Abstract
The tumor microenvironment (TME) is heterogeneous and contains a multiple cell population with surrounded immune cells, which plays a major role in regulating metastasis. The multifunctional pathways, Hedgehog (Hh), Wnt, Notch, and NF-kB, cross-regulates metastasis in breast cancer. This review presents substantial evidence for cross-regulation of TME components and signaling pathways, which makes breast TME more heterogeneous and complex, promoting breast cancer progression and metastasis as a highly aggressive form. We discoursed the importance of stromal and immune cells as well as their crosstalk in bridging the metastasis. We also discussed the role of Hh and Notch pathways in the intervention between breast cancer cells and macrophages to support TME; Notch signaling in the bidirectional communication between cancer cells and components of TME; Wnt signal pathway in controlling the factors responsible for EMT and NF-κB pathway in the regulation of genes controlling the inflammatory response. We also present the role of exosomes and their miRNAs in the cross-regulation of TME cells as well as pathways in the reprogramming of breast TME to support metastasis. Finally, we examined and discussed the targeted small molecule inhibitors and natural compounds targeting developmental pathways and proposed small molecule natural compounds as potential therapeutics of TME based on the multitargeting ability. In conclusion, the understanding of the molecular basis of the cross-regulation of TME pathways and their inhibitors helps identify molecular targets for rational drug discovery to treat breast cancers.
Keywords: Breast cancer, Cross-regulation, Hedgehog, NF-κB, Notch, Tumor microenvironment, Wnt
Breast cancer
Breast cancer (BC) is one of the most common deadliest malignancies affecting women worldwide. It accounts for 30% of total cancer incidences and half a million deaths in the last two decades. The subpopulation of BCs with characteristic self-renewal capacity and differentiation is responsible for tumorigenicity and high metastatic and cancer recurrence.1 Currently, BCs are being treated by surgical, chemo, and radiation therapy, but most BC patients remain incurable and lead to high mortality in women.2 The progression and development of the metastatic stage are one of the major death-related causes of BCs. In addition, dysregulated signaling pathways also responsible for excessive cell proliferation and survival. The major signaling pathways which dysregulated are the Hedgehog pathway (Hh), Notch, Wnt, and NF-kB.3 These pathways play a central role in the proliferation, differentiation, as well as evasion of apoptosis. Recent studies have shown the importance of these pathways in the breast tumor microenvironment (TME). The breast TME has high inflammation reinforced by the infiltrated immune cells, growth factors (GFs) and cytokines. Breast tumors along with immune infiltration shows different patterns based on the presence of ER. Regulatory T-cells with negative immune feature associated with both ER-positive and ER-negative breast tumors and confess an immunosuppressive environment.4 This characteristic feature has an importance in the immune TME of breast cancer.
Tumor microenvironment
The tumor microenvironment (TME) contains multiple cell populations, such as the tumor cells, surrounded by immune cells and stromal cells, which play a critical role in promoting tumor growth and progression.1 The TME is continuously changing during the progression of tumor growth. The subtypes of breast tumors differ in the TME. The stroma consists of different connective tissues, which acts as a framework for tissues and many organs.1 Among all, fibroblasts are important in synthesizing extracellular matrix. The primary tumor mass surrounding the TME encompasses the metastatic destination, which resembles a similar network of communication. The connection between cells of TME acts as a signaling bridge, which activates angiogenesis, migration, cell adhesion, and invasiveness.5 The cell population of TME, which are non-malignant, but resides in the extracellular matrix (ECM) and constituents of matrices, are cancer-associated fibroblasts (CAFs), tumor-associated endothelial cells (TAEs), and tumor-associated osteoblasts and osteoclasts (TAOs). The cancer cell behavior is influenced by stromal cells through the secretion of ECM proteins, growth factors, cytokines, and chemokines. Stromal cells influence cancer cells by secreting various proteins via activating autocrine and paracrine loops. The stromal cell interaction and underlying genomic instability in cancer cells govern the tumor cell morphology, growth, and metastasis. Within the breast TME, the stromal cells with alterations in the molecular and aberrant signaling pathways have prognostic significance in the clinical setting.6
Intratumoral hypoxia increases the risk of metastasis and drug resistance by promoting neovascularization. It also influences metastasis and drug resistance by recruiting stromal cells, remodeling ECM, forming premetastatic niche, inducing cell migration and local invasion, and maintaining the cancer stem cell phenotype in breast TME.7 Inflamed TME promotes metastasis of breast cancer cells to bone or lung, or the brain depends on their interaction with TME cells. It also induces cancer cell proliferation, angiogenesis, invasion of apoptosis, and suppression of anti-tumor immunity. Additionally, TME frequently imparts a niche for the stay and interaction of cancer cells with surrounding stromal and immune cells. The cancer cells and stromal cells' interactions predispose breast cancer metastasis by remodeling TME cellular components, ECM, and neovascularization. TME promotes metastasis and drug resistance by inducing epithelial cell transition to stem cells.8 TME induces oncogenic metabolic reprogramming, including increased glycolytic activity, TCA cycle, glutaminolysis, and biosynthesis of fatty acids that influence breast cancer progression.9 TME also actively contribute drug resistance and heterogeneity by providing an environment for the interaction of breast cancer cells with surrounding stromal cells. Theses interactions tightly regulate vital steps of invasiveness by various signaling pathways. Recent studies explored significant alterations in gene expression in TME cells, which helps cancer progression by promoting cancer hallmarks.10 In addition to stromal cells, major immune cells infiltrate into TME and associate with the negative prognosis of BCs. They include tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), tumor-associated dendritic cells (TADCs), myeloid-derived suppressor cells (MDSCs), and regulatory T-cells (Tregs). The stromal and immune cells with unique functions shape breast TME (Fig. 1).
Tumor-associated macrophages
A subclass of myeloid cells differentiates into TAMs by the tumors within TME. The TAMs mediate different opposing function based on tumor-derived cytokines.11 They include M1 (classically activated) as well as M2 (alternatively activated) macrophages. M1 macrophages activate the target genes associated with anti-tumor immune response,12 while M2macrophages mediate an anti-inflammatory activity in response to cytokines.13 The TAMs are major mediators of the TME remodeling.
Cancer-associated fibroblasts
Fibroblasts are phenotypically similar to mesenchymal cells, form a tumor stroma. They play an important role in the secretion and remodeling of ECM.14 CAF's are the specialized group of fibroblasts, which promotes the growth and invasion of tumor cells by providing unique TME.15,16 The CAFs maintain the integrity of ECM and promotes the remodeling of ECM by secreting many proteases.17 The activated fibroblasts, which are stimulated by inflammatory condition, also promotes wound healing.
Tumor-associated endothelial cells
Proangiogenic behavior of tumor-associated endothelial cells (TAEs) is highly influenced by the hedgehog pathway, where the ligand of Hh encourages migration, invasion, and proliferation of endothelial cells. It is believed that the nuclear translocation of Gli facilitates through the activation of integrin in the presence of Hh ligands.18 The expression of MMP9 and Osteopontin, in ECs causes the phosphorylation of focal adhesion kinases.19 The TAEs are mostly responsible for tumor cell invasion, metastasis, stem cell maintenance, promotion of neutrophil infiltration, and drug resistance.20 Together, TAEs assist cancer progression and metastasis in the TME.
Tumor-associated osteoblasts and osteoclasts
The cancer-associated osteoblasts and osteoclasts (TOAs) are the centerpiece of the metastatic bone microenvironment (BME). Bone is made of mineralized matrices, and mineralized tissues promote the development of bone metastasis.21 Mineralized bone matrices contain many GFs such as IGF's, TGFβ, and FGF-1 etc.21 The most abundant GF stored in bone is IGF-1, which stimulates cell growth of breast cancer. Overexpression of IGF-1 increases the bone metastasis and reduces the apoptotic breast cancer cells.22 The IGF-1 is released from bone as a result of osteoclastic bone resorption, which plays an important role in the proliferation and inhibition of apoptosis in metastatic breast cancer cells. TGFβ is also stored in bone in large amounts and released during osteoclastic bone resorption. A subpopulation of TAOs is edified via crosstalk with BCs by secreting inflammatory cytokines in the bone TME.23 The BME has a unique biological feature, through which cancer cells circulate and proliferate.
Myeloid-derived suppressor cells
Myeloid-derived suppressor cells (MDSCs) are specialized immune cells characterized by the presence of high CD33 and CD14 and low HLA-DR in tumors. In TME, breast cancer cells induce MDSCs for infiltration and secretion of IL-6, which triggers metastasis in BCs.24 These cells are formed in the bone marrow and then move to tumor tissues. They have an important role in tumor progression by suppressing anti-tumor immunity.25 These myeloid-derived suppressor cells form a differentiation pattern, which results in the rapid differentiation of TAMs. Mostly in relation to these cells in peripheral lymphoid organs, these cells suppress the T-cell in a non-specific manner.26 They are major tumor favoring and immune suppressor cells, promotes tumor metastasis.
Tumor-associated neutrophils
Neutrophils are major kinds of myeloid cells derived as white cells and circulating in the human blood.27 The N1 neutrophils counteract the cancer cells by cytotoxicity, tumor rejection, and anti-tumor immunity. Recent studies reported that N1 neutrophils are polarized to N2 or Tumor-associated neutrophils (TANs) in the TMEby TGF-β and IFN-β. The N2 neutrophils are harmful to the host but beneficial for tumor growth, invasion, and metastasis via ECM proteolysis.28 Furthermore, ROS and proteinases released by neutrophils impact regulation of cancer cell proliferation, angiogenesis, and metastasis.29 Neutrophils significantly influence the TME by releasing cytokines and chemokines, which promotes recruitment and activation of inflammatory cells.
