Open Access

Aryl hydrocarbon receptor: An emerging player in breast cancer pathogenesis and its potential as a drug target (Review)

  • Authors:
    • Cong Chen
    • Zhiying Wang
    • Zhihong Liao
    • Yuanqi Zhang
    • Wei Lei
    • Xiaorong Shui
  • View Affiliations

  • Published online on: November 21, 2023     https://doi.org/10.3892/mmr.2023.13134
  • Article Number: 11
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Breast cancer is the most common malignancy in women. Metastatic breast cancer is incurable and is a major cause of shortened patient survival. The different molecular types of breast cancer make targeted therapy difficult and a complex challenge. Aryl hydrocarbon receptor (AhR) is an evolutionarily conserved transcription factor that has been implicated in the metabolism of xenobiotic ligands. AhR is activated by numerous exogenous and endogenous ligands and participates in multiple physiological processes, including proliferation, migration, invasion and apoptosis. AhR expression is upregulated in certain breast cancer subtypes, including estrogen receptor‑positive breast cancer, and has been implicated in the development and progression of breast cancer. Over the last two decades, AhR and its ligands have emerged as novel biological targets for the treatment of breast cancer. Both AhR agonists and antagonists may be effective in inhibiting critical activities of breast cancer. The present review evaluates the role and underlying mechanisms of AhR and its ligands in breast cancer and demonstrates the potential of exploiting AhR as a novel target for breast cancer therapy.

Introduction

Breast cancer is the most common cancer in women (1), with an estimated 2.3 million new cases (accounting for 11.7% of the new cancer cases worldwide) in 2020 (2). Following the development of early diagnostic and treatment strategies over the last 10 years, the 5-year survival rate increased from 75% of patients diagnosed in the mid-1970s to 90% of patients diagnosed from 2011 to 2017 (3). However, the incidence and mortality rates remain high and the survival of patients with distant metastasis has decreased (1,35). In the United States, from 2011 to 2017, the 5-year relative survival rate of patients diagnosed with stage I breast cancer was close to 100%, while the 5-year relative survival rate of patients diagnosed with stage IV breast cancer (also known as metastatic breast cancer) decreased to 29% (1,3). Additionally, the treatment of different molecular subtypes of breast cancer remains a complex challenge (68).

Aryl hydrocarbon receptor (AhR) is a ligand-activated nuclear transcription factor and a member of the basic helix-loop-helix/Per AhR nuclear translocator (ARNT)-Sim transcription factor family (912). AhR has a complex ligand-binding domain that is activated by numerous exogenous and endogenous ligands and natural compounds with different structures and binding affinities (1315). Following ligand binding, AhR translocates into the nucleus (16) to form a heterodimer with ARNT and subsequently transactivate target genes (16,17). AhR is activated by numerous ligands, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and β-naphthoflavone (β-NF), and regulates different target genes depending on the type of ligand (Fig. 1). Exogenous AhR ligands, endogenous ligands and natural products activate AhR through genomic and non-genomic pathways (18). In the genomic pathways, activated AhR acts as a transcription factor to bind dioxin reaction elements (DREs) in promoters and regulate the expression of genes encoding xenobiotic metabolic enzymes, such as cytochrome P450 family 1 subfamily A member 1 (CYP1A1), cytochrome P450 family 1 subfamily A member 2 (CYP1A2), cytochrome P450 family 1 subfamily B member 1 (CYP1B1) and glutathione transferase and aldehyde dehydrogenase (ALDH) (1923). In non-genomic pathways, AhR exerts non-transcriptional activities via the involvement of other transcriptional regulators or signal transducers, such as c-Src, NF-κB or estrogen receptor α (ERα) (2428).

Figure 1.

AhR ligand-activated pathways mediate anticancer activities in breast cancer cells. In triple-negative breast cancer cells, the tumor suppressor characteristics exhibited by different types of AhR ligand may be related to the downregulation of CXCR4, the miR-212/132/SOX4 signaling axis, the JAG1/NOTCH1 signaling pathway and the TGF-β signaling pathway. In estrogen receptor-positive breast cancer cells, the tumor-suppressive characteristics of different types of AhR ligand may be associated with the activation of the MAPK/ERK signaling pathway and the inhibition of the PI3K/AKT signaling pathway, FAK and the expression of MMPs. In addition, a number of AhR ligands may disrupt the interaction between CDK4 and AhR to induce cell cycle arrest in breast cancer cells. (a) The signal pathway involved in AhR ligand in TNBC cells, such as MDA-MB-231 cells. (b) The signal pathway involved in AhR ligand in ER-positive breast cancer cells, such as MCF-7 cells. β-NF, β-naphthoflavone; AF, aminoflavone; AhR, aryl hydrocarbon receptor; ARNT, AhR nuclear translocator; CXCR4, C-X-C motif chemokine receptor 4; CYP1A1, cytochrome P450 family 1 subfamily A member 1; CYP1A2, cytochrome P450 family 1 subfamily A member 2; CYP1B1, cytochrome P450 family 1 subfamily B member 1; DIM, 3,3-diindolylmethane; FAK, focal adhesion kinase; FICZ, 6-formylindolo(3,2-b) carbazole; I3C, indole-3-carbinol; ICZ, indole (3,2-b) carbazole; ITE, 2-(1′h-indole-3′-carbonyl)-thiazole-4-carboxylic acid; JAG1, jagged canonical Notch ligand 1; miR, microRNA; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; XRE, xenobiotic response elements.

AhR regulates numerous important physiological and pathological processes, such as immune response inhibition (16,17,2931), homeostasis of the liver, vascular and cardiovascular systems (3234), tumor induction (11,31), inflammation (17,31,35) and intestinal barrier function (17,36). AhR is also activated by environmental pollutants, such as polycyclic aromatic hydrocarbons (PAHs) and halogenated aromatic hydrocarbons (HAHs), which affect tumor formation (3739).

Previous studies have reported the functional interactions of AhR with certain signaling pathways, including the ERα and the TGF-β pathways (25,40), and its physiological role in regulating a number of cellular processes related to cancer development, including cell proliferation, the cell cycle, cell migration, pluripotency and stemness (40). AhR expression is upregulated in multiple types of cancer, including breast, lung, liver, stomach, head, neck, cervical and ovarian cancer (4145), and its expression in these types of cancer is associated with the stage of the disease (44,45). Research has demonstrated that AhR mediates either pro- or anticancer activities in breast cancer cells, with conflicting evidence linking AhR to breast cancer progression or inhibition (15,4648). A number of structural AhR ligands, such as aminoflavone (AF) and tranilast, can inhibit various aspects of breast carcinogenesis (15). Conversely, AhR ligands such as TCDD have also been reported to enhance the growth and development of breast cancer (4648).

In the present review, the roles of AhR and its ligands as breast cancer inhibitors and promoters in vitro and in vivo are summarized. The potential role of AhR as a novel target for breast cancer therapy is also evaluated.

Association between AhR and breast cancer

Breast cancer is a heterogeneous disease with different molecular subtypes, and the molecular type of breast cancer is closely related to prognosis (49,50). Breast cancer is classified into three types on the basis of the expression of specific hormone and growth factor receptors: Hormone receptor (HR)-positive breast cancer, HER-2-positive breast cancer and triple-negative breast cancer (TNBC; HR-negative and HER-2 negative) (4952). HRs include ER and progesterone receptor (PR).

Current treatments for breast cancer include surgery, chemotherapy, radiotherapy, endocrine therapy and targeted therapy. The treatment regimen depends on the tumor subtype, the expression of HRs and HER-2, and whether the tumor is non-invasive (carcinoma in situ) or invasive (4,49,50). The choice of chemotherapy and endocrine therapy depends on the presence of ER, PR and HER-2. HER-2-positive breast cancer can often be successfully treated with trastuzumab, pertuzumab and lapatinib (53). The most common form of endocrine therapy for HR-positive cancer is selective ER modulators (SERMs), such as tamoxifen and aromatase inhibitors, both of which can inhibit estrogen biosynthesis (54). Aromatase inhibitors can improve cancer-associated outcomes in the management of HR-positive breast cancer, which may reduce the incidence of new primary breast cancer but have less of an effect on more severe distant recurrences (55). These drugs are usually administered with CDK4/6 inhibitors to increase the sensitivity of HR-positive and HER-2 negative metastatic breast tumors to chemotherapy (56).

The role of AhR in the development of breast cancer has been widely studied (Table I). AhR is involved in normal mammary gland development (57), but is upregulated in certain breast cancer subtypes and has a prognostic role (58,59). For example, compared with ER-negative breast cancer, the upregulation of AhR expression in ER-positive breast cancer is associated with higher rates of survival, including increased overall survival, distant metastasis-free survival at admission and recurrence-free survival (58). By contrast, inflammatory breast cancer (IBC) tissues have higher expression levels of AhR compared with non-IBC tissues, and upregulation of AhR expression is positively associated with poor clinical prognosis, including lymphovascular invasion and lymph node metastasis (60). A meta-analysis on the prognostic impact of AhR in breast cancer indicated that AhR is also a marker of poor outcome in patients with node-negative breast cancer (61). In a cohort study, compared with normal breast tissue, the high expression of AhR in breast tumors is associated with inflammation and the expression of endogenous tryptophan metabolism genes, but is only weakly related to a classic diagnostic factor (age) (62). Another cohort study found that low cytoplasmic AhR levels are associated with more aggressive ER negative tumors (63). The impact of AhR on the prognosis of primary breast cancer depends on the type of SERMs (63). Additionally, knockdown of AhR serves a key role in the malignant characteristics of breast cancer, with functions in proliferation, migration, invasion, apoptosis and angiogenesis (64,65). A number of environmental toxicants, such as TCDD, are AhR ligands (66). Chronic exposure to low doses of environmental pollutants promotes cancer metastasis and generates chemoresistance through epithelial-mesenchymal transition (EMT) and cancer stemness (the stem cell-like phenotype in tumors) (66). However, another meta-analysis demonstrates that AhR (rs2066853) polymorphism does not modify the risk of breast cancer (67). These findings suggest that AhR may be a promising potential drug target for breast cancer treatment.

Table I.

Effect of AhR expression on breast cancer outcomes.

Table I.

Effect of AhR expression on breast cancer outcomes.

First author/s, yearSamplesAhR expressionClinical prognosis(Refs.)
O'Donnell et al, 2014Online tool KMPLOT based on the updated 2012 datasetHigh AhR expressionThe prognosis of ER-positive breast cancer was improved compared with ER-negative breast cancer. ER-positive breast cancer: Overall, distant metastasis-free and relapse-free survival was increased. Basal subtype breast cancer: Relapse-free survival was improved.(58)
Romagnolo et al, 2015Human normal and breast tumor tissue sectionsIncreased AhR expression in TNBC (~3.0-fold of control)-(59)
Mohamed et al, 2018Cohort study of 14 healthy volunteers undergoing breast reduction mammoplasty and 61 patients with breast cancer (33 non-IBC and 28 IBC)AhR and CYP1B1 mRNA and protein levels higher in IBC compared with in non-IBC tissuesAhR and CYP1B1 mRNA expression were positively associated with the number of metastatic lymph nodes and with tumor grade, lymphovascular invasion and Ki-67 expression in IBC.(60)
Jeschke et al, 2019Meta-analysis of 302 paraffin-embedded breast tumor tissue samples from 297 patients with primary breast cancer (5 were bilateral breast cancer)High AhR expressionTotal and nuclear expression were associated with poor outcomes in LN-negative disease. Total AhR was an independent prognostic marker only in the sub-group of LN-negative patients. LN-positive patients: Overall survival was increased.(61)
Vacher et al, 2018Cohort study of 439 primary unilateral invasive breast tumors excised from women managed at the Institut Curie-René Huguenin Hospital (Saint-Cloud, France) between 1978 and 2008AhR mRNA levels higher in tumor specimens compared with in normal breast tissue samplesAhR mRNA expression was weakly associated only with one classical prognostic factor (age).(62)
Tryggvadottir et al, 2021Cohort study of 1,116 patients with breast cancer between October 2002 and June 2012 in Sweden-Low cytosolic AhR levels were associated with more aggressive ER-negative tumors. The prognostic impact of AhR was substantially modified by the treatment of different types of SERMs such as tamoxifen, raloxifene and aromatase inhibitors.(63)
Li et al, 2015Meta-analysis of 2,999 patients and 3,050 controls from three related case-control studies-AhR (rs2066853) polymorphism did not modify the risk of breast cancer.(67)

[i] AhR, aryl hydrocarbon receptor; CYP1B1, cytochrome P450 family 1 subfamily B member 1; ER, estrogen receptor; IBC, inflammatory breast cancer; KMPLOT, Kaplan-Meier plotter; LN, lymph node; TNBC, triple-negative breast cancer; SERMs, selective estrogen receptor modulators.

AhR pathways in breast cancer

Crosstalk between AhR and ERα

ERα is a transcription factor that is activated in >70% of patients with breast cancer (68,69). Tamoxifen, an antagonist of ERα, is the first-line treatment for HR-positive breast cancer. It inhibits the activity of ERα, thereby interfering with aberrant ERα transcriptional activity and prolonging patient survival (70,71). AhR is expressed in ER-positive and -negative breast cancer cells (58,59). Certain studies have reported that the AhR target gene CYP1A1 is activated by TCDD only in ER-positive breast cancer cells (24,25,72). TCDD is a well-studied environmental pollutant, an effective immunosuppressant and one of the most potent exogenous agonists of AhR (9,11,73,74). Transfection into MDA-MB-231 cells with an ERα overexpression vector confers AhR ligand sensitivity, whereas ERα knockdown in MCF-7 cells confers resistance to the same ligand (72,75). Therefore, ER expression may affect the activity of AhR in breast cancer. Additionally, several studies on the anticancer effects of AhR ligands have reported that the crosstalk between the AhR pathway and ERα can influence the selectivity and resistance of different molecular subtypes of breast cancer cells, such as MCF-7 and MDA-MB-231 cells, to AhR ligands (25,7577).

ARNT functions as a modulator of ERs (78). Dioxin-type environmental pollutants, such as TCDD and 3-methylcholanthrene (3MC), are AhR agonists that modulate ER-mediated estrogen signaling by activating AhR/ARNT, leading to estrogen-associated adverse effects, such as endometrial hyperplasia (24,79). In an animal study, it was reported that rats chronically exposed to TCDD have a lower incidence of mammary and uterine tumors compared with that in rats not exposed to TCDD (80). The mechanism may involve TCDD interference with the ERα signaling pathway and activation of AhR, thereby affecting the metabolism of estrogen through a mechanism involving CYP1A1 and CYP1B1 (25,26,76).

Botanical estrogens (BEs), although not estrogens, are natural phytocompounds that bind to ER and are commonly used in hormone replacement therapy for menopausal women (81). A study on the effects of BEs and estradiol (E2) on liver cells and ER-positive breast cancer cells reported that both treatments cause the upregulation of ERα activity and enhance the proliferation of breast cancer cells, whilst E2 has no significant effect on the stimulation of AhR (82). Additionally, it has been reported that BEs act via the AhR pathway to bind to xenobiotic response elements and upregulate CYP1A1 and CYP1B1, whereas E2 only acts through ER (82). This indicates that the crosstalk between AhR and ER is ligand- and cell-specific.

Crosstalk between AhR and BRCA1

Among the genetic factors that drive breast cancer, the BRCA1 and BRCA2 tumor suppressor genes serve an important role in breast cancer susceptibility (27,83). BRCA proteins are involved in cell cycle progression, apoptosis, DNA repair and transcription (84). BRCA1 interacts with the estrogen pathway at the transcriptional and post-transcriptional levels to limit the effects of estrogen on the promotion of mammary gland growth. BRCA-regulated transcription occurs via protein-protein interactions, the most important of which is via a complex formation with ERα, leading to the transactivation of ERs (27,85). The absence of this control, through a BRCA1 gene mutation, is a well-known risk factor for TNBC development (84).

In ER-positive breast cancer cells, BRCA1 has been associated with the AhR pathway. A study reported that upon ligand activation, BRCA1 was recruited to the promoter regions of CYP1A1 and CYP1B1, together with ARNT and AhR. However, this was not observed in ER-negative cells, suggesting an association between ER and BRCA1 presence (27,86,87). In the mammary glands, BRCA1 limits aromatase expression, and thus estrogen production, and AhR ligand-induced BRCA1 inhibition results in the increase of aromatase and E2 in tumor cells, thereby maintaining cell proliferation (84,85). In breast cancer cells treated with AhR agonists, activation of the aromatase gene and an increase in E2 production have been reported (88). Previous studies have reported that AhR inhibits ER-dependent signaling by recruiting the proteasome complex (79,89). Furthermore, BRCA1 activates the ESR1 gene, which encodes ERα (90). Therefore, the paradoxical effects of activated AhR on the inhibition of ERα and the increased expression of E2 induced by activated AhR may be associated with the inhibition of BRCA1 by AhR.

AhR ligands exhibit tumor-suppressive properties

TCDD and structurally related HAHs

The anticancer effects of the AhR agonist TCDD and structurally related HAHs in breast cancer in vivo and in vitro models are summarized in Table II. In one study, seven ER-negative breast cancer cell lines were treated with six ligands, including TCDD and 6-methyl-1,3,8-trichlorodibenzofuran (MCDF), and it was reported that these ligands inhibited the proliferation of ER-negative breast cancer cells (91). Other studies reported that TCDD inhibited the invasion of different types of breast cancer cell lines, including ER-positive (MCF-7 and ZR75), ER-negative (MDA-MB-231) and HER-2-positive (BT474 and SKBR3) breast cancer cells (9294). In a study of AhR regulation of cell cycle progression in human breast cancer cells, disruption of the interaction of AhR with CDK4 by TCDD inhibited cell cycle progression in MCF-7 and MDA-MB-231 cells (95). In another study of 4T1 murine breast cancer cells in a syngeneic mouse model, TCDD was reported to inhibit lung metastasis of the primary tumor but did not impact primary tumor growth (96). MCDF, a partial AhR antagonist, has been reported to inhibit CYP1A1 induction by TCDD in cell culture (97). Zhang et al (92) reported that MCDF inhibited the proliferation and invasion of HER-2-positive (BT474) and ER-negative (MDA-MB-231) cells and inhibited lung metastasis in an athymic nude mouse xenograft model bearing tumors from MDA-MB-231 cells. Additionally, MCDF has been reported to inhibit tumor growth in an athymic nude mouse xenograft model bearing tumors from MDA-MB-468 cells (91). These results obtained using TCDD and structurally related HAHs as AhR ligands suggest that these compounds exhibit anticancer effects in breast cancer.

Table II.

TCDD and structurally related halogenated aromatic hydrocarbons as inhibitors of breast cancer progression.

Table II.

TCDD and structurally related halogenated aromatic hydrocarbons as inhibitors of breast cancer progression.

First author/s, yearLigandStudy typeCell line/animal modelResponseTargeted signaling pathway(Refs.)
Narasimhan et al,TCDDIn vitroSUM149, Hs578T,↓Irregular colony-(48)
2018 BP1growth
Zhang et al, 2009TCDDIn vitroBT474, HCC-38,↓Proliferation-(91)
MDA-MB-157,
MDA-MB-435,
MDA-MB-436,
MDA-MB-453,
MDA-MB-468
Zhang et al,TCDDIn vitroMDA-MB-231↓Migration,-(92)
2012 ↓invasion
BT474↓Invasion-
Hanieh, 2015TCDDIn vitroT47D↓Migration-(93)
MDA-MB-231↓Migration,AhR-miR-212/
↓invasion132-SOX4 pathway
In vivoMDA-MB-231 in↓Spontaneous-
athymic nude micemetastasis
Hall et al, 2010TCDDIn vitroMDA-MB-231,↓Invasion,-(94)
MCF-7, ZR75,↓colony
SKBR3formation
Barhoover et al,TCDDIn vitroMCF-7,↓Cell cycleInteraction of AhR(95)
2010 MDA-MB-231progressionand CDK4
Wang et al, 2011TCDDIn vitro4T1 ↔Proliferation,-(96)
↔migration,
↔colony formation
In vivo4T1 in Balb/c mice↓Lung metastasis,-
↔tumor growth
Zhang et al, 2009MCDFIn vitroBT474, HCC-38,↓Proliferation-(91)
MDA-MB-157,
MDA-MB-435,
MDA-MB-436,
MDA-MB-453,
MDA-MB-468
In vivoMDA-MB-468 in↓Tumor growth-
athymic nude mice
Zhang et al, 2012MCDFIn vitroMDA-MB-231↓Migration,-(92)
↓invasion
BT474↓Invasion-
BT474,↓Proliferation-
MDA-MB-231 -
MDA-MB-231 in↓Lung metastasis
athymic nude mice
Zhang et al, 2009TCDF, PCDD,In vitroBT474,↓Proliferation-(91)
PCDF, PCB MDA-MB-468

[i] ↑, increase; ↓, decrease; ↔, no change. AhR, aryl hydrocarbon receptor; MCDF, 6-methyl-1,3,8-trichlorodibenzofuran; miR, microRNA; PCB, 3,3′,4,4′,5-pentachlorobiphenyl; PCDD, 1,2,3,7,8-pentachlorodibenzo-p-dioxin; PCDF, 2,3,4,7,8-pentachlorodibenzofuran; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; TCDF, 2,3,7,8-tetrachlorodibenzofuran.

Endogenous and natural AhR ligands

Most endogenous AhR ligands exhibit anticancer activity in breast cancer (Table III). Cruciferous vegetables contain a number of compounds that function as AhR ligands with anticancer activity, including indole-3-carbinol (I3C), indole (3,2-b) carbazole (ICZ) and 3,3-diindolylmethane (DIM) (48,93,98,99). For example, in one study, DIM suppressed the invasive and metastatic activities of ER-positive (MCF-7 and ZR-75), ER-negative (MDA-MB-231) and HER-2-positive (SKBR3) breast cancer cells, and knockdown of AhR reversed this effect (94). Another study reported that DIM activated AhR and inhibited the migration and invasion of MDA-MB-231 and T47D cells via the AhR-microRNA (miRNA/miR)-212/132-SOX4 signaling axis (93). A further in vivo study reported that DIM inhibited orthotopic tumor growth and spontaneous lung metastasis in a xenograft model (93). A prospective clinical trial of healthy women with positive BRCA expression also revealed that DIM reduced the risk of breast cancer development (100). Nguyen et al (99) established a lymphatic barrier model using three-dimensional lymphatic endothelial cells as a monolayer co-cultured with spheroids of MDA-MB-231 cells. AhR silencing and an AhR antagonist (DIM) or an endogenous AhR ligand [6-formylindolo(3,2-b) carbazole (FICZ)] reduced or increased lymphatic barrier invasion, respectively. FICZ also exerted antiproliferative and anti-migratory effects on MCF-7 cells possibly by regulating the expression of miRNAs (101). Another endogenous ligand, 2-(1′h-indole-3′-carbonyl)-thiazole-4-carboxylic acid, acts as an AhR agonist to inhibit the proliferation, migration and invasion of MDA-MB-231 cells, possibly through jagged 1/NOTCH1 signaling; however, this was not observed in MCF-7 cells (102). I3C and ICZ inhibit the migration of breast cancer cells by inhibiting focal adhesion kinase expression to reduce MMP activity and inhibit the EMT process (98). Tryptophan metabolites, indoxyl sulfate (IS) and indole propionic acid (IPA), which are AhR ligands, suppress EMT and cancer stem cell (CSC) numbers by inducing oxidative stress, and thus inhibit the colony formation of 4T1 cells and the metastasis of 4T1 cells in mice. AhR antagonist CH223191 has also been reported to inhibit IS- and IPA-evoked effects (103,104). These results indicate that these compounds inhibit some pro-cancerous activities in breast cancer.

Table III.

Endogenous AhR ligands exhibit anticancer activity in breast cancer.

Table III.

Endogenous AhR ligands exhibit anticancer activity in breast cancer.

First author/s, yearLigandStudy typeCell line/animal modelResponseTargeted signaling pathway(Refs.)
Piwarski et al,ITEIn vitroMCF-7↓AhR, ERα, ↔proliferation,-(102)
2020 ↔invasion, ↔migration
MDA-MB-231↓JAG1, AhR, NICD1,JAG1-NOTCH1
phospho-STAT3,signaling pathway
↓proliferation, ↓invasion,
↓migration
MCF-7,↓JAG1-
MDA-MB-231,
MDA-MB-157,
MDA-MB-436
Bekki et al, 2015KynIn vitroP20E↓Apoptosis (Dox-treated)-(150)
Novikov et al,Kyn,In vitroSUM149↑MigrationTDO2-AhR signaling(153)
2016XA pathway
D'Amato et al,KynIn vitroBT549↑AhR transcriptionalTDO2-AhR signaling(166)
2015 activitypathway
BT549 (forced↑AhR transcriptional-
suspension culture)activity, ↑Resistance to
anoikis
SUM159 (forced↑Resistance to anoikis-
suspension culture)
Narasimhan et al,DIMIn vitroSUM149, Hs578T,↑Migration, ↓irregular-(48)
2018 BP1colony growth
Hanieh, 2015DIMIn vitroMDA-MB-231↓Migration, ↓invasionAhR-miR-212/(93)
132-SOX4 pathway
T47D↓Migration,-
↓proliferation, ↓invasion
In vivoMDA-MB-231 in↓Lung metastasis-
athymic mice
T47D in athymic↓Lung metastasis,-
mice↓tumor growth
Hall et al, 2010DIMIn vitroMCF-7,↓Invasion, ↓colony-(94)
MDA-MB-231formation
SKBR3, ZR-75-1↓Colony formation-
Nguyen et al,DIMIn vitroMDA-MB-231↓CCIDs formation,-(99)
2016 ↓lymphatic barrier
invasion, ↓12(S)-HETE
Nguyen et al,FICZIn vitroMDA-MB-231↑CCIDs formation,-(99)
2016 ↑lymphatic barrier
invasion, ↑12(S)-HETE
Ho et al, 2013I3CIn vitroMCF-7,↓Migration-(98)
MDA-MB-231
Ho et al, 2013ICZIn vitroMCF-7↓Migration-(98)
Sári et al, 2020ISIn vitro4T1↓Colony formation,-(103)
↓migration
In vivo4T1 in BALB/c mice↓Metastasis-
Sári et al, 2020IPAIn vitro4T1↓Colony-(104)
formation
In vivo4T1 in BALB/c mice↓Metastasis-

[i] ↑, increase; ↓, decrease; ↔, no change. 12(S)-HETE, 12(S)-hydroxy eicosatetraenoic acid; AhR, aryl hydrocarbon receptor; CCIDs, circular chemorepellent induced defects; DIM, 3,3-diindolylmethane; Dox, doxorubicin; ERα, estrogen receptor α; FICZ, 6-formylindolo(3,2-b) carbazole; ICZ, indole (3,2-b) carbazole; ITE, 2-(1′h-indole-3′-carbonyl)-thiazole-4-carboxylic acid; IS, indoxyl-sulfate; IPA, indole propionic acid; I3C, indole-3-carbinol; JAG1, jagged canonical Notch ligand 1; Kyn, kynurenine; miR, microRNA; NICD1, NOTCH1 intracellular domain 1; P20E, 1 nM E2 selection process for MCF-10AT1 cells, the AhR-overexpressing breast cancer cells; TDO2, tryptophan-2,3-dioxygenase; XA, xanthurenic acid.

Synthetic and pharmaceutical AhR ligands

Several potential AhR ligands have been identified and designed. Some of these compounds are categorized as selective AhR modulators (105), exhibiting low to medium affinity to AhR. These ligands activate AhR in both genomic and non-genomic pathways to influence breast cancer tumorigenesis and metastasis (15) (Table IV).

Table IV.

Anticancer effects of synthetic and pharmaceutical aryl hydrocarbon receptor ligands in breast cancer.

Table IV.

Anticancer effects of synthetic and pharmaceutical aryl hydrocarbon receptor ligands in breast cancer.

First author/s, yearLigandStudy typeCell line/animal modelResponseTargeted signaling pathway(Refs.)
Campbell et al,AFIn vitroTamR MCF-7↓Mammosphere formation, α6-integrin-Src-AKT(114)
2018 ↓proliferation (tamoxifen-pathway
induced), ↑sensitivity to
tamoxifen, ↓α6-integrin
(α6A and α6B), ↑BAX
ZR-75-30, BT474↓Mammosphere formation-
BT474↓α6-integrin, ↑sensitivity to-
tamoxifen,
Zhao et al, 2012β-NFIn vitroMCF-7↓Mammosphere formation,-(117)
↓secondary mammosphere
formation, ↓the proportion
of cells with high ALDH
activity
Wang et al, 2014β-NFIn vitroMCF-7↓Proliferation, ↓cell cyclePI3K/AKT and(118)
progression, ↓cyclin D1 andMAPK/ERK
CDK4signaling
MDA-MB-231↔Proliferation, ↔cell cycle-
progression
Fukasawa et al,NK150460In vitroMCF-7, T47D,↓Proliferation-(119)
2015 MDA-MB-453,
MDA-MB-468,
SK-BR-3
In vivoZR-75-1 in a nude↓Tumor growth-
rat xenograft model
Gilbert et al,ANI-7In vitroMCF-7, MCF-10A,↓Proliferation-(121)
2018 MDA-MB-231,
MDA-MB-468,
BT20, BT474,
T47D, ZR-75-1,
SKBR3
MDA-MB-468↓Cell cycle progression,-
↑survival (siRNA-AhR),
↑XRE promotor activity,
↑AhR, CYP1A1, CYP1A2,
CYP1B1
O'Donnell et al, 2014RaloxifeneIn vitroMDA-MB-231↑Apoptosis-(58)
Ning et al, 2007Y134,In vitroMCF-7, T47D,↓Proliferation-(132)
Raloxifene MDA-MB-231
Jang et al, 2017Y134In vitroMDA-MB-231,↑Apoptosis-(133)
MDA-MB-436
Jin et al, 2012OmeprazoleIn vitroMDA-MB-468↓Migration, ↓AhR,-(125)
↑CYP1A1 mRNA,
CYP1B1 mRNA,
↑CYP1A1, CYP1B1,
↑DRE promotor activity
In vitroBT474↓AhR, ↑CYP1A1 mRNA,-
CYP1B1 mRNA,
↑CYP1A1, ↔ CYP1A1
Jin et al, 2014OmeprazoleIn vitroMDA-MB-231↓Migration, ↓invasion,-(138)
↓MMP9, CXCR4
In vivoMDA-MB-231 in↓Lung metastasis-
athymic nude mice
Prud'hommeTranilastIn vitroMDA-MB-231↓Proliferation, ↓colony-(124)
et al, 2010 formation, ↓Mammosphere
formation, ↓secondary
mammosphere formation,
↓CD133, Oct-4, ↑CYP1A1
BT474↓Proliferation, ↓colony-
formation, ↓mammosphere
formation, ↓secondary
mammosphere formation
In vivoMitoxantrone-↓Tumor growth; ↓lung-
selected MDA-metastasis
MB-231 in NOD
scid gamma mice
Jin et al, 2012TranilastIn vitroMDA-MB-468↓Migration, ↑AhR,-(125)
↔CYP1A1 mRNA,
↑CYP1B1 mRNA,
↔CYP1A1, ↑CYP1B1
BT474↑AhR, ↑CYP1A1-
mRNA, ↔CYP1B1
mRNA, ↑CYP1A1,
↔CYP1B1
ChakrabartiTranilastIn vitro4T1, LA7, MDA-↓Proliferation-(142)
et al, 2009 MB-231, MCF-7
4T1↓EMT, ↓cell-cycleTGF-β signaling
progressionpathway
In vivo4T1 in BALB/c↓Tumor growth,-
nude mice↓lung metastasis

[i] ↑, increase; ↓, decrease; ↔, no change. β-NF, β-naphthoflavone; AF, aminoflavone; ALDH, aldehyde dehydrogenase; ANI-7, (Z)-2-(3,4-dichlorophenyl)-3-(1H-pyrrol-2-yl) acrylonitrile; CXCR4, C-X-C motif chemokine receptor 4; CYP1A1, cytochrome P450 family 1 subfamily A member 1; EMT, epithelial-to-mesenchymal transition; NK150460, 5,6,7,8-tetrahydrocarcinolin-5-ol; TamR, tamoxifen-resistant.

Aminoflavone (AF) has been reported to potently inhibit the proliferation of ER-positive MCF-7 cells (106,107), and it has been clinically tested in patients with breast cancer (108). Mechanistic studies reported that AF activated the AhR pathway and induced the expression of CYP1A1 and CYP1A2 (109,110), forming metabolites that covalently bonded to DNA. These metabolites inhibited DNA synthesis by inducing S-phase arrest and phosphorylation of H2AX (a replication dependent histone), leading to DNA double-strand breaks and ultimately cytotoxicity (77,111,112). Previous studies have reported that AF inhibits α6-integrin expression (113115). Upregulation of this cell adhesion molecule is associated with tumor-initiating cell proliferation, malignant breast cancer progression and poor prognosis (115). Furthermore, α6-integrin upregulation is associated with radiotherapy resistance and tamoxifen resistance in breast cancer (114,116). In tamoxifen-resistant MCF-7 and BT474 cells, AF has been reported to decrease α6-integrin expression, inhibit α6-integrin-Src-AKT signaling and inhibit tamoxifen resistance of the ER-positive cells (114). A study reported that β-NF, a strong inducer of CYP1A1, had antitumor activity in vitro against ER-positive (MCF-7) (117). A study reported that β-NF mediates cell cycle arrest in ER-positive breast cancer cells via AhR-dependent regulation of PI3K/AKT and MAPK/ERK signaling, leading to cellular senescence (118). This inhibition of proliferation was not observed in MDA-MB-231 cells and was reported to be AhR-dependent (118). Furthermore, a report identified 5,6,7,8-tetrahydrocarcinolin-5-ol (NK150460) as a noncompetitive inhibitor of E2-dependent transcriptional regulation for the potential treatment of ER-positive breast cancer (119). Simultaneous treatment of MCF-7 cells with NK150460 and AhR antagonists demonstrated that inhibition of ER transcriptional regulation by NK150460 was mediated by AhR pathway modulation and CYP1A1 induction. In addition, the study also reported that NK150460 not only inhibited the proliferation of several ER-positive breast cancer cell lines such as MCF-7 and T47D cells, but also some ER-negative cell lines such as MDA-MB-453, MDA-MB-468 and SKBR3 cells (119). Another study reported that (Z)-2-(3,4-dichlorophenyl)-3-(1H-pyrrol-2-yl) acrylonitrile (ANI-7), a member of the acrylonitrile family, exhibited good cytotoxic activity (120). This compound inhibited the proliferation of different breast cancer cell lines, including ER-positive (MCF-7) breast cancer cells, TNBC (MDA-MB-231) cells and HER-2-positive (SKBR3) breast cancer cells (121). MDA-MB-468 cells treated with ANI-7 exhibited S phase and G2 + M phase cell cycle arrest, and this effect was mediated by the AhR pathway and specifically increased CYP1A1 expression levels (121).

Several studies have assessed the repurposing of drugs to identify novel compounds to target the AhR pathway. Consequently, drugs with agonistic activities for AhR have been identified (22,122). These drugs included the antiosteoporosis drug raloxifene (58), the proton pump inhibitor omeprazole (123) and the antiallergen drug tranilast (124,125).

Raloxifene is a selective ER-targeted drug that is a second generation SERM and has been approved for the prevention of osteoporosis in postmenopausal women (54), to whom it is frequently administered. It has been reported to reduce the risk of breast cancer, and it has been reported to have high efficacy, comparable to that of tamoxifen (126129). Raloxifene exhibits estrogenic properties at low concentrations, and in vivo studies have reported that the administration of 1–20 mg/kg/day raloxifene inhibits mammary tumor growth in rats (130,131). In a study that screened novel activators of the AhR pathway, raloxifene was reported to induce AhR nuclear localization in MDA-MB-231 (ER-negative) and Hepa1 cells at levels similar to that of TCDD and induce apoptosis in ER-negative breast cancer cells in an AhR-dependent manner (58). In a previous study, the raloxifene analog Y134, which serves as an AhR ligand, induced apoptosis in TNBC (MDA-MB-231 and MDA-MB-436) cells in an AhR-dependent manner, and also inhibited the proliferation of ER-positive (MCF-7, T47D) breast cancer cells and ER-negative (MDA-MB-231) breast cancer cells (132,133). The study also reported a low toxicity profile in a zebrafish embryo model (133). As aforementioned, SERMs, such as raloxifene and aromatase inhibitors, are currently also used to treat osteoporosis; however, this does not interfere with the role of SERMs as cancer drugs (54,134). Thus, there is potential for the use of raloxifene and Y134 via the AhR pathway for the treatment of breast cancer.

The proton pump inhibitor omeprazole is used clinically to primarily treat peptic ulcers. A number of studies have reported that omeprazole acts on the AhR pathway (22,123,135), while in liver and pancreatic cancer cells, it does not bind directly to AhR but activates it through a nongenomic pathway (14,136,137). Omeprazole, identified as an AhR activator and inducer of CYP1A1, promotes the expression of AhR-induced DREs (22). The propensity of omeprazole to displace TCDD has additionally been demonstrated by AhR competitive ligand binding experiments (22). Another study reported the upregulation of CYP1A1 via the AhR pathway in omeprazole-treated BT474 and MDA-MB-468 cells (125). Another study by the same group reported that omeprazole inhibited the lung metastasis of MDA-MB-231 cells in athymic nude mice (138). Treatment of MDA-MB-231 cells with omeprazole in vitro inhibited cell migration and invasion by upregulating CYP1A1 and downregulating C-X-C motif chemokine receptor 4 via the AhR pathway (138). Omeprazole is the most commonly used drug in digestive diseases and its good overall safety profile, combined with its inhibition of breast cancer invasion and metastasis via the AhR pathway, suggests that it may be a promising targeted breast cancer drug (15,125,138).

The anti-allergic drug tranilast is commonly used to treat bronchial asthma and allergic rhinitis (139,140). Its AhR-inducing activity was first revealed in a study that reported its inhibition of the activity of breast CSCs. The study also reported that tranilast was effective in vivo, as it inhibited lung metastasis in mice injected with triple-negative (MDA-MB-231) mitoxantrone-selected cells (124). AhR knockdown or treatment with the AhR antagonist α-naphthoflavone (α-NF) completely abolished the anti-CSC activity of tranilast (124). CSCs are a type of pluripotent cell that express stem cell marker genes, such as the OCT4 and ALDH genes, and exhibit self-renewal ability, making them immortal (141). Chakrabarti et al (142) reported that tranilast has no cytotoxicity on 4T1 cells (an estrogen-independent mouse breast cancer cell) and inhibited the proliferation of certain breast cancer cells, such as MDA-MB-231 and MCF-7 cells, in vitro. The study also reported that tranilast inhibited tumor growth and lung metastasis in vivo, while tranilast inhibited EMT and cell cycle progression of 4T1 cells through the TGF-β signaling pathway in vitro. In another study, tranilast inhibited the proliferation, migration and colony formation, and stimulated apoptosis of HER-2-positive (BT474) and triple-negative (MDA-MB-231) cells. BT474 cells were more responsive to treatment with tranilast than MDA-MB-231 cells (143). Following treatment with tranilast, the expression of CYP1A1 in BT474 cells was induced, whereas CYP1B1 expression was induced in MDA-MB-468 cells (125). CSCs serve key roles in tumor metastasis, and AhR may be a potential target for the inhibition of CSCs (41,124). Thus, the use of tranilast for the treatment of breast cancer requires further evaluation.

Other drugs, such as the nonsteroidal antiandrogen drug flutamide (22), the nonsteroidal anti-inflammatory drug sulindac (144), the calcium ion antagonist nimodipine (22) and the antiarrhythmic drug mexiletine (22), also exhibit AhR-inducing activity. These may all affect breast cancer development and metastasis via the AhR pathway (15,22,125).

In summary, structurally diverse AhR ligands have been reported to inhibit breast carcinogenesis in multiple breast cancer cell lines and xenograft models (Table II, Table III, Table IV). AhR ligands have distinct actions that may be governed by different mechanisms (Figs. 1 and 2), depending on the ligand structure and cell context. For example, the AhR ligand mediated antitumor effect is associated with the TGF-β signaling pathway or PI3K/AKT signaling pathway or MAPK/ERK signaling pathway. However, AhR ligand mediated pro-cancer activity is associated with the Wnt/β-catenin signaling pathway or PTEN-PI3K/AKT signaling pathway or several molecules such as the glucocorticoid receptor (GR). The binding affinity of AhR for the same AhR ligand varies among species, likely due to species-specific biochemical and physiological properties of AhR resulting from differences in the amino acid sequence of the ligand-binding domain (145). Furthermore, both AhR inhibitors and agonists may lead to similar outcomes if the inhibitors block signaling pathways driven by the endogenous ligands, whereas the exogenous ligands drive different pathways, effectively ‘diverting’ the signaling (146).

Figure 2.

AhR ligand-activated pathways mediate pro-cancer activities in breast cancer cells. HAHs (such as 2,3,7,8-tetrachlorodibenzo-p-dioxin), PAHs (such as benzo[a]pyrene) and structural analogues (such as chlorpyrifos) may enhance cell motility and promote cell migration and invasion via the HDAC6/c-Myc, c-Src/HER1/STAT5b, HER1/ERK1/2, Wnt/β-Catenin and PTEN/PI3K/AKT signaling pathways. (A) In triple-negative breast cancer cells, endogenous tryptophan metabolites as AhR ligands also participate in distant metastasis of cells. (B) Ligand-activated AhR interacts with a variety of signaling molecules to mediate the cancer promoting effect of breast cancer cells. (a) The interaction of AhR and GR inhibits apoptosis by inducing the expression of Brk. (b) The interaction of AhR with both ERα and NOS promotes the expression of VEGF, while the interaction of AhR with NOS promotes the expression of COX-2. Subsequently, the increased expression levels of VEGF and COX-2 promotes angiogenesis. (c) The interaction of AhR and TNF-α promotes the production of inflammatory factors such as IL-6. (d) The interaction of AhR and IL-6 induces DNA damage via the miR-27b-CYP1B1 signaling pathway. AhR, aryl hydrocarbon receptor; AP1, activator protein 1; ARNT, AhR nuclear translocator; Brk, breast tumor kinase; C/EBPβ, CCAAT/enhancer binding protein β; COX-2, cyclooxygenase-2; CREB1, cAMP responsive element binding protein 1; CYP1A1, cytochrome P450 family 1 subfamily A member 1; CYP1B1, cytochrome P450 family 1 subfamily B member 1; ER, estrogen receptor; GR, glucocorticoid receptor; HDAC6, histone deacetylase 6; HER1, human epidermal receptor; HIF-1α, hypoxia-inducible factor 1α; IL-6, interleukin-6; Kyn, kynurenine; KYNU, kynureninase; miR, microRNA; NOS, nitric oxide synthase; PKA, protein kinase A; ROS, reactive oxygen species; Samd3, sterile α motif domain containing 3; TDO2, tryptophan 2,3-dioxygenase; TNF-α, tumor necrosis factor α; VEGF, vascular endothelial growth factor; XRE, xenobiotic response elements; HAHs, halogenated aromatic hydrocarbons; PAHs, polycyclic aromatic hydrocarbons.

AhR and its ligands promote mammary carcinogenesis

Although AhR ligands of different structures exhibit anticancer effects, the expression and function of AhR in breast cancer cells are variable (Fig. 2). Several studies have reported that exogenous AhR ligands, such as PAHs and HAHs, act as AhR agonists to exert cancer-promoting effects in breast cancer (Table V). These ligands, which exhibit AhR agonistic activity, maintain or even promote malignant transformation phenotypes in breast cancer cells and xenograft models by activating the AhR pathway (48,62,102,117,147164). The ligands exert numerous effects, such as enhancing the migratory and invasive capacity of breast cancer cells, inhibiting apoptosis, stimulating CSC generation, promoting angiogenesis and inducing inflammatory responses.

Table V.

Pro-breast tumorigenic role of exogenous AhR agonists.

Table V.

Pro-breast tumorigenic role of exogenous AhR agonists.

First author/s, yearLigandStudy typeCell line/animal modelResponseTargeted signaling pathway(Refs.)
NarasimhanTCDDIn vitroSUM149, Hs578T,↑Migration, ↓irregularAhR signaling(48)
et al, 2018 BP1colony growthpathway
Vacher et al, 2018TCDDIn vitroMDA-MB-436↑IL-1B, IL-6-(62)
Piwarski et al,TCDDIn vitroMCF-7,↓AhR, ERα-(102)
2020 MDA-MB-231↓JAG1, AhR-
Vogel et al, 2021TCDDIn vivoE0771 in C57BL/6↑Tumor growth-(147)
Shan et al, 2020TCDDIn vitroMCF-7↑Migration, ↑invasion-(149)
In vivoMCF-7 in BALB/c↑AhR, CYP1A1-
nude mice
Bekki et al, 2015TCDDIn vitroP20C, P20E, MDA-↓Apoptosis (UV-treated),-(150)
MB-231, P35E↓Apoptosis (Dox-treated),
↓Apoptosis (Lap-treated)
P20C, MDA-MB-↓Apoptosis (Pac-treated)-
231, P35C, SKBR3
P20C, P20E↑COX-2 (Dox-treated),C/EBPβ alternative
↑RelB (Dox-treated),AhR pathway
↑IDO1, IDO2
Vogel et al, 2011TCDDIn vitroMCF-7, MDA-MB-↑IL-8-(152)
436
NarasimhanTCDDIn vitroSUM149↑Migration-(48,
et al, 2018; 153)
Novikov et al, 2016
Miret et al, 2016TCDDIn vitroMDA-MB-231↑TGF-β1-(155)
Al-Dhfyan et al,TCDDIn vitroMCF-7↑Mammosphere formation,Wnt/β-catenin(157)
2017 ↑ALDH, ↑side populationsignaling pathway,
cells, ↑CYP1A1, β-catenin,PTEN-PI3K/AKT
cyclin D1, ↑β-cateninsignaling pathway
cellular content, and nuclear
translocation, ↓PTEN,
↑AKT, p-AKT
Hs578T, T47D↑ALDH-
Jung et al, 2011TCDDIn vitroMCF-7↑Mammosphere formation,-(158)
↑OCT4
Pontillo et al,HCBIn vitroHMEC-1↑VEGF, AhR, COX-2,ERK/VEGFR2(151)
2015 p-ERK1-2/ERK1-2,signaling pathway
p-p38/p38, ↑migration,
↑neovasculogenesis
In vivoMDA-MB-231↑Angiogenesis (VEGF)-
xenograft model in
female nude mice
Miret et al, 2016HCBIn vitroMDA-MB-231↓AhR, ↑TGF-β1,Modulation of the(155)
↑migration, ↑invasioncrosstalk between
AhR and TGF-β1
signaling
Pontillo et al,HCBIn vitroMDA-MB-231↑Migration, ↑invasion, c-Src/HER1/STAT5b(156)
2013 ↑MMP2, MMP9,and ERK1/2
signaling pathways
In vivoMDA-MB-231 in↑Tumor growth-
BALB/c nude mice
C4-HI in BALB/c↑Tumor growth, ↑liver or-
nude micelung metastasis
LM3 in BALB/c↑Tumor growth-
nude mice
Zárate et al,HCBIn vivoMCF-7 in nude↑VEGF-A, ↑number ofAhR and ER(160)
2020 Swiss micevesselssignaling pathways
In vitroMCF-7↑VEGF-A, ↑COX-2-
EA. hy926 (media↑Neovasculogenesis, ↑total-
from MCF-7tube length, ↑number of
treated with HCB)branch points
Pontillo et al,HCBIn vitroMDA-MB-231↑Migration c-Src/HER1/STAT5b(161)
2011 and HER1/ERK1/2
signaling pathways
Hsieh et al, 2012PhthalatesIn vitroMDA-MB-231↑Proliferation, ↑migration,AhR/HDAC6/(154)
(BBP/DBP) ↑invasionc-Myc signaling
pathway
In vivoMDA-MB-231 in↑Tumor growth, ↑distant-
BALB/c nude micemetastasis
Shan et al, 2020MEHPIn vitroMCF-7↑Migration; ↑invasion,AhR-MMP2(149)
↑AhR, CYP1A1, ↓CYP1A1pathway
(TCDD-treated), ↓migration
(TCDD-treated), ↓invasion
(TCDD-treated)
In vivoMCF-7 in BALB/c↓AhR-
nude mice
Vacher et al, 2018B[a]PIn vitroMDA-MB-436↑IL-1B, IL-6-(62)
Novikov et al, 2016B[a]PIn vitroSUM149↑Migration-(153)
Castillo-SanchezB[a]PIn vitroMDA-MB-231,↑Migration, ↑αvβ3Activation of FAK,(162)
et al, 2013 MCF-7integrin-cell surface levels,Src and extracellular
↑MMP-2, MMP-9 signal-regulated
kinase 2
Guo et al, 2015B[a]PIn vitroMCF-7↑Migration, ↑invasion,Upregulation of(163)
↑MMP9ROS-induced ERK
signaling pathway
In vivoMCF-7 in BALB/c↑Tumor growth, ↑liver and-
nude micelung metastasis
Malik et al, 2019B[a]P, PhIPIn vitroMCF-7, MDA-↑MN formationIL-6-miR-27b-(159)
MB-231(IL-6-treated), ↓miR-27bCYP1B1 signaling
pathway
Kolasa et al,BZA, B[a]P,In vitroMCF-7↑IL-6 (TNF-α-treated)NF-κB signaling(148)
2013TCDD, pathway
3MC
Zhao et al, 20123MCIn vitroMCF-7 ↓Proliferation,Wnt/β-catenin and(117)
↓mammosphere formation,Notch signaling
↓the proportion of cells withpathway
high ALDH activity
Cirillo et al,3MCIn vitroSkBr3↑Proliferation, ↑CYP1B1,Crosstalk between(164)
2019 cyclin D1AhR and GPER
Al-Dhfyan et al,DMBAIn vitroMCF-7↑Mammosphere formation,Wnt/β-catenin(157)
2017 ↑ALDH, ↑side populationsignaling pathway,
cells, ↑CYP1A1, β-catenin,PTEN-PI3K/AKT
cyclin D1, ↓PTEN, ↑AKT,signaling pathway
p-AKT
T47D↑ALDH-
In vivoBALB/c nude mice↑CYP1A1, ↑ALDH,-
↑β-catenin, ↓PTEN, ↑AKT,
p-AKT
Zárate et al,CPFIn vivoMCF-7 in nude↑VEGF-A, ↑number of-(160)
2020 Swiss micevessels
In vitroMCF-7↑VEGF-A, ↑COX-2-
EA. hy926 (media ↑Neovasculogenesis,-
from MCF-7 treated↑total tube length, ↑number
with HCB)of branch points

[i] ↑, increase; ↓, decrease; ↔, no change. 3MC, 3-methylchoanthrene; AhR, aryl hydrocarbon receptor; ALDH, aldehyde dehydrogenase-1; B[a]P, benzo[a]pyrene; BBP, butyl benzyl phthalate; BZA, benzanthracene; C/EBPβ, CCAAT/enhancer-binding protein β; COX-2, cyclooxygenase 2; CPF, chlorpyrifos; DBP, di-(n-butyl) phthalate; CYP1A1, cytochrome P450 family 1 subfamily A member 1; CYP1B1, cytochrome P450 family 1 subfamily B member 1; DMBA, 7,12-dimethybenz(a)anthracene; Dox, doxorubicin; ERα, estrogen receptor α; FAK, focal adhesion kinase; HCB, hexachlorobenzene; HDAC6, histone deacetylase 6; HER1, human epidermal receptor; GPER, G protein-coupled estrogen receptor 1; IDO, indoleamine 2, 3-dioxygenase; JAG1, jagged canonical Notch ligand 1; Lap, lapatinib; MEHP, mono 2-ethylhexyl phthalate; miR, microRNA; p-, phosphorylated; MN, micronuclei; Pac, paclitaxel; PhIP, 2-amino-1-methyl-6-phenylimidazo [4, 5-b] pyridine; P20C, 1 nM E2 selection process for MCF-10AT1 cells, mock selected control breast cancer cells; P20E, 1 nM E2 selection process for MCF-10AT1 cells, AhR-overexpressing breast cancer cells; P35C, 1 nM E2 selection process for MCF-7 cells, mock selected control breast cancer cells; P35E, 1 nM E2 selection process for MCF-7 cells, AhR-overexpressing breast cancer cells; ROS, reactive oxygen species; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; VEGF, vascular endothelial growth factor.

AhR ligands increase the motility of breast cancer cells and promote metastasis

Numerous environmental toxicants, such as TCDD, butyl benzyl phthalate (BBP), di-n-butyl phthalate (DBP), hexachlorobenzene (HCB) and benzo[a]pyrene (B[a]P) enhance cell motility by activating AhR, which promotes cell migration and organ invasion (48,94,149,151,153156,161163). Most of these environmental toxicants serve as AhR agonists in different breast cancer cell lines, including ER-positive (T47D, MCF-7 and ZR-75), ER-negative (MDA-MB-231, MDA-MB-436, MCF-10A, SUM149 and Hs578T) and HER-2-positive (SKBR3) cells. However, the use of AhR antagonists, AhR silencing or AhR knockout reversed this effect (48,153,165167). Among them, the effects of AhR antagonists used in a number of studies are listed in Table VI. These synthetic antagonists suppress the effect of AhR activation on breast cancer cells (48,124,149,150,152,153,157,165167). In a breast cancer cell xenograft zebrafish model, AhR knockdown or AhR antagonist (CH223191) impaired cell invasion and migration, and suppressed metastasis of TNBC and IBC cells by decreasing the expression of invasion-associated genes and increasing the expression of E-cadherin (48). However, AhR agonists may also exhibit anti-tumorigenic effects, for example, TCDD inhibited the formation of irregular colonies of TNBC cells, including SUM149, Hs578T and BP1 cells, as presented in Table V (48). In another study, MCF-7 cells were co-treated with TCDD and mono 2-ethylhexyl phosphate (MEHP). MEHP was reported to be a potential AhR agonist, and MEHP and TCDD alone both induced cell migration and invasion. The promotion was partially dependent on AhR, and this effect mediated by MEHP may be related to the AhR-MMP2 pathway (153). Another study also reported that following the co-treatment with MEPH and TCDD, MEHP antagonized TCDD to reduce AhR-mediated CYP1A1 expression and inhibit the migration and invasion of in MCF-7 cells (Table V) (149).

Table VI.

Anti-breast cancer role of synthetic AhR ligands.

Table VI.

Anti-breast cancer role of synthetic AhR ligands.

First author/s, yearLigandStudy typeCell line/animal modelResponseTargeted signaling pathway(Refs.)
Narasimhan et al, 2018CB7993113In vitroHs578T, SUM149↓Migration-(48)
In vivoRFP-MDA-MB-231 xenograft↓Metastasis-
model in zebrafish
Yamashita et al, 2018CB7993113In vitroBP1↓Invasion-(165)
In vivoC57BL/6J↓DMBA-induced toxicity-
NarasimhanCH223191In vitroBP1↓Irregular colony growth-(48)
et al, 2018 In vivoRFP-MDA-MB-231 xenograft↓Metastasis-
model in zebrafish
Shan et al, 2020CH223191In vitroSUM149↓Migration (Kyn-treated),TDO2-AhR(149)
↓migration (XA-treated)signaling pathway
Cirillo et al, 2019CH223191In vitroHER2-5↓Migration (HRG-treated)-(164)
D'Amato et al, 2015CH223191In vitroBT549↓Proliferation, ↓migration-(166)
BT549 (forced↓CYP1A1, CYP1B1,-
suspension culture) ↓anchorage-independent
growth, ↓resistance to anoikis
SUM159 (forced ↓Anchorage-independent-
suspension culture)growth, ↓resistance to anoikis
MDA-MB-231↓Proliferation, ↓migration-
Narasimhan et al, 2018;CH223191In vitroSUM149↓Migration-(48,153,
Novikov et al, 2016; 165)
Yamashita et al, 2018
Narasimhan et al, 2018;CH223191In vitroHs578T↓Migration-(48,
Yamashita et al, 2018 165)
Prud'homme et al, 2010α-NFIn vitroMDA-MB-231↑Proliferation (tranilast-treated)-(124)
Pontillo et al, 2015α-NFIn vitroHMEC-1↓p-ERK1-2/ERK1-2 (HCB-treated),ERK/VEGFR2 signaling(151)
p-p38/p38 (HCB-treated)pathway
Al-Dhfyan et al, 2017α-NFIn vitroMCF-7↓Side population cells, ↑apoptosis-(157)
Al-Dhfyan et al, 2017α-NFIn vitroMCF-7↓ALDH (TCDD-treated; DMBA-treated),-(157)
↓side population cells (TCDD-treated;
DMBA-treated), ↑apoptosis
(Dox-treated), ↑apoptosis inside
population cells (Dox-treated)
D'Amato et al, 2015α-NFIn vitroBT549, BT549 (suspended-culture),↓AhR transcriptional activity-(166)
MDA-MB-231 (suspended-culture)
Bekki et al, 2015MNFIn vitroP20C, P20E, MDA-MB-231↑Apoptosis (UV and TCDD-treated),-(150)
↑apoptosis (Dox and TCDD-treated),
↑apoptosis (Lap and TCDD-treated)
P35E, SKBR3↑Apoptosis (Pac and TCDD-treated)-
P20C, P20E↓COX-2 (Dox and TCDD-treated),C/EBPβ alternative AhR
↓RelB (Dox and TCDD-treated)pathway
Vogel et al, 2011MNFIn vitroP20C, P20E↑Apoptosis (UV-treated)-(152)

[i] ↑, increase; ↓, decrease; ↔, no change. α-NF, α-naphthoflavone; AhR, aryl hydrocarbon receptor; ALDH, aldehyde dehydrogenase; C/EBPβ, CCAAT/enhancer-binding protein β; COX-2, cyclooxygenase 2; CYP1A1, cytochrome P450 family 1 subfamily A member 1; CYP1B1, cytochrome P450 family 1 subfamily B member 1; DMBA, 7,12-dimethybenz(a)anthracene; Dox, doxorubicin; HRG, heregulin; Kyn, kynurenine; Lap, lapatinib; MNF, 3′methoxy-4′nitroflavone; Pac, paclitaxel; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; TDO, tryptophan 2,3-dioxygenase; XA, xanthurenic acid.

In another study on environmental toxicant phthalates (AhR agonists), Hsieh et al (154) reported that phthalates induced the proliferation and invasiveness of ER-negative (MDA-MB-231) cells via the AhR/histone deacetylase 6/c-Myc signaling pathway, and AhR was activated via a non-genomic pathway. An increase in breast cancer cell migration and invasion as a result of AhR activation may promote these features through an alternative pathway (independent of AhR). For example, the organochlorine pesticide HCB, an AhR ligand, may activate AhR and promote breast cancer cell migration and invasion via the c-Src/HER1/STAT5b and HER1/ERK1/2 signaling pathways (151,156,161). However, the crosstalk between AhR and TGF-β1 signaling may also promote ER-negative breast cancer cell migration and invasion (155). Additionally, studies have reported that B[a]P induces the metastasis of ER-positive (MCF-7) cells via lipoxygenase- and Src-dependent pathways and the reactive oxygen species/ERK/MMP9 signaling pathway (162,163). Increased tumor growth and more distant metastasis were also observed in a xenograft nude mouse model after AhR activation with different exogenous ligands (BBP, DBP, HCB and B[a]P) (154,156,163). Narasimhan et al (48) injected MDA-MB-231 cells into zebrafish and treated them with AhR antagonists (CB7993133 or CH223191), reporting that AhR inhibitors blocked metastasis. Furthermore, Novikov et al (153) reported that endogenous ligands [kynurenine (Kyn) and xanthurenic acid] activated AhR to promote the migration of ER-negative (SUM149) cells. These two tryptophan-derived ligands are generated via the Kyn pathway (153). In another study, AhR knockdown or AhR antagonist (CH223191) treatment reduced the proliferation and migration of ER-negative (MDA-MB-231 and BT549) cells (166).

AhR ligands promote CSC emergence and proliferation

CSCs drive tumorigenesis, progression and metastasis (141,168,169). A study reported that TCDD treatment stimulated the expression of OCT4 and promoted the self-renewal of breast CSCs in ER-positive (MCF-7) breast cancer cells (158), suggesting that AhR ligands may promote tumorigenesis by promoting the proliferation of CSCs. This is supported by another study in which TCDD and 7,12-dimethylbenz(a)anthracene (DMBA) activated the AhR/CYP1A1 signaling pathway through inhibition of PTEN and activation of β-catenin and AKT pathways to enhance the proliferation, development, self-renewal and chemoresistance of breast CSCs (Table V) (157). Similarly, suppression of PTEN expression is observed in the mammary tissue of the DMBA-treated mouse model, accompanied by increased phosphorylated-AKT, β-catenin and ALDH expression. Inhibition of the AhR/CYP1A1 pathway by an AhR antagonist, α-NF, blocks the increase of ALDH activity and blocks the increase to the proportion of side population cells that is mediated by TCDD and DMBA. Furthermore, α-NF treatment alone reduces the percentage of side population cells (157). This side population cell sorting is considered to be a valuable technology for CSCs identification and sorting, and breast CSCs can be identified and isolated by a side population phenotype (157). However, Zhao et al (117) reported that the AhR agonists β-NF and 3MC activated AhR, suppressed mammosphere formation and decreased the proportion of cells with high ALDH activity in MCF-7 cells (Tables IV and V, respectively). The study also reported that AhR activation regulated self-renewal signals by downregulating Wnt/β-catenin and Notch.

AhR ligands inhibit apoptosis in breast cancer cells

Several studies have reported that exogenous ligands that activate AhR, such as TCDD, suppress apoptosis induced by stimuli, including chemotherapeutic drugs in ER-positive (MCF-7 and T47D), ER-negative (MDA-MB-231, Hs578T and MCF-10A) and HER-2 positive (SKBR3) cells (65,150,157,170). When the AhR pathway was blocked using AhR silencing (RNA interference), AhR knockout cell lines or AhR antagonists (CH223191 or α-NF or 3′ methoxy-4′nitroflavone), an increase in cell death was observed (150,157). Treatment with an endogenous AhR ligand (Kyn) inhibited anoikis (a type of epithelial cell programmed death) in ER-negative (BT549 and SUM159) cells in forced suspension culture (Table III), whereas AhR knockdown or AhR inhibitor (CH223191) treatment promoted anoikis (Table VI) (166). Similarly, Kyn inhibited apoptosis in ER-negative breast cancer cells (150). Goode et al (65) reported that AhR knockout in athymic nude mouse xenograft models reduced tumor growth by increasing apoptosis. However, the exact biological mechanism linking the activation of AhR and the reduction of apoptosis remains unclear. Bekki et al (150) proposed that TCDD induces the expression of inflammatory genes, such as the genes encoding cyclooxygenase 2 (COX-2) and NF-κB subunit RelB, to prevent apoptosis. Another possible mechanism, proposed by Anderson et al (170), is that exposure of TNBC cells to chemotherapeutic agents, such as paclitaxel, induces the expression of breast tumor kinase via the AhR/GR/hypoxia-inducible factor signaling axis, which is involved in the inhibition of apoptosis.

AhR ligands promote angiogenesis in breast cancer models

The environmental pollutants TCDD, HCB and chlorpyrifos (CPF) promote angiogenesis in breast cancer models by activating AhR (150,151,160). TCDD induces expression of the inflammatory marker COX-2 in mammalian cells through an alternative AhR pathway involving CCAAT/enhancer binding protein β (150). This promotes angiogenesis by upregulating vascular endothelial growth factor (VEGF) (171,172). Pontillo et al (151) reported that HCB stimulated angiogenesis and increased VEGF expression in a breast cancer xenograft mouse model. HCB also induced neovasculogenesis of the HMEC-1 human microvascular endothelial cell line in vitro, enhanced the expression of VEGF-receptor 2 and activated the downstream pathways p38 and ERK1/2 (Table V), whereas an AhR inhibitor (α-NF) suppressed these effects (Table VI) (151). Another study reported that VEGF-A expression, induced by HCB and CPF, was mediated by ER and nitric oxide (NO), whilst the increase of COX-2 was mediated via AhR and the NO pathway in MCF-7 cells. In vivo, HCB and CPF stimulated the angiogenic switch (160).

AhR ligands induce an increase in the inflammatory response

A number of studies have reported that activation of AhR leads to increased expression of numerous inflammatory markers, including COX-2, IL-6 and IL-8, in numerous tumors and cancer types (46,62,150,152,160), including breast cancer (46,173). Furthermore, a more aggressive, chemoresistant breast cancer phenotype in ER-positive cells can reside in the inflammatory microenvironment (174,175). Epidemiological evidence indicates that IBC has a poor prognosis and that patients with IBC have a shortened survival compared with patients with non-IBC (176,177). The NF-κB pathway is a key pathway linking AhR activation to cellular inflammation (28,148,152,178). NF-κB is a hub molecular in a number of inflammatory pathways. Kim et al (179) reported that the activity of NF-κB (p65/p50) increased during neoplastic transformation in murine normal mammary cells treated with DMBA and in human non-transformed mammary cells treated with DMBA or B[a]P. When non-transformed breast cells MCF-10F (ER-negative, PR-negative and HER-2 negative) were treated with DMBA or B[a]P, the AhR interacted with NF-κB subunit RelA to activate transcription of the proto-oncogene and expression of the protein c-Myc, a master regulator of cell proliferation and neoplastic transformation (180). Furthermore, in DMBA-induced murine mammary tumors, high expression of AhR, c-Myc and cyclin D1 was associated with NF-κB and Wnt signaling pathways (181). Another inflammatory marker, IL-6, which is regulated by NF-κB, is involved in the immune function, hematopoiesis, acute phase response and inflammatory response (182). In mammary tissue, IL-6, along with its downstream transcription factor, STAT3, has been reported to stimulate cell proliferation and migration during ontogeny, and be involved in gland remodeling during aging (173,183). A previous study reported that IL-6 enhances B[a]P and 2-amino-1-methyl-6-phenylimidazo [4,5-b] pyridine-induced micronuclei (MN) formation in breast cancer cells via the miR-27b-CYP1B1 signaling pathway, which leads to DNA damage (159).

Discussion

AhR is a receptor with complex functions, as its activation is ligand- and cell-dependent. AhR is not only bound by multiple ligands, but it can also interact and function with numerous molecules. A number of AhR agonists also target other molecules; therefore, ligand-induced activation of AhR leads to the altered expression of hundreds of genes. Additionally, breast cancer cells of the same molecular subtype have different regulatory effects and mechanisms associated with the same AhR ligand (Fig. 3). Moreover, breast cancer has different molecular subtypes, which makes the studies more complicated. Most AhR ligands demonstrate tumor-suppressive characteristics; however, under specific circumstances, different types of ligands also show different regulation patterns. For example, the exogenous AhR ligand TCDD is involved in the promotion of mammosphere formation, and inhibition of migration/invasion and cell cycle progression in ER-positive (MCF-7) breast cancer cells, whilst it inhibits proliferation and invasion in HER-2-positive (BT474) breast cancer cells (Tables II and V). Additionally, TCDD exhibits dual regulatory roles in proliferation and migration/invasion in TNBC cells (Fig. 3A). Numerous endogenous, synthetic and medicinal AhR ligands also exhibit tumor-suppressive characteristics (Fig. 3B). All three categories of AhR ligands exhibit mainly inhibitory roles in TNBC cell proliferation and promote apoptosis in the three types of breast cancer, while their regulatory roles in cell migration/invasion are inconsistent. AhR mediates either pro-cancer or anticancer activities in breast cancer cells (Fig. 4); however, the underlying mechanisms are still unclear. AhR regulation in cancer appears to be dependent on the types and levels of AhR ligands. However, quantifying each of these ligands in patients with different types of cancer, including breast cancer, may be challenging as patients may be exposed to a number of AhR ligands over a prolonged period (66). For example, the half-life of the environmental carcinogen TCDD in humans is 7–10 years (184). Thus, directly targeting AhR rather than AhR ligands may be a more feasible option for cancer treatment. However, a number of AhR agonists are too toxic at high levels to be used clinically or have not been tested in humans, necessitating the development and exploration of non-toxic, clinically applicable alternatives, such as omeprazole and tranilast.

Figure 3.

Effects of AhR ligands on different types of breast cancer cells. (A) TCDD, as a typical exogenous AhR ligand, affects the proliferation, apoptosis and migration/invasion via different pathways in different types of breast cancer cells. It promotes proliferation and inhibits migration/invasion in ER-positive cells, inhibits proliferation, apoptosis and migration/invasion in HER-2-positive cells, and inhibits apoptosis and promotes or inhibits proliferation and migration/invasion in TNBC cells. (B) Other PAHs and HAHs, endogenous and synthetic and pharmaceutical AhR ligands mainly exhibit tumor-suppressive characteristics. However, PAHs and HAHs not only promote the proliferation of ER-positive and HER-2-positive cells, but also promote the migration/invasion of TNBC and ER-positive cells. Endogenous ligands not only inhibit the proliferation of TNBC and ER-positive cells, but also inhibit the migration/invasion of TNBC and HER-2-positive cells. Moreover, synthetic and pharmaceutical ligands not only inhibit the proliferation and promote apoptosis of the different types of breast cancer cells, but also inhibit the migration/invasion of TNBC cells. (a) Effects of PAHs and HAHs on different types of breast cancer cells. (b) Effects of endogenous AhR ligands on different types of breast cancer cells. (c) Effects of synthetic and pharmaceutical AhR ligands on different types of breast cancer cells. AF, aminoflavone; AhR, aryl hydrocarbon receptor; ANI-7, (Z)-2-(3,4-dichlorophenyl)-3-(1H-pyrrol-2-yl) acrylonitrile; B[a]P, benzo[a]pyrene; DIM, 3,3-diindolylmethane; ER, estrogen receptor; FICZ, 6-formylindolo(3,2-b) carbazole; HAH, halogenated aromatic hydrocarbon; HCB, hexachlorobenzene; HER-2, human epidermal growth factor receptor 2; ICZ, indole (3,2-b) carbazole; ITE, 2-(1′h-indole-3′-carbonyl)-thiazole-4-carboxylic acid; IS, indoxyl-sulfate; IPA, indole propionic acid; I3C, indole-3-carbinol; kyn, kynurenine; NK150460, 5,6,7,8-tetrahydrocarcinolin-5-ol; PAH, polycyclic aromatic hydrocarbon; PCB, 3,3′,4,4′,5-pentachlorobiphenyl; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin; ER, estrogen receptor; TNBC, triple-negative breast cancer; α-NF, α-naphthoflavone; β-NF, β-naphthoflavone.

Conclusion

Current research has demonstrated that AhR and its ligands serve important roles in breast cancer progression and metastasis, possibly via the regulation of apoptosis, migration, invasion, inflammation and angiogenesis. Several synthetic AhR ligands and widely used drugs with affinity to AhR exhibit selectivity for breast cancer cells with different molecular types, and regulate breast cancer cell functions. This suggests their potential as novel strategies for breast cancer therapy.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Characteristic Innovation Projects of Universities in Guangdong Province (grant no. 2021KTSCX037) and Zhