (Review), Mol. Med. Rep. 19 (2019), 4529–4535. https:// breast cancer cell epithelial-mesenchymal transition, prolifera-
doi.org/10.3892/mmr.2019.10121 tion and apoptosis, Riv. Eur. Sci. Med. Farmacol. 24 (2020),
36. B. Vanhaesebroeck, L. Stephens, and P. Hawkins, PI3K sig- 3085–3096. https://doi.org/10.26355/eurrev_202003_20673
nalling: The path to discovery and understanding, Nat. Rev. Mol. 49. J.-H. Ma, L. Qin, and X. Li, Role of STAT3 signaling pathway in
Cell Biol. 13 (2012), 195–203. https://doi.org/10.1038/nrm3290 breast cancer, Cell Commun. Signal. 18 (2020), 33. https://doi.
37. M. Beyens, T. Vandamme, M. Peeters, G. Van Camp, and K. Op org/10.1186/s12964-020-0527-z
De Beeck, Resistance to targeted treatment of gastroenteropan- 50. P. A. Johnston, and J. R. Grandis, STAT3 signaling: Anticancer
creatic neuroendocrine tumors, Endocr. Relat. Cancer. 26 (2019), strategies and challenges, Mol. Interv. 11 (2011), 18–26. https://
R109–R130. https://doi.org/10.1530/erc-18-0420 doi.org/10.1124/mi.11.1.4
38. A. Nicolini, P. Ferrari, and M. J. Duffy, Prognostic and pre- 51. S. Hashimoto, A. Hashimoto, R. Muromoto, Y. Kitai, K.
dictive biomarkers in breast cancer: Past, present and future, Oritani, and T. Matsuda, Central roles of STAT3-mediated sig-
Semin. Cancer Biol. 52 (2017), 56–73. https://doi.org/10.1016/j. nals in onset and development of cancers: Tumorigenesis and
semcancer.2017.08.010 immunosurveillance, Cells. 11 (2022), 2618.
39. C.-H. Zhan, D.-S. Ding, W. Zhang, H.-L. Wang, Z.-Y. Mao, 52. A. B. D’assoro, Abstract 3163: PR/STAT3 nuclear transcriptional
and G.-J. Liu, The cancer-testis antigen a-kinase anchor pro- complexes mediate aurora-A kinase-induced stemness plasticity
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CHATTERJEE et al. 13 of 15

in ER+ breast cancer, Cancer Res. 82 (2022), 3163–3163. https:// enchymal glioblastoma, Oncogene. 39 (2020), 6990–7004.
doi.org/10.1158/1538-7445.Am2022-3163 https://doi.org/10.1038/s41388-020-01506-6
53. A. N. Gargalionis, K. A. Papavassiliou, and A. G. Papavassiliou, 64. T. Srinivasan, J. Walters, P. Bu, E. B. Than, K.-L. Tung, K.-
Targeting STAT3 signaling pathway in colorectal cancer, Y. Chen, N. Panarelli, J. Milsom, L. Augenlicht, S. M. Lipkin,
Biomedicines. 9 (2021), 1016. and X. Shen, NOTCH signaling regulates asymmetric cell fate of
54. P. Polakis, Wnt signaling in cancer, Cold Spring Harb. Perspect. fast- and slow-cycling colon cancer–initiating cells, Cancer Res.
Biol. 4 (2012), no. 5, a008052–a008052. https://doi.org/10.1101/ 76 (2016), 3411–3421. https://doi.org/10.1158/0008-5472.Can-15-
cshperspect.a008052 3198
55. M. Katoh, and M. Katoh, WNT signaling and cancer stemness, 65. A. W. Fender, J. M. Nutter, T. L. Fitzgerald, F. E. Bertrand,
Essays Biochem. 66 (2022), 319–331. https://doi.org/10.1042/ and G. Sigounas, Notch-1 promotes stemness and epithe-
ebc20220016 lial to mesenchymal transition in colorectal cancer, J. Cell.
56. J. Guinney, R. Dienstmann, X. Wang, A. De Reyniès, Biochem. 116 (2015), 2517–2527. https://doi.org/10.1002/jcb.
A. Schlicker, C. Soneson, L. Marisa, P. Roepman, G. 25196
Nyamundanda, P. Angelino, B. M. Bot, J. S. Morris, I. M. 66. T. Ishida, H. Hijioka, K. Kume, A. Miyawaki, and N. Nakamura,
Simon, S. Gerster, E. Fessler, F. De Sousa E Melo, E. Missiaglia, Notch signaling induces EMT in OSCC cell lines in a hypoxic
H. Ramay, D. Barras, K. Homicsko, D. Maru, G. C. Manyam, environment, Oncol. Lett. 6 (2013), 1201–1206, https://doi.org/
B. Broom, V. Boige, B. Perez-Villamil, T. Laderas, R. Salazar, J. 10.3892/ol.2013.1549
W. Gray, D. Hanahan, J. Tabernero, R. Bernards, S. H. Friend, 67. K. O. Alfarouk, C.-M. Stock, S. Taylor, M. Walsh, A. K.
P. Laurent-Puig, J. P. Medema, A. Sadanandam, L. Wessels, Muddathir, D. Verduzco, A. H. H. Bashir, O. Y. Mohammed,
M. Delorenzi, S. Kopetz, L. Vermeulen, and S. Tejpar, The G. O. Elhassan, S. Harguindey, S. J. Reshkin, M. E. Ibrahim,
consensus molecular subtypes of colorectal cancer, Nat. Med. 21 and C. Rauch, Resistance to cancer chemotherapy: Failure in
(2015), 1350–1356. https://doi.org/10.1038/nm.3967 drug response from ADME to P-gp, Cancer Cell Int. 15 (2015),
57. S. Papanikolaou, A. Vourda, S. Syggelos, and K. Gyftopoulos, 71. https://doi.org/10.1186/s12935-015-0221-1
Cell plasticity and prostate cancer: The role of epithelial– 68. C. Sheridan, H. Kishimoto, R. K. Fuchs, S. Mehrotra, P. Bhat-
mesenchymal transition in tumor progression, invasion, metas- Nakshatri, C. H. Turner, R. Goulet, S. Badve, and H. Nakshatri,
tasis and cancer therapy resistance, Cancers. 13 (2021), 2795. CD44+/CD24- breast cancer cells exhibit enhanced invasive
58. Q. Li, D. Xia, Z. Wang, B. Liu, J. Zhang, P. Peng, Q. Tang, J. properties: An early step necessary for metastasis, Breast Cancer
Dong, J. Guo, D. Kuang, W. Chen, J. Mao, Q. Li, and X. Chen, Res. 8 (2006), R59. https://doi.org/10.1186/bcr1610
Circadian rhythm gene PER3 negatively regulates stemness of 69. S. Ricardo, A. F. Vieira, R. Gerhard, D. Leitao, R. Pinto, J.
prostate cancer stem cells via WNT/β-catenin signaling in tumor F. Cameselle-Teijeiro, F. Milanezi, F. Schmitt, and J. Paredes,
microenvironment, Front. Cell Dev. Biol. 9 (2021), 656981. Breast cancer stem cell markers CD44, CD24 and ALDH1:
59. T. Zhan, G. Ambrosi, A. M. Wandmacher, B. Rauscher, J. Betge, Expression distribution within intrinsic molecular subtype, J.
N. Rindtorff, R. S. Häussler, I. Hinsenkamp, L. Bamberg, B. Clin. Pathol. 64 (2011), 937–946. https://doi.org/10.1136/jcp.
Hessling, K. Müller-Decker, G. Erdmann, E. Burgermeister, 2011.090456
M. P. Ebert, and M. Boutros, MEK inhibitors activate Wnt sig- 70. A. Jaggupilli, and E. Elkord, Significance of CD44 and CD24
nalling and induce stem cell plasticity in colorectal cancer, Nat. as cancer stem cell markers: An enduring ambiguity, Clin. Dev.
Commun. 10 (2019), 2197. https://doi.org/10.1038/s41467-019- Immunol. 2012 (2012), 708036. https://doi.org/10.1155/2012/
09898-0 708036
60. R. Zhu, O. Gires, L. Zhu, J. Liu, J. Li, H. Yang, G. Ju, J. Huang, 71. X. Qiao, Y. Zhang, L. Sun, Q. Ma, J. Yang, L. Ai, J. Xue, G. Chen,
W. Ge, Y. Chen, Z. Lu, and H. Wang, TSPAN8 promotes can- H. Zhang, C. Ji, X. Gu, H. Lei, Y. Yang, and C. Liu, Associ-
cer cell stemness via activation of sonic Hedgehog signaling, Nat. ation of human breast cancer CD44-/CD24- cells with delayed
Commun. 10 (2019), 2863. https://doi.org/10.1038/s41467-019- distant metastasis, eLife. 10 (2021), e65418. https://doi.org/10.
10739-3 7554/eLife.65418
61. A. S. Cazet, M. N. Hui, B. L. Elsworth, S. Z. Wu, D. Roden, C.-L. 72. I. Malanchi, A. Santamaria-Martínez, E. Susanto, H. Peng, H.-
Chan, J. N. Skhinas, R. Collot, J. Yang, K. Harvey, M. Z. Johan, A. Lehr, J.-F. Delaloye, and J. Huelsken, Interactions between
C. Cooper, R. Nair, D. Herrmann, A. Mcfarland, N. Deng, M. cancer stem cells and their niche govern metastatic colonization,
Ruiz-Borrego, F. Rojo, J. M. Trigo, S. Bezares, R. Caballero, E. Nature. 481 (2012), 85–89.
Lim, P. Timpson, S. O’toole, D. N. Watkins, T. R. Cox, M. S. 73. H. W. Huth, T. Castro-Gomes, A. M. De Goes, and C. Ropert,
Samuel, M. Martín, and A. Swarbrick, Targeting stromal remod- Translocation of intracellular CD24 constitutes a triggering event
eling and cancer stem cell plasticity overcomes chemoresistance for drug resistance in breast cancer, Sci. Rep. 11 (2021), 17077.
in triple negative breast cancer, Nat. Commun. 9 (2018), 2897. https://doi.org/10.1038/s41598-021-96449-7
https://doi.org/10.1038/s41467-018-05220-6 74. J. E. Jiménez-Salazar, R. Damian-Ferrara, M. Arteaga, N.
62. S. Sarkar, R. Mirzaei, F. J. Zemp, W. Wei, D. L. Senger, S. Batina, and P. Damián-Matsumura, Non-genomic actions of
M. Robbins, and V. W. Yong, Activation of NOTCH signaling estrogens on the DNA repair pathways are associated with
by tenascin-c promotes growth of human brain tumor-initiating chemotherapy resistance in breast cancer, Front. Oncol. 11
cells, Cancer Res. 77 (2017), 3231–3243. https://doi.org/10.1158/ (2021), 631007. https://doi.org/10.3389/fonc.2021.631007
0008-5472.Can-16-2171 75. A. Mal, A. B. Bukhari, R. K. Singh, A. Kapoor, A. Barai,
63. I. Angel, O. Pilo Kerman, L. Rousso-Noori, and D. Friedmann- I. Deshpande, T. Wadasadawala, P. Ray, S. Sen, and A.
Morvinski, Tenascin C promotes cancer cell plasticity in mes- De, EpCAM-mediated cellular plasticity promotes radiation
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
14 of 15 CHATTERJEE et al.

resistance and metastasis in breast cancer, Front. Cell Dev. Biol. survival pathway, Nat. Commun. 12 (2021), 432. https://doi.org/
8 (2020), 597673. https://doi.org/10.3389/fcell.2020.597673 10.1038/s41467-020-20664-5
76. K. M. Arnold, L. M. Opdenaker, N. J. Flynn, D. K. Appeah, 87. M. Below, and C. Osipo, Notch signaling in breast cancer: A role
and J. Sims-Mourtada, Radiation induces an inflammatory in drug resistance, Cells. 9 (2021), no. 10, 2204. https://doi.org/
response that results in STAT3-dependent changes in cellular 10.3390/cells9102204
plasticity and radioresistance of breast cancer stem-like cells, 88. K. G. Leong, K. Niessen, I. Kulic, A. Raouf, C. Eaves,
Int. J. Radiat. Biol. 96 (2020), 434–447. https://doi.org/10.1080/ I. Pollet, and A. Karsan, Jagged1-mediated Notch activa-
09553002.2020.1705423 tion induces epithelial-to-mesenchymal transition through Slug-
77. S. Ahmed, H. T. Mohamed, N. El-Husseiny, M. M. El Mahdy, induced repression of E-cadherin, J. Exp. Med. 204 (2007),
G. Safwat, A. A. Diab, A. A. El-Sherif, M. El-Shinawi, and M. M. 2935–2948. https://doi.org/10.1084/jem.20071082
Mohamed, IL-8 secreted by tumor associated macrophages con- 89. M. Wada, M. Horinaka, T. Yamazaki, N. Katoh, and T. Sakai,
tribute to lapatinib resistance in HER2-positive locally advanced The dual RAF/MEK inhibitor CH5126766/RO5126766 may be
breast cancer via activation of Src/STAT3/ERK1/2-mediated a potential therapy for RAS-mutated tumor cells, PLoS One. 9
EGFR signaling, Biochim. Biophys. Acta. Mol. Cell Res. 1868 (2014), e113217. https://doi.org/10.1371/journal.pone.0113217
(2021), 118995. https://doi.org/10.1016/j.bbamcr.2021.118995 90. V. T. Hoang, M. D. Matossian, J. La, K. Hoang, D. A. Ucar,
78. L. Lu, J. Dong, L. Wang, Q. Xia, D. Zhang, H. Kim, T. Yin, S. S. Elliott, H. E. Burks, T. D. Wright, S. Patel, A. Bhatt, T.
Fan, and Q. Shen, Activation of STAT3 and Bcl-2 and reduction Phamduy, D. Chrisey, A. Buechlein, D. B. Rusch, K. P. Nephew,
of reactive oxygen species (ROS) promote radioresistance in breast M. Anbalagan, B. Rowan, J. E. Cavanaugh, P. T. Flaherty, L.
cancer and overcome of radioresistance with niclosamide, Onco- Miele, B. M. Collins-Burow, and M. E. Burow, Dual inhibi-
gene. 37 (2018), 5292–5304. https://doi.org/10.1038/s41388-018- tion of MEK1/2 and MEK5 suppresses the EMT/migration axis
0340-y in triple-negative breast cancer through FRA-1 regulation, J. Cell.
79. L. Ng, P. Kaur, N. Bunnag, J. Suresh, I. Sung, Q. Tan, J. Gruber, Biochem. 122 (2021), 835–850. https://doi.org/10.1002/jcb.29916
and N. Tolwinski, WNT signaling in disease, Cells. 8 (2019), no. 91. C. D. Mohan, Y. Y. Liew, Y. Y. Jung, S. Rangappa, H. D.
8, 826. https://doi.org/10.3390/cells8080826 Preetham, A. Chinnathambi, T. A. Alahmadi, S. A. Alharbi, Z.-
80. L. T. H. Phi, I. N. Sari, Y.-G. Yang, S.-H. Lee, N. Jun, K. S. Kim, X. Lin, K. S. Rangappa, and K. S. Ahn, Brucein D modulates
Y. K. Lee, and H. Y. Kwon, Cancer stem cells (CSCs) in drug MAPK signaling cascade to exert multi-faceted anti-neoplastic
resistance and their therapeutic implications in cancer treatment, actions against breast cancer cells, Biochimie. 182 (2021), 140–
Stem Cells Int. 2018 (2018), 5416923. https://doi.org/10.1155/ 151. https://doi.org/10.1016/j.biochi.2021.01.009
2018/5416923 92. H. Li, L. Prever, E. Hirsch, and F. Gulluni, Targeting
81. W. A. Abreu De Oliveira, S. Moens, Y. El Laithy, B. K. Van Der PI3K/AKT/mTOR signaling pathway in breast cancer, Cancers.
Veer, P. Athanasouli, E. E. Cortesi, M. F. Baietti, K. P. Koh, J.- 13 (2021), 3517.
J. Ventura, F. Amant, D. Annibali, and F. Lluis, Wnt/β-Catenin 93. J. Huang, C. Liu, S. Duan, J. Lin, Y. Luo, S. Tao, S. Xing, X.
inhibition disrupts carboplatin resistance in isogenic models of Zhang, H. Du, H. Wang, C. Huang, and G. Wei, Gigantol inhibits
triple-negative breast cancer, Front. Oncol. 11 (2021), 705384. proliferation and enhances DDP-induced apoptosis in breast-
https://doi.org/10.3389/fonc.2021.705384 cancer cells by downregulating the PI3K/Akt/mTOR signaling
82. H. Wan, Z. Li, H. Wang, F. Cai, and L. Wang, ST8SIA1 inhibi- pathway, Life Sci. 274 (2021), 119354. https://doi.org/10.1016/j.
tion sensitizes triple negative breast cancer to chemotherapy via lfs.2021.119354
suppressing Wnt/β-catenin and FAK/Akt/mTOR, Clin. Transl. 94. Y. Ma, X. Yang, H. Han, Z. Wen, M. Yang, Y. Zhang, J.
Oncol. 23 (2021), 902–910. https://doi.org/10.1007/s12094-020- Fu, X. Wang, T. Yin, G. Lu, J. Qi, H. Lin, X. Wang, and
02484-7 Y. Yang, Design, synthesis and biological evaluation of anilide
83. J. Y. Chai, V. Sugumar, A. F. Alshanon, W. F. Wong, S. Y. Fung, (dicarboxylic acid) shikonin esters as antitumor agents through
and C. Y. Looi, Defining the role of GLI/Hedgehog signaling in targeting PI3K/Akt/mTOR signaling pathway, Bioorg. Chem.
chemoresistance: Implications in therapeutic approaches, Can- 111 (2021), 104872. https://doi.org/10.1016/j.bioorg.2021.104872
cers (Basel). 13 (2021), no. 19, 4746. https://doi.org/10.3390/ 95. K. Zhu, X. Liu, C. Liu, Y. Xu, Y. Fu, W. Dong, Y. Yan, W. Wang,
cancers13194746 and C. Qian, AKT inhibitor AZD5363 suppresses stemness and
84. M. Prieto-Vila, R.-U. Takahashi, W. Usuba, I. Kohama, and T. promotes anti-cancer activity of 3,3’-diindolylmethane in human
Ochiya, Drug resistance driven by cancer stem cells and their breast cancer cells, Toxicol. Appl. Pharmacol. 429 (2021), 115700.
niche, Int. J. Mol. Sci. 18 (2017), 2574. https://doi.org/10.1016/j.taap.2021.115700
85. P. Magistri, C. Battistelli, R. Strippoli, N. Petrucciani, T. 96. Z. Kohandel, T. Farkhondeh, M. Aschner, A. M. Pourbagher-
Pellinen, L. Rossi, L. Mangogna, P. Aurello, F. D’angelo, M. Shahri, and S. Samarghandian, STAT3 pathway as a molecular
Tripodi, G. Ramacciato, and G. Nigri, SMO inhibition mod- target for resveratrol in breast cancer treatment, Cancer Cell Int.
ulates cellular plasticity and invasiveness in colorectal cancer, 21 (2021), 468. https://doi.org/10.1186/s12935-021-02179-1.
Front. Pharmacol. 8 (2017), 956. https://doi.org/10.3389/fphar. 97. M. Behroozaghdam, M. Dehghani, A. Zabolian, D. Kamali, S.
2017.00956 Javanshir, F. Hasani Sadi, M. Hashemi, T. Tabari, M. Rashidi, S.
86. S. Kumar, A. Nandi, S. Singh, R. Regulapati, N. Li, J. W. Tobias, Mirzaei, A. Zarepour, A. Zarrabi, D. De Greef, and A. Bishayee,
C. W. Siebel, M. A. Blanco, A. J. Klein-Szanto, C. Lengner, A. Resveratrol in breast cancer treatment: From cellular effects to
L. Welm, Y. Kang, and R. Chakrabarti, Dll1(+) quiescent tumor molecular mechanisms of action, Cell. Mol. Life Sci. 79 (2022),
stem cells drive chemoresistance in breast cancer through NF-κB 539. https://doi.org/10.1007/s00018-022-04551-4
26899655, 2023, 3, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/cso2.1051 by Nat Prov Indonesia, Wiley Online Library on [19/05/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
CHATTERJEE et al. 15 of 15

98. A. Hosseinzadeh, P. Merikhian, N. Naseri, M. R. Eisavand, T. J. Ley, C. A. Maher, and R. C. Fields, The clonal evolution of
and L. Farahmand, MUC1 is a potential target to overcome metastatic colorectal cancer, Sci. Adv. 6 (2020), eaay9691.
trastuzumab resistance in breast cancer therapy, Cancer Cell Int. 112. H. D. Jung, Y. J. Sung, and H. U. Kim, Omics and computational
22 (2022), 110. https://doi.org/10.1186/s12935-022-02523-z modeling approaches for the effective treatment of drug-resistant
99. Z. Liu, X. Ge, Y. Gu, Y. Huang, H. Liu, M. Yu, and Y. Liu, cancer cells, Front. Genet. 12 (2021), 742902, https://doi.org/10.
Small molecule STAT3 inhibitor, 6Br-6a suppresses breast cancer 3389/fgene.2021.742902
growth in vitro and in vivo, Biomed. Pharmacother. 121 (2020), 113. P. Juárez, and T. A. Guise, TGF-β in cancer and bone: Implica-
109502. https://doi.org/10.1016/j.biopha.2019.109502 tions for treatment of bone metastases, Bone. 48 (2011), 23–29.
100. D. Barua, and W. S. Hlavacek, Modeling the effect of APC trun- https://doi.org/10.1016/j.bone.2010.08.004
cation on destruction complex function in colorectal cancer cells, 114. T. A. Guise, K. S. Mohammad, G. Clines, E. G. Stebbins, D. H.
PLoS Comput. Biol. 9 (2013), e1003217. https://doi.org/10.1371/ Wong, L. S. Higgins, R. Vessella, E. Corey, S. Padalecki, L. Suva,
journal.pcbi.1003217 and J. M. Chirgwin, Basic mechanisms responsible for osteolytic
101. Z. Zhong, and D. M. Virshup, Wnt signaling and drug resistance and osteoblastic bone metastases, Clin. Cancer Res. 12 (2006),
in cancer, Mol. Pharmacol. 97 (2020), 72–89. https://doi.org/10. 6213s–6216s. https://doi.org/10.1158/1078-0432.Ccr-06-1007
1124/mol.119.117978 115. L. M. Cook, A. Araujo, J. M. Pow-Sang, M. M. Budzevich,
102. N. M. Nguyen, and J. Cho, Hedgehog pathway inhibitors as tar- D. Basanta, and C. C. Lynch, Predictive computational mod-
geted cancer therapy and strategies to overcome drug resistance, eling to define effective treatment strategies for bone metastatic
Int. J. Mol. Sci. 23 (2022), no. 3, 1733. https://doi.org/10.3390/ prostate cancer, Sci. Rep. 6 (2016), 29384. https://doi.org/10.
ijms23031733 1038/srep29384
103. A.-N. Tsao, Y.-S. Chuang, Y.-C. Lin, Y. Su, and T.-C. Chao, 116. M. H. Zangooei, R. Margolis, and K. Hoyt, Multiscale compu-
Dinaciclib inhibits the stemness of two subtypes of human breast tational modeling of cancer growth using features derived from
cancer cells by targeting the FoxM1 and Hedgehog signaling path- microCT images, Sci. Rep. 11 (2021), 18524. https://doi.org/10.
way, Oncol. Rep. 47 (2022), no. 5, 105. https://doi.org/10.3892/ 1038/s41598-021-97966-1
or.2022.8316 117. S. Gu, S. Lheureux, A. Sayad, P. Cybulska, L. Hogen, I.
104. K. Brennan, and R. B. Clarke, Combining Notch inhibition with Vyarvelska, D. Tu, W. R. Parulekar, M. Nankivell, S. Kehoe,
current therapies for breast cancer treatment, Ther. Adv. Med. D. S. Chi, D. A. Levine, M. Q. Bernardini, B. Rosen, A. Oza,
Oncol. 5 (2013), 17–24. https://doi.org/10.1177/175883401245 M. Brown, and B. G. Neel, Computational modeling of ovar-
7437 ian cancer dynamics suggests optimal strategies for therapy
105. A. Jaiswal, K. Murakami, A. Elia, Y. Shibahara, S. J. Done, S. A. and screening, Proc. Nat. Acad. Sci. U.S.A. 118 (2021), no. 25,
Wood, N. J. Donato, P. S. Ohashi, and M. Reedijk, Therapeutic e2026663118. https://doi.org/10.1073/pnas.2026663118
inhibition of USP9x-mediated Notch signaling in triple-negative 118. J. M. Greene, J. L. Gevertz, and E. D. Sontag, Mathematical
breast cancer, Proc. Natl. Acad. Sci. 118 (2021), e2101592118. approach to differentiate spontaneous and induced evolution
https://doi.org/10.1073/pnas.2101592118 to drug resistance during cancer treatment, JCO Clin. Cancer
106. S. R. Torborg, Z. Li, J. E. Chan, and T. Tammela, Cellular and Inform. 3 (2019), 1–20. https://doi.org/10.1200/cci.18.00087
molecular mechanisms of plasticity in cancer, Trends Cancer. 8 119. K. S. Prajapati, S. Gupta, and S. Kumar, Targeting breast cancer-
(2022), 735–746. https://doi.org/10.1016/j.trecan.2022.04.007 derived stem cells by dietary phytochemicals: A strategy for cancer
107. S. Qin, J. Jiang, Y. Lu, E. C. Nice, C. Huang, J. Zhang, and W. prevention and treatment, Cancers (Basel). 14 (2022), no. 12,
He, Emerging role of tumor cell plasticity in modifying therapeu- 2864. https://doi.org/10.3390/cancers14122864
tic response, Signal Transduction Targeted Ther. 5 (2020), 228. 120. W.-C. Yen, M. M. Fischer, F. Axelrod, C. Bond, J. Cain, B.
https://doi.org/10.1038/s41392-020-00313-5 Cancilla, W. R. Henner, R. Meisner, A. Sato, J. Shah, T. Tang,
108. F. Lüönd, S. Tiede, and G. Christofori, Breast cancer as an B. Wallace, M. Wang, C. Zhang, A. M. Kapoun, J. Lewicki,
example of tumour heterogeneity and tumour cell plasticity dur- A. Gurney, and T. Hoey, Targeting notch signaling with a
ing malignant progression, Br. J. Cancer. 125 (2021), 164–175. notch2/notch3 antagonist (tarextumab) inhibits tumor growth
https://doi.org/10.1038/s41416-021-01328-7 and decreases tumor-initiating cell frequency, Clin. Cancer Res.
109. H. Hayashi, E. Nadal, J. E. Gray, A. Ardizzoni, N. Caria, T. 21 (2015), 2084–2095. https://doi.org/10.1158/1078-0432.Ccr-14-
Puri, and C. Grohe, Overall treatment strategy for patients with 2808
metastatic NSCLC with activating EGFR mutations, Clin. Lung
Cancer. 23 (2022), e69–e82.
110. A. S, A. Chakraborty, and S. Patnaik, Clonal evolution and
expansion associated with therapy resistance and relapse of col- How to cite this article: N. Chatterjee, B.
orectal cancer, Mutat Res. Rev. Mutat. Res. 790 (2022), 108445, Pulipaka, A. R. Subbalakshmi, M. K. Jolly, and R.
https://doi.org/10.1016/j.mrrev.2022.108445
Nair, Unraveling the dangerous duet between cancer
111. H. X. Dang, B. A. Krasnick, B. S. White, J. G. Grossman, M. S.
Strand, J. Zhang, C. R. Cabanski, C. A. Miller, R. S. Fulton, S.
cell plasticity and drug resistance, Comput Syst
P. Goedegebuure, C. C. Fronick, M. Griffith, D. E. Larson, B. Oncol. 3 (2023), e1051.
D. Goetz, J. R. Walker, W. G. Hawkins, S. M. Strasberg, D. C. https://doi.org/10.1002/cso2.1051
Linehan, K. H. Lim, A. C. Lockhart, E. R. Mardis, R. K. Wilson,

You might also like