Abstract
Breast cancer (BC) is the most prevalent cancer in women globally. Anti-cancer advancements have enabled the killing of BC cells through various therapies; however, cancer relapse is still a major limitation and decreases patient survival and quality of life. Epithelial-to-mesenchymal transition (EMT) is responsible for tumor relapse in several cancers. This highly regulated event causes phenotypic, genetic, and epigenetic changes in the tumor microenvironment (TME). This review summarizes the recent advancements regarding EMT using de-differentiation and partial EMT theories. We extensively review the mechanistic pathways, TME components, and various anti-cancer adjuvant and neo-adjuvant therapies responsible for triggering EMT in BC tumors. Information regarding essential clinical studies and trials is also discussed. Furthermore, we also highlight the recent strategies targeting various EMT pathways. This review provides a holistic picture of BC biology, molecular pathways, and recent advances in therapeutic strategies.
Keywords: Breast cancer, Epithelial-to-mesenchymal transition, Metastases, Cancer stem cells, Epigenetics
Graphical abstract
Highlights
-
•
Epithelial-to-mesenchymal transition (EMT) induces phenotypic changes necessary for tumor cell survival and progression.
-
•
Other cell populations, such as fibroblasts, immune and hypoxic cells, introduce EMT-promoting factors.
-
•
Stress-related stimuli such as hypoxia, chemotherapy, and nutrient deprivation promote EMT in tumors.
Introduction
Breast cancer (BC) is the most common type of cancer diagnosed in women. In 2020, approximately 2.26 million women were diagnosed with BC, with 685,000 deaths globally. BC leads to the most disability-adjusted life years in women compared to other cancers. By 2040, new BC cases will increase by 31.2%.1,2
BC can arise in the duct epithelium (85%) or the lobules in glandular tissues (15%).3 As with other solid tumors, BC metastasis is primarily responsible for patient mortality.4 Epithelial-to-mesenchymal transition (EMT) is crucial in tumor pathophysiology, where epithelial cells lose their polarity, morphology, and cell–cell adhesion. BC cells gain migratory and invasive phenotypes to transform into mesenchymal cells.5 EMT also induces stem-like phenotypes such as self-renewal, pluripotency, and tumorigenicity to circulating tumor cells (CTCs) responsible for the tumorigenic potential. The tumorigenic potential is essential for CTCs to adapt and develop secondary tumors in a new environment.6 Owing to the massive impact of EMT on BC progression, various clinical trials have been initiated to target EMT in BC progression [Table 1].
Table 1.
Drugs and combinations in various phases of clinical trials to target EMT-mediated tumor progression in breast cancer.
| NCT No. | Title | Therapeutic intervention | Sponsor/collaborators | Phase | Reference |
|---|---|---|---|---|---|
| NCT01861054 | Pilot study to evaluate safety & biological effects of orally administered reparixin in early breast cancer | Reparixin | Dompé Farmaceutici S.p.A | Phase 2 | 7 |
| NCT01952054 | Denosumab for breast cancer with bone mets | Denosumab | M.D. Anderson Cancer Center, Amgen | Phase 2 | 8 |
| NCT02001974 | Pilot study to evaluate reparixin with weekly paclitaxel in patients with HER2-negative metastatic breast cancer (MBC) | Paclitaxel + reparixin | Dompé Farmaceutici S.p.A, PRA Health Sciences | Phase 1 | 9 |
| NCT03285607 | MCS110 combined with neoadjuvant doxorubicin, cyclophosphamide, and weekly paclitaxel in patients with hormone-receptor Positive and HER2- breast cancer | Biological: MCS110, doxorubicin, cyclophosphamide, paclitaxel | Washington University School of Medicine, Novartis Pharmaceuticals | Phase 1 | 10 |
| NCT04664829 | The role of bexarotene in inducing susceptibility to chemotherapy in metastatic TNBC | Bexarotene, capecitabine | National Cancer Centre, Singapore, National Medical Research Council (NMRC), Singapore | Phase 1 | 11 |
| NCT03306472 | A pre-operative window study of letrozole plus PR agonist (megestrol acetate) versus letrozole alone in post-menopausal patients with ER-positive breast cancer | Megestrol acetate, letrozole | Cambridge University Hospitals NHS Foundation Trust, Anticancer Fund, Belgium | Phase 2 | 12 |
| NCT02602938 | Aspirin on CTCs of advanced breast and colorectal cancer | Aspirin | Zhejiang Provincial People's Hospital | Phase 2 | 13 |
CTC: Circulating tumor cells; ER: Estrogen receptor; HER: Human epithelial growth factor receptor; MBC: Metastatic breast cancer; MCS110: Lacnotuzumab; NCT: National Clinical Trial; NHS: National Health Service; NMRC: National Medical Research Council; PR: Progesterone receptor agonist; TNBC: Triple-negative breast cancer.
EMT is responsible for differentiating and de-differentiating cancer cells in the tumor microenvironment (TME). EMT partially dedifferentiates cancer cells from their progenitors, breaking the barriers to differentiate into new cell types.14 During EMT, terminally differentiated cells are reprogrammed to acquire new phenotypes required for invasion, migration, and stem cell-like behavior.15
However, whether EMT leads to CSC development is unknown. Cancer epithelial cells rarely achieve complete mesenchymal phenotypes after EMT and only attain partial mesenchymal characteristics.16 However, EMT transforms epithelial cells to express epithelial and mesenchymal characters17; Grosse-Wilde et al18 reported epithelial and mesenchymal phenotypes in the same cell after EMT. These hybrid cells form during the mesenchymal conversion of epithelial cells during EMT or the epithelial conversion of mesenchymal cells during the mesenchymal-to-epithelial transition (MET). Hybrid cells demonstrate better stemness, self-renewability, and spheroid-forming abilities than completely differentiated epithelial or mesenchymal cells.18 This event is known as partial EMT and is responsible for developing stemness in the cancer cells.19
In this article, we reviewed the role of EMT in tumor progression, metastasis, chemoresistance, and cancer cell differentiation. We also discussed the molecular mechanisms and epigenetic modulations responsible for triggering EMT in BC cells. The phenotypic changes caused by EMT in the cancer cells and the various strategies to inhibit EMT-causing pathways were elucidated. We also discussed the importance of phenotypic changes caused by EMT in tumor progression and metastasis. The role of the TME in triggering metastasis is mechanistically reviewed, and we discuss the role of cancer-associated fibroblasts (CAFs) and immune cells such as macrophages and neutrophils. Additionally, regarding the mechanistic role of EMT in inducing the events above, we summarize recent drugs and potential strategies to inhibit metastasis and EMT-induced chemoresistance in BC.
Phenotypical changes caused by epithelial-to-mesenchymal transition in breast cancer cells
EMT is a reversible event that converts epithelial cells to quasi-mesenchymal cells, causing them to lose their cobble-shaped morphology to become spindle-shaped mesenchymal cells.20 Actin stress fibers, a crucial part of the cellular cytoskeleton, are reorganized to transform the shape into quasi-mesenchymal cells.21 Adherens, tight junctions, gap junctions, and desmosomes tightly hold cells in epithelial sheets. These junctions are formed with the assistance of E-cadherin molecules, and epithelial cells demonstrate apical-basal polarity and interact with the underlying basal membranes via α6β4 integrins and hemidesmosomes.22 EMT-mediated gene alteration includes downregulating genes associated with the epithelial cell phenotypes and upregulating mesenchymal genes. The expression of genes associated with epithelial adhesion proteins such as occludin, E-cadherin, and Traffic jam 1 are underexpressed with subsequent increases in mesenchymal adhesion protein (N-cadherin), fibronectin, and vimentin.23 Basal membrane degradation is necessary for transformed cancer cell migration and dissemination.24
Studies have also supported EMT-related transcription factor (TF)-mediated dedifferentiation of cells. During embryogenesis, Slug expression converts mammary luminal cells to stem cells.25 Twist overexpression in mesenchymal cells prevents their maturation to the cartilage and bones and maintains their immature phenotypes.26 The signals for EMT originate from the tumor stroma surrounding cancer epithelial cells. Signals from the TME induce the expression of EMT-associated TFs in the epithelial cells. These TFs are responsible for developing EMT-associated machinery inside the epithelial cells.27 Paracrine signaling by CAFs28 and macrophages29 induces EMT in primary tumors.27,30
The epithelial phenotype demonstrates high proliferation capabilities, whereas the mesenchymal phenotype demonstrates migration and invasion capabilities. Hence, converting epithelial cells to mesenchymal cells and vice versa is crucial for secondary tumor generation and proliferation.31 Spheroid formation is positively correlated with CSCs and their tumorigenic potential.32 Primary tumors that undergo EMT demonstrate a 10-fold higher spheroid forming potential. Twist1 and Snail overexpression are correlated with a 30- and 2-fold increase in the tumor spheroid forming and tumor-initiating potential, respectively.33,34 Furthermore, CD24− cancer cells demonstrate greater stemness and migratory potentials, and downregulating CD24 is achieved by transforming growth factor-β (TGF-β)-triggered EMT signaling.35,36 Figure 1 represents the phenotypic and metastatic changes caused in epithelial cells after EMT.22
Figure 1.
Representation of phenotypic changes induced by EMT. The epithelial cells maintain polarity and interact with each other and the basal membrane through junctions. However, cancer cells lose their polarity and interactions with neighboring cells after losing the epithelial phenotype. The variation in invasive, tumorigenic, and chemoresistant potential when epithelial cells transform during EMT is also epitomized. EMT: Epithelial-to-mesenchymal transition; MET: Mesenchymal-to-epithelial transition.
Role of epithelial-to-mesenchymal transition in tumor migration and initiation at distant anatomical sites
EMT is a pleiotropic event affecting several TME processes, including cell migration, morphological changes, chemoresistance, and genetic alterations.37 Changes in metastatic cells are closely associated with modifications caused by EMT. However, whether EMT is responsible for tumor metastasis is still unclear. Nonetheless, ZEB1 upregulation is reportedly associated with cell migration and tumor initiation in adenocarcinoma.38 Additionally, Snail was necessary for cancer cell dissemination in a mouse model.39 Both these markers are upregulated during EMT in cancer cells.39
However, CTCs present as single cells and cell clusters. These cell clusters express E-cadherin; hence, the theory of EMT, i.e., that “The expression of E-cadherin is inhibited after EMT takes place” is contradicted.40,41 In contrast, the hypothesis of partial EMT is strengthened by this observation. However, cells leading the invading cell clusters demonstrate gene expressions agreeing with activation by EMT.42
However, EMT alone fails to explain colonization in new tissues after cell shedding.43 Subsequent steps after cell shedding involve seeding disseminated clusters in the parenchyma of the destination tissue and forming secondary tumors. After arrival at the new tissue environment, the cells must undergo adaptive mechanisms to survive in the new environment. However, the environment in new tissues is likely unfavorable for secondary tumor growth.44,45 Thus, the chances of successful colonization are extremely low. Therefore, after migration, the migratory mesenchymal cells revert to epithelial cells to acquire the required epithelial phenotypes to grow new tumors successfully.46 This event is known as MET. MET is characterized by inhibiting EMT-related TF release. Messengers such as cyclic adenosine monophosphate (cAMP) are released, assisting in MET.47 Furthermore, epithelial cell markers such as E-cadherin are re-expressed. This process is responsible for forming secondary tumors, in which cells are organized hierarchically in their epithelial and stromal states to form new tumors.22 The absence of the signals produced by the TME of primary tumors is also hypothesized to be responsible for restoring the epithelial phenotypes in migratory mesenchymal cells.48
Mechanistic pathways involved in triggering epithelial-to-mesenchymal transition
The signals from tumor stroma initiate EMT pathways in epithelial cells, resulting in them developing mesenchymal phenotypes. EMT is performed by several TFs, such as Twist, Snai1 (Snail), Snai2 (Slug), Zeb1, and Zeb2.49 These TFs downregulate epithelial gene expression in epithelial cancer cells.50
Transforming growth factor-β pathway
The TGF-β pathway is one of the most important pathways in cancer EMT.51 This pathway is triggered by binding specific ligands to TGF-β receptor type 1 (TGF-βR1) and TGF-βR2 [Figure 2]. The specific ligands activating this pathway include TGF-β (three isoforms), bone morphogenetic proteins, and activins.52 TGF-βs binding to TGF-βR1 and TGF-βR2 leads to SMAD2 and SMAD3 phosphorylation, further complexing with SMAD4. Activating TGF-β receptors with bone morphogenetic proteins leads to SMAD1 and SMAD5 phosphorylation, which is also complex with SMAD4.22 This SMAD trimeric translocates to the cell nucleus and is a transcription regulator for acquiring mesenchymal phenotypes in epithelial cells. SMAD complexes downregulate E-cadherin and upregulate mesenchymal proteins N-cadherin, vimentin, and fibronectin.53 Various TFs associated with EMT, such as Twist, Slug, Snail, and Zeb1, are overexpressed in the transforming epithelial cells. These TFs increase TGF-β receptor expression through a positive feedback mechanism.54
Figure 2.
Mechanistic representation of the WNT, PI3K/AKT, NOTCH, Hedgehog, and TGF-β pathways responsible for EMT induction in BC. AKT: Protein kinase B; BC: Breast cancer; EMT: Epithelial-to-mesenchymal transition; H/E: Hematoxylin and eosin; NF-κB: Nuclear factor kappa B; PI3K: Phosphoinositide 3-kinase; TGF-β: Transforming growth factor-β; WNT: Wingless-related integration site.
Another member of the TGF-β family, growth differentiation factor 10 (GDF-10), is associated with BC progression. Zhou et al55 conducted micro ribonucleic acid (miRNA) sequencing of specimens obtained from human triple-negative BC (TNBC) and observed a negative correlation between GDF-10 and BC progression. In vitro studies on TNBC cells revealed downregulating GDF-10 upregulated EMT phenotypes through the TGF-β pathway. GDF-10 expression inhibited cell proliferation and EMT in BC cells by upregulating SMAD7 and blocking nuclear SMAD4 translocation. The anti-metastatic activity of GDF-10 is also translated in in vivo BC xenograft models, thus showing its potential for clinical translation.55
MHP-1, a polysaccharide isolated from Mortierella hepiali, inhibited BC metastasis in vitro and in vivo by selectively inhibiting the TGF-β pathway. MHP-1 re-sensitized the topotecan-resistant MCF-7 cell line with mesenchymal phenotypes. MHP-1 also decreased MDA-MB-231 cell metastasis in the xenografts model and reduced vimentin and ALK5 expression.56 Alpha-lipoic acid suppresses metastatic MDA-MB-231 and 4T1 cell migration and inhibits TGF-β-induced angiopoietin-like 4.57
Wingless-related integration site signaling pathway
Nineteen ligands activate the wingless-related integration site (Wnt) signaling pathway upon binding with the frizzled family of Wnt membrane receptors. The Wnt pathway includes canonical and non-canonical signaling pathways. Activating the canonical pathway leads to β-catenin nuclear translocation.22 Nuclear β-catenin is a cofactor for expressing various genes related to cell proliferation, differentiation, and tumorigenesis [Figure 2].58 E-Cadherin uses β-catenin to form adherens complexes. β-Catenin also connects the cytoskeleton with cell surface adherens junctions. Hence, when the epithelial character is lost in epithelial cells, β-catenin is free for nuclear translocation.59
Activating Wnt pathways is further responsible for upregulating EMT-inducing TFs.60 Canonical Wnt pathway activation through recombinant Wnt3A led to Slug and Twist upregulation in BC.61 To increase their expression, β-catenin directly interacts with the promoters of EMT-inducing TFs, such as Twist, Zeb1, and Slug.48 Inhibiting secreted frizzled-related protein-1 (SFRP1), a Wnt signaling pathway inhibitor, induces EMT in BC cells, confirming the central role of Wnt signaling in EMT.62 In contrast, the non-canonical Wnt pathway induces EMT through the protein kinase-C-dependent route. Activating the non-canonical pathway leads to vimentin and Snail upregulation and E-cadherin downregulation.63,64
Manganese-12 acetate potentially blocks the Wnt/β-catenin pathway by suppressing AKT and phosphoinositide 3-kinase (PI3K) phosphorylation. Cell migration and invasion capabilities in MCF-7 and MDA-MB-231 cell lines were significantly reduced. Furthermore, N-cadherin downregulation and E-cadherin upregulation were confirmed through western blotting. Real-time-polymerase chain reaction (PCR) confirmed the downregulation of EMT-related TFs such as Zeb1, Slug, Snail, and Twist1. Notably, the messenger RNA (mRNA) and protein levels of programmed death-ligand 1 (PD-L1) were also suppressed. Thus, manganese-12 acetate showcased a huge potential to manage BC metastasis by targeting major metastatic pathways.65
The long noncoding RNA (lncRNA) HOTTIP also triggers EMT and induces migration and invasion in BC cells. Silencing HOTTIP using short hairpin RNA inhibited the activity of the lncRNA in MDA-MB-231 and MDA-MB-468 cell lines. This EMT inhibitory activity was also translated in in vivo BC models by blocking the Wnt/β-catenin pathway.66
NOTCH pathway
The NOTCH pathway controls cell proliferation and differentiation. Jagged family and delta-like ligands bind to four NOTCH receptor isoforms (NOTCH1–NOTCH4).67,68 After ligands bind to NOTCH receptors, a cleavage process frees an intracellular NOTCH fragment. This fragment travels to the nucleus, causing EMT-specific genetic expressions [Figure 2].69 The NOTCH pathway promotes Slug and Snail and downregulates E-cadherin expression.70 This pathway also causes CD44 receptor overexpression in cancer cells.71 However, NUMB represses the NOTCH pathway by metabolizing cleaved NOTCH fragments and inhibiting EMT in BC cells.72 The epigenetic regulations performed by miRNAs and lncRNAs on the NOTCH pathway were previously reviewed.73
Phosphoinositide 3-kinase/protein kinase B signaling pathway
The PI3K/protein kinase B (AKT) signaling pathway involves different cellular physiological processes such as cell cycle, growth, proliferation, and apoptosis via effector molecules acting downstream of the pathway.74 PI3K/AKT is a major regulatory factor during cell survival in stressful environments, such as in tumors; therefore, PI3K/AKT plays a crucial role in cancer development.74,75 The association between PI3K/AKT and EMT has been established in different cancers. Twist overexpresses the AKT2 gene, leading to invasion and drug resistance in BC cells. However, Twist-induced metastasis, invasion, and resistance to paclitaxel were inhibited by silencing AKT. Thus, Twist and AKT maintain a positive feedback loop in enhancing pro-EMT function [Figure 2].76 PI3K and phosphoinositide-dependent kinase-1 (PDK) expression increase AKT activity and p65 subunit expression of nuclear factor kappa B (NF-κB), participants of EMT induction in BC.77 Cyclin-dependent kinase-6 (CDK-6) upregulates PI3K expression and silences CDK-6-regulated PI3K, further downregulating the efflux P-gp protein and leading to chemo-sensitization of cancer cells.78
Hedgehog signaling
The Hedgehog (Hh) signaling pathway plays a major role during embryonic development. This signaling cascade activates after binding the Hh protein with a transmembrane receptor called “patched.” Patched activation leads to glioma-associated oncogene (GLI) activation via another intermediate protein called “smoothened.” Subsequently, GLI mediates the expression of downstream target genes. Canonical and non-canonical Hh signaling is crucial in BC development.79 In the case of estrogen-positive (ER+) BC, estrogen increases SHH and GLI1, leading to an increase in invasive characteristics of T47D (HER2−) and BT-474 (HER2+) cells.80 Furthermore, more than one pathway leads to GLI activation in TNBC. Apart from GLI activation via the Hh pathway, NF-κB and forkhead box C1 protein (FOXC1) reportedly activate GLI1 and GLI2 in TNBC. Moreover, GLI1 increases the metastatic potential of BC cells by increasing the levels of the C-X-C chemokine receptors CXCR4/CXCR7.79 LCP1/L-PLASTIN, a chemokine ligand 12 (CXCL12)/CXCR4 signaling mediator, is expressed by GLI1.81 These results indicate that inhibiting GLI1 could be a rational therapeutic approach to reducing the metastatic burden in BC. Figure 2 pictorially represents the major mechanistic pathways responsible for EMT in BC.
Role of the tumor microenvironment in epithelial-to-mesenchymal transition
Tumorigenesis is associated with disrupting the normal balance in surrounding tissues and developing a microenvironment more suitable for rapid cell growth. EMT in the neoplastic cells is controlled by stromal cells in TME. CAFs, immune cells, and vascular cells are primarily responsible for regulating EMT in epithelial cells.
Cancer-associated fibroblasts
CAFs involve various neoplastic cells crucial in tumor progression.82 CAFs are differentiated from several cells, including CSCs, cancer fibroblasts, endothelial cells, and stellate cells.83 The TME also contains non-neoplastic fibroblasts; however, they do not participate in tumor signaling and management like CAFs. CAFs express higher α-smooth muscle actin than normal breast fibroblasts (NBFs). CAFs release various cytokines and chemokines such as tumor necrosis factor (TNF)-β, TGF-β, interleukin-8 (IL-8), and CC motif chemokine ligand 17 (CCL17), which remodel the TME and cause phenotypic changes in BC cells [Tables 2 and 3].82,84 TGF-β1 is also responsible for apoptotic resistance in BC CAFs via the p-44/42/mitogen-activated protein kinase (MAPK) signaling pathway.85 CAFs co-cultured with MCF-7 cell lines induced higher N-cadherin and vimentin expression while reducing E-cadherin expression. CAFs increased the invasiveness of MCF-7 cells by inducing such changes, whereas NBFs could not.86
Table 2.
Pathways for CAF-induced EMT for its prevention.
| Biomarker | Mechanistic pathway | Pharmacological action | Reference |
|---|---|---|---|
| TGF-β1 | p-44/43/MAPK pathway | Resistance to apoptosis in BC CAFs | 85 |
| DNMT3B | TGF-β1/miR-200s/miR-221/DNMT3B regulatory loop | Transformation of NBFs to CAFs | 87 |
| TGF-β1 | Autophagy and FAP-α overexpression | EMT and pulmonary metastasis | 28 |
| IL-32 | p-38/MAPK signaling pathway | EMT | 88 |
| Exosomal survivin | Upregulation of SOD1 | Transformation of NBFs to CAFs | 89 |
| TGF-β1 | Upregulation of lncRNA HOTAIR | EMT | 90 |
| IL-8 | Upregulation of S100A8 | Enhanced invasive capabilities | 91 |
| Exosomal miR-181d-5p | Downregulation of CDX2 and HOXA5 | EMT | 92 |
| miR-146a | Suppressing TXNIP that potentiated Wnt signaling | EMT | 93 |
| miR-335 | Downregulation of CDH11 | Downregulation of EMT | 94 |
| miRNA Let-7b | IL-8 inhibition | Inhibition of CAF-mediated EMT | 95 |
| CCL17 | HIC1 inhibition | Tumor progression and metastasis | 96 |
BC: Breast cancer; CAF: Cancer-associated fibroblast; CCL17: CC motif chemokine ligand 17; EMT: Epithelial-to-mesenchymal transition; FAP-α: Fibroblast activation protein-alpha; HOTAIR: HOX antisense intergenic ribonucleic acid; IL: Interleukin; lncRNA: Long noncoding ribonucleic acid; MAPK: Mitogen-activated protein kinase; miRNA: Micro ribonucleic acid; NBF: Normal breast fibroblast; SOD1: Superoxide dismutase 1; TGF-β1: Transforming growth factor beta 1; Wnt: Wingless-related integration site.
Table 3.
Recent targets and strategies to target CAF-mediated EMT in BC.
| Therapeutic entity | Target | Mechanism of action | Reference |
|---|---|---|---|
| Pirfenidone | Repression of PD-L1 expressions | Inhibiting cytokines | 97 |
| Pirfenidone | Suppression of YAP and CD44 | Reduction of EMT and stemness in BC cells | 98 |
| Artemisinin | Suppression of TGF-β | Inhibition of EMT and inactivation of CAFs | 99 |
| Inhibition of TGF-β1 by siRNA | Suppression of p-44/42 leading to suppression of MAPK signaling pathway | Sensitization of CAFs to 5-FU and tamoxifen | 85 |
| NBFs released OPG | Blocking the Wnt/β-catenin pathway | Inhibition of EMT and metastasis | 100 |
| miR-4516 | Expression of FOSL1 | Suppression of CAF metastatic abilities | 101 |
| Valsartan | Decrease in IL-6 mRNA levels | Inhibition of pulmonary metastasis | 102 |
5-FU: 5-Fluorouracil; BC: Breast cancer; CAF: Cancer-associated fibroblast; EMT: Epithelial-to-mesenchymal transition; IL: Interleukin; MAPK: Mitogen-activated protein kinase; NBF: Normal breast fibroblast; OPG: Osteoprotegerin; PD-L1: Programmed death-ligand 1; siRNA: Small interfering RNA; TGF-β1: Transforming growth factor beta 1; Wnt: Wingless-related integration site.
Mechanistic pathway for the continuous active state of cancer-associated fibroblasts
Tang et al87 reported an essential pathway responsible for the continuous active state of CAFs to fuel BC progression and metastasis: the autocrine TGF-β1/miR-200s/miR-221/DNMT3B regulatory loop signaling pathway. CAFs could remain active without TME and cancer epithelial cells in vitro.103,104 DNMT3B decreases tumor suppressor miRNA-200b, miRNA-200c, and miRNA-221 levels through DNA methylation of their promoters. This continuous suppression transformed the NBFs into CAFs. Inhibiting the TGF-β1/miR-200s/miR-221/DNMT3B regulatory loop led to miRNA-200b and miRNA-200c demethylation, restoring the NBFs phenotypes in CAFs. Thus, the epigenetic modulations performed by this feedback loop are critical to maintaining an active CAF status in BC cells.87
Conversion of normal breast fibroblasts to cancer-associated fibroblasts
Li et al89 reported the role of BC cells in transforming NBFs into CAFs. BC epithelial cells release exosomes enriched with survivin. The survivin-rich exosomes activate NBFs into CAFs by upregulating superoxide dismutase 1 (SOD1). These activated fibroblasts induced BC progression and metastasis.89 Furthermore, Soon et al105 isolated CAFs and NBFs from patients with BC; CAFs had increased resistance towards doxorubicin compared to NBFs. CAFs also demonstrated an increased level of pro-invasion biomarkers such as IL-6, CCL2, fibroblast growth factor 7 (FGF7), matrix metalloproteinase 2 (MMP2), MMP9, and MMP11 compared to the NBFs.105
TGF-β1 orchestrates EMT in BC cells via autophagy; TGF-β1 stimulates CAF-orchestrated tumor progression, EMT, and pulmonary metastasis through autophagy and overexpression of fibroblast activation protein-alpha (FAP-α). However, autophagy inhibitor 3-methyladenine blocked TGF-β1 mediated metastasis in BC.28 IL-32, another cytokine overexpressed in the CAFs, binds to BC cells via integrin β3 receptors overexpressed on the BC epithelial cells. IL-32 overexpression causes EMT-related changes in the epithelial cells by acting through the p-38/MAPK signaling pathway.88
Mechanistic pathways responsible for cancer-associated fibroblast-induced epithelial-to-mesenchymal transition in breast cancer
Tumor suppressor breast cancer gene 1 (BRCA1) is essential in orchestrating CAF-induced metastasis. Mutated BRCA1 cells were co-cultured with HCC1937 cells, and CAFs demonstrated increased EMT-specific biomarkers. These transformed CAFs triggered EMT in BC cells.106 Furthermore, TGF-β1 is responsible for inducing EMT in BC cells. The CAFs-conditioned culture medium enhanced lncRNA HOX antisense intergenic RNA (HOTAIR) in BC cells, and the promoter site of HOTAIR is directly bound to downstream TGF-β proteins, i.e., SMAD2, 3, and 4. Hence, TGF-β pathway stimulation leads to HOTAIR upregulation. Downregulating HOTAIR inhibits EMT in BC cells.90 The mechanistic pathways and therapeutic targets to prevent CAF-induced EMT are summarized in Tables 2 and 3.
CAFs also induce EMT in breast epithelial cells by upregulating IL-8. IL-8 upregulates S100A8 expression in non-invasive BC cells to render them invasive characteristics. S100A8 is a calcium-binding protein belonging to the S-100 family. S100A8 overexpression is also associated with cell migration in colon, breast, and lung tumors. S100A8 is upregulated by IL-8-mediated TFs p65, NF-κB, and CEBPβ upregulation.91 BC CAFs treated with 20 ng/mL of docetaxel upregulated IL-8 in BC CAFs. Hence, IL-8 may be important in chemoresistance and tumor survival.107
CAFs also play a role in mediating epigenetic modulations via miRNAs. CAFs secrete miR-181d-5p-enriched exosomes that potentiated cell proliferation and EMT in BC cells. miR-181d-5p promoted BC cell proliferation and EMT by downregulating CDX2 and HOXA5. HOXA5 overexpression in BC cells increases proliferation, migration, invasion, and EMT.92 Furthermore, BC cell-derived miR-146a can enrich exosomes to convert NBFs to CAFs and induce EMT in the BC epithelial cells. miR-146a orchestrated this action by suppressing TXNIP, which stimulated the Wnt pathway.93 CDH11 genes in CAFs are associated with metastasis in BC cells. CDH11 overexpression leads to vimentin, fibronectin, and β-catenin overexpression in BC cells.108 miR-335 expression downregulates CDH-11 and the biomarkers above in BC cells. Hence, miR-335 epigenetically suppresses EMT in BC cells. Furthermore, anti-CDH11 antibodies suppressed the EMT in BC cells and inhibited BC metastasis.94
Let-7b miRNA of the Let-7 family is an IL-8-mediated BC metastasis inhibitor.109 Hence, Let-7b is mechanistically inhibited in CAFs to continue IL-8-mediated metastasis in BC cells.95 However, NBFs demonstrate higher Let-7b expression than CAFs. Inhibiting Let-7b in NBFs activates CAFs through the IL-6-dependent positive feedback loop. Increased Let-7b expression in CAFs led to CAF inactivation by blocking the IL-6-dependent positive feedback loop. Furthermore, an increase in Let-7b also suppressed IL-8 expression, suppressing BC metastasis and tumor growth.95
CAFs release CCL17 in high concentrations, leading to EMT and BC progression.110 Inhibiting the HIC1 protein in pre-metastatic BC cells is essential for CCL17-dependent metastasis. HIC1− epithelial cells released CXCL14, which bound to its cognate GPR85 receptors on the mammary fibroblasts to transform them to CAFs following the AKT, extracellular signal-regulated kinase 1/2 (ERK1/2), and neddylation pathways. These activated CAFs then release CCL17 for tumor progression and metastasis in BC.96
Strategies to prevent cancer-associated fibroblast-induced epithelial-to-mesenchymal transition in breast cancer
Pirfenidone (PFD), a small pyridine class molecule, has recently gained attention for its ability to target CAFs. PFD is an orally active U.S. Food and Drug Administration-approved drug commercially used to treat lung fibrosis.111 PFD inhibited IL-6, IL-8, TNF-β, CCL17, and CXCL14 release from CAFs and downregulated PD-L1 expression by downregulating inflammatory mediators. PD-L1 overexpression inactivates T cells in the BC TME.97 The potential of PFD to suppress EMT-induced stemness in BC cells has also been reported. Overexpressing yes-associated protein-1 (YAP1) was associated with increased stromal characteristics and induced EMT. A positive correlation was also observed between YAP1 and EMT-associated gene expression, such as ZEB and vimentin in BC cells. PFD also reduced the invasiveness of invasive BC cells and CAFs. The treatment could reduce EMT-associated biomarkers. Notably, stemness-associated proteins, such as YAP1 and CD44, were also reduced.98 Hence, PFD shows potential to be used with first-line chemotherapeutic and immunotherapeutic drugs in BC therapy.
Artemisinin (ART), a sesquiterpene lactone, and its derivatives were tested for their anti-CAF activity. ART has anti-malarial and anticancer activities.112 ART suppressed the TGF-β signaling pathway in TME. Because TGF-β-mediated signaling is crucial for the trans-differentiation of CAFs in the BC TME,99 inhibiting TGF-β inactivated CAFs in BC cells. ART and its derivatives also inhibited TGF-β-mediated metastasis and BC growth in orthotopic models in vivo.113 Furthermore, small interfering RNA (siRNA)-mediated genetic inhibition of TGF-β1 sensitized CAFs to 5-fluorouracil (5-FU) and tamoxifen treatment. Suppressing p-44/42 proteins inhibited the MAPK signaling pathway. Notably, the genetic knockdown of TGF-β1 also orchestrated a 3-fold decrease in the anti-apoptotic B cell lymphoma 2 protein (Bcl-2), leading to apoptosis in drug-resistant BC CAFs.85 Alrouji et al100 recently demonstrated the suppressing ability of normal NBFs. NBFs release osteoprotegerin (OPG), a decoy receptor for the NF-κB ligand-receptor activator. BC cells were treated with human recombinant OPG in vitro and in vivo. Recombinant OPG suppressed the EMT, metastasis, and proliferation in the BC tumor in vivo. Recombinant OPG also inhibited the metastasis in BC tumors by blocking the Wnt/β-catenin pathway.100
miR-4156 has tumor-suppressing activities.114 CAFs in patients with TNBC demonstrated 5-fold lower concentrations of miR-4156 compared to NBFs. Exosomal miR-4156 expression is inversely related to FOSL1 expression in patients with TNBC. FOSL1 expression is associated with poor prognosis, high proliferation, and metastasis in TNBC cells. Hence, miR-4156 and FOSL1 inhibitors could soon be used as an anticancer agent.101
Angiotensin II, an essential peptide of the renin-angiotensin system, plays an important role in vasoconstriction, blood volume, aldosterone secretion, and maintaining water and sodium in the body.115 Angiotensin II potentiates cancer progression, metastasis, migration, and progression.116 Takiguchi et al102 studied the potential of the angiotensin blocker valsartan to block BC progression and pulmonary metastasis. Angiotensin II significantly increased BC proliferation, EMT, and pulmonary metastasis in the presence of CAFs in vivo. The mRNA expression of IL-6 was significantly increased in xenograft tumors. Furthermore, valsartan significantly reduced the pulmonary metastasis of BC cells.102
Role of immune cells and inflammation in epithelial-to-mesenchymal transition
Primary tumors release pro-inflammatory mediators such as TGF-β, TNF-α, IL-1, IL-6, and IL-8 recruit infiltrating immune cells into the TME.117 Rather than performing anti-tumor actions, immune cells, mainly lymphocytes, are chemically programmed by the TME to aid tumor progression and metastasis [Tables 4 and 5].118 Inflammatory exogenous chemicals such as lipopolysaccharides also induce EMT by downregulating E-cadherin by upregulating mesenchymal markers such as smooth muscle actin and S100A by binding to Toll-like receptor 4.119 Immune cells and inflammation-related mechanistic pathways and therapeutic targets are reviewed in Tables 4 and 5.
Table 4.
Immune cells and inflammation-related pathways in BC EMT.
| Biomarker | Mechanistic pathway | Pharmacological action | Reference |
|---|---|---|---|
| CD163 expression in TAMs | – | Lymph node metastasis | 120 |
| CXCL8 | CXCL2 binds to CXCR2 | EMT and increased stemness | 121 |
| IL-8 | Binds to IL-8Rα and IL-8Rβ | Conversion of neutrophils to TANs | 122 |
| Nicotine | Recruitment of TANs in TME | EMT by releasing lipocalin | 123 |
| Histidine decarboxylase expressed MDSCs | Potentiating Wnt/β-catenin pathway | Increased metastatic potential | 124 |
| Operative stress | Increased infiltration of MDSCs in tumors | Increased EMT | 125 |
| CXCL17 | Chemotaxis of MDSCs and upregulation of platelet growth factor-BB | Increased lung metastasis | 126 |
| CCL3 | Activation of the PI3K-AkT-mTOR signaling pathway | Infiltration of MDSCs in the TME | 127 |
| MDSCs released NO and IL-6 | Activation of NOTCH and STAT3 signaling | Increased TGF-β signaling and stemness in BC cells | 128 |
| Overexpression of transcription factor ΔNp63 | Upregulation of CXCL2 and CCL22 | Enhanced recruitment of MDSCs the breast TME | 129 |
AkT: Protein kinase B; BC: Breast cancer; CCL: CC motif chemokine ligand; CXCL: Chemokine ligand; CXCR: C-X-C chemokine receptor; EMT: Epithelial-to-mesenchymal transition; MDSC: Myeloid-derived stem cell; NO: Nitric oxide; PI3K: Phosphoinositide 3-kinase; TAM: Tumor-associated macrophage; TAN: Tumor-associated neutrophil; TGF-β: Transforming growth factor beta; TME: Tumor microenvironment; Wnt: Wingless-related integration site; –: No data.
Table 5.
Recent drugs and targets to suppress immune cell-mediated EMT.
| Therapeutic entity | Mechanistic pathway | Pharmacological activity | Reference |
|---|---|---|---|
| Ginsenoside RG3 | Inhibition of STAT3 pathway, NOTCH pathway, and downregulation of chemokines | Downregulation of EMT and MDSCs infiltration in TME | 130 |
| BML-111 | Inhibition of inflammatory mediators. Suppression of ILK signaling pathway | Inhibition of EMT | 131 |
| Danirixin | Suppression of stem-cell phenotypes | Inhibition of cell proliferation and lung metastasis | 121 |
| Ruyiping | Inhibition of STAT6-mediated macrophage activation | Inhibition of EMT and lung metastasis | 132 |
| Progranulin | Downregulation of exosomal miR-5100, which suppressed CXCL2 levels | Inhibition of metastasis | 133 |
| OVOL2 | Inhibition of TGF-β signaling by epigenetically inhibiting SMAD4/7 levels | Reduced EMT | 134 |
| OVOL2 | Suppressing IL-10 | Inactivation of TAM2 | 29 |
| Salidroside | Suppression of STAT3 activation | Conversion of pro-tumor TAN2 to anti-tumor TAN1 | 123 |
CXCL: Chemokine ligand; EMT: Epithelial-to-mesenchymal transition; ILK: Integrin-linked kinase; MDSC: Myeloid-derived stem cell; miR: Micro ribose nucleic acid; TAM: Tumor-associated macrophage; TAN: Tumor-associated neutrophil; TGF-β: Transforming growth factor beta; TME: Tumor microenvironment.
Effect of smoking in triggering epithelial-to-mesenchymal transition in breast cancer by increasing cytokine levels
Pham et al135 studied the effect of electronic (E)-cigarette smoke on BC cell metastasis. The vapor condensate increased CCL5, vascular cell adhesion protein 1 (VCAM-1), and other major pro-inflammatory cytokine concentrations in BC cells. CCL5 mechanistically binds to CXCR1 receptors to mediate EMT, migration, and invasion in BC cells.136 E-cigarette smoke also increased the survival of BC cells by regulating the tumor-invading macrophages through VCAM-1 and integrin α4β1. Immunohistochemical studies showed that E-cigarette smoke increased macrophage infiltration into the BC cells and increased BC cell proliferation, EMT, and lung metastasis in vivo.135
Role of tumor-associated macrophages in regulating epithelial-to-mesenchymal transition in breast cancer
Tumor-associated macrophages (TAMs) are the most extensively explored recruited immune cells. TAMs release TNF-α, which mediates EMT, activating the p38 MAPK pathway.137,138 TNF-α mediated EMT is regulated by NF-κB in BC cells.139 TAMs also secrete vascular endothelial growth factor (VEGF), leading to tumor angiogenesis.140 Zhang et al141 studied samples from 278 patients with TNBC and reported CD163+ TAMs in the TME. Cell infiltration was associated with lymph node metastasis and tumor recurrence. They also observed lower E-cadherin expressions in BC cells; however, multivariate analysis showed that TAM infiltration in the TNBC and E-cadherin downregulation were independent prognostic markers.141 BC cells were incubated with activated M1 macrophages in a conditioned media, inducing metastasis and EMT in the epithelial cells, indicating that the M1 macrophages communicate signals to trigger EMT.142 Additionally, Nie et al121 investigated clinical BC tissues and observed high TAM-released CXCL8 expression. CXCL8 overexpression was also associated with migration, invasion, and EMT in patients with BC via binding to CXCR2. CXCL8 also improves the self-renewal capability of BC stem-like cells.121 Immune cells infiltrating the tumor induce EMT by releasing MMP enzymes, urokinases, and cathepsins.143,144
Targeting tumor-associated macrophages to prevent epithelial-to-mesenchymal transition in breast cancer
Targeting the immune system in the BC TME is an effective strategy to inhibit cancer progression. BML-111, an analog of lipoxin A4, is an effective inhibitor of inflammatory mediators and is used to manage inflammation-mediated cancer metastasis,145 and Lin et al131 used BML-111 to target TAMs in the TNBC TME. BML-111 inhibited EMT in MDA-MB-231 and 4T1 cells. A concentration of 800 ng/mL of BML-111 induced significant cell death and reduced metastasis in BC cells by inhibiting the integrin-linked kinase (ILK) pathway and downregulating ILK, p-AKT, and p-GSK3β protein expression.131 CXCR2, a CXCL2 receptor, is overexpressed in BC cells and TAMs. Nie et al121 used danirixin, a CXCR2 antagonist, to inhibit metastasis in BC cells. Danirixin suppressed the cell proliferation rate and lung metastasis of BC cells, suppressing BC stem cell activity in vivo.121 Ruyiping is a traditional Chinese medicine to treat BC and inhibits TAM2-mediated EMT of TNBC cells by preventing STAT6-mediated macrophage activation. Furthermore, in vivo studies using ruyiping demonstrated a reduced TAM2:TAM1 ratio and lung metastasis of TNBC.132
Progranulin (PGN), a glycoprotein secreted by several cells, is a metastasis biomarker in BC. High PGN expression on TAMs is associated with EMT and metastasis.146 Yue et al133 knocked out PGN from TAMs, and the PGN−/− TAMs inhibited BC migration and invasion. PGN−/− released exosomes enriched with miR-5100, which downregulated CXCL12. Thus, the CXCL12/CXCR4 pathway was suppressed, inhibiting BC lung metastasis in PGN−/− mice in vivo.133
Zinc finger TF OVOL2 has gained interest in BC research due to its ability to inhibit the TGF-β pathway.134 OVOL2 has activity by binding to the DNA sequences in the promoter region.147 OVOL2 blocked the TGF-β pathway at multiple levels by inhibiting SMAD4 and SMAD7 mRNA expression, blocking complex formation between SMAD4 and SMAD2/3, and inhibiting SMAD4 binding to target DNA.134 Additionally, OVOL2 can inhibit M2 macrophage activation by antagonizing IL-10. IL-10 was downregulated by OVOL2 directly binding to the promoter.29
Role of neutrophil infiltration in regulating epithelial-to-mesenchymal transition in breast cancer
IL-8 plays a major role in the chemo-attraction of neutrophils towards TME. IL-8 binds to two receptors, IL-8Rα and IL-8Rβ, expressed on neutrophils. Neutrophils convert into tumor-associated neutrophils (TANs) when responding to immune mediators.148,149 Like TAMs, TANs can behave as anti-tumor (TAN1) or pro-tumor (TAN2).150 Neutrophils orchestrate EMT in BC by releasing neutrophil extracellular traps (NETs).151 Factors present in the TME, such as IL-8 and granulocyte colony-stimulating factor, trigger the formation of NETs from TANs. NETs comprise double-stranded DNA and nuclear and granular neutrophil proteins. NETs can capture CTCs and encourage secondary tumor formation.151 Martins-Cardoso et al152 studied the effects of isolated NETs on MCF-7 cell lines and found EMT-related gene alteration in vitro. They also observed a positive correlation between NETs and metastasis in the cancer genome atlas.152
Prevention of tumor-associated neutrophil-induced epithelial-to-mesenchymal transition by salidroside
Nicotine can recruit TAN2 in the pre-metastatic TME. Recruited TANs released lipocalin 2 via STAT3 activation, inducing EMT in the BC cells. Elevated lipocalin 2 blood and urine levels are observed in early-stage patients with BC, thus making it a promising prognostic marker for BC disease.123 Salidroside, a natural antioxidant isolated from Rhodiola rosea, has anti-cancer potential at high concentrations.153 However, low-dose salidroside inhibited neutrophil conversion to TAN2 and enhanced antitumor TAN1 phenotypes by blocking STAT3 activation. Thus, salidroside treatment inhibited BC cell metastasis to the lungs in vivo.123
Induction of epithelial-to-mesenchymal transition by myeloid-derived suppressor cells in breast cancer
Myeloid-derived suppressor cells (MDSCs) are crucial for suppressing immune action against tumors.154 MDSCs trigger cancer cell metastasis by releasing MMP9, epidermal growth factor (EGF), and TGF-β.155 MDSCs are a heterogeneous population of cells comprising immature macrophages, dendritic cells, and granulocytes.156 The genesis of MDSCs occurs through altered myelopoiesis during the existence of a tumor.157 MDSCs play an important role in tumor progression and metastasis by remodeling the TME by promoting EMT, stem-like phenotypes in tumor cells, releasing metalloproteases and angiogenic factors, and suppressing the immune response.158 Furthermore, MDSCs promote tumor growth by suppressing the immune action exhibited by natural killer, CD4+ T, and CD8+ T cells.159,160 MDSCs are characterized by Gr-1, Ly6G, and Ly6C molecules on their surface in mice. In humans, MDSCs are characterized by CD33 receptor expression on their membranes and histamine receptors.
Role of myeloid-derived suppressor cells in colonizing migratory breast cancer cells
MDSCs play a crucial role in BC metastasis. A subpopulation of BC cells with overexpressed cytokeratin 14 colonized and raised secondary tumors. Histidine decarboxylase-labeled MSDCs have a close spatial relationship with cytokeratin 14-expressing BC cells by promoting Wnts expression, potentiating Wnt/β-catenin signaling in BC cells.124
Myeloid-derived suppressor cells induce epithelial-to-mesenchymal transition after surgical ablation of breast cancer tumors
Surgical ablation is a major treatment for BC. However, MDSCs promote BC metastasis after the primary tumor is removed through surgery. The operative stress possibly increased MDSCs infiltration in the TME. Infiltrated MDSCs were isolated and incubated with 4T1 cells in vitro. The co-incubation led to metastatic changes in epithelial cells via TGF-β, VEGF, and IL-10 upregulation.125 Thus, post-operative MDSC downregulation can potentially overcome BC metastasis.
Mechanistic and epigenetic pathways responsible for myeloid-derived suppressor cell-induced epithelial-to-mesenchymal transition
Chemokine CXCL17 orchestrates CD11b+Gr-1+ MDSC chemotaxis to secondary tumor sites in the lungs and aids in colonizing BC cells by releasing platelet growth factor-BB.126 CCL3 infiltrates MDSCs in TME, and CCL3-activated MDSCs trigger EMT in epithelial cells via the PI3K-AKT-mTOR signaling pathway.127 MDSCs induce nitric oxide-mediated NOTCH pathway activation and IL-6-mediated STAT3 phosphorylation. These two pharmacological modulations lead to increased TGF-β signaling pathway activation via prolonged STAT3 activation, inducing stem cell-like phenotypes in BC cells and suppressing T cell-mediated immune action.128
High levels of TF ΔNp63 are observed in samples from patients with TNBC. ΔNp63 mediates MDSC chemotaxis by upregulating CXCL2 and CCL22. ΔNp63 levels correlated positively with MDSC infiltration in breast tumors, and ΔNp63 also increased BC cell proliferation, metastasis, and colonization in the lungs. Owing to its roles in immunosuppression and metastasis, blocking ΔNp63 can inhibit BC progression.129
RG3 prevents myeloid-derived suppressor cell-induced epithelial-to-mesenchymal transition
Ginsenoside RG3 has been studied for its role in inhibiting MDSC-mediated EMT in BC.161 RG3 inhibited MDSC-mediated EMT by blocking the STAT3 and NOTCH pathways and suppressing chemokines in the BC TME. In a mouse model, RG3 delayed MDSC-mediated tumor growth and metastasis and downregulated MDSCs in the blood samples at a non-cytotoxic dose. Furthermore, in vitro studies demonstrated the potential of RG3 to inhibit the stem-cell-like phenotypes in BC cells. Thus, RG3 demonstrated strong potential as an anti-metastatic agent for BC management by blocking multiple pathways essential for EMT.130
Role of programmed death-ligand 1 in triggering epithelial-to-mesenchymal transition and recent therapeutic strategies
PD-L1 is a crucial transmembrane protein suppressing acquired immune cells. PD-L1 binds to the inhibitory checkpoint receptor programmed cell death 1 (PD1) and transmits an inhibitory signal to the immune response, antagonizing it.162 PD1 antagonization leads to antigen-specific cytotoxic T cell proliferation suppression, simultaneously decreasing apoptosis in anti-inflammatory regulatory T cells.162 EMT is associated with PD-L1 overexpression in the BC TME by activating the PI3K/AKT pathway.163 A study on 67 patients reported a high correlation between PD-L1 overexpression and EMT markers in the BC TME (p = 0.005).164 Furthermore, EMT triggers PD-L1 expression by upregulating the CMTM6 protein.165 Penisuloxazine A is a C-terminal terminal inhibitor of heat shock protein 90 that downregulated PD-L1 in invasive BC tumors. Penisuloxazine A also destabilized the epidermal- (EGFR) and FGF receptors (FGFR), blocking their downstream signaling. Penisuloxazine A significantly reduced EMT and its biomarkers in MCF-7, MDA-MB-231, and trastuzumab-resistant JIMT-1 cells in vitro.166
Noman et al167 schematically silenced Zeb1, Snail, Slug, and Twist in BC cell lines in vitro to investigate the TFs involved in EMT-induced PD-L1 upregulation; silencing Zeb1 decreased PD-L1 expression significantly in BC cells. Overexpressing the miR-200 family also resulted in EMT-mediated PD-L1 overexpression.167 PD-L1 was inhibited using the monoclonal antibody atezolizumab, and atezolizumab downregulated EMT-associated genes and upregulated apoptosis-inducing genes in MDA-MB-231 cells.168 Sativan, an isoflavone isolated from Spatholobus subrectus, is a Chinese herbal medicine traditionally used for treating BC. Sativan demonstrated excellent apoptosis-inducing capabilities in MD-MB-231 cell lines by upregulating BCL-2-associated X (BAX) and downregulating BCL-2 expressions in vitro. Furthermore, a decrease in EMT-inducing protein was also reported. Sativan upregulated miR-200c expression in BC cells, epigenetically suppressing PD-L1 expression. Sativan also inhibited tumor growth and metastasis in BC in vivo.169
Poly (Adenosine diphosphate-ribose) polymerase inhibitors induce epithelial-to-mesenchymal transition in breast cancer gene-mutated breast cancer and the role of metformin
Poly (adenosine diphosphate [ADP]-ribose) polymerase (PARP) proteins are associated with spindle pole formation, inflammation, cell cycle regulation, and DNA repair. PARPs also play an important role in base excision repair and recruit repair enzymes at the site of single-strand breaks. PARP inhibitors act by blocking DNA double-strand break repair.170 However, clinical and pre-clinical evidence shows that BRCA-mutated BC does not respond to PARP inhibitors, and the resistance to PARP inhibitors in BC is associated with EMT and chemoresistance.171
PD-L1 was upregulated in BC cells via p-AKT S473 activation after treatment with PARP inhibitors. PD-L1 upregulation also potentiated EMT through the B-catenin/STT3-mediated transcriptional pathway. However, PARP inhibitors could still initiate EMT after PD-L1 knockout. Hence, anti-PD-L1 monotherapy was ineffective against PARP inhibitor-induced EMT. Metformin blocks the mTOR pathway responsible for tumor proliferation and chemoresistance and was used to treat PARP inhibitor-induced EMT. Metformin blocked p-AKT S473 phosphorylation to inhibit PARP inhibitor-induced EMT and subsequent chemoresistance. Therefore, co-administering metformin and PARP inhibitors holds potential in BRCA-mutated BC therapy.172
Role of hypoxia in epithelial-to-mesenchymal transition
Hypoxia plays an important role in BC progression and metastasis. Hypoxic tumors are characterized by more aggressive phenotypes, metastasis, and resistance to chemotherapy and radiotherapy.173 The reasons for a hypoxic TME include a high metabolic rate and poor vascular network, causing insufficient oxygen supplies and poor dissemination of waste products from the tumor.174 Hypoxia-inducible factor (HIF)-1 is a key regulator of hypoxia-induced gene expression in the TME. HIF-1 modulates gene expression related to hypoxia tolerance, apoptosis escape, chemo-resistance, and angiogenesis [Figure 3].175 HIF-1 comprises two sub-units: HIF-1α and HIF-1β. Under normal oxygen concentrations, HIF-1α is readily ubiquitinated. In contrast, HIF-1α is more stable under hypoxic conditions, leading to increased expression.176 Inhibiting HIF-1 in a triple TNBC orthotopic tumor model in mice resulted in growth arrest in the primary tumor and inhibited metastasis to lymph nodes and lungs.177 Furthermore, a hypoxic environment upregulates vimentin in TNBC with E-cadherin downregulation. A zinc finger protein known as Gli-1 is upregulated in the hypoxic TNBC microenvironment. Gli-1 knockdown downregulated hypoxia-induced EMT.178 Extracellular adenosine triphosphate (ATP) also plays an important role in BC tumor metastasis by upregulating HIF-1/2α signaling. HIF-2α knockdown in BC cells decreased ATP-driven EMT. A chromatin immunoprecipitation (ChIP) assay revealed that downstream HIF-2α proteins orchestrate the ATP-associated EMT, among which MMP-9 and LOXL-2 play significant roles.179
Figure 3.
Role of hypoxia in promoting EMT in tumors. The cells undergo various metabolic and signaling changes due to various factors attributing to hypoxia in the tumor microenvironment, leading to EMT in the cancer cells and subsequent invasion and metastasis. EMT: Epithelial-to-mesenchymal transition.
Heme oxygenase-1 (HO-1) is crucial for cancer cell survival in hypoxia-induced oxidative stress. HO-1 is an antioxidant resulting from two TFs: activating TF-4 (ATF-4) and nuclear factor erythroid 2-related factor 2 (NRF-2). The cumulative cytoprotective actions induced by these two TFs include antioxidant enzyme upregulation and autophagy induction. ATF-4 and NRF-2 repression is positively related to anoikis and the inability to develop lung metastatic nodules.180 ATF-4 upregulation leads to ZEB1 activation and E-cadherin downregulation.181 A study in patients with TNBC concluded that ATF-4 promoted EMT through the TGF-β/SMAD pathway.182
Epigenetic modulations responsible for hypoxia-induced epithelial-to-mesenchymal transition
HIF-1α and hypoxia directly upregulate lncRNARP11-390F4.3, and lncRNARP11-390F4.3 expression is crucial for hypoxia-induced EMT and subsequent metastasis. Overexpression of lncRNARP11-390F4.3 overexpression leads to the expression of EMT-related phenotypes in BC cells.183 The epigenetic modulation orchestrates the hypoxia-mediated suppression of apoptosis-promoting protein p52-2 via miR-205. miR-205 overexpression is also accompanied by the appearance of EMT-related markers in BC cells.184
Role of obesity in hypoxia-mediated epithelial-to-mesenchymal transition in breast cancer
Along with tumors, hypoxia in the adjacent adipose tissues causes EMT in BC cells. Obesity is a major cause of incidence and metastasis of BC in postmenopausal women.185 Co-culturing MCF-7 cells and adipocytes led to upregulating TGF-β, HIF-1α, and lectin-like oxidized low-density lipoprotein receptor 1 (LOX1) receptor mRNA levels in MCF-7 cells. The levels of pro-EMT TFs Twist1 and FOXC2 were also increased. Notably, estrogen receptors on the surface of MCF-7 cells were downregulated, causing hormone therapy resistance. Adipose tissue hypoxia accompanies obesity; therefore, obesity is a significant risk factor for EMT and BC progression.186 Adipokine resistin induces EMT and stemness in BC cells by binding to adenylyl cyclase-activated protein 1 (CAP1) receptors. Resistin-induced EMT in the MDA-MB-231 and MCF-7 cell lines by re-organizing F-actin filaments to form protrusions in the BC cells. Furthermore, the levels of epithelial markers (E-cadherin and claudin-1) and mesenchymal markers (Twist1, Zeb1, vimentin, Snail, and Slug) were downregulated and upregulated, respectively.187
Targets to prevent hypoxia-mediated epithelial-to-mesenchymal transition
Owing to the important role of hypoxia in tumor progression, various strategies have been designed to target hypoxia [Table 6, Table 7]. Paeoniflorin, a monoterpene glycoside, suppresses HIF-1α upregulation in hypoxic conditions. Paeoniflorin suppresses EMT in hypoxic MDA-MB-231 cell lines by repressing phosphorylated PI3K and AKT expressions in TNBC cells.188 Fucoidan, a sulfated polysaccharide, downregulates the HIF-1α protein and hypoxia-induced EMT in MDA-MB-231 cells.189
Table 6.
Mechanistic pathways to prevent hypoxia-mediated EMT in BC.
| Biomarker | Mechanistic pathway | Pharmacological activity | Reference |
|---|---|---|---|
| Resistin | Binding to CAP1 receptors | Induction of EMT and stemness | 187 |
| Gli-1 | Upregulation of hypoxia | Induction of EMT in TNBC | 178 |
| Extracellular ATP | Upregulation of HIF-1/2α signaling | EMT | 179 |
| lncRNARP11-390F4.3 | lncRNARP11-390F4.3 is upregulated by hypoxic conditions | Hypoxia-induced EMT | 183 |
| miR-205 | Suppression of apoptosis-promoting protein p52-2 in hypoxic conditions | EMT | 184 |
ATP: Adenosine triphosphate; BC: Breast cancer; CAP1: Cyclase-activated protein 1; EMT: Epithelial-to-mesenchymal transition; HIF: Hypoxia-inducible factor; lncRNA: Long noncoding ribonucleic acid; miR: Micro ribose nucleic acid; TNBC: Triple-negative breast cancer.
Table 7.
Recent drugs and targets to inhibit hypoxia-mediated EMT.
| Therapeutic entity | Mechanistic pathway | Pharmacological response | Reference |
|---|---|---|---|
| Paeoniflorin | HIF-1α suppression by repressing phosphorylated PI3K and Akt | EMT suppression | 188 |
| Fucoidan | HIF-1α downregulation | Repression of hypoxia-induced EMT | 189 |
| Escin la | LOXL2 suppression | Suppression of TGF-β-mediated EMT | 190 |
Akt: Protein kinase B; EMT: Epithelial-to-mesenchymal transition; HIF: Hypoxia-inducible factor; PI3K: Phosphoinositide 3-kinase.
A saponin, Escin la, inhibits LOXL2, a downstream protein of HIF-2α. Escin la suppressed LOXL2 mRNA expression. Escin la prevented E-cadherin downregulation and suppressed EMT in the TGF-β-overexpressed MCF-7 cells. However, Escin la could not prevent hypoxia in the BC cells; hence, the mode of action can be credited to downregulating LOXL2 expression.190 Mechanistic pathways in hypoxia-mediated EMT are represented in Table 6, Table 7.
Relationship between epithelial-to-mesenchymal transition and breast cancer stem cells
Apart from the role of EMT in BC metastasis, EMT also transforms epithelial cells to give them stem cell-like properties [Figure 4].191 These properties include CD44 glycoprotein overexpression, CD24 glycoprotein downregulation, developing spheres in suspension culture, and tumorigenesis in mice or human systems.192 However, CSCs, by EMT definition, should be able to differentiate into epithelial cells through MET. Epithelial cells can dedifferentiate into several alternative mesenchymal states according to the situation, one of which is BC stem cells (BCSCs). Thus, BCSCs can form new metastatic tumors when disseminated to distant tissues.14
Figure 4.
Schematic representation of EMT-induced breast cancer stem cells in the TME and the pathways involved in generating differentiated tumor cells at distant anatomical sites. EMT: Epithelial-to-mesenchymal transition; TME: Tumor microenvironment; TGF-β: Transforming growth factor-β; Wnt: Wingless-related integration site.
Migratory cells undergo MET before forming the macrometastasis.193,194 During EMT transformation, CSCs develop mature adhesion plaques (macromolecular structures comprising grouped integrins). The plaques contribute to the differentiation ability of CSCs by activating the focal adhesion kinase (FAK).195
Molecular and epigenetic modulations responsible for epithelial-to-mesenchymal transition-triggered breast cancer stem cell generation
EMT activation leads to the activation of the autocrine WnT/β-catenin and TGF-β/SMAD signaling pathways. Intracellular expression of Snail reduces the expression of the tumor suppressor p53 gene through p53 deacetylation. p53 knockdown potentiated the tumorigenic potential of BC cells.196 Epithelial cell adhesion molecule (EpCAM) is a transmembrane glycoprotein that plays an important role in tumor progression and metastasis and induces stemness in hypoxic BC tumors.197 EpCAM overexpression leads to upregulating stemness-related markers, including NaNOG, OCT4, and SOX2.198 N-glycosylation is an important pre-step for EpCAM to regulate stemness and EMT in breast tumors by modulating NF-κB in tumor cells.199
The expression of cytokeratin 18 (CK18), a cytoskeleton protein, is important in BC metastasis and BCSC development. CK18 expression is downregulated in aggressive, metastatic, and TGF-β-treated MCF-7 cells. In contrast, CK18 depletion upregulates EpCAM expression by activating the Wnt-β-catenin pathway.200 Moreover, radio-resistant BC cells isolated from the MDA-MB-231 cell line reportedly demonstrate BCSC markers such as notch-1, CD44, and aldehyde dehydrogenase 1.201
BCSCs have high telomerase activity associated with stemness,202 EMT, and BC cell migration. Inhibiting telomerase reverse transcriptase (TRT) transforms BCSCs into cells with more epithelial phenotypes.203 Telomerase inhibitor BIBR1532 exhibited apoptosis in BCSCs with an IC50 value of 38.71 μM at 72 h. BIBR1532 also decreased the sphere-forming and migration abilities of BCSCs in vitro.204
Role of partial epithelial-to-mesenchymal transition in breast cancer stem cell generation and tumorigenic potential
BCSCs are present in two alternative epithelial- and mesenchymal-like stem cell states. These states are capable of inducing new tumors.31,205 TNBC cells promote stemness under the influence of chemotherapeutic stress.206 Activated CD8+ T cells have also induced BCSC-like features in mammary epithelial cells by inducing EMT and giving them tumorigenic potential.207 IL-6-mediated EMT led to BCSC generation from mammary epithelial cells.208 Furthermore, IL-6-mediated EMT and subsequent stemness led to resistance against PI3K inhibitors.209 The extent of EMT is crucial. Overactivating EMT leads to detrimental effects on the tumorigenic ability of cells. In low-claudin BC, cells with epithelial phenotypes expressed higher capabilities to generate new tumors than those without epithelial characteristics.210 Thus, partial EMT leading to CSC generation is focused upon.211
Role of yes-associated protein/TAZ in breast cancer stem cell generation
YAP/TAZ transcriptional regulator overexpression is associated with increased metastasis and stemness in BC. YAP/TAZ promotes malignancy and stemness by directly interacting with transcriptional-enhanced associate domain (TEAD) TFs.212 Various YAP/TAZ inhibitors, such as verteporfin,213 dasatinib,214 and celastrol,215 inhibited cancer growth. However, luteolin, a natural flavonoid, exhibits YAP/TAZ inhibition in TNBC cells. Luteolin inhibits CYR61 and CTGF expression, two major YAP/TAZ-targeted genes. Luteolin also inhibited YAP/TAZ nuclear translocation and transcription in TNBC. However, luteolin increased YAP/TAZ phosphorylation via the non-canonical Hippo pathway, thus increasing its proteasome degradation and inhibiting its nuclear translocation.216
Targets and strategies to target breast cancer stem cells
Downregulating integrin α3 promotes stemness in BC cells. Integrin interacts with VASP and regulates the PI3K/AKT pathway, inhibiting BC promotion, EMT, and stemness. Metastatic BC cells expressed a low level of integrin α3. Integrin α3 treatment of metastatic BC cells reduced wound healing potential and spheroid formation abilities. Thus, integrin α3 should be explored as a possible anti-metastatic agent for BC management.217 Another marker, phosphatase and tensin homolog (PTEN), is downregulated in BC. PTEN suppresses tumors by inhibiting PI3K/AKT pathway-mediated proliferation, metastasis, and tumorigenesis.218 PTEN was downregulated in BCSCs, and PTEN expression prevented Matrigel invasion by BCSCs. Furthermore, immunoprecipitation studies found that Abi1, a core adaptor protein in the WAVE regulatory complex, was downregulated by PTEN. Upregulation of Abi1 downregulated PTEN to induce EMT and BCSC phenotypes in BC cells.219 Efflux proteins such as BC resistance protein (BCRP) have been implicated in secreting chemotherapeutic drugs out of cells, leading to drug-resistant phenotypes in CSCs.220
Effects of hyaluronic acid on breast cancer stem cells
Hyaluronic acid (HA), a common pharmaceutical ingredient, is a crucial targeting moiety in BC-targeted nanoscale formulations.221 However, HA also plays a critical role in BC cell migration.222 Jariyal et al223 reported the effects of high molecular weight (HMW) and low molecular weight (LMW) HA on different sub-populations of BC cells and BCSCs. LMWHA induced EMT in the MCF-7 cell line in vitro, whereas HMWHA did not. LMWHA and HMWHA downregulated the stemness-regulating EpCAM glycoprotein in MCF-7 cell lines. In MDA-MB-231 cells, treating LMW and HMW HA downregulated stem cell-specific CD44 receptors.223
Epithelial-to-mesenchymal transition orchestrates chemoresistance in breast cancer
EMT was thought to be limited to the loss of epithelial phenotypes and the gain of mesenchymal phenotypes to migrate and colonize in distant tissues. However, studies indicate EMT leads to immune escape, chemotherapy resistance, and BCSC generation [Table 8].6,224 The net effects of these phenotypic changes caused by EMT include chemoresistance and tumor relapse.225 Chemoresistant BC samples were enriched with EMT-related genes.226 Furthermore, cancer cell dormancy, EMT, and expression of multidrug resistance proteins such as P-gp go hand-in-hand.227 We have summarized biomarkers and epigenetic modulators that induce chemoresistance via EMT-related pathways in Table 8.
Table 8.
EMT-based mechanisms for chemoresistance in the BC cells.
| Biomarker | Mechanism | Pharmacological effects | Reference |
|---|---|---|---|
| Downregulation of miR-200b | Fibronectin 1 and increased luciferase activity overexpression | EMT and DOX resistance | 228 |
| Snail | Snail downregulation | 5-FU resistance | 229 |
| AMP-activated protein kinase | Deactivation of AMP-activated protein kinase | EMT induced 5-FU, ADR, and PTX resistance in MCF-7 and MDA-MB-231 cell lines | 230 |
| Terminal differentiation-induced noncoding RNA (TINCR) | Overexpression of TINCR inhibits miR-125b | EMT and trastuzumab resistance | 231 |
| Downregulation of miR-489 | SMAD3 upregulation | EMT and ADR resistance | 232 |
| S-phase kinase-associated protein-2 upregulation | p27 inhibition | EMT and PTX resistance | 233 |
| Downregulation of miR-137 | Upregulation of dual-specific phosphatase 4 | EMT and DOX resistance | 234 |
| Downregulation of miR-125b | Sema4C upregulation | EMT and PTX resistance | 235 |
| Upregulation of lncRNA H19 | Snail upregulation | EMT and tamoxifen resistance. Curcumin prevented H19-mediated EMT and resistance | 236 |
| SIRT1 | Wnt/β-catenin pathway activation | EMT and DOX resistance. Resveratrol blocked DOX resistance by blocking the SIRT1-β-catenin pathway | 237 |
| Ki67 upregulation | Upregulation of mesenchymal phenotypes | EMT and cisplatin resistance. Niclosamide suppressed Ki67 to inhibit EMT and cisplatin resistance | 238 |
| ADR treatment mediated EMT and subsequent chemoresistance | P53 triggered p21 activation. Disrupting the connection of p53 with mdm2 | EMT by upregulation of Twist1 and ADR resistance | 239 |
| 5-FU treatment mediated EMT and chemoresistance | Upregulation of stemness and EMT markers | 5-FU resistance. Salinomycin re-sensitized the resistant BC cells to 5-FU | 240 |
| Zeb1 | Increase in the autophagic genes resulting in LC3-II overexpression | DOX resistance | 241 |
| Lactate dehydrogenase A | Increase in pro-survival autophagy and downregulation of apoptosis | EMT and ADR resistance. Downregulation in lactate dehydrogenase A level inhibited autophagy and triggered apoptosis | 242 |
5-FU: 5-fluorouracil; ADR: Adriamycin; AMP: Adenosine monophosphate; BC: Breast cancer; DOX: Doxorubicin; EMT: Epithelial-to-mesenchymal transition; LC3-II: Microtubule-associated protein 1A/1B-light chain 3 – phosphatidylethanolamine conjugate; lncRNA: Long noncoding ribonucleic acid; MCF-7: Michigan Cancer Foundation-7; MDA-MB-231: M.D. Anderson metastatic breast cancer cells; miR: Micro ribose nucleic acid; PTX: Paclitaxel; TINCR: Terminal differentiation-induced noncoding RNA; Wnt: Wingless-related integration site.
Conclusion and perspective
This review describes the importance of EMT in tumor progression and promotion. We mechanistically discussed the pathways responsible for orchestrating EMT in cancer cells, the role of the TME in promoting EMT, and the role of EMT in tumor progression, metastasis, and chemoresistance. We also summarized various therapeutic targets to counter EMT in cancer cells.
EMT is essential in BC progression, metastasis, relapse, and chemo-resistance. EMT is also crucial for generating various mesenchymal cells and BCSCs, aggravating BC, and raising secondary tumors. The highly regulated nature of EMT by various TME components demonstrates its utmost importance. Scientists have targeted various EMT-involved pathways, and several studies inhibited stemness in BC cells by targeting EMT. This approach can prevent the tumorigenic potential of BC cells, ultimately preventing disease relapse.
However, stress-induced metastasis due to chemotherapy or surgical ablation should be countered. Targeted delivery of TGF-β inhibitors and agents such as all-trans retinoic acid, which differentiate immature MDSCs,243 should be explored to block metastasis post-approval, thus inhibiting tumor relapse. Lifestyle-associated factors, including obesity and smoking, also trigger EMT through cytokine-associated pathways. Thus, targeted delivery of TGF-β inhibitors should be explored for patients with these risk factors. Because several pathways can trigger metastasis, targeted delivery systems are required to neutralize disseminated migratory cell clusters in the systemic circulation. However, conventional nanocarriers cannot accumulate in the TME owing to high intra-tumor pressure, stiff extracellular matrix, poor extravasation of nanocarriers, and high cell density.244,245 In contrast, cell-based drug delivery systems have recently demonstrated superior tumor-targeting properties.246, 247, 248 Such systems should target primary and secondary tumors and CTCs. Furthermore, re-purposed drugs such as statins with cytotoxic and anti-metastatic pharmacological activities should be used to treat BC.249 Lastly, co-delivering anti-metastatic drugs and chemotherapeutic agents should be studied for enhanced antitumor effects.250
This review provides a comprehensive summary of BC biology, molecular pathways, and recent advancements in therapeutic strategies, molecular pathways, and recent advances in therapeutic strategies. We also highlight the need to investigate the pathophysiology of BC to elucidate novel targets for which therapeutic agents can be developed to treat BC.
Authors contribution
Paras Famta, Saurabh Shah, and Biswajit Dey – conceptualization, data curation, manuscript writing, data visualization, editing; Kondasingh Charan Kumar, Deepkumar Bagasariya, Ganesh Vambhurkar, Dadi A. Srinivasarao, Anamika Sharma, and Rahul Kumar – data curation; Santosh Kumar Guru, and Rajeev Singh Raghuvanshi – supervision and validation; Saurabh Srivastava – supervision, editing, and conceptualization. All the authors have read and approved the final version of the manuscript.
Ethics statement
Not applicable.
Declaration of generative AI and AI-assisted technologies in the writing process
The authors declare that generative artificial intelligence (AI) and AI assisted technologies were not used in the writing process or any other process during the preparation of this manuscript.
Funding
None.
Conflict of interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Data availability statement
No data was used for the research described in the manuscript.
Acknowledgments
The authors thank the research funding support by the Department of Pharmaceuticals (DoP), Ministry of Chemicals and Fertilizers, Government of India to “Pharmaceutical Innovation and Translational Research Lab” (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, India.
Managing Editor: Peng Lyu
References
- 1.Lévy C. Cancer du sein. Oncologie. 2011;13:510–513. doi: 10.1007/s10269-011-2047-3. [DOI] [Google Scholar]
- 2.Pandey G., Phatale V., Khairnar P., et al. Supramolecular self-assembled peptide-engineered nanofibers: a propitious proposition for cancer therapy. Int J Biol Macromol. 2024;256 doi: 10.1016/J.IJBIOMAC.2023.128452. [DOI] [PubMed] [Google Scholar]
- 3.Loibl S., Poortmans P., Morrow M., Denkert C., Curigliano G. Breast cancer. Lancet. 2021;397:1750–1769. doi: 10.1016/S0140-6736(20)32381-3. [DOI] [PubMed] [Google Scholar]
- 4.Yousefi M., Nosrati R., Salmaninejad A., Dehghani S., Shahryari A., Saberi A. Organ-specific metastasis of breast cancer: molecular and cellular mechanisms underlying lung metastasis. Cell Oncol. 2018;41:123–140. doi: 10.1007/s13402-018-0376-6. [DOI] [PubMed] [Google Scholar]
- 5.Kyuno D., Takasawa A., Kikuchi S., Takemasa I., Osanai M., Kojima T. Role of tight junctions in the epithelial-to-mesenchymal transition of cancer cells. Biochim Biophys Acta Biomembr. 2021;1863 doi: 10.1016/j.bbamem.2020.183503. [DOI] [PubMed] [Google Scholar]
- 6.Richard V., Kumar T.R.S., Pillai R.M. Transitional dynamics of cancer stem cells in invasion and metastasis. Transl Oncol. 2021;14 doi: 10.1016/j.tranon.2020.100909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Goldstein L.J., Perez R.P., Yardley D., et al. A window-of-opportunity trial of the CXCR1/2 inhibitor reparixin in operable HER-2-negative breast cancer. Breast Cancer Res. 2020;22:4. doi: 10.1186/S13058-019-1243-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.M.D. Anderson Cancer Center. Denosumab for Breast Cancer with Bone Mets. ClinicalTrials.gov ID NCT01952054.
- 9.Dompé Farmaceutici S.p.A. Pilot Study to Evaluate Reparixin with Weekly Paclitaxel in Patients with HER 2 Negative Metastatic Breast Cancer (MBC). ClinicalTrials.gov ID NCT02001974.
- 10.Washington University School of Medicine. MCS110 Combined with Neoadjuvant Doxorubicin, Cyclophosphamide, and Weekly Paclitaxel in Patients with Hormone-Receptor Positive and HER2-Breast Cancer.ClinicalTrials.gov ID NCT03285607.
- 11.National Cancer Centre, Singapore. The Role of Bexarotene in Inducing Susceptibility to Chemotherapy in Metastatic TNBC. ClinicalTrials.gov ID NCT04664829.
- 12.Cambridge University Hospitals NHS Foundation Trust. A Pre-operative Window Study of Letrozole Plus PR Agonist (Megestrol Acetate) Versus Letrozole Alone in Post-menopausal Patients with ER-positive Breast Cancer. ClinicalTrials.gov ID NCT03306472.
- 13.Zhejiang Provincial People’s Hospital. Aspirin on CTCs of Advanced Breast and Colorectal Cancer. ClinicalTrials.gov ID NCT02602938.
- 14.Wang H., Unternaehrer J.J. Epithelial-mesenchymal transition and cancer stem cells: at the crossroads of differentiation and dedifferentiation. Dev Dyn. 2019;248:10–20. doi: 10.1002/dvdy.24678. [DOI] [PubMed] [Google Scholar]
- 15.Ottevanger P.B. Ovarian cancer stem cells more questions than answers. Semin Cancer Biol. 2017;44:67–71. doi: 10.1016/j.semcancer.2017.04.009. [DOI] [PubMed] [Google Scholar]
- 16.Saitoh M. JB special review-cellular plasticity in epithelial homeostasis and diseases: involvement of partial EMT in cancer progression. J Biochem. 2018;164:257–264. doi: 10.1093/jb/mvy047. [DOI] [PubMed] [Google Scholar]
- 17.Grigore A.D., Jolly M.K., Jia D., Farach-Carson M.C., Levine H. Tumor budding: the name is EMT. partial EMT. J Clin Med. 2016;5:51. doi: 10.3390/jcm5050051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Grosse-Wilde A., D’Hérouël A.F., McIntosh E., et al. Stemness of the hybrid epithelial/mesenchymal state in breast cancer and its association with poor survival. PLoS One. 2015;10 doi: 10.1371/journal.pone.0126522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jolly M.K., Huang B., Lu M., Mani S.A., Levine H., Ben-Jacob E. Towards elucidating the connection between epithelial-mesenchymal transitions and stemness. J R Soc Interface. 2014;11 doi: 10.1098/rsif.2014.0962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Aiello N.M., Maddipati R., Norgard R.J., et al. EMT subtype influences epithelial plasticity and mode of cell migration. Dev Cell. 2018;45:681–695.e4. doi: 10.1016/j.devcel.2018.05.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lamouille S., Xu J., Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15:178–196. doi: 10.1038/nrm3758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Shibue T., Weinberg R.A. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol. 2017;14:611–629. doi: 10.1038/nrclinonc.2017.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Jayachandran J., Srinivasan H., Mani K.P. Molecular mechanism involved in epithelial to mesenchymal transition. Arch Biochem Biophys. 2021;710 doi: 10.1016/j.abb.2021.108984. [DOI] [PubMed] [Google Scholar]
- 24.Thiery J.P., Acloque H., Huang R.Y.J., Nieto M.A. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–890. doi: 10.1016/j.cell.2009.11.007. [DOI] [PubMed] [Google Scholar]
- 25.Guo W., Keckesova Z., Donaher J.L., et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell. 2012;148:1015–1028. doi: 10.1016/j.cell.2012.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Isenmann S., Arthur A., Zannettino A.C.W., et al. TWIST family of basic helix-loop-helix transcription factors mediate human mesenchymal stem cell growth and commitment. Stem Cell. 2009;27:2457–2468. doi: 10.1002/stem.181. [DOI] [PubMed] [Google Scholar]
- 27.Famta P., Shah S., Khatri D.K., Guru S.K., Singh S.B., Srivastava S. Enigmatic role of exosomes in breast cancer progression and therapy. Life Sci. 2022;289 doi: 10.1016/j.lfs.2021.120210. [DOI] [PubMed] [Google Scholar]
- 28.Huang M., Fu M., Wang J., et al. TGF-β1-activated cancer-associated fibroblasts promote breast cancer invasion, metastasis and epithelial-mesenchymal transition by autophagy or overexpression of FAP-α. Biochem Pharmacol. 2021;188 doi: 10.1016/j.bcp.2021.114527. [DOI] [PubMed] [Google Scholar]
- 29.Wu R.S., Lin J., Xing Y.M., et al. OVOL2 inhibits macrophage M2 polarization by regulating IL-10 transcription, and thus inhibits the tumor metastasis by modulating the tumor microenvironment. Immunol Lett. 2022;242:17–26. doi: 10.1016/j.imlet.2021.05.004. [DOI] [PubMed] [Google Scholar]
- 30.Quail D.F., Joyce J.A. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–1437. doi: 10.1038/nm.3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Liu S., Cong Y., Wang D., et al. Breast cancer stem cells transition between epithelial and mesenchymal states reflective of their normal counterparts. Stem Cell Rep. 2014;2:78–91. doi: 10.1016/j.stemcr.2013.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Vesuna F., Lisok A., Kimble B., Raman V. Twist modulates breast cancer stem cells by transcriptional regulation of CD24 expression. Neoplasia. 2009;11:1318–1328. doi: 10.1593/neo.91084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hollier B.G., Tinnirello A.A., Werden S.J., et al. FOXC2 expression links epithelial-mesenchymal transition and stem cell properties in breast cancer. Cancer Res. 2013;73:1981–1992. doi: 10.1158/0008-5472.CAN-12-2962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Mani S.A., Guo W., Liao M.J., et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704–715. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Morel A.P., Lièvre M., Thomas C., Hinkal G., Ansieau S., Puisieux A. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS One. 2008;3 doi: 10.1371/journal.pone.0002888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ghuwalewala S., Ghatak D., Das P., et al. CD44 high CD24 low molecular signature determines the cancer stem cell and EMT phenotype in oral squamous cell carcinoma. Stem Cell Res. 2016;16:405–417. doi: 10.1016/j.scr.2016.02.028. [DOI] [PubMed] [Google Scholar]
- 37.Erin N., Grahovac J., Brozovic A., Efferth T. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updat. 2020;53 doi: 10.1016/j.drup.2020.100715. [DOI] [PubMed] [Google Scholar]
- 38.Ye X., Tam W.L., Shibue T., et al. Distinct EMT programs control normal mammary stem cells and tumour-initiating cells. Nature. 2015;525:256–260. doi: 10.1038/nature14897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Krebs A.M., Mitschke J., Losada M.L., et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat Cell Biol. 2017;19:518–529. doi: 10.1038/ncb3513. [DOI] [PubMed] [Google Scholar]
- 40.Piacentino M.L., Li Y., Bronner M.E. Epithelial-to-mesenchymal transition and different migration strategies as viewed from the neural crest. Curr Opin Cell Biol. 2020;66:43–50. doi: 10.1016/j.ceb.2020.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yu M., Bardia A., Wittner B.S., et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339:580–584. doi: 10.1126/science.1228522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Cheung K.J., Gabrielson E., Werb Z., Ewald A.J. Collective invasion in breast cancer requires a conserved basal epithelial program. Cell. 2013;155:1639–1651. doi: 10.1016/j.cell.2013.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Liao T.T., Yang M.H. Revisiting epithelial-mesenchymal transition in cancer metastasis: the connection between epithelial plasticity and stemness. Mol Oncol. 2017;11:792–804. doi: 10.1002/1878-0261.12096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liu H., Zhang X., Li J., Sun B., Qian H., Yin Z. The biological and clinical importance of epithelial–mesenchymal transition in circulating tumor cells. J Cancer Res Clin Oncol. 2015;141:189–201. doi: 10.1007/s00432-014-1752-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mittal V. Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol Mech Dis. 2018;13:395–412. doi: 10.1146/annurev-pathol-020117-043854. [DOI] [PubMed] [Google Scholar]
- 46.Lambert A.W., Pattabiraman D.R., Weinberg R.A. Emerging biological principles of metastasis. Cell. 2017;168:670–691. doi: 10.1016/j.cell.2016.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pattabiraman D.R., Bierie B., Kober K.I., et al. Activation of PKA leads to mesenchymal-to-epithelial transition and loss of tumor-initiating ability. Science. 2016;351 doi: 10.1126/science.aad3680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Dongre A., Weinberg R.A. New insights into the mechanisms of epithelial–mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol. 2019;20:69–84. doi: 10.1038/s41580-018-0080-4. [DOI] [PubMed] [Google Scholar]
- 49.Goossens S., Vandamme N., Van Vlierberghe P., Berx G. EMT transcription factors in cancer development re-evaluated: beyond EMT and MET. Biochim Biophys Acta Rev Cancer. 2017;1868:584–591. doi: 10.1016/j.bbcan.2017.06.006. [DOI] [PubMed] [Google Scholar]
- 50.Venhuizen J.H., Jacobs F.J.C., Span P.N., Zegers M.M. P120 and E-cadherin: double-edged swords in tumor metastasis. Semin Cancer Biol. 2020;60:107–120. doi: 10.1016/j.semcancer.2019.07.020. [DOI] [PubMed] [Google Scholar]
- 51.Katsuno Y., Derynck R. Epithelial plasticity, epithelial-mesenchymal transition, and the TGF-β family. Dev Cell. 2021;56:726–746. doi: 10.1016/j.devcel.2021.02.028. [DOI] [PubMed] [Google Scholar]
- 52.Wendt M.K., Allington T.M., Schiemann W.P. Mechanisms of the epithelial-mesenchymal transition by TGF-β. Futur Oncol. 2009;5:1145–1168. doi: 10.2217/fon.09.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Feldkoren B., Hutchinson R., Rapaport Y., Mahajan A., Margulis V. Integrin signaling potentiates transforming growth factor-beta 1 (TGF-β1) dependent down-regulation of E-Cadherin expression – important implications for epithelial to mesenchymal transition (EMT) in renal cell carcinoma. Exp Cell Res. 2017;355:57–66. doi: 10.1016/j.yexcr.2017.03.051. [DOI] [PubMed] [Google Scholar]
- 54.Lin Y. Te, Wu K.J. Epigenetic regulation of epithelial-mesenchymal transition: focusing on hypoxia and TGF-β signaling. J Biomed Sci. 2020;27:1–10. doi: 10.1186/s12929-020-00632-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Roux-Mallouf T. Le, Pelen F., Vallejo A., Halimaoui I., Doutreleau S., Verges S. Effect of chronic nitrate and citrulline supplementation on vascular function and exercise performance in older individuals. Aging. 2019;11:3315–3332. doi: 10.18632/aging.101983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lin S., Lyu X., Yu J., et al. MHP-1 inhibits cancer metastasis and restores topotecan sensitivity via regulating epithelial-mesenchymal transition and TGF-β signaling in human breast cancer cells. Phytomedicine. 2016;23:1053–1063. doi: 10.1016/j.phymed.2016.06.013. [DOI] [PubMed] [Google Scholar]
- 57.Tripathy J., Tripathy A., Thangaraju M., Suar M., Elangovan S. α-Lipoic acid inhibits the migration and invasion of breast cancer cells through inhibition of TGFβ signaling. Life Sci. 2018;207:15–22. doi: 10.1016/j.lfs.2018.05.039. [DOI] [PubMed] [Google Scholar]
- 58.Tewari D., Bawari S., Sharma S., DeLiberto L.K., Bishayee A. Targeting the crosstalk between canonical Wnt/β-catenin and inflammatory signaling cascades: a novel strategy for cancer prevention and therapy. Pharmacol Ther. 2021;227 doi: 10.1016/j.pharmthera.2021.107876. [DOI] [PubMed] [Google Scholar]
- 59.Choi Y., Lee H.J., Jang M.H., et al. Epithelial-mesenchymal transition increases during the progression of in situ to invasive basal-like breast cancer. Hum Pathol. 2013;44:2581–2589. doi: 10.1016/j.humpath.2013.07.003. [DOI] [PubMed] [Google Scholar]
- 60.Farooqi A.A., Naureen H., Attar R. Regulation of cell signaling pathways by circular RNAs and microRNAs in different cancers: spotlight on Wnt/β-catenin, JAK/STAT, TGF/SMAD, SHH/GLI, NOTCH and Hippo pathways. Semin Cell Dev Biol. 2022;124:72–81. doi: 10.1016/j.semcdb.2021.04.002. https://doi.org/10.1016/j.semcdb.2021.04.002 [DOI] [PubMed] [Google Scholar]
- 61.Wu Y., Ginther C., Kim J., et al. Expression of Wnt3 activates Wnt/β-catenin pathway and promotes EMT-like phenotype in trastuzumab-resistant HER2-overexpressing breast cancer cells. Mol Cancer Res. 2012;10:1597–1606. doi: 10.1158/1541-7786.MCR-12-0155-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Gauger K.J., Chenausky K.L., Murray M.E., Schneider S.S. SFRP1 reduction results in an increased sensitivity to TGF-β signaling. BMC Cancer. 2011;11:1–10. doi: 10.1186/1471-2407-11-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kestler H.A., Kühl M. From individual Wnt pathways towards a Wnt signalling network. Philos Trans R Soc B Biol Sci. 2008;363:1333–1347. doi: 10.1098/rstb.2007.2251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Remo A., Sina S., Barbi S., et al. Wnt (canonical and non canonical) pathways in breast carcinoma with extensive vascular invasion and inflammatory breast carcinoma. Pathol Res Pract. 2021;219 doi: 10.1016/j.prp.2021.153347. [DOI] [PubMed] [Google Scholar]
- 65.Ju H., Li Y., Xing X., et al. Manganese-12 acetate suppresses the migration, invasion, and epithelial–mesenchymal transition by inhibiting Wnt/β-catenin and PI3K/AKT signaling pathways in breast cancer cells. Thorac Cancer. 2018;9:353–359. doi: 10.1111/1759-7714.12584. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 66.Han S., Jin X., Liu Z., et al. The long noncoding RNA HOTTIP promotes breast cancer cell migration, invasiveness, and epithelial–mesenchymal transition via the Wnt–β-catenin signaling pathway. Biochem Cell Biol. 2019;97:655–664. doi: 10.1139/bcb-2018-0313. [DOI] [PubMed] [Google Scholar]
- 67.Alves-Guerra MC, Capobianco AJ, Demarest RM. Notch/jagged signaling. In: Encycl Cancer. Published online 2011:2559–2565. https://doi.org/10.1007/978-3-642-16483-5_4132.
- 68.Wang M.M. Notch signaling and Notch signaling modifiers. Int J Biochem Cell Biol. 2011;43:1550–1562. doi: 10.1016/j.biocel.2011.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Krishna B.M., Jana S., Singhal J., et al. Notch signaling in breast cancer: from pathway analysis to therapy. Cancer Lett. 2019;461:123–131. doi: 10.1016/j.canlet.2019.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wang Z., Li Y., Kong D., Sarkar F H. The role of notch signaling pathway in epithelial-mesenchymal transition (EMT) during development and tumor aggressiveness. Curr Drug Targets. 2010;11:745–751. doi: 10.2174/138945010791170860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Apostolou P., Toloudi M., Ioannou E., et al. Study of the interaction among Notch pathway receptors, correlation with stemness, as well as their interaction with CD44, dipeptidyl peptidase-IV, hepatocyte growth factor receptor and the SETMAR transferase, in colon cancer stem cells. J Recept Signal Transduct. 2013;33:353–358. doi: 10.3109/10799893.2013.828072. [DOI] [PubMed] [Google Scholar]
- 72.Luo Z., Mu L., Zheng Y., et al. NUMB enhances Notch signaling by repressing ubiquitination of NOTCH1 intracellular domain. J Mol Cell Biol. 2020;12:345–358. doi: 10.1093/jmcb/mjz088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ghafouri-Fard S., Glassy M.C., Abak A., Hussen B.M., Niazi V., Taheri M. The interaction between miRNAs/lncRNAs and Notch pathway in human disorders. Biomed Pharmacother. 2021;138 doi: 10.1016/j.biopha.2021.111496. [DOI] [PubMed] [Google Scholar]
- 74.Shi X., Wang J., Lei Y., Cong C., Tan D., Zhou X. Research progress on the PI3K/AKT signaling pathway in gynecological cancer. Mol Med Rep. 2019;19:4529–4535. doi: 10.3892/mmr.2019.10121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Porta C., Paglino C., Mosca A. Targeting PI3K/Akt/mTOR signaling in cancer. Front Oncol. 2014;4:64. doi: 10.3389/fonc.2014.00064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Xu W., Yang Z., Lu N. A new role for the PI3K/Akt signaling pathway in the epithelial-mesenchymal transition. Cell Adhes Migr. 2015;9:317–324. doi: 10.1080/19336918.2015.1016686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Maier H.J., Schmidt-Straßburger U., Huber M.A., Wiedemann E.M., Beug H., Wirth T. NF-κB promotes epithelial-mesenchymal transition, migration and invasion of pancreatic carcinoma cells. Cancer Lett. 2010;295:214–228. doi: 10.1016/j.canlet.2010.03.003. [DOI] [PubMed] [Google Scholar]
- 78.Zhang L., Li Y., Hu C., et al. CDK6-PI3K signaling axis is an efficient target for attenuating ABCB1/P-gp mediated multi-drug resistance (MDR) in cancer cells. Mol Cancer. 2022;21:1–26. doi: 10.1186/S12943-022-01524-W/FIGURES/8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Riobo-Del Galdo N., Lara Montero Á., Wertheimer E. Role of hedgehog signaling in breast cancer: pathogenesis and therapeutics. Cells. 2019;8:375. doi: 10.3390/cells8040375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Bhateja P., Cherian M., Majumder S., Ramaswamy B. The hedgehog signaling pathway: a viable target in breast cancer? Cancers. 2019;11:1126. doi: 10.3390/cancers11081126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Dubovsky J.A., Chappell D.L., Harrington B.K., et al. Lymphocyte cytosolic protein 1 is a chronic lymphocytic leukemia membrane-associated antigen critical to niche homing. Blood. 2013;122:3308–3316. doi: 10.1182/blood-2013-05-504597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sahai E., Astsaturov I., Cukierman E., et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20:174–186. doi: 10.1038/s41568-019-0238-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Biffi G., Tuveson D.A. Diversity and biology of cancerassociated fibroblasts. Physiol Rev. 2021;101:147–176. doi: 10.1152/physrev.00048.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Shiga K., Hara M., Nagasaki T., Sato T., Takahashi H., Takeyama H. Cancer-associated fibroblasts: their characteristics and their roles in tumor growth. Cancers. 2015;7:2443–2458. doi: 10.3390/cancers7040902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Chandra Jena B., Kanta Das C., Banerjee I., et al. Paracrine TGF-β1 from breast cancer contributes to chemoresistance in cancer associated fibroblasts via upregulation of the p44/42 MAPK signaling pathway. Biochem Pharmacol. 2021;186 doi: 10.1016/j.bcp.2021.114474. [DOI] [PubMed] [Google Scholar]
- 86.Soon P.S.H., Kim E., Pon C.K., et al. Breast cancer-associated fibroblasts induce epithelial-to-mesenchymal transition in breast cancer cells. Endocr Relat Cancer. 2013;20:1–12. doi: 10.1530/ERC-12-0227. [DOI] [PubMed] [Google Scholar]
- 87.Tang X., Tu G., Yang G., et al. Autocrine TGF-β1/miR-200s/miR-221/DNMT3B regulatory loop maintains CAF status to fuel breast cancer cell proliferation. Cancer Lett. 2019;452:79–89. doi: 10.1016/j.canlet.2019.02.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Wen S., Hou Y., Fu L., et al. Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3–p38 MAPK signalling. Cancer Lett. 2019;442:320–332. doi: 10.1016/j.canlet.2018.10.015. [DOI] [PubMed] [Google Scholar]
- 89.Li K., Liu T., Chen J., Ni H., Li W. Survivin in breast cancer-derived exosomes activates fibroblasts by up-regulating SOD1, whose feedback promotes cancer proliferation and metastasis. J Biol Chem. 2020;295:13737–13752. doi: 10.1074/jbc.RA120.013805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ren Y., huan Jia H., qi Xu Y., et al. Paracrine and epigenetic control of CAF-induced metastasis: the role of HOTAIR stimulated by TGF-ß1 secretion. Mol Cancer. 2018;17:5. doi: 10.1186/s12943-018-0758-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lim H., Koh M., Jin H., et al. Cancer-associated fibroblasts induce an aggressive phenotypic shift in non-malignant breast epithelial cells via interleukin-8 and S100A8. J Cell Physiol. 2021;236:7014–7032. doi: 10.1002/jcp.30364. [DOI] [PubMed] [Google Scholar]
- 92.Wang H., Wei H., Wang J., Li L., Chen A., Li Z. MicroRNA-181d-5p-containing exosomes derived from CAFs promote EMT by regulating CDX2/HOXA5 in breast cancer. Mol Ther Nucleic Acids. 2020;19:654–667. doi: 10.1016/j.omtn.2019.11.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yang S.S., Ma S., Dou H., et al. Breast cancer-derived exosomes regulate cell invasion and metastasis in breast cancer via miR-146a to activate cancer associated fibroblasts in tumor microenvironment. Exp Cell Res. 2020;391:111983. doi: 10.1016/j.yexcr.2020.111983. [DOI] [PubMed] [Google Scholar]
- 94.Chen J.H., Huang W.C., Bamodu O.A., Chang P.M.H., Chao T.Y., Huang T.H. Monospecific antibody targeting of CDH11 inhibits epithelial-to-mesenchymal transition and represses cancer stem cell-like phenotype by up-regulating miR-335 in metastatic breast cancer, in vitro and in vivo. BMC Cancer. 2019;19:634. doi: 10.1186/s12885-019-5811-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Al-Harbi B., Hendrayani S.F., Silva G., Aboussekhra A. Let-7b inhibits cancer-promoting effects of breast cancerassociated fibroblasts through IL-8 repression. Oncotarget. 2018;9:17825–17838. doi: 10.18632/oncotarget.24895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wang Y., Weng X., Wang L., et al. HIC1 deletion promotes breast cancer progression by activating tumor cell/fibroblast crosstalk. J Clin Investig. 2018;128:5235–5250. doi: 10.1172/JCI99974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Aboulkheyr Es H., Zhand S., Thiery J.P., Warkiani M.E. Pirfenidone reduces immune-suppressive capacity of cancer-associated fibroblasts through targeting CCL17 and TNF-beta. Integr Biol. 2020;12:188–197. doi: 10.1093/intbio/zyaa014. [DOI] [PubMed] [Google Scholar]
- 98.Es H.A., Cox T.R., Sarafraz-Yazdi E., Thiery J.P., Warkiani M.E. Pirfenidone reduces epithelial–mesenchymal transition and spheroid formation in breast carcinoma through targeting cancer-associated fibroblasts (Cafs) Cancers. 2021;13:5118. doi: 10.3390/cancers13205118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ghahremanifard P., Chanda A., Bonni S., Bose P. TGF-β mediated immune evasion in cancer—spotlight on cancer-associated fibroblasts. Cancers. 2020;12:1–11. doi: 10.3390/cancers12123650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Alraouji N.N., Hendrayani S.F., Ghebeh H., Al-Mohanna F.H., Aboussekhra A. Osteoprotegerin (OPG) mediates the anti-carcinogenic effects of normal breast fibroblasts and targets cancer stem cells through inhibition of the β-catenin pathway. Cancer Lett. 2021;520:374–384. doi: 10.1016/j.canlet.2021.08.013. [DOI] [PubMed] [Google Scholar]
- 101.Kim J.E., Kim B.G., Jang Y., Kang S., Lee J.H., Cho N.H. The stromal loss of miR-4516 promotes the FOSL1-dependent proliferation and malignancy of triple negative breast cancer. Cancer Lett. 2020;469:256–265. doi: 10.1016/j.canlet.2019.10.039. [DOI] [PubMed] [Google Scholar]
- 102.Takiguchi T., Takahashi-Yanaga F., Ishikane S., et al. Angiotensin II promotes primary tumor growth and metastasis formation of murine TNBC 4T1 cells through the fibroblasts around cancer cells. Eur J Pharmacol. 2021;909 doi: 10.1016/j.ejphar.2021.174415. [DOI] [PubMed] [Google Scholar]
- 103.Karakasheva T.A., Lin E.W., Tang Q., et al. IL-6 mediates cross-talk between tumor cells and activated fibroblasts in the tumor microenvironment. Cancer Res. 2018;78:4957–4970. doi: 10.1158/0008-5472.CAN-17-2268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wang L., Hou Y., Sun Y., et al. c-Ski activates cancer-associated fibroblasts to regulate breast cancer cell invasion. Mol Oncol. 2013;7:1116–1128. doi: 10.1016/j.molonc.2013.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.González L., Eiro N., Fernandez-Garcia B., González L.O., Dominguez F., Vizoso F.J. Gene expression profile of normal and cancer-associated fibroblasts according to intratumoral inflammatory cells phenotype from breast cancer tissue. Mol Carcinog. 2016;55:1489–1502. doi: 10.1002/mc.22403. [DOI] [PubMed] [Google Scholar]
- 106.Hemalatha S.K., Sengodan S.K., Nadhan R., et al. Brcal defective breast cancer cells induce in vitro transformation of cancer associated fibroblasts (CAFs) to metastasis associated fibroblasts (MAF) Sci Rep. 2018;8:13903. doi: 10.1038/s41598-018-32370-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Li Y., Rong G., Kang H. Taxotere-induced elevated expression of IL8 in carcinoma-associated fibroblasts of breast invasive ductal cancer. Oncol Lett. 2017;13:1856–1860. doi: 10.3892/ol.2017.5612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Li L., Ying J., Li H., et al. The human cadherin 11 is a pro-apoptotic tumor suppressor modulating cell stemness through Wnt/Β-catenin signaling and silenced in common carcinomas. Oncogene. 2012;31:3901–3912. doi: 10.1038/onc.2011.541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Thammaiah C.K., Jayaram S. Role of let-7 family microRNA in breast cancer. Noncoding RNA Res. 2016;1:77–82. doi: 10.1016/j.ncrna.2016.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Omland S.H., Wettergren E.E., Mourier T., et al. Cancer associated fibroblasts (CAFs) are activated in cutaneous basal cell carcinoma and in the peritumoural skin. BMC Cancer. 2017;17:675. doi: 10.1186/s12885-017-3663-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Noble P.W., Albera C., Bradford W.Z., et al. Pirfenidone for idiopathic pulmonary fibrosis: analysis of pooled data from three multinational phase 3 trials. Eur Respir J. 2016;47:243–253. doi: 10.1183/13993003.00026-2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Augustin Y., Staines H.M., Krishna S. Artemisinins as a novel anti-cancer therapy: targeting a global cancer pandemic through drug repurposing. Pharmacol Ther. 2020;216 doi: 10.1016/j.pharmthera.2020.107706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Yao Y., Guo Q., Cao Y., et al. Artemisinin derivatives inactivate cancer-associated fibroblasts through suppressing TGF-β signaling in breast cancer. J Exp Clin Cancer Res. 2018;37:1–14. doi: 10.1186/s13046-018-0960-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Shen Y., Yang Y., Li Y. MiR-133a acts as a tumor suppressor in lung cancer progression by regulating the LASP1 and TGF-β/Smad3 signaling pathway. Thorac Cancer. 2020;11:3473–3481. doi: 10.1111/1759-7714.13678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Fountain J.H., Lappin S.L. StatPearls. StatPearls Publishing; Treasure Island (FL): March 12, 2023. Physiology, renin angiotensin system. [PubMed] [Google Scholar]
- 116.wei Xu Z., xue Yan S., xun Wu H., et al. The influence of TNF-α and Ang II on the proliferation, migration and invasion of HepG2 cells by regulating the expression of GRK2. Cancer Chemother Pharmacol. 2017;79:747–758. doi: 10.1007/s00280-017-3267-z. [DOI] [PubMed] [Google Scholar]
- 117.Shah S., Famta P., Tiwari V., et al. Instigation of the epoch of nanovaccines in cancer immunotherapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2023;15 doi: 10.1002/WNAN.1870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Li L., Yu R., Cai T., et al. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int Immunopharmacol. 2020;88 doi: 10.1016/j.intimp.2020.106939. [DOI] [PubMed] [Google Scholar]
- 119.Jing Y.Y., Han Z.P., Sun K., et al. Toll-like receptor 4 signaling promotes epithelial-mesenchymal transition in human hepatocellular carcinoma induced by lipopolysaccharide. BMC Med. 2012;10:1–12. doi: 10.1186/1741-7015-10-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Ramos R.N., Rodriguez C., Hubert M., et al. CD163+ tumor-associated macrophage accumulation in breast cancer patients reflects both local differentiation signals and systemic skewing of monocytes. Clin Transl Immunol. 2020;9:e1108. doi: 10.1002/cti2.1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Nie G., Cao X., Mao Y., et al. Tumor-associated macrophages-mediated CXCL8 infiltration enhances breast cancer metastasis: suppression by danirixin. Int Immunopharmacol. 2021;95 doi: 10.1016/j.intimp.2020.107153. [DOI] [PubMed] [Google Scholar]
- 122.Zhu B., Zhang X., Sun S., Fu Y., Xie L., Ai P. NF-κB and neutrophil extracellular traps cooperate to promote breast cancer progression and metastasis. Exp Cell Res. 2021;405 doi: 10.1016/j.yexcr.2021.112707. [DOI] [PubMed] [Google Scholar]
- 123.Tyagi A., Sharma S., Wu K., et al. Nicotine promotes breast cancer metastasis by stimulating N2 neutrophils and generating pre-metastatic niche in lung. Nat Commun. 2021;12:1–18. doi: 10.1038/s41467-020-20733-9. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 124.Chen N., Feng Q., Deng J., et al. Hdc-expressing myeloid-derived suppressor cells promote basal-like transition and metastasis of breast cancer. Int J Clin Exp Pathol. 2020;13:1431–1443. [PMC free article] [PubMed] [Google Scholar]
- 125.Ma X., Wang M., Yin T., Zhao Y., Wei X. Myeloid-derived suppressor cells promote metastasis in breast cancer after the stress of operative removal of the primary cancer. Front Oncol. 2019;9:855. doi: 10.3389/fonc.2019.00855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hsu Y.L., Yen M.C., Chang W.A., et al. CXCL17-derived CD11b+Gr-1+ myeloid-derived suppressor cells contribute to lung metastasis of breast cancer through platelet-derived growth factor-BB. Breast Cancer Res. 2019;21:1–13. doi: 10.1186/s13058-019-1114-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Luo A., Meng M., Wang G., et al. Myeloid-derived suppressor cells recruited by chemokine (C-c motif) ligand 3 promote the progression of breast cancer via phosphoinositide 3-kinase-protein kinase b-mammalian target of rapamycin signaling. J Breast Cancer. 2020;23:141–161. doi: 10.4048/jbc.2020.23.e26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Peng D., Tanikawa T., Li W., et al. Myeloid-derived suppressor cells endow stem-like qualities to breast cancer cells through IL6/STAT3 and NO/NOTCH cross-talk signaling. Cancer Res. 2016;76:3156–3165. doi: 10.1158/0008-5472.CAN-15-2528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Kumar S., Wilkes D.W., Samuel N., et al. Δnp63-Driven recruitment of myeloid-derived suppressor cells promotes metastasis in triple-negative breast cancer. J Clin Investig. 2018;128:5095–5109. doi: 10.1172/JCI99673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Song J.H., Eum D.Y., Park S.Y., et al. Inhibitory effect of ginsenoside Rg3 on cancer stemness and mesenchymal transition in breast cancer via regulation of myeloidderived suppressor cells. PLoS One. 2020;15 doi: 10.1371/journal.pone.0240533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lin L., Luo X., Wang L., et al. BML-111 inhibits EMT, migration and metastasis of TAMs-stimulated triple-negative breast cancer cells via ILK pathway. Int Immunopharmacol. 2020;85 doi: 10.1016/j.intimp.2020.106625. [DOI] [PubMed] [Google Scholar]
- 132.Yang R., Xie Y., Li Q., et al. Ruyiping extract reduces lung metastasis in triple negative breast cancer by regulating macrophage polarization. Biomed Pharmacother. 2021;141 doi: 10.1016/j.biopha.2021.111883. [DOI] [PubMed] [Google Scholar]
- 133.Yue S., Ye X., Zhou T., et al. PGRN−/− TAMs-derived exosomes inhibit breast cancer cell invasion and migration and its mechanism exploration. Life Sci. 2021;264:118687. doi: 10.1016/j.lfs.2020.118687. [DOI] [PubMed] [Google Scholar]
- 134.Wu R.S., Hong J.J., Wu J.F., et al. OVOL2 antagonizes TGF-β signaling to regulate epithelial to mesenchymal transition during mammary tumor metastasis. Oncotarget. 2017;8:39401–39416. doi: 10.18632/oncotarget.17031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Pham K., Huynh D., Le L., et al. E-cigarette promotes breast carcinoma progression and lung metastasis: macrophage-tumor cells crosstalk and the role of CCL5 and VCAM-1. Cancer Lett. 2020;491:132–145. doi: 10.1016/j.canlet.2020.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Khalid A., Wolfram J., Ferrari I., et al. Recent advances in discovering the role of CCL5 in metastatic breast cancer. Mini Rev Med Chem. 2015;15:1063–1072. doi: 10.2174/138955751513150923094709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Neamatallah T. Mitogen-activated protein kinase pathway: a critical regulator in tumor-associated macrophage polarization. J Microsc Ultrastruct. 2019;7:53–56. doi: 10.4103/jmau.jmau_68_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Jain N., Srinivasarao D.A., Famta P., et al. The portrayal of macrophages as tools and targets: a paradigm shift in cancer management. Life Sci. 2023;316 doi: 10.1016/J.LFS.2023.121399. [DOI] [PubMed] [Google Scholar]
- 139.Wu Y., Zhou B.P. TNF-α/NFκ-B/Snail pathway in cancer cell migration and invasion. Br J Cancer. 2010;102:639–644. doi: 10.1038/sj.bjc.6605530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Fu L.Q., Du W.L., Cai M.H., Yao J.Y., Zhao Y.Y., Mou X.Z. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol. 2020;353 doi: 10.1016/j.cellimm.2020.104119. [DOI] [PubMed] [Google Scholar]
- 141.Zhang W. jie, hua Wang X., Gao S. ting, et al. Tumor-associated macrophages correlate with phenomenon of epithelial-mesenchymal transition and contribute to poor prognosis in triple-negative breast cancer patients. J Surg Res. 2018;222:93–101. doi: 10.1016/j.jss.2017.09.035. [DOI] [PubMed] [Google Scholar]
- 142.Bednarczyk R.B., Tuli N.Y., Hanly E.K., et al. Macrophage inflammatory factors promote epithelial-mesenchymal transition in breast cancer. Oncotarget. 2018;9:24272–24282. doi: 10.18632/oncotarget.24917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Zhang S.C., Hu Z.Q., Long J.H., et al. Clinical implications of tumor-infiltrating immune cells in breast cancer. J Cancer. 2019;10:6175–6184. doi: 10.7150/jca.35901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Tan G.J., Peng Z.K., Lu J.P., Tang F.Q. Cathepsins mediate tumor metastasis. World J Biol Chem. 2013;4:91–101. doi: 10.4331/wjbc.v4.i4.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Wu X., Pan C., Chen R., Zhang S., Zhai Y., Guo H. BML-111 attenuates high glucose-induced inflammation, oxidative stress and reduces extracellular matrix accumulation via targeting Nrf2 in rat glomerular mesangial cells. Int Immunopharmacol. 2020;79 doi: 10.1016/j.intimp.2019.106108. [DOI] [PubMed] [Google Scholar]
- 146.Serrero G. Autocrine growth factor revisited: PC-cell-derived growth factor (progranulin), a critical player in breast cancer tumorigenesis. Biochem Biophys Res Commun. 2003;308:409–413. doi: 10.1016/S0006-291X(03)01452-9. [DOI] [PubMed] [Google Scholar]
- 147.Tsuji G., Ito T., Chiba T., et al. The role of the OVOL1–OVOL2 axis in normal and diseased human skin. J Dermatol Sci. 2018;90:227–231. doi: 10.1016/j.jdermsci.2018.02.005. [DOI] [PubMed] [Google Scholar]
- 148.David J.M., Dominguez C., Hamilton D.H., Palena C. The IL-8/IL-8R axis: a double agent in tumor immune resistance. Vaccines. 2016;4:22. doi: 10.3390/vaccines4030022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Phatale V., Famta P., Srinivasarao D.A., et al. Neutrophil membrane-based nanotherapeutics: propitious paradigm shift in the management of cancer. Life Sci. 2023;331 doi: 10.1016/J.LFS.2023.122021. [DOI] [PubMed] [Google Scholar]
- 150.Masucci M.T., Minopoli M., Carriero M.V. Tumor associated neutrophils. Their role in tumorigenesis, metastasis, prognosis and therapy. Front Oncol. 2019;9:1146. doi: 10.3389/fonc.2019.01146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Masucci M.T., Minopoli M., Del Vecchio S., Carriero M.V. The emerging role of neutrophil extracellular traps (NETs) in tumor progression and metastasis. Front Immunol. 2020;11:1749. doi: 10.3389/fimmu.2020.01749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Martins-Cardoso K., Almeida V.H., Bagri K.M., et al. Neutrophil extracellular traps (Nets) promote pro-metastatic phenotype in human breast cancer cells through epithelial–mesenchymal transition. Cancers. 2020;12:1–16. doi: 10.3390/cancers12061542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Wang J., Li J.Z., Lu A.X., Zhang K.F., Li B.J. Anticancer effect of salidroside on A549 lung cancer cells through inhibition of oxidative stress and phospho-p38 expression. Oncol Lett. 2014;7:1159–1164. doi: 10.3892/ol.2014.1863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Cai J., Cui Y., Yang J., Wang S. Epithelial-mesenchymal transition: when tumor cells meet myeloid-derived suppressor cells. Biochim Biophys Acta Rev Cancer. 2021;1876 doi: 10.1016/j.bbcan.2021.188564. [DOI] [PubMed] [Google Scholar]
- 155.Won W.J., Deshane J.S., Leavenworth J.W., Oliva C.R., Griguer C.E. Metabolic and functional reprogramming of myeloid-derived suppressor cells and their therapeutic control in glioblastoma. Cell Stress. 2019;3:47–65. doi: 10.15698/cst2019.02.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Hegde S., Leader A.M., Merad M. MDSC: markers, development, states, and unaddressed complexity. Immunity. 2021;54:875–884. doi: 10.1016/j.immuni.2021.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Pastaki Khoshbin A., Eskian M., Keshavarz-Fathi M., Rezaei N. Roles of myeloid-derived suppressor cells in cancer metastasis: immunosuppression and beyond. Arch Immunol Ther Exp. 2019;67:89–102. doi: 10.1007/s00005-018-0531-9. [DOI] [PubMed] [Google Scholar]
- 158.Chiodoni C., Sangaletti S., Colombo M.P. Matricellular proteins tune myeloid-derived suppressor cell recruitment and function in breast cancer. J Leukoc Biol. 2017;102:287–292. doi: 10.1189/jlb.3mr1016-447r. [DOI] [PubMed] [Google Scholar]
- 159.Ma J., Xu H., Wang S. Immunosuppressive role of myeloid-derived suppressor cells and therapeutic targeting in lung cancer. J Immunol Res. 2018;2018 doi: 10.1155/2018/6319649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Zhu H., Gu Y., Xue Y., Yuan M., Cao X., Liu Q. CXCR2+ MDSCs promote breast cancer progression by inducing EMT and activated T cell exhaustion. Oncotarget. 2017;8:114554–114567. doi: 10.18632/oncotarget.23020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Sun M., Ye Y., Xiao L., Duan X., Zhang Y., Zhang H. Anticancer effects of ginsenoside Rg3. Int J Mol Med. 2017;39:507–518. doi: 10.3892/ijmm.2017.2857. [DOI] [PubMed] [Google Scholar]
- 162.Annibali O., Crescenzi A., Tomarchio V., et al. PD-1/PD-L1 checkpoint in hematological malignancies. Leuk Res. 2018;67:45–55. doi: 10.1016/j.leukres.2018.01.014. [DOI] [PubMed] [Google Scholar]
- 163.Almozyan S., Colak D., Mansour F., et al. PD-L1 promotes OCT4 and Nanog expression in breast cancer stem cells by sustaining PI3K/AKT pathway activation. Int J Cancer. 2017;141:1402–1412. doi: 10.1002/ijc.30834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Alsuliman A., Colak D., Al-Harazi O., et al. Bidirectional crosstalk between PD-L1 expression and epithelial to mesenchymal transition: significance in claudin-low breast cancer cells. Mol Cancer. 2015;14:1–13. doi: 10.1186/s12943-015-0421-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Xiao M., Hasmim M., Lequeux A., et al. Epithelial to mesenchymal transition regulates surface pd-l1 via cmtm6 and cmtm7 induction in breast cancer. Cancers. 2021;13:1–12. doi: 10.3390/cancers13051165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Zhang A., Qi X., Du F., Zhang G., Li D., Li J. PNSA, a novel C-terminal inhibitor of HSP90, reverses epithelial-mesenchymal transition and suppresses metastasis of breast cancer cells in vitro. Mar Drugs. 2021;19:117. doi: 10.3390/md19020117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Noman M.Z., Janji B., Abdou A., et al. The immune checkpoint ligand PD-l1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. OncoImmunology. 2017;6 doi: 10.1080/2162402X.2016.1263412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Saleh R., Taha R.Z., Nair V.S., Alajez N.M., Elkord E. PD-L1 blockade by atezolizumab downregulates signaling pathways associated with tumor growth, metastasis, and hypoxia in human triple negative breast cancer. Cancers. 2019;11:1050. doi: 10.3390/cancers11081050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Peng F., Xiong L., Peng C. (-)-Sativan inhibits tumor development and regulates miR-200c/PD-L1 in triple negative breast cancer cells. Front Pharmacol. 2020;11:251. doi: 10.3389/fphar.2020.00251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Rizvi A., Merlin M.A., Shah G.M. Poly (ADP-ribose) polymerase (PARP) inhibition in cancer: potential impact in cancer stem cells and therapeutic implications. Eur J Pharmacol. 2021;911 doi: 10.1016/j.ejphar.2021.174546. [DOI] [PubMed] [Google Scholar]
- 171.Sun X., Wang X., Zhang J., et al. Efficacy and safety of PARP inhibitors in patients with BRCA-mutated advanced breast cancer: a meta-analysis and systematic review. Breast. 2021;60:26–34. doi: 10.1016/j.breast.2021.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Han Y., Li C.W., Hsu J.M., et al. Metformin reverses PARP inhibitors-induced epithelial-mesenchymal transition and PD-L1 upregulation in triple-negative breast cancer. Am J Cancer Res. 2019;9:800–815. [PMC free article] [PubMed] [Google Scholar]
- 173.Hao X., Ren Y., Feng M., Wang Q., Wang Y. Metabolic reprogramming due to hypoxia in pancreatic cancer: implications for tumor formation, immunity, and more. Biomed Pharmacother. 2021;141 doi: 10.1016/j.biopha.2021.111798. [DOI] [PubMed] [Google Scholar]
- 174.Xiong Q., Liu B., Ding M., Zhou J., Yang C., Chen Y. Hypoxia and cancer related pathology. Cancer Lett. 2020;486:1–7. doi: 10.1016/j.canlet.2020.05.002. [DOI] [PubMed] [Google Scholar]
- 175.Hajizadeh F., Okoye I., Esmaily M., et al. Hypoxia inducible factors in the tumor microenvironment as therapeutic targets of cancer stem cells. Life Sci. 2019;237 doi: 10.1016/j.lfs.2019.116952. [DOI] [PubMed] [Google Scholar]
- 176.Pan Z., Ma G., Kong L., Du G. Hypoxia-inducible factor-1: regulatory mechanisms and drug development in stroke. Pharmacol Res. 2021;170 doi: 10.1016/j.phrs.2021.105742. [DOI] [PubMed] [Google Scholar]
- 177.Liu Z. Ji, Semenza G.L., Zhang H. Feng. Hypoxia-inducible factor 1 and breast cancer metastasis. J Zhejiang Univ Sci B. 2015;16:32–43. doi: 10.1631/jzus.B1400221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Lei J., Fan L., Wei G., et al. Gli-1 is crucial for hypoxia-induced epithelial-mesenchymal transition and invasion of breast cancer. Tumor Biol. 2015;36:3119–3126. doi: 10.1007/s13277-014-2948-z. [DOI] [PubMed] [Google Scholar]
- 179.Yang H., Geng Y.H., Wang P., et al. Extracellular ATP promotes breast cancer invasion and epithelial-mesenchymal transition via hypoxia-inducible factor 2α signaling. Cancer Sci. 2019;110:2456–2470. doi: 10.1111/cas.14086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Dey S., Sayers C.M., Verginadis, et al. ATF4-dependent induction of heme oxygenase 1 prevents anoikis and promotes metastasis. J Clin Investig. 2015;125:2592–2608. doi: 10.1172/JCI78031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Zeng P., Sun S., Li R., Xiao Z.X., Chen H. HER2 upregulates ATF4 to promote cell migration via activation of ZEB1 and downregulation of E-cadherin. Int J Mol Sci. 2019;20:2223. doi: 10.3390/IJMS20092223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Gonzalez-González A., Muñoz-Muela E., Marchal J.A., et al. Activating transcription factor 4 modulates TGFb-induced aggressiveness in triple-negative breast cancer via SMad2/3/4 and mTORC2 signaling. Clin Cancer Res. 2018;24:5697–5709. doi: 10.1158/1078-0432.CCR-17-3125/274568/AM/ACTIVATING-TRANSCRIPTION-FACTOR-4-MODULATES-TGF. [DOI] [PubMed] [Google Scholar]
- 183.Peng P.H., Chieh-Yu Lai J., Hsu K.W., Wu K.J. Hypoxia-induced lncRNA RP11-390F4.3 promotes epithelial-mesenchymal transition (EMT) and metastasis through upregulating EMT regulators. Cancer Lett. 2020;483:35–45. doi: 10.1016/j.canlet.2020.04.014. [DOI] [PubMed] [Google Scholar]
- 184.Wang X., Yu M., Zhao K., et al. Upregulation of miR-205 under hypoxia promotes epithelial–mesenchymal transition by targeting ASPP2. Cell Death Dis. 2016;7 doi: 10.1038/cddis.2016.412. e2517–e2517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.sha Wang S., Gu Q., Liu N., Li J., Liu X. Aerobic exercise attenuates ectopic renal sinus adipose tissue accumulation-related renal hypoxia injury in obese mice. Life Sci. 2021;279 doi: 10.1016/j.lfs.2021.119106. [DOI] [PubMed] [Google Scholar]
- 186.Aiwei Y.B., Behjatolah M.K., Hedges R.A., Rogers L.J., Kadlubar S.A., Thomas K.E. Adipocyte hypoxia promotes epithelial-mesenchymal transition-related gene expression and estrogen receptor-negative phenotype in breast cancer cells. Oncol Rep. 2015;33:2689–2694. doi: 10.3892/or.2015.3880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Avtanski D., Garcia A., Caraballo B., et al. Resistin induces breast cancer cells epithelial to mesenchymal transition (EMT) and stemness through both adenylyl cyclase-associated protein 1 (CAP1)-dependent and CAP1-independent mechanisms. Cytokine. 2019;120:155–164. doi: 10.1016/j.cyto.2019.04.016. [DOI] [PubMed] [Google Scholar]
- 188.Zhou Z., Wang S., Song C., Hu Z. Paeoniflorin prevents hypoxia-induced epithelial-mesenchymal transition in human breast cancer cells. OncoTargets Ther. 2016;9:2511–2518. doi: 10.2147/OTT.S102422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Li W., Xue D., Xue M., et al. Fucoidan inhibits epithelial-to-mesenchymal transition via regulation of the HIF-1α pathway in mammary cancer cells under hypoxia. Oncol Lett. 2019;18:330–338. doi: 10.3892/ol.2019.10283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Wang Y., Xu X., Zhao P., Tong B., Wei Z., Dai Y. Escin Ia suppresses the metastasis of triple-negative breast cancer by inhibiting epithelial mesenchymal transition via down-regulating LOXL2 expression. Oncotarget. 2016;7:23684–23699. doi: 10.18632/oncotarget.8152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Hong D., Fritz A.J., Zaidi S.K., et al. Epithelial-to-mesenchymal transition and cancer stem cells contribute to breast cancer heterogeneity. J Cell Physiol. 2018;233:9136–9144. doi: 10.1002/jcp.26847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Prager B.C., Xie Q., Bao S., Rich J.N. Cancer stem cells: the architects of the tumor ecosystem. Cell Stem Cell. 2019;24:41–53. doi: 10.1016/j.stem.2018.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Lawson D.A., Bhakta N.R., Kessenbrock K., et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature. 2015;526:131–135. doi: 10.1038/nature15260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.del Pozo Martin Y., Park D., Ramachandran A., et al. Mesenchymal cancer cell-stroma crosstalk promotes niche activation, epithelial reversion, and metastatic colonization. Cell Rep. 2015;13:2456–2469. doi: 10.1016/j.celrep.2015.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Zhou J., Yi Q., Tang L. The roles of nuclear focal adhesion kinase (FAK) on Cancer: a focused review. J Exp Clin Cancer Res. 2019;38:1–11. doi: 10.1186/s13046-019-1265-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Ni T., Li X.Y., Lu N., et al. Snail1-dependent p53 repression regulates expansion and activity of tumour-initiating cells in breast cancer. Nat Cell Biol. 2016;18:1221–1232. doi: 10.1038/ncb3425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Mal A., Bukhari A.B., Singh R.K., et al. EpCAM-mediated cellular plasticity promotes radiation resistance and metastasis in breast cancer. Front Cell Dev Biol. 2021;8:1637. doi: 10.3389/fcell.2020.597673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Keller L., Werner S., Pantel K. Biology and clinical relevance of EpCAM. Cell Stress. 2019;3:165–180. doi: 10.15698/cst2019.06.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Zhang D., Yang L., Liu X., et al. Hypoxia modulates stem cell properties and induces EMT through N-glycosylation of EpCAM in breast cancer cells. J Cell Physiol. 2020;235:3626–3633. doi: 10.1002/jcp.29252. [DOI] [PubMed] [Google Scholar]
- 200.Shi R., Liu L., Wang F., et al. Downregulation of cytokeratin 18 induces cellular partial EMT and stemness through increasing EpCAM expression in breast cancer. Cell Signal. 2020;76 doi: 10.1016/j.cellsig.2020.109810. [DOI] [PubMed] [Google Scholar]
- 201.Ko Y.S., Jin H., Lee J.S., et al. Radioresistant breast cancer cells exhibit increased resistance to chemotherapy and enhanced invasive properties due to cancer stem cells. Oncol Rep. 2018;40:3752–3762. doi: 10.3892/or.2018.6714. [DOI] [PubMed] [Google Scholar]
- 202.Hiyama E., Hiyama K. Telomere and telomerase in stem cells. Br J Cancer. 2007;96:1020–1024. doi: 10.1038/sj.bjc.6603671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.El-Badawy A., Ghoneim N.I., Nasr M.A., et al. Telomerase reverse transcriptase coordinates with the epithelial-to-mesenchymal transition through a feedback loop to define properties of breast cancer stem cells. Biol Open. 2018;7 doi: 10.1242/bio.034181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Kusoglu A., Goker Bagca B., Ozates Ay N.P., Gunduz C., Biray Avci C. Telomerase inhibition regulates EMT mechanism in breast cancer stem cells. Gene. 2020;759 doi: 10.1016/j.gene.2020.145001. [DOI] [PubMed] [Google Scholar]
- 205.Kim J., Villadsen R., Sørlie T., et al. Tumor initiating but differentiated luminal-like breast cancer cells are highly invasive in the absence of basal-like activity. Proc Natl Acad Sci U S A. 2012;109:6124–6129. doi: 10.1073/pnas.1203203109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Li X., Strietz J., Bleilevens A., Stickeler E., Maurer J. Chemotherapeutic stress influences epithelial– mesenchymal transition and stemness in cancer stem cells of triple-negative breast cancer. Int J Mol Sci. 2020;21:404. doi: 10.3390/ijms21020404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Reiman J.M., Knutson K.L., Radisky D.C. Immune promotion of epithelial-mesenchymal transition and generation of breast cancer stem cells. Cancer Res. 2010;70:3005–3008. doi: 10.1158/0008-5472.CAN-09-4041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Xie G., Yao Q., Liu Y., et al. IL-6-induced epithelial-mesenchymal transition promotes the generation of breast cancer stem-like cells analogous to mammosphere cultures. Int J Oncol. 2012;40:1171–1179. doi: 10.3892/ijo.2011.1275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Yang L., Han S., Sun Y. An IL6-STAT3 loop mediates resistance to PI3K inhibitors by inducing epithelial-mesenchymal transition and cancer stem cell expansion in human breast cancer cells. Biochem Biophys Res Commun. 2014;453:582–587. doi: 10.1016/j.bbrc.2014.09.129. [DOI] [PubMed] [Google Scholar]
- 210.Bierie B., Pierce S.E., Kroeger C., et al. Integrin-β4 identifies cancer stem cell-enriched populations of partially mesenchymal carcinoma cells. Proc Natl Acad Sci U S A. 2017;114:E2337–E2346. doi: 10.1073/pnas.1618298114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Jolly M.K., Jia D., Boareto M., et al. Coupling the modules of EMT and stemness: a tunable “stemness window” model. Oncotarget. 2015;6:25161–25174. doi: 10.18632/oncotarget.4629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Zhao W., Wang M., Cai M., et al. Transcriptional co-activators YAP/TAZ: potential therapeutic targets for metastatic breast cancer. Biomed Pharmacother. 2021;133 doi: 10.1016/j.biopha.2020.110956. [DOI] [PubMed] [Google Scholar]
- 213.Liu-Chittenden Y., Huang B., Shim J.S., et al. Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev. 2012;26:1300–1305. doi: 10.1101/gad.192856.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Oku Y., Nishiya N., Shito T., et al. Small molecules inhibiting the nuclear localization of YAP/TAZ for chemotherapeutics and chemosensitizers against breast cancers. FEBS Open Bio. 2015;5:542–549. doi: 10.1016/j.fob.2015.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Nouri K., Azad T., Ling M., et al. Identification of celastrol as a Novel YAP-TEAD inhibitor for cancer therapy by high throughput screening with ultrasensitive YAP/TAZ-TEAD biosensors. Cancers. 2019;11:1596. doi: 10.3390/cancers11101596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Cao D., Zhu G.Y., Lu Y., et al. Luteolin suppresses epithelial-mesenchymal transition and migration of triple-negative breast cancer cells by inhibiting YAP/TAZ activity. Biomed Pharmacother. 2020;129 doi: 10.1016/j.biopha.2020.110462. [DOI] [PubMed] [Google Scholar]
- 217.Zhang H., Cui X., Cao A., Li X., Li L. ITGA3 interacts with VASP to regulate stemness and epithelial-mesenchymal transition of breast cancer cells. Gene. 2020;734 doi: 10.1016/j.gene.2020.144396. [DOI] [PubMed] [Google Scholar]
- 218.Carbognin L., Miglietta F., Paris I., Dieci M.V. Prognostic and predictive implications of PTEN in breast cancer: unfulfilled promises but intriguing perspectives. Cancers. 2019;11:1401. doi: 10.3390/cancers11091401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Chao J.C., Qi Y., Liu J., Scheuerman M.P., Lee L.Y., Li S. PTEN suppresses epithelial-mesenchymal transition and breast cancer stem cell activity by down regulating Abi1. J Am Coll Surg. 2020;231:e86–e87. doi: 10.1016/j.jamcollsurg.2020.08.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Natarajan K., Xie Y., Baer M.R., Ross D.D. Role of breast cancer resistance protein (BCRP/ABCG2) in cancer drug resistance. Biochem Pharmacol. 2012;83:1084–1103. doi: 10.1016/J.BCP.2012.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Kim J.H., Moon M.J., Kim D.Y., Heo S.H., Jeong Y.Y. Hyaluronic acid-based nanomaterials for cancer therapy. Polymers. 2018;10:1133. doi: 10.3390/polym10101133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Wu W., Chen L., Wang Y., Jin J., Xie X., Zhang J. Hyaluronic acid predicts poor prognosis in breast cancer patients: a protocol for systematic review and meta analysis. Medicine. 2020;99 doi: 10.1097/MD.0000000000020438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Jariyal H., Gupta C., Srivastava A. Hyaluronic acid induction on breast cancer stem cells unfolds subtype specific variations in stemness and epithelial-to-mesenchymal transition. Int J Biol Macromol. 2020;160:1078–1089. doi: 10.1016/j.ijbiomac.2020.05.236. [DOI] [PubMed] [Google Scholar]
- 224.Famta P., Shah S., Jain N., et al. Tumor-promoting aftermath post-chemotherapy: a focus on breast cancer. Life Sci. 2022;310 doi: 10.1016/J.LFS.2022.121125. [DOI] [PubMed] [Google Scholar]
- 225.Gooding A.J., Schiemann W.P. Epithelial-mesenchymal transition programs and cancer stem cell phenotypes: mediators of breast cancer therapy resistance. Mol Cancer Res. 2020;18:1257–1270. doi: 10.1158/1541-7786.MCR-20-0067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Kaur B., Mukhlis Y., Natesh J., Penta D., Musthapa Meeran S. Identification of hub genes associated with EMT-induced chemoresistance in breast cancer using integrated bioinformatics analysis. Gene. 2022;809 doi: 10.1016/j.gene.2021.146016. [DOI] [PubMed] [Google Scholar]
- 227.Szebényi K., Füredi A., Bajtai E., et al. Effective targeting of breast cancer by the inhibition of P-glycoprotein mediated removal of toxic lipid peroxidation byproducts from drug tolerant persister cells. Drug Resist Updat. 2023;71 doi: 10.1016/J.DRUP.2023.101007. [DOI] [PubMed] [Google Scholar]
- 228.Yang X., Hu Q., Hu L.X., et al. miR-200b regulates epithelial-mesenchymal transition of chemo-resistant breast cancer cells by targeting FN1. Discov Med. 2017;24:75–85. [PubMed] [Google Scholar]
- 229.Zhang W., Feng M., Zheng G., et al. Chemoresistance to 5-fluorouracil induces epithelial-mesenchymal transition via up-regulation of Snail in MCF7 human breast cancer cells. Biochem Biophys Res Commun. 2012;417:679–685. doi: 10.1016/j.bbrc.2011.11.142. [DOI] [PubMed] [Google Scholar]
- 230.Qu C., Zhang W., Zheng G., Zhang Z., Yin J., He Z. Metformin reverses multidrug resistance and epithelial-mesenchymal transition (EMT) via activating AMP-activated protein kinase (AMPK) in human breast cancer cells. Mol Cell Biochem. 2014;386:63–71. doi: 10.1007/s11010-013-1845-x. [DOI] [PubMed] [Google Scholar]
- 231.Dong H., Hu J., Zou K., et al. Activation of LncRNA TINCR by H3K27 acetylation promotes trastuzumab resistance and epithelial-mesenchymal transition by targeting MicroRNA-125b in breast cancer 11 medical and health sciences 1112 oncology and carcinogenesis. Mol Cancer. 2019;18:1–18. doi: 10.1186/s12943-018-0931-9. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 232.Jiang L., He D., Yang D., et al. MiR-489 regulates chemoresistance in breast cancer via epithelial mesenchymal transition pathway. FEBS Lett. 2014;588:2009–2015. doi: 10.1016/j.febslet.2014.04.024. [DOI] [PubMed] [Google Scholar]
- 233.Yang Q., Huang J., Wu Q., et al. Acquisition of epithelial-mesenchymal transition is associated with Skp2 expression in paclitaxel-resistant breast cancer cells. Br J Cancer. 2014;110:1958–1967. doi: 10.1038/bjc.2014.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Du F., Yu L., Wu Y., et al. miR-137 alleviates doxorubicin resistance in breast cancer through inhibition of epithelial-mesenchymal transition by targeting DUSP4. Cell Death Dis. 2019;10:922. doi: 10.1038/s41419-019-2164-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Yang Q., Wang Y., Lu X., et al. MiR-125b regulates epithelial-mesenchymal transition via targeting Sema4C in paclitaxel-resistant breast cancer cells. Oncotarget. 2015;6:3268–3279. doi: 10.18632/oncotarget.3065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Cai J., Sun H., Zheng B., et al. Curcumin attenuates lncRNA H19-induced epithelial-mesenchymal transition in tamoxifen-resistant breast cancer cells. Mol Med Rep. 2021;23:13. doi: 10.3892/mmr.2020.11651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Jin X., Wei Y., Liu Y., et al. Resveratrol promotes sensitization to doxorubicin by inhibiting epithelial-mesenchymal transition and modulating SIRT1/β-catenin signaling pathway in breast cancer. Cancer Med. 2019;8:1246–1257. doi: 10.1002/cam4.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Liu J., Chen X., Ward T., Pegram M., Shen K. Combined niclosamide with cisplatin inhibits epithelial-mesenchymal transition and tumor growth in cisplatin-resistant triple-negative breast cancer. Tumor Biol. 2016;37:9825–9835. doi: 10.1007/s13277-015-4650-1. [DOI] [PubMed] [Google Scholar]
- 239.Li Q.Q., Xu J. Da, Wang W.J., et al. Twist1-mediated adriamycin-induced epithelial-mesenchymal transition relates to multidrug resistance and invasive potential in breast cancer cells. Clin Cancer Res. 2009;15:2657–2665. doi: 10.1158/1078-0432.CCR-08-2372. [DOI] [PubMed] [Google Scholar]
- 240.Sivanesan D., Arun R.P., Verma R.S. Effect of salinomycin on EMT and stemness pathways in 5-FU-resistant breast cancer. Adv Cancer Biol Metastasis. 2021;1 doi: 10.1016/j.adcanc.2021.100004. [DOI] [Google Scholar]
- 241.Fedorova O., Daks A., Parfenyev S., et al. Zeb1-mediated autophagy enhances resistance of breast cancer cells to genotoxic drugs. Biochem Biophys Res Commun. 2022;589:29–34. doi: 10.1016/j.bbrc.2021.11.088. [DOI] [PubMed] [Google Scholar]
- 242.Das C.K., Parekh A., Parida P.K., Bhutia S.K., Mandal M. Lactate dehydrogenase A regulates autophagy and tamoxifen resistance in breast cancer. Biochim Biophys Acta Mol Cell Res. 2019;1866:1004–1018. doi: 10.1016/j.bbamcr.2019.03.004. [DOI] [PubMed] [Google Scholar]
- 243.Bauer R., Udonta F., Wroblewski M., et al. Blockade of myeloid-derived suppressor cell expansion with all-trans retinoic acid increases the efficacy of antiangiogenic therapy. Cancer Res. 2018;78:3220–3232. doi: 10.1158/0008-5472.CAN-17-3415. [DOI] [PubMed] [Google Scholar]
- 244.Famta P., Shah S., Jain N., et al. Nanocarrier-based drug delivery via cell-hitchhiking: emphasizing pharmacokinetic perspective towards taming the “big-old” tumors. J Drug Deliv Sci Technol. 2023;89 doi: 10.1016/J.JDDST.2023.105050. [DOI] [Google Scholar]
- 245.Famta P., Shah S., Jain N., et al. Albumin-hitchhiking: fostering the pharmacokinetics and anticancer therapeutics. J Control Release. 2023;353:166–185. doi: 10.1016/j.jconrel.2022.11.034. [DOI] [PubMed] [Google Scholar]
- 246.Jain N., Shahrukh S., Famta P., et al. Immune cell–camouflaged surface-engineered nanotherapeutics for cancer management. Acta Biomater. 2023;155:57–79. doi: 10.1016/J.ACTBIO.2022.11.001. [DOI] [PubMed] [Google Scholar]
- 247.Bagasariya D., Charankumar K., Shah S., et al. Biomimetic nanotherapeutics: employing nanoghosts to fight melanoma. Eur J Pharm Biopharm. 2022;177:157–174. doi: 10.1016/J.EJPB.2022.06.014. [DOI] [PubMed] [Google Scholar]
- 248.Shah S., Famta P., Srinivasarao D.A., et al. Unravelling the potential of microbots in cancer therapy. Appl Mater Today. 2023;34 doi: 10.1016/J.APMT.2023.101887. [DOI] [Google Scholar]
- 249.Famta P., Shah S., Fernandes V., et al. Quality by design (QbD) assisted fabrication & evaluation of simvastatin loaded nano-enabled thermogel for melanoma therapy. Int J Pharm. 2022;628 doi: 10.1016/J.IJPHARM.2022.122270. [DOI] [PubMed] [Google Scholar]
- 250.Famta P., Shah S., Jain N., Shahrukh S., Bala Singh S., Srivastava S. Strategic combinatorial delivery of tranilast and paclitaxel using differently functionalized PLGA nanoparticles for enhanced penetration and accumulation in breast tumor. Med Hypotheses. 2022;169 doi: 10.1016/J.MEHY.2022.110981. [DOI] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No data was used for the research described in the manuscript.