Tumor-associated dendritic cells (TADCs)
Dendritic cells (DCs) are compelling antigen-presenting cells, recognized as critical mediators of anti-tumor immunity. The prostaglandin E2 and β-catenin mediates the impairment of the function of tumor-associated DCs.30 Recently, regulatory DCs with immune suppressive activity are reported in TME by MDSCs.31 The TADCs interfere with the activation of NK-cell pathways or receptors that control NK-cells' activation and reduce anti-tumor activity.32 Thus, identification of TME mediated immune cells together with stromal cells, represent key questions for the design of effective therapies of breast cancers.
Regulatory T-cells
The regulatory T cells (Tregs) are essential for the maintenance of T-cell tolerance to self-antigens. High infiltration into tumors is frequently associated with poor prognosis. The Tregs have distinct characteristics, including high expression of chemokine receptor (CCR4) and CTLA-4 in breast cancers.33,34 A subsets of Tregs with high expression of immune checkpoint molecules support tumor progression.35 They diminish T-cell immunity to tumor-associated antigens.36 TME promotes differentiation and proliferation of Tregs as well as secretion of immunosuppressive signals for the recruitment of immunosuppressive cells.37 CAFs increase the ability of regulatory T cells to inhibit the proliferation of T effector cells.38 However, the proliferation of Tregs is distinctly inhibited by CDK4/6 inhibitors.39 These distinct characters offer an opportunity for the design of targeted immunotherapies.
Crosstalk between TME cells
Crosstalk between cancer cells and stromal cells
The TME promotes the progression as well as metastasis of tumor cells by acquiring specific crosstalk between stromal and cancer cells. For example, under hormonal influence, stromal cells promote tumor progression by releasing chemokines and cytokines.40 Cancer cells co-opt with reactive stromal cells by releasing pro-tumorigenic factors, which recruit other pro-tumorigenic cells for architecting TME. The interaction of stromal cells and TAMs promotes metastasis via the IL-33-ST2-dependent pathway.41 The crosstalking of cancer cells with ECM and stromal as well as immune cells, promote the polarization of monocytes into TAM. CAFs recruit granulocytes to tumors by releasing chemokines CSF1,42 suggesting that crosstalk between cancer cells and stromal cells mediates the tumor progression by signaling molecules.
Crosstalk between cancer cells and immune cells
The role of tumor-associated immune cells in the remodeling of TME is countless. TAMs are abundantly rich in TME communicate with tumor cells and stromal cells through complex signaling mediated by chemokines, cytokines, GFs, and proteases.43 The TAMs also have cross-talk with immune cells and tumor cells to induce epithelial to mesenchymal transition (EMT) via JAK2/STAT3/miR-506-3p/FoxQ1 axis.44 TAMs communicate with tumor cells for the formation of vasculature by secreting IL-1α and activating NF-kB.45 TAMs mediate tumor-induced angiogenesis, a critical metastasis phenomenon by cross-talking innate immunity and humoral response cells with cancer cells in the TME.46 The crosstalk of TAMs with tumor cells promotes metastasis by increasing migration of tumor cells,47 suggesting that TAMs are the major mediators of crosstalk between BCs and immune cells in promoting metastasis.
Exosome mediated crosstalk between TME cells
Exosomes are nanovesicles recognized recently as mediators of intercellular crosstalk. CAFs have dynamic crosstalk with tumor-associated immune cells, TAEs via exosomal proteins, and activates the CSC within TME.48 CAFs communicate with cancer cells by transmitting exosomal microRNAs, lncRNAs, and proteins. CAFs induce stemness and drug resistance by delivering lncRNA H19 containing exosomes49 indicating that key tumor-associated stromal cells contribute to tumor progression by modulating TME.
Tumor cells cross-talk with TAEs for the formation of new blood vessels by secreting VEGF-enriched exosomes.50 Cancer cells disseminate through the pro-metastatic crosstalk with endothelial cells by providing exosomal miRNAs.51 The crosstalk of cancer cells with surrounding TME cells activates oncogenic signaling and abnormal expression of angiogenesis-associated genes via tumor-derived exosomal lncRNAs.52 Tumor cells interact with TAEs to regulate angiogenesis via derived exosomal microRNA-194.53 Cancer-derived exosome-mediated cross-talk of stem cells with immune cells and blocks their anti-cancer activity by promoting transform them into suppressor cells via transferring miRNAs.54 Exosome mediated cross-talk induces tumor phenotypes in mesenchymal stem cells by activating glycolysis.55 Even, exosomes derived from M2 -macrophage induces migration and invasion of cancer cells. Exosomes enriched with lncRNAs from CAFs reinforcing their crosstalk with tumor cells.56 These studies propose that blocking the interaction of exosomes and the components of TME has potential in therapeutic approaches.
Multifunctional pathways of TME
The highly conserved ancient and multifunctional developmental signaling pathways, including Hh, Notch, and WNT, as well as NF-kB pathways, are hyperactive in TME and regulate the basic cellular mechanisms such as proliferation, differentiation, and survival and trigger most of the critical cell fate decisions (Table 1).
Table 1.
Developmental pathway | Biological function | Target of activity |
---|---|---|
Hedgehog pathway |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Notch pathway |
|
|
|
|
|
|
|
|
|
|
|
Wnt pathway |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
NF-kB pathway |
|
|
|
|
|
|
|
|
|
|
Hedgehog signaling pathway
The Hh pathway plays a significant role in tissue regeneration, self-renewal, and embryonic development. The Hh protein family comprises Sonic hedgehog (Shh), Desert hedgehog (Dhh), and Indian hedgehog (Ihh). The Ihh is primarily involved in the differentiation of bone, Dhh intricates in the differentiation of gonads, and Shh is broadly expressed and best studied in the Hh pathway.3,57 These proteins activate the signaling cascade that binds to the 12-pass transmembrane (TM) receptorsPatched1 and 2 (PTCH1/PTCH2). These receptors differ in amino acid composition in the human population. In the cell membrane, SMO activates the TFs of Gli via Intraflagellar transport proteins (IFT). These proteins are important for the functioning of cilia.58 The Gli- TFs are the zinc finger family members, consisting of Gli-1,-2, and -3. Gli-1 plays a role in gene activation, whereas Gli-3 is repressive, and Gli-2 plays a role in both activation and repression. Hh pathway also intervene between breast cancer and macrophages that support the TME.59,60
For tumor initiation and metastasis, the tumors supplant many signaling pathways, which participate in normal growth and tissue repair. The mammalian embryos involve many signaling pathways in development, regulating cell proliferation, growth, vascularization, and tissue patterning.61 The range of Hh signaling in TME is extensive. The TME contains non-tumor cells, which are important for the promotion of cancers.62,63 Shh pathway directly targets fibroblasts.64 Further, sustained synthesis of Shh leads to genetic instability and accumulation of mutations. Hh signaling regulates several proteins such as TGF-β and PDGF,65,66 which activate fibroblasts to produce proliferative and pro-survival molecules.
Notch signaling pathway
Notch signaling, a highly conserved pathway, is important for the development of the normal embryo as well as tissue homeostasis.67 It is related to diverse types of cancer. The Notch receptor is cleaved into two fragments and is placed in the plasma membrane, which is linked to a heterodimer of the N- and C-terminal fragments. After binding with the ligand, the receptor is a cleavage in two steps, one is mediated by ϒ-secretase complex, which releases the active intracellular domain, and the mastermind mediates the other step like protein, which activates the transcription of the Notch target gene. Notch signaling acts as a promising target for BC treatment. High levels of Jagged and Notch correlates with the mortality of BCs.68,69
Notch signaling was recognized as an important pathway in BCs development by a series of experiments. The first indication was on the study of mouse mammary tumor virus (MMTV).70,71 The MMTV activates protooncogenes by flanking, which causes the malignant transformation of the mammary gland that results in truncated Notch 4 proteins. Cancer progression and metastasis depend on bidirectional communication between cancer cells and the environment, thus forming TME.72,73 The role of this pathway in cancer acts as a tumor-suppressive pathway. Notch signaling in TME expresses various ligands and receptors that induce between sending a signal and receiving a signal. Notch signaling in TME is involved in pro and anti-tumor effects.
Wnt signaling
Wnt signaling is a regulatory pathway that governs the cellular and developmental process. The research on the Wnt pathway has established gene products of wingless in drosophila.74 Wnt pathway consists of a hydrophobic and cysteine family -rich glycolipoprotein, which directs the tissue homeostasis and stem cell proliferation. Humans contain 19 genes that encode Wnt, which connects to receptors and stimulate the intracellular signal transduction pathway.75,76 This pathway is either canonical, which depends on β-catenin, and non-canonical, which is independent of β-catenin.
The canonical pathway is triggered by the interaction of the frizzled receptor (Fz) with Lipoprotein receptor protein (LRP5). When Wnt bounds this receptor, the receptor complex stimulates the canonical pathway.77 When this pathway gets activated, Fz gets to interact with a cytoplasmic protein called Dishevelled (Dsh), which acts as an upstream regulator of β-catenin.78,79 The non-canonical pathway utilizes downstream signaling, which evokes the response in transcription. This pathway activates the Ca2+ signaling cascade. Binding of Fz to Wnt ligand causes the activation of membrane-bound phospholipase C80, indicating that the pathway results in cancer progression and neurodegenerative diseases.
EMT is an essential process in which the epithelial cells lose their contact and acquire properties that resemble mesenchymal cells. The TME also assists in metastasis of cancer, which induces EMT.81,82 The GFs of TME is also responsible for Wnt signal activation. Platelet-derived growth factor (PDGF) is one of the growth factors that activate Wnt signaling, Phosphorylation of P68 led by PDGF promotes β-catenin signaling. VEGF is another GF, which stabilizes β-catenin levels in endothelial cells. PGE2, which is secreted by TAEs of TME.83,84 The regulation of Wnt signaling is associate with aggressive and hyper-proliferative behavior of BCs. The knowledge expanded in the recent past provides an opportunity for the rational design of novel BC therapeutics.
NF-κB signaling pathway
The nuclear factor kappa-light chain enhancer of activated B cells (NF-κB) involve in the cytokine production. It mostly regulates immune response, which is linked to cancer, inflammation etc.85 NF-κB is a superfamily of transcription factors (TFs) discovered in 1986includesReIA (p65), C-ReI,ReIB,NF-κB1 (p50)and NF-κB (p52).86,87 The N terminal region contain ReI, which is responsible for the binding of specific DNA, while the C-terminal is responsible for binding with other TFs.88,89 NF-κB is present in the cytoplasm as inactive form by complexing with inhibitor subunits IKB-α,-β and-ϒ.90,91 The dissociation of inhibitory subunits (IkBs) results in the activation and rapid translocation of KF-kB to the nucleus.
Increased expression of NF-κB genes results in tumor progression and metastasis.92,93 The role of NF-κB in tumor growth with reference to TME is very complex because of different functions.94,95 Human breast cancers form the nuclear accumulation in classic form KF-kB dimer p50/p65. The activated NF-κB dimer increases the proliferation of cancer cells and decreases apoptosis.96 Stimulation of NF-κB in solid tumors results in the loss of mutations, which results in inflammatory TME.97,98 The chronic inflammation develops many other changes in the cellular environment such as metabolic changes, DNA damage etc.99 The inflammation response involved in the release of cytokines and activation of the canonical NF-κB signaling pathway. However, the activated pathway destroys transformed cells.100,101 This is mostly owing to the protective mechanism against cancer cells. In breast cancer, leukocytes infiltrating tumors in the stroma may lead to cancer progression and result in the increase of malignancies in breast epithelial cells involving NF-kB signaling.102,103 Activation of this pathway regulates the expression of various cytokine genes.104,105 NF-κB is a major player of chronic disease, which includes cancer. Therefore, it can be used to predict the prognosis of breast cancers.
Cross regulation of TME signaling pathways
The TME is the birthplace for the activation of various developmental pathways and is prompted by crosstalk's mediated by signaling (Fig. 2). The Hh pathway is required for embryonic development as well as in the formation of different tumors.106,107 NF-κB recognized as the TF in response to inflammation results in the overexpression of Shh in cancer. Out of three ligands in Hh, Shh plays an important role.108,109 The Shh is function through two transmembrane proteins, such as SMO, patched, and TFs related to the Gli family. Cancer development and the chronic inflammation association is recognized for many years. Different cytokines, i.e., TNF-α and IL-1, majorly contribute to the inflammatory response. The Shh pathway, which is induced by NF-κB, increases proliferation of cancer cells in a ligand dependent manner.110
The intracellular pathways mediated Raf/MK/ERK associate with Gli 1/2 proteins and regulated the transcription of Jagged 2, a Notch ligand, indicating a link between Hh and Notch signaling.111 Hes1, a Notch target protein, promptly binds to Gli1 promoter and suppresses its transcription triggering the low activity of Hh.112 Besides, expression of Jagged 1 has a promising association with Notch and WNT pathways.113,114 Lately, Notch 2 was recognized as the target of the WNT/β-catenin pathway.115 Unexpectedly, Notch 1 has suppressive activity on WNT/β-catenin target genes.116 Wnt and Hh pathways regulate among each other that affect their outcome, which results in the TFs. The activation of Hh signaling is a key step for tumorigenicity in breast cancer cells. There is existence for the positive results between the Hh and Wnt, which are present in the epithelial transformation, where there is an activation of Gli expression throughout the signaling. One side β-catenin enhances directly with the luciferase activity and results in the induction of post transcription, which stabilizes Gli mRNA, which upregulates the CRD-BP (coding region determinant-binding protein).117 On the other hand, induction of Gli through activation of Wnt2b, Wnt 4, Wnt7b, which triggers the Wnt pathway that promotes the β-catenin.
In relation to Hh, Notch signaling is also involved in the differentiation of multiple tissues. Notch pathway also increases the survival and self-renewal capacity of hematopoietic progenitors that controls the regulation of T-cell expansion.118 Hh and Notch pathways play similar roles during the development of T-cell. Both Hh and Notch pathways maintain the balance of intracellular levels through the integration of other signaling pathways.119 Hh signaling results in the formation of tumor growth and cancer stem cells.
β-catenin interacts and is inhibited by NF-κB that functions in the breast cancer cells. β-catenin results in the formation of the complex with ReIA and p50 that decreases NF-κB binding to DNA and increases transactivation activity and targets the gene expression. β-catenin activation is inhibited by the expression of NF-κB target genes.120 Many studies have revealed the negative regulation of NF-κB that has stimulatory effects of β-catenin. Activation of the Wnt pathway results in the overexpression of β-catenin that increase βTrCP expression. βTrCP induction by β-catenin accelerates the degradation of β-catenin that serves as negative feedback.121,122
Activation of NF-κB is controlled via κB kinase, which forms a complex with two inhibitory subunits IKK1 and IKK2, and regulatory protein such as IKK3. The NF-κB signaling includes cytokines.123,124 Notch and NF-κB pathways are triggered by the ligation of notch receptors, leading to the proteolytic cleavage of Notch intracellular domain, subsequent translocation into the nucleus binds to the DNA –binding protein.125 NF-κB is a putative target gene of activated Notch-1.126 Notch and Wnt pathways closely relate to cells' differentiation. Activation of the notch pathway inhibits the specific markers results in the influence on β-catenin translocation.127,128 Activation of notch signaling in the differentiated cells indicates upregulated markers and activates the canonical pathway. These findings suggest that cross-regulation of developmental pathways initiates a prometastatic phenotype in TME.
Exosomal and non-exosomal miRNAs significantly affect the cross regulation of signaling pathways by transporting oncogenic miRNAs between different cells of TME. miRNAs crosstalk with Wnt, TG-F-β, and Notch signaling pathways and control stem cell phenotype.129 The regulatory crosstalk of miRNAs with Wnt/β-catenin, Notch, and Hedgehog signaling pathways control cancer stem cell reprogramming.130 Further, miR-146 regulates NF-κB via the Notch-mediated signaling pathway of immunity and cancer.131 Coregulatory signals of Notch and NF-kB promotes miR-223 dependent suppression of cancer cells.132 GSK-3 regulates Wnt, Hh, and Notch signaling crosstalk via miRNAs.133 Taken together, exosomes mediate cross-regulation of pathways involved in TME.
Inhibitors of TME pathways
Targeting TME is an attractive strategy for the treatment of metastatic BCs. Small molecules and natural products, which are potentially important in targeting either components or signaling pathways of TME, can overcome the immune escape and drug resistance. The small molecule, cyclopamine was first described as the hedgehog pathway inhibitor.134 It is a steroid alkaloid produced by corn lilies, which has teratogenic and anti-tumor activities (Fig. 3). Cyclopamine breast cancer by directly binding to the hydrophobic core of the SMO.135,136 It suppresses the proliferation of MCF-7 and MDA-MB231 cells137 and reduces invasiveness.138 Cyclopamine sensitizes the BC cells to Paclitaxel, EGFR inhibitors, and tamoxifen.139 Cyclopamine inhibits estrogen-receptor (ER) positive as well as ER-negative tumorigenic BCs at higher doses.140 Co-delivery with BSA nanoparticles reverses Dox resistance by downregulating P-glycoprotein.141
SMO antagonists inhibits breast cancer growth and drug resistance (Fig. 4). Sonidegib, which is antagonistic of SMO118 is also well studied in clinical trials against different cancers.142 Vismodegib is a synthetic SMO inhibitor.143 It is under clinical trial for TNBC.144 The SMO inhibitor Vismodegib directly binds to the SMO145, which results in the inhibition of cancer cell progression.146 Hh pathway mediates self-renewal of stem cells; therefore, the rationale of inhibiting the Hh pathway leads to the dismissal of cancer stem cells and prevent the recurrence. Itraconazole is an antifungal triazole compound, which inhibits the Hh pathway by acting on SMO.147,148 However, its inhibitory effect on Hh signaling appears to be different.149 It binds to SMO at a distinct site, which is different from other SMP inhibitors. When administered into mice, the drug suppresses the growth of medulloblastoma and reduces Hh activation markers.118,150 Itraconazole reduces drug resistance by inhibiting P-glycoprotein and angiogenesis.151,152 Some of the small molecule SMO inhibitors that target Hh signaling are approved by the FDA to treat cancers.
The small molecule inhibitors inhibit breast cancer by targeting different developmental pathways (Fig. 5). XAV 939 inhibits Wnt pathway by blocking degradation of Axin 1 and IWR-1 inhibit the Wnt pathway by stimulating the degradation of β-catenin via stabilizing Axin1.153 However, small molecule Dishevelled inhibitors NSC668036 and FJ9 bind to the frizzled receptors on C-terminal inhibit the Wnt pathway.154,155 Wnt pathway is closely related to the other pathways, such as Notch. The gamma-secretase inhibitors (GSI) indirectly target Wnt pathways in CD44+ cancer stem cells (Fig. 6).156 As Notch signaling is associated with breast cancer progression, it is recognized as a promising therapeutic target.157,158 MRK-003 is s new generation GSI inhibits tumor-initiating cells transplanted mouse model.159 BMS-906024 is the only GSI that equipotent in inhibiting all the four NOTCH isoforms, which is under clinical trial for breast cancer.160 Therefore, small molecule inhibitors that are blocking Wnt and Notch pathways could be implicated in targeting breast cancer stem cells.
The target-specific anti-cancer agents are failed to accomplish the anticipated results; therefore, new multi-targeted natural products have become significant. Mounting evidence has established that several natural small molecules could inhibit developmental pathways.161,162 Genistein (Gn) is an isoflavone present in soy, and other plants are reported to have activity against breast and prostate cancers (Fig. 7).163 It affects many signaling pathways, which suppress tumor growth. Gn shows the reduction in SMO and Gli expression, which results in the reduction of cancer stem cells.164 It also inactivates NF-kB via the Akt pathway.165 Gn inhibits expression of Notch 1166 and Wnt.167
Curcumin has biological activity against inflammation, ischemia, and cancer (Fig. 8).168,169 Hh is also affected by curcumin.170 Wnt/β catenin, Notch and MAPK are also the targets of curcumin.171 Curcumin induced GSK-3β causes downregulation of nuclear β-catenin.172 It downregulates the expression of SMO and Gli-1 in breast cancer stem cells.18 It also inhibits oncogenic NF-kB activity.173 The Hh pathway stimulation by overexpression of Gli-1 increases cancer stem cell markers such as CD44, CD24, BMI-1 etc. and suppresses the effect of curcumin.174 It is under clinical trial for breast cancer along with radiation treatment and chemotherapy.
Green tea, a widely consumed beverages, contains EGCG, reported preventing many cancers including breast cancer (Fig. 9).175 It mostly inhibits the Shh pathway by suppressing Gli-1 mRNA expression and downregulation of Gli-1 activity,176 Wnt pathway by downregulating the expression of β-catenin,177 decreased activation of NF-κB178 and transcriptional levels of Notch.179 The Hedgehog pathway cross talks with the Notch, Wnt, and NF-kB can be implicated for therapeutic interventions in breast cancers.
Conclusion
The ancient developmental pathways are important for tumor progression. Cross regulation is reported to involve many cellular mechanisms, especially in the development and maintenance of cancer stem cell phenotype and metastasis. The hedgehog pathway is a predominant regulator of the paracrine network in TME; therefore, its inhibitors target both tumor cells as well as other supporting cells. Notch signaling also plays a role in oncogenic solid tumors like breast cancer, especially in tumor angiogenesis; therefore, it is a viable target for breast cancer treatment. Wnt signaling promotes tumor stem cells, provides a large platform for rational drug discovery. NF-κB is a key regulator of inflammatory pathways prevalent in TME and mediates chemo-radio resistance, also the principal target of breast cancers. Because target-specific anti-cancer agents fail to present anticipated results, multi-targeted natural products like curcumin, genistein, and EGCGs are significant in targeting TME to develop therapeutics for breast cancers.
Author contributions
Prof. RamaRao Malla has substantial contributions to the conception and design of the work as well as drafting a part, interpretation of data and revising it critically for important intellectual content. Ms.P.Kiran has contributed to drafting the part of the manuscript. Final approval of the version to be published; AN.
Conflict of interests
The authors declared that there is no conflict of interest.
Funding
This work was supported by ICMR, India (No.05/13/4/2014-NCD-III dt.22.12.2017).
Acknowledgements
The authors would like to thank the management of GITAM (Deemed to be University) for providing additional facilities. and Mr. Manas Malla, Visakha Valley School for language corrections and proofreading.
Footnotes
Peer review under responsibility of Chongqing Medical University.
References
- 1.Key T.J., Verkasalo P.K., Banks E. Epidemiology of breast cancer. Lancet Oncol. 2001;2(3):133–140. doi: 10.1016/S1470-2045(00)00254-0. [DOI] [PubMed] [Google Scholar]
- 2.Hui M., Cazet A., Nair R., Watkins D.N., O’Toole S.A., Swarbrick A. The Hedgehog signalling pathway in breast development, carcinogenesis and cancer therapy. Breast Cancer Res. 2013;15(2):203. doi: 10.1186/bcr3401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ingham P.W., McMahon A.P. Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 2001;15(23):3059–3087. doi: 10.1101/gad.938601. [DOI] [PubMed] [Google Scholar]
- 4.Barth S.D., Schulze J.J., Kühn T., et al. Treg-mediated immune tolerance and the risk of solid cancers: findings from EPIC-Heidelberg. J Natl Cancer Inst. 2015;107(11):djv224. doi: 10.1093/jnci/djv224. [DOI] [PubMed] [Google Scholar]
- 5.Linke F., Harenberg M., Nietert M.M., et al. Microenvironmental interactions between endothelial and lymphoma cells: a role for the canonical WNT pathway in Hodgkin lymphoma. Leukemia. 2017;31(2):361–372. doi: 10.1038/leu.2016.232. [DOI] [PubMed] [Google Scholar]
- 6.Soysal S.D., Tzankov A., Muenst S.E. Role of the tumor microenvironment in breast cancer. Pathobiology. 2015;82(3–4):142–152. doi: 10.1159/000430499. [DOI] [PubMed] [Google Scholar]
- 7.Malla R.R., Deepak K., Merchant N., Dasari V.R. Breast tumor microenvironment: emerging target of therapeutic phytochemicals. Phytomedicine. 2020;70:153227. doi: 10.1016/j.phymed.2020.153227. [DOI] [PubMed] [Google Scholar]
- 8.Deepak K.G.K., Vempati R., Nagaraju G.P., et al. Tumor microenvironment: challenges and opportunities in targeting metastasis of triple negative breast cancer. Pharmacol Res. 2020;153:104683. doi: 10.1016/j.phrs.2020.104683. [DOI] [PubMed] [Google Scholar]
- 9.Dias A.S., Almeida C.R., Helguero L.A., Duarte I.F. Metabolic crosstalk in the breast cancer microenvironment. Eur J Cancer. 2019;121:154–171. doi: 10.1016/j.ejca.2019.09.002. [DOI] [PubMed] [Google Scholar]
- 10.Martins D., Schmitt F. Microenvironment in breast tumorigenesis: friend or foe? Histol Histopathol. 2019;34(1):13–24. doi: 10.14670/HH-18-021. [DOI] [PubMed] [Google Scholar]
- 11.Wang N., Liu W., Zheng Y., et al. CXCL1 derived from tumor-associated macrophages promotes breast cancer metastasis via activating NF-κB/SOX4 signaling. Cell Death Dis. 2018;9(9):880. doi: 10.1038/s41419-018-0876-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wang G., Zhao J., Zhang M., et al. Ferumoxytol and CpG oligodeoxynucleotide 2395 synergistically enhance antitumor activity of macrophages against NSCLC with EGFR(L858R/T790M) mutation. Int J Nanomed. 2019;14:4503–4515. doi: 10.2147/IJN.S193583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Chen Y., Wen H., Zhou C., et al. TNF-α derived from M2 tumor-associated macrophages promotes epithelial-mesenchymal transition and cancer stemness through the Wnt/β-catenin pathway in SMMC-7721 hepatocellular carcinoma cells. Exp Cell Res. 2019;378(1):41–50. doi: 10.1016/j.yexcr.2019.03.005. [DOI] [PubMed] [Google Scholar]
- 14.Shinde A.V., Humeres C., Frangogiannis N.G. The role of α-smooth muscle actin in fibroblast-mediated matrix contraction and remodeling. Biochim Biophys Acta (BBA) - Mol Basis Dis. 2017;1863(1):298–309. doi: 10.1016/j.bbadis.2016.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Erdogan B., Webb D.J. Cancer-associated fibroblasts modulate growth factor signaling and extracellular matrix remodeling to regulate tumor metastasis. Biochem Soc Trans. 2017;45(1):229–236. doi: 10.1042/BST20160387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Takahashi H., Sakakura K., Kudo T., et al. Cancer-associated fibroblasts promote an immunosuppressive microenvironment through the induction and accumulation of protumoral macrophages. Oncotar. 2017;8(5):8633–8647. doi: 10.18632/oncotarget.14374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Schmidt A., von Woedtke T., Vollmar B., Hasse S., Bekeschus S. Nrf2 signaling and inflammation are key events in physical plasma-spurred wound healing. Theranostics. 2019;9(4):1066–1084. doi: 10.7150/thno.29754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Sterling J.A., Oyajobi B.O., Grubbs B., et al. The hedgehog signaling molecule Gli2 induces parathyroid hormone-related peptide expression and osteolysis in metastatic human breast cancer cells. Cancer Res. 2006;66(15):7548–7553. doi: 10.1158/0008-5472.CAN-06-0452. [DOI] [PubMed] [Google Scholar]
- 19.Zeng Y.F., Xiao Y.S., Liu Y., et al. Formin-like 3 regulates RhoC/FAK pathway and actin assembly to promote cell invasion in colorectal carcinoma. World J Gastroenterol. 2018;24(34):3884–3897. doi: 10.3748/wjg.v24.i34.3884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hida K., Maishi N., Annan D.A., Hida Y. Contribution of tumor endothelial cells in cancer progression. Int J Mol Sci. 2018;19(5):1272. doi: 10.3390/ijms19051272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hughes-Fulford M., Li C.F. The role of FGF-2 and BMP-2 in regulation of gene induction, cell proliferation and mineralization. J Orthop Surg Res. 2011;6:8. doi: 10.1186/1749-799X-6-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Christopoulos P.F., Msaouel P., Koutsilieris M. The role of the insulin-like growth factor-1 system in breast cancer. Mol Canc. 2015;14:43. doi: 10.1186/s12943-015-0291-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kolb A.D., Shupp A.B., Mukhopadhyay D., Marini F.C., Bussard K.M. Osteoblasts are “educated” by crosstalk with metastatic breast cancer cells in the bone tumor microenvironment. Breast Cancer Res. 2019;21(1):31. doi: 10.1186/s13058-019-1117-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Oh K., Lee O.Y., Shon S.Y., et al. A mutual activation loop between breast cancer cells and myeloid-derived suppressor cells facilitates spontaneous metastasis through IL-6 trans-signaling in a murine model. Breast Cancer Res. 2013;15(5):R79. doi: 10.1186/bcr3473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yu J., Du W., Yan F., et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190(7):3783–3797. doi: 10.4049/jimmunol.1201449. [DOI] [PubMed] [Google Scholar]
- 26.Condamine T., Ramachandran I., Youn J.I., Gabrilovich D.I. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu Rev Med. 2015;66:97–110. doi: 10.1146/annurev-med-051013-052304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Borregaard N. Neutrophils, from marrow to microbes. Immunity. 2010;33(5):657–670. doi: 10.1016/j.immuni.2010.11.011. [DOI] [PubMed] [Google Scholar]
- 28.Piccard H., Muschel R.J., Opdenakker G. On the dual roles and polarized phenotypes of neutrophils in tumor development and progression. Crit Rev Oncol-Hematol. 2012;82(3):296–309. doi: 10.1016/j.critrevonc.2011.06.004. [DOI] [PubMed] [Google Scholar]
- 29.Gregory A.D., Houghton A.M. Tumor-associated neutrophils: new targets for cancer therapy. Canc Res. 2011;71(7):2411–2416. doi: 10.1158/0008-5472.CAN-10-2583. [DOI] [PubMed] [Google Scholar]
- 30.Hansen M., Andersen M.H. The role of dendritic cells in cancer. Semin Immunopathol. 2017;39(3):307–316. doi: 10.1007/s00281-016-0592-y. [DOI] [PubMed] [Google Scholar]
- 31.Veglia F., Gabrilovich D.I. Dendritic cells in cancer: the role revisited. Curr Opin Immunol. 2017;45:43–51. doi: 10.1016/j.coi.2017.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Vitale M., Cantoni C., Pietra G., Mingari M.C., Moretta L. Effect of tumor cells and tumor microenvironment on NK-cell function. Eur J Immunol. 2014;44(6):1582–1592. doi: 10.1002/eji.201344272. [DOI] [PubMed] [Google Scholar]
- 33.Plitas G., Konopacki C., Wu K., et al. Regulatory T cells exhibit distinct features in human breast cancer. Immunity. 2016;45(5):1122–1134. doi: 10.1016/j.immuni.2016.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Tanaka A., Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017;27(1):109–118. doi: 10.1038/cr.2016.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sasidharan Nair V., Elkord E. Immune checkpoint inhibitors in cancer therapy: a focus on T-regulatory cells. Immunol Cell Biol. 2018;96(1):21–33. doi: 10.1111/imcb.1003. [DOI] [PubMed] [Google Scholar]
- 36.Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006;6(4):295–307. doi: 10.1038/nri1806. [DOI] [PubMed] [Google Scholar]
- 37.Wang Y.A., Li X.L., Mo Y.Z., et al. Effects of tumor metabolic microenvironment on regulatory T cells. Mol Cancer. 2018;17(1):168. doi: 10.1186/s12943-018-0913-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Costa A., Kieffer Y., Scholer-Dahirel A., et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Canc Cell. 2018;33(3):463–479. doi: 10.1016/j.ccell.2018.01.011. [DOI] [PubMed] [Google Scholar]
- 39.Goel S., DeCristo M.J., Watt A.C., et al. CDK4/6 inhibition triggers anti-tumour immunity. Nat. 2017;548(7668):471–475. doi: 10.1038/nature23465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.De Nola R., Menga A. The crowded crosstalk between cancer cells and stromal microenvironment in gynecological malignancies: biological pathways and therapeutic implication. Int J Mol Sci. 2019;20(10) doi: 10.3390/ijms20102401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Yang Y., Andersson P., Hosaka K., et al. The PDGF-BB-SOX7 axis-modulated IL-33 in pericytes and stromal cells promotes metastasis through tumour-associated macrophages. Nat Commun. 2016;7:11385. doi: 10.1038/ncomms11385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kumar V., Donthireddy L., Marvel D., et al. Cancer-associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell. 2017;32(5):654–668. doi: 10.1016/j.ccell.2017.10.005. e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wang J., Li D., Cang H., Guo B. Crosstalk between cancer and immune cells: role of tumor-associated macrophages in the tumor microenvironment. Cancer Med. 2019;8(10):4709–4721. doi: 10.1002/cam4.2327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wei C., Yang C., Wang S., et al. Crosstalk between cancer cells and tumor associated macrophages is required for mesenchymal circulating tumor cell-mediated colorectal cancer metastasis. Mol Cancer. 2019;18(1):64. doi: 10.1186/s12943-019-0976-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Georgouli M., Herraiz C., Crosas-Molist E., et al. Regional activation of myosin II in cancer cells drives tumor progression via a secretory cross-talk with the immune microenvironment. Cell. 2019;176(4):757–774. doi: 10.1016/j.cell.2018.12.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Guadagno E., Presta I. Role of macrophages in brain tumor growth and progression. Int J Mol Sci. 2018;19(4):1005. doi: 10.3390/ijms19041005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Li H., Huang N., Zhu W., et al. Modulation the crosstalk between tumor-associated macrophages and non-small cell lung cancer to inhibit tumor migration and invasion by ginsenoside Rh2. BMC Cancer. 2018;18(1):579. doi: 10.1186/s12885-018-4299-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Alguacil-Núñez C., Ferrer-Ortiz I., García-Verdú E., López-Pirez P., Llorente-Cortijo I.M., Sainz B., Jr. Current perspectives on the crosstalk between lung cancer stem cells and cancer-associated fibroblasts. Crit Rev Oncol-Hematol. 2018;125:102–110. doi: 10.1016/j.critrevonc.2018.02.015. [DOI] [PubMed] [Google Scholar]
- 49.Ren J., Ding L., Zhang D., et al. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8(14):3932–3948. doi: 10.7150/thno.25541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zeng Y., Yao X., Liu X., et al. Anti-angiogenesis triggers exosomes release from endothelial cells to promote tumor vasculogenesis. J Extracell Vesicles. 2019;8(1):1629865. doi: 10.1080/20013078.2019.1629865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kim K.S., Park J.I., Oh N., Cho H.J., Park J.H., Park K.S. ELK3 expressed in lymphatic endothelial cells promotes breast cancer progression and metastasis through exosomal miRNAs. Sci Rep. 2019;9(1):8418. doi: 10.1038/s41598-019-44828-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zhao J., Li L., Han Z.Y., Wang Z.X., Qin L.X. Long noncoding RNAs, emerging and versatile regulators of tumor-induced angiogenesis. Am J Canc Res. 2019;9(7):1367–1381. [PMC free article] [PubMed] [Google Scholar]
- 53.Chen L., Han Y., Li Y., et al. Hepatocyte-derived exosomal MiR-194 activates PMVECs and promotes angiogenesis in hepatopulmonary syndrome. Cell Death Dis. 2019;10(11):853. doi: 10.1038/s41419-019-2087-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Whiteside T.L. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin Immunol. 2018;35:69–79. doi: 10.1016/j.smim.2017.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ma Z., Cui X., Lu L., et al. Exosomes from glioma cells induce a tumor-like phenotype in mesenchymal stem cells by activating glycolysis. Stem Cell Res Ther. 2019;10(1):60. doi: 10.1186/s13287-019-1149-5. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 56.Herrera M., Llorens C., Rodríguez M., et al. Differential distribution and enrichment of non-coding RNAs in exosomes from normal and cancer-associated fibroblasts in colorectal cancer. Mol Cancer. 2018;17(1):114. doi: 10.1186/s12943-018-0863-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nagase T., Nagase M., Machida M., Fujita T. Hedgehog signalling in vascular development. Rev Angiogene. 2008;11(1):71–77. doi: 10.1007/s10456-008-9105-5. [DOI] [PubMed] [Google Scholar]
- 58.Beachy P.A., Hymowitz S.G., Lazarus R.A., Leahy D.J., Siebold C. Interactions between hedgehog proteins and their binding partners come into view. Genes Dev. 2010;24(18):2001–2012. doi: 10.1101/gad.1951710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Cohen M.M., Jr. Hedgehog signaling update. Am J Med Genet. 2010;152a(8):1875–1914. doi: 10.1002/ajmg.a.32909. [DOI] [PubMed] [Google Scholar]
- 60.Stecca B., Ruiz I.A.A. Context-dependent regulation of the GLI code in cancer by Hedgehog and non-Hedgehog signals. J Mol Cell Biol. 2010;2(2):84–95. doi: 10.1093/jmcb/mjp052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 62.Theunissen J.W., de Sauvage F.J. Paracrine Hedgehog signaling in cancer. Canc Res. 2009;69(15):6007–6010. doi: 10.1158/0008-5472.CAN-09-0756. [DOI] [PubMed] [Google Scholar]
- 63.Yang L., Wang Y., Mao H., et al. Sonic hedgehog is an autocrine viability factor for myofibroblastic hepatic stellate cells. J Hepatol. 2008;48(1):98–106. doi: 10.1016/j.jhep.2007.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lipinski R.J., Gipp J.J., Zhang J., Doles J.D., Bushman W. Unique and complimentary activities of the Gli transcription factors in Hedgehog signaling. Exp Cell Res. 2006;312(11):1925–1938. doi: 10.1016/j.yexcr.2006.02.019. [DOI] [PubMed] [Google Scholar]
- 65.Li X., Wang Z., Jiang Z., et al. Regulation of seminiferous tubule-associated stem Leydig cells in adult rat testes. Proceed Nat Acad Sci USA. 2016;113(10):2666–2671. doi: 10.1073/pnas.1519395113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Luo K. Signaling cross talk between TGF-β/smad and other signaling pathways. Cold Spring Harbor Pers Biol. 2017;9(1):a022137. doi: 10.1101/cshperspect.a022137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Amankulor N.M., Hambardzumyan D., Pyonteck S.M., Becher O.J., Joyce J.A., Holland E.C. Sonic hedgehog pathway activation is induced by acute brain injury and regulated by injury-related inflammation. J Neurosci. 2009;29(33):10299–10308. doi: 10.1523/JNEUROSCI.2500-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Yamasaki A., Kameda C., Xu R., et al. Nuclear factor kappaB-activated monocytes contribute to pancreatic cancer progression through the production of Shh. Cancer Immunol Immunother. 2010;59(5):675–686. doi: 10.1007/s00262-009-0783-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kopan R., Ilagan M.X. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137(2):216–233. doi: 10.1016/j.cell.2009.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Das C., Berezovska O., Diehl T.S., et al. Designed helical peptides inhibit an intramembrane protease. J Am Chem Soc. 2003;125(39):11794–11795. doi: 10.1021/ja037131v. [DOI] [PubMed] [Google Scholar]
- 71.Dovey H.F., John V., Anderson J.P., et al. Functional gamma-secretase inhibitors reduce beta-amyloid peptide levels in brain. J Neurochem. 2001;76(1):173–181. doi: 10.1046/j.1471-4159.2001.00012.x. [DOI] [PubMed] [Google Scholar]
- 72.Churcher I., Ashton K., Butcher J.W., et al. A new series of potent benzodiazepine gamma-secretase inhibitors. Bioorg Med Chem Lett. 2003;13(2):179–183. doi: 10.1016/s0960-894x(02)00909-5. [DOI] [PubMed] [Google Scholar]
- 73.Yang M.G., Shi J.L., Modi D.P., et al. Design and synthesis of benzoazepinone-derived cyclic malonamides and aminoamides as potent gamma-secretase inhibitors. Bioorg Med Chem Lett. 2007;17(14):3910–3915. doi: 10.1016/j.bmcl.2007.04.102. [DOI] [PubMed] [Google Scholar]
- 74.Jenny F.H., Basler K. Powerful Drosophila screens that paved the wingless pathway. Fly (Austin) 2014;8(4):218–225. doi: 10.4161/19336934.2014.985988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Nüsslein-Volhard C., Wieschaus E. Mutations affecting segment number and polarity in Drosophila. Nature. 1980;287(5785):795–801. doi: 10.1038/287795a0. [DOI] [PubMed] [Google Scholar]
- 76.Rijsewijk F., Schuermann M., Wagenaar E., Parren P., Weigel D., Nusse R. The Drosophila homolog of the mouse mammary oncogene int-1 is identical to the segment polarity gene wingless. Cell. 1987;50(4):649–657. doi: 10.1016/0092-8674(87)90038-9. [DOI] [PubMed] [Google Scholar]
- 77.Tamai K., Zeng X., Liu C., et al. A mechanism for Wnt coreceptor activation. Mol Cell. 2004;13(1):149–156. doi: 10.1016/s1097-2765(03)00484-2. [DOI] [PubMed] [Google Scholar]
- 78.van Amerongen R., Nusse R. Towards an integrated view of Wnt signaling in development. Dev. 2009;136(19):3205–3214. doi: 10.1242/dev.033910. [DOI] [PubMed] [Google Scholar]
- 79.Kikuchi A., Yamamoto H., Sato A., Matsumoto S. New insights into the mechanism of Wnt signaling pathway activation. Inter Rev Cell Mol Biol. 2011;291:21–71. doi: 10.1016/B978-0-12-386035-4.00002-1. [DOI] [PubMed] [Google Scholar]
- 80.Niehrs C. The complex world of WNT receptor signalling. Rev Nat Rev Mol Cell Biol. 2012;13(12):767–779. doi: 10.1038/nrm3470. [DOI] [PubMed] [Google Scholar]
- 81.Wong G.T., Gavin B.J., McMahon A.P. Differential transformation of mammary epithelial cells by Wnt genes. Mole Cell Biol. 1994;14(9):6278–6286. doi: 10.1128/mcb.14.9.6278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mao J., Wang J., Liu B., et al. Low-density lipoprotein receptor-related protein-5 binds to Axin and regulates the canonical Wnt signaling pathway. Mol Cell. 2001;7(4):801–809. doi: 10.1016/s1097-2765(01)00224-6. [DOI] [PubMed] [Google Scholar]
- 83.Davidson G., Wu W., Shen J., et al. Casein kinase 1 gamma couples Wnt receptor activation to cytoplasmic signal transduction. Nat. 2005;438(7069):867–872. doi: 10.1038/nature04170. [DOI] [PubMed] [Google Scholar]
- 84.Katoh M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review) Int J Oncol. 2017;51(5):1357–1369. doi: 10.3892/ijo.2017.4129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Ho S.Y., Keller T.H. The use of porcupine inhibitors to target Wnt-driven cancers. Review. Bioorg Med Chem Lett. 2015;25(23):5472–5476. doi: 10.1016/j.bmcl.2015.10.032. [DOI] [PubMed] [Google Scholar]
- 86.Kim M.K. Novel insight into the function of tankyrase. Oncol Lett. 2018;16(6):6895–6902. doi: 10.3892/ol.2018.9551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Haikarainen T., Krauss S., Lehtio L. Tankyrases: structure, function and therapeutic implications in cancer. Research Support, Non-U S Gov't Review. Curr Pharmaceut Des. 2014;20(41):6472–6488. doi: 10.2174/1381612820666140630101525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Yang K., Wang X., Zhang H., et al. The evolving roles of canonical WNT signaling in stem cells and tumorigenesis: implications in targeted cancer therapies. Lab Invest J Tech Method Pathol. 2016;96(2):116–136. doi: 10.1038/labinvest.2015.144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Piha-Paul S.A., Munster P.N., Hollebecque A., et al. Results of a phase 1 trial combining ridaforolimus and MK-0752 in patients with advanced solid tumours. Eur J Canc. 2015;51(14):1865–1873. doi: 10.1016/j.ejca.2015.06.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yamaguchi N., Ito T., Azuma S., et al. Constitutive activation of nuclear factor-kappaB is preferentially involved in the proliferation of basal-like subtype breast cancer cell lines. Cancer Sci. 2009;100(9):1668–1674. doi: 10.1111/j.1349-7006.2009.01228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Gilmore T.D. The Re1/NF-kappa B/I kappa B signal transduction pathway and cancer. Canc Treat Res. 2003;115:241–265. [PubMed] [Google Scholar]
- 92.Romieu-Mourez R., Kim D.W., Shin S.M., et al. Mouse mammary tumor virus c-rel transgenic mice develop mammary tumors. Mole Cell Biol. 2003;23(16):5738–5754. doi: 10.1128/MCB.23.16.5738-5754.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Liu M., Sakamaki T., Casimiro M.C., et al. The canonical NF-kappaB pathway governs mammary tumorigenesis in transgenic mice and tumor stem cell expansion. Canc Res. 2010;70(24):10464–10473. doi: 10.1158/0008-5472.CAN-10-0732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Shen R.R., Hahn W.C. Emerging roles for the non-canonical IKKs in cancer. Oncogene. 2011;30(6):631–641. doi: 10.1038/onc.2010.493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Hagemann T., Biswas S.K., Lawrence T., Sica A., Lewis C.E. Regulation of macrophage function in tumors: the multifaceted role of NF-kappaB. Blood. 2009;113(14):3139–3146. doi: 10.1182/blood-2008-12-172825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Giuliani C., Napolitano G., Bucci I., Montani V., Monaco F. [Nf-kB transcription factor: role in the pathogenesis of inflammatory, autoimmune, and neoplastic diseases and therapy implications] Clin Ter. 2001;152(4):249–253. [PubMed] [Google Scholar]
- 97.Schott A.F., Landis M.D., Dontu G., et al. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clinic Canc Res : J Am Ass Canc Res. 2013;19(6):1512–1524. doi: 10.1158/1078-0432.CCR-11-3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Atwood S.X., Whitson R.J., Oro A.E. Advanced treatment for basal cell carcinomas. Cold Spring Harbor Perspect Med. 2014;4(7):a013581. doi: 10.1101/cshperspect.a013581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Shacter E., Weitzman S.A. Chronic inflammation and cancer. Oncology (Williston Park) 2002;16(2):217–226. 229; discussion 230-2. [PubMed] [Google Scholar]
- 100.Ling J., Kumar R. Crosstalk between NFkB and glucocorticoid signaling: a potential target of breast cancer therapy. Canc Lett. 2012;322(2):119–126. doi: 10.1016/j.canlet.2012.02.033. [DOI] [PubMed] [Google Scholar]
- 101.Sen R., Baltimore D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell. 1986;46(5):705–716. doi: 10.1016/0092-8674(86)90346-6. [DOI] [PubMed] [Google Scholar]
- 102.Ghosh S., May M.J., Kopp E.B. NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol. 1998;16:225–260. doi: 10.1146/annurev.immunol.16.1.225. [DOI] [PubMed] [Google Scholar]
- 103.Gilmore T.D. Multiple myeloma: lusting for NF-kappaB. Cancer Cell. 2007;12(2):95–97. doi: 10.1016/j.ccr.2007.07.010. [DOI] [PubMed] [Google Scholar]
- 104.Huber M.A., Azoitei N., Baumann B., et al. NF-kappaB is essential for epithelial-mesenchymal transition and metastasis in a model of breast cancer progression. J Clin Invest. 2004;114(4):569–581. doi: 10.1172/JCI21358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Keats J.J., Fonseca R., Chesi M., et al. Promiscuous mutations activate the noncanonical NF-kappaB pathway in multiple myeloma. Cancer Cell. 2007;12(2):131–144. doi: 10.1016/j.ccr.2007.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Dunn G.P., Old L.J., Schreiber R.D. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–360. doi: 10.1146/annurev.immunol.22.012703.104803. [DOI] [PubMed] [Google Scholar]
- 107.DeNardo D.G., Coussens L.M. Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res. 2007;9(4):212. doi: 10.1186/bcr1746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Thayer S.P., di Magliano M.P., Heiser P.W., et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nat. 2003;425(6960):851–856. doi: 10.1038/nature02009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Berman D.M., Karhadkar S.S., Maitra A., et al. Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours. Nat. 2003;425(6960):846–851. doi: 10.1038/nature01972. [DOI] [PubMed] [Google Scholar]
- 110.He J., Sheng T., Stelter A.A., et al. Suppressing Wnt signaling by the hedgehog pathway through sFRP-1. J Biol Chem. 2006;281(47):35598–35602. doi: 10.1074/jbc.C600200200. [DOI] [PubMed] [Google Scholar]
- 111.Kasper M., Schnidar H., Neill G.W., et al. Selective modulation of Hedgehog/GLI target gene expression by epidermal growth factor signaling in human keratinocytes. Mole Cell Biol. 2006;26(16):6283–6298. doi: 10.1128/MCB.02317-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Schreck K.C., Taylor P., Marchionni L., et al. The Notch target Hes1 directly modulates Gli1 expression and Hedgehog signaling: a potential mechanism of therapeutic resistance. Clin Cancer Res. 2010;16(24):6060–6070. doi: 10.1158/1078-0432.CCR-10-1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Chen X., Stoeck A., Lee S.J., Shih Ie M., Wang M.M., Wang T.L. Jagged1 expression regulated by Notch3 and Wnt/β-catenin signaling pathways in ovarian cancer. Oncotar. 2010;1(3):210–218. doi: 10.18632/oncotarget.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Rodilla V., Villanueva A., Obrador-Hevia A., et al. Jagged1 is the pathological link between Wnt and Notch pathways in colorectal cancer. Proceed Nat Acad Sci USA. 2009;106(15):6315–6320. doi: 10.1073/pnas.0813221106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Ungerbäck J., Elander N., Grünberg J., Sigvardsson M., Söderkvist P. The Notch-2 gene is regulated by Wnt signaling in cultured colorectal cancer cells. PLoS One. 2011;6(3):e17957. doi: 10.1371/journal.pone.0017957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kim H.A., Koo B.K., Cho J.H., et al. Notch1 counteracts WNT/β-catenin signaling through chromatin modification in colorectal cancer. J Clin Invest. 2012;122(9):3248–3259. doi: 10.1172/JCI61216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Akiyoshi T., Nakamura M., Koga K., et al. Gli1, downregulated in colorectal cancers, inhibits proliferation of colon cancer cells involving Wnt signalling activation. Gut. 2006;55(7):991–999. doi: 10.1136/gut.2005.080333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Campese A.F., Bellavia D., Gulino A., Screpanti I. Notch signalling at the crossroads of T cell development and leukemogenesis. Semin Cell Dev Biol. 2003;14(2):151–157. doi: 10.1016/s1084-9521(02)00184-2. [DOI] [PubMed] [Google Scholar]
- 119.Ezratty E.J., Stokes N., Chai S., Shah A.S., Williams S.E., Fuchs E. A role for the primary cilium in Notch signaling and epidermal differentiation during skin development. Cell. 2011;145(7):1129–1141. doi: 10.1016/j.cell.2011.05.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Thyssen G., Li T.H., Lehmann L., Zhuo M., Sharma M., Sun Z. LZTS2 is a novel beta-catenin-interacting protein and regulates the nuclear export of beta-catenin. Mole Cell Biol. 2006;26(23):8857–8867. doi: 10.1128/MCB.01031-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Gwak J., Park S., Cho M., et al. Polysiphonia japonica extract suppresses the Wnt/beta-catenin pathway in colon cancer cells by activation of NF-kappaB. Int J Mol Med. 2006;17(6):1005–1010. [PubMed] [Google Scholar]
- 122.Schwitalla S., Fingerle A.A., Cammareri P., et al. Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell. 2013;152(1–2):25–38. doi: 10.1016/j.cell.2012.12.012. [DOI] [PubMed] [Google Scholar]
- 123.Li D., Beisswenger C., Herr C., et al. Myeloid cell RelA/p65 promotes lung cancer proliferation through Wnt/β-catenin signaling in murine and human tumor cells. Oncogene. 2014;33(10):1239–1248. doi: 10.1038/onc.2013.75. [DOI] [PubMed] [Google Scholar]
- 124.Deng J., Miller S.A., Wang H.Y., et al. beta-catenin interacts with and inhibits NF-kappa B in human colon and breast cancer. Cancer Cell. 2002;2(4):323–334. doi: 10.1016/s1535-6108(02)00154-x. [DOI] [PubMed] [Google Scholar]
- 125.Du Q., Zhang X., Cardinal J., et al. Wnt/beta-catenin signaling regulates cytokine-induced human inducible nitric oxide synthase expression by inhibiting nuclear factor-kappaB activation in cancer cells. Cancer Res. 2009;69(9):3764–3771. doi: 10.1158/0008-5472.CAN-09-0014. [DOI] [PubMed] [Google Scholar]
- 126.Oswald F., Liptay S., Adler G., Schmid R.M. NF-kappaB2 is a putative target gene of activated Notch-1 via RBP-Jkappa. Mole Cell Biol. 1998;18(4):2077–2088. doi: 10.1128/mcb.18.4.2077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Lu Z., Li Y., Takwi A., et al. miR-301a as an NF-κB activator in pancreatic cancer cells. EMBO J. 2011;30(1):57–67. doi: 10.1038/emboj.2010.296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Miyamoto Y., Maitra A., Ghosh B., et al. Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003;3(6):565–576. doi: 10.1016/s1535-6108(03)00140-5. [DOI] [PubMed] [Google Scholar]
- 129.Onyido E.K., Sweeney E., Nateri A.S. Wnt-signalling pathways and microRNAs network in carcinogenesis: experimental and bioinformatics approaches. Mol Canc. 2016;15(1):56. doi: 10.1186/s12943-016-0541-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Prokopi M., Kousparou C.A., Epenetos A.A. The secret role of microRNAs in cancer stem cell development and potential therapy: a notch-pathway approach. Front Oncol. 2015;4:389. doi: 10.3389/fonc.2014.00389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Osipo C., Golde T.E., Osborne B.A., Miele L.A. Off the beaten pathway: the complex cross talk between Notch and NF-kappaB. Lab Invest J Tech Method Pathol. 2008;88(1):11–17. doi: 10.1038/labinvest.3700700. [DOI] [PubMed] [Google Scholar]
- 132.Kumar V., Palermo R., Talora C., et al. Notch and NF-kB signaling pathways regulate miR-223/FBXW7 axis in T-cell acute lymphoblastic leukemia. Leukemia. 2014;28(12):2324–2335. doi: 10.1038/leu.2014.133. [DOI] [PubMed] [Google Scholar]
- 133.McCubrey J.A., Rakus D., Gizak A., et al. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity—diverse effects on cell growth, metabolism and cancer. Biochem Biophys Acta Mol Cell Res. 2016;1863(12):2942–2976. doi: 10.1016/j.bbamcr.2016.09.004. [DOI] [PubMed] [Google Scholar]
- 134.Qualtrough D., Rees P., Speight B., Williams A.C., Paraskeva C. The hedgehog inhibitor cyclopamine reduces β-catenin-Tcf transcriptional activity, induces E-cadherin expression, and reduces invasion in colorectal cancer cells. Canc. 2015;7(3):1885–1899. doi: 10.3390/cancers7030867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Bariwal J., Kumar V., Dong Y., Mahato R.I. Design of Hedgehog pathway inhibitors for cancer treatment. Med Res Rev. 2019;39(3):1137–1204. doi: 10.1002/med.21555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Chen J.K. I only have eye for Ewe: the discovery of cyclopamine and development of Hedgehog pathway-targeting drugs. Nat Prod Rep. 2016;33(5):595–601. doi: 10.1039/c5np00153f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Xu X., Ye J., Huang C., Yan Y., Li J. M2 macrophage-derived IL6 mediates resistance of breast cancer cells to hedgehog inhibition. Toxicol Appl Pharmacol. 2019;364:77–82. doi: 10.1016/j.taap.2018.12.013. [DOI] [PubMed] [Google Scholar]
- 138.Che J., Zhang F.-Z., Zhao C.-Q., Hu X.-D., Fan S.-J. Cyclopamine is a novel Hedgehog signaling inhibitor with significant anti-proliferative, anti-invasive and anti-estrogenic potency in human breast cancer cells. Oncol Lett. 2013;5(4):1417–1421. doi: 10.3892/ol.2013.1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Chai F., Zhou J., Chen C., et al. The Hedgehog inhibitor cyclopamine antagonizes chemoresistance of breast cancer cells. Onco Targets Ther. 2013;6:1643–1647. doi: 10.2147/OTT.S51914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Zhang X., Harrington N., Moraes R.C., Wu M.F., Hilsenbeck S.G., Lewis M.T. Cyclopamine inhibition of human breast cancer cell growth independent of Smoothened (Smo) Breast Canc Res Treat. 2009;115(3):505–521. doi: 10.1007/s10549-008-0093-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Lu Y.L., Ma Y.B., Feng C., et al. Co-delivery of cyclopamine and doxorubicin mediated by bovine serum albumin nanoparticles reverses doxorubicin resistance in breast cancer by down-regulating P-glycoprotein expression. J Cancer. 2019;10(10):2357–2368. doi: 10.7150/jca.30323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Jing Y., Han Z., Zhang S., Liu Y., Wei L. Epithelial-Mesenchymal Transition in tumor microenvironment. Cell Biosci. 2011;1:29. doi: 10.1186/2045-3701-1-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Cucchi D., Occhione M.A., Gulino A., De Smaele E. Hedgehog signaling pathway and its targets for treatment in basal cell carcinoma. J Exp Pharmacol. 2012;4:173–185. doi: 10.2147/JEP.S28553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Belani C.P., Dahlberg S.E., Rudin C.M., et al. Vismodegib or cixutumumab in combination with standard chemotherapy for patients with extensive-stage small cell lung cancer: a trial of the ECOG-ACRIN Cancer Research Group (E1508) Cancer. 2016;122(15):2371–2378. doi: 10.1002/cncr.30062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Liao S., Floyd C., Verratti N., Leung L., Wu C. Analysis of vismodegib resistance in D473G and W535L mutants of SMO receptor and design of novel drug derivatives using molecular dynamics simulations. Life Sci. 2020;244:117302. doi: 10.1016/j.lfs.2020.117302. [DOI] [PubMed] [Google Scholar]
- 146.Tong W., Qiu L., Qi M., et al. GANT-61 and GDC-0449 induce apoptosis of prostate cancer stem cells through a GLI-dependent mechanism. J Cell Biochem. 2018;119(4):3641–3652. doi: 10.1002/jcb.26572. [DOI] [PubMed] [Google Scholar]
- 147.Pace J.R., Jog R., Burgess D.J., Hadden M.K. Formulation and evaluation of itraconazole liposomes for Hedgehog pathway inhibition. J Liposome Res. 2019;30(3):305–311. doi: 10.1080/08982104.2019.1668011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hu Q., Hou Y.C., Huang J., Fang J.Y., Xiong H. Itraconazole induces apoptosis and cell cycle arrest via inhibiting Hedgehog signaling in gastric cancer cells. J Exp Clin Cancer Res. 2017;36(1):50. doi: 10.1186/s13046-017-0526-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Chen B., Trang V., Lee A., et al. Posaconazole, a second-generation triazole antifungal drug, inhibits the hedgehog signaling pathway and progression of basal cell carcinoma. Mol Canc Therapeut. 2016;15(5):866–876. doi: 10.1158/1535-7163.MCT-15-0729-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Kim J., Aftab B.T., Tang J.Y., et al. Itraconazole and arsenic trioxide inhibit Hedgehog pathway activation and tumor growth associated with acquired resistance to smoothened antagonists. Cancer Cell. 2013;23(1):23–34. doi: 10.1016/j.ccr.2012.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lam A., Hoang J.D., Singleton A., Han X., Bleier B.S. Itraconazole and clarithromycin inhibit P-glycoprotein activity in primary human sinonasal epithelial cells. Inter Forum Allergy Rhinol. 2015;5(6):477–480. doi: 10.1002/alr.21454. [DOI] [PubMed] [Google Scholar]
- 152.van Beusekom C.D., Lange R., Schrickx J.A. A functional model for feline P-glycoprotein. J Vet Pharmacol Therapeut. 2016;39(1):95–97. doi: 10.1111/jvp.12248. [DOI] [PubMed] [Google Scholar]
- 153.Huang S.-M.A., Mishina Y.M., Liu S., et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nat. 2009;461(7264):614–620. doi: 10.1038/nature08356. [DOI] [PubMed] [Google Scholar]
- 154.Boutros M., Paricio N., Strutt D.I., Mlodzik M. Dishevelled activates JNK and discriminates between JNK pathways in planar polarity and wingless signaling. Cell. 1998;94(1):109–118. doi: 10.1016/s0092-8674(00)81226-x. [DOI] [PubMed] [Google Scholar]
- 155.Ishitani T., Kishida S., Hyodo-Miura J., et al. The TAK1-NLK mitogen-activated protein kinase cascade functions in the Wnt-5a/Ca(2+) pathway to antagonize Wnt/beta-catenin signaling. Mole Cell Biol. 2003;23(1):131–139. doi: 10.1128/MCB.23.1.131-139.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Schott A.F., Landis M.D., Dontu G., et al. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013;19(6):1512–1524. doi: 10.1158/1078-0432.CCR-11-3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Han J., Shen Q. Targeting γ-secretase in breast cancer. Breast Canc. 2012;4:83–90. doi: 10.2147/BCTT.S26437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Al-Hussaini H., Subramanyam D., Reedijk M., Sridhar S.S. Notch signaling pathway as a therapeutic target in breast cancer. Mol Canc Therapeut. 2011;10(1):9–15. doi: 10.1158/1535-7163.MCT-10-0677. [DOI] [PubMed] [Google Scholar]
- 159.Kondratyev M., Kreso A., Hallett R.M., et al. Gamma-secretase inhibitors target tumor-initiating cells in a mouse model of ERBB2 breast cancer. Oncogene. 2012;31(1):93–103. doi: 10.1038/onc.2011.212. [DOI] [PubMed] [Google Scholar]
- 160.Ran Y., Hossain F., Pannuti A., et al. γ-Secretase inhibitors in cancer clinical trials are pharmacologically and functionally distinct. EMBO Mol Med. 2017;9(7):950–966. doi: 10.15252/emmm.201607265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Liu D., Chen L., Zhao H., Vaziri N.D., Ma S.C., Zhao Y.Y. Small molecules from natural products targeting the Wnt/β-catenin pathway as a therapeutic strategy. Biomed Pharmacother = Biomed Pharmacotherapie. 2019;117:108990. doi: 10.1016/j.biopha.2019.108990. [DOI] [PubMed] [Google Scholar]
- 162.Dutta S., Mahalanobish S., Saha S., Ghosh S., Sil P.C. Natural products: an upcoming therapeutic approach to cancer. Food Chem Toxicol : J Pub British Ind Biol Res Ass. 2019;128:240–255. doi: 10.1016/j.fct.2019.04.012. [DOI] [PubMed] [Google Scholar]
- 163.Phan V., Walters J., Brownlow B., Elbayoumi T. Enhanced cytotoxicity of optimized liposomal genistein via specific induction of apoptosis in breast, ovarian and prostate carcinomas. J Drug Target. 2013;21(10):1001–1011. doi: 10.3109/1061186X.2013.847099. [DOI] [PubMed] [Google Scholar]
- 164.Fan P., Fan S., Wang H., et al. Genistein decreases the breast cancer stem-like cell population through Hedgehog pathway. Stem Cell Res Ther. 2013;4(6):146. doi: 10.1186/scrt357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Gong L., Li Y., Nedeljkovic-Kurepa A., Sarkar F.H. Inactivation of NF-κB by genistein is mediated via Akt signaling pathway in breast cancer cells. Oncogene. 2003;22(30):4702–4709. doi: 10.1038/sj.onc.1206583. [DOI] [PubMed] [Google Scholar]
- 166.Pan H., Zhou W., He W., et al. Genistein inhibits MDA-MB-231 triple-negative breast cancer cell growth by inhibiting NF-κB activity via the Notch-1 pathway. Int J Mol Med. 2012;30(2):337–343. doi: 10.3892/ijmm.2012.990. [DOI] [PubMed] [Google Scholar]
- 167.Yeh C.-C., Fan Y., Jiang L., et al. Genistein suppresses growth of human uterine sarcoma cell lines via multiple mechanisms. Anticanc Res. 2015;35:3167–3173. [PubMed] [Google Scholar]
- 168.Zhang Y., Fang M., Sun Y., et al. Curcumin attenuates cerebral ischemia injury in Sprague-Dawley rats and PC12 cells by suppressing overactivated autophagy. J Photochem Photobiol B Biol. 2018;184:1–6. doi: 10.1016/j.jphotobiol.2018.05.010. [DOI] [PubMed] [Google Scholar]
- 169.Zendedel E., Butler A.E., Atkin S.L., Sahebkar A. Imp Curcumin Sirtuin: Rev. 2018;119(12):10291–10300. doi: 10.1002/jcb.27371. [DOI] [PubMed] [Google Scholar]
- 170.Cao L., Xiao X., Lei J., Duan W., Ma Q., Li W. Curcumin inhibits hypoxia-induced epithelial-mesenchymal transition in pancreatic cancer cells via suppression of the hedgehog signaling pathway. Oncol Rep. 2016;35(6):3728–3734. doi: 10.3892/or.2016.4709. [DOI] [PubMed] [Google Scholar]
- 171.Varghese E., Samuel S.M., Abotaleb M., Cheema S., Mamtani R., Büsselberg D. The “Yin and Yang” of natural compounds in anticancer therapy of triple-negative breast cancers. Cancers (Basel) 2018;10(10):346. doi: 10.3390/cancers10100346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Vallée A., Lecarpentier Y., Vallée J.-N. Curcumin: a therapeutic strategy in cancers by inhibiting the canonical WNT/β-catenin pathway. J Exp Clin Canc Res. 2019;38(1):323. doi: 10.1186/s13046-019-1320-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Marquardt J.U., Gomez-Quiroz L., Arreguin Camacho L.O., et al. Curcumin effectively inhibits oncogenic NF-κB signaling and restrains stemness features in liver cancer. J Hepatol. 2015;63(3):661–669. doi: 10.1016/j.jhep.2015.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Dandawate P.R., Subramaniam D., Jensen R.A., Anant S. Targeting cancer stem cells and signaling pathways by phytochemicals: novel approach for breast cancer therapy. Semin Cancer Biol. 2016;40–41:192–208. doi: 10.1016/j.semcancer.2016.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Yang C.S., Wang X. Green tea and cancer prevention. Nutr Cancer. 2010;62(7):931–937. doi: 10.1080/01635581.2010.509536. [DOI] [PubMed] [Google Scholar]
- 176.Farahmand L., Darvishi B., Majidzadeh-A K., Madjid Ansari A. Naturally occurring compounds acting as potent anti-metastatic agents and their suppressing effects on Hedgehog and WNT/β-catenin signalling pathways. Cell Prolif. 2017;50(1):e12299. doi: 10.1111/cpr.12299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Hong O.Y., Noh E.M., Jang H.Y., et al. Epigallocatechin gallate inhibits the growth of MDA-MB-231 breast cancer cells via inactivation of the β-catenin signaling pathway. Oncol Lett. 2017;14(1):441–446. doi: 10.3892/ol.2017.6108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Gu J.W., Makey K.L., Tucker K.B., et al. EGCG, a major green tea catechin suppresses breast tumor angiogenesis and growth via inhibiting the activation of HIF-1α and NFκB, and VEGF expression. Vasc Cell. 2013;5(1):9. doi: 10.1186/2045-824X-5-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Lee S.H., Nam H.J., Kang H.J., Kwon H.W., Lim Y.C. Epigallocatechin-3-gallate attenuates head and neck cancer stem cell traits through suppression of Notch pathway. Eur J Canc. 2013;49(15):3210–3218. doi: 10.1016/j.ejca.2013.06.025. [DOI] [PubMed] [Google Scholar]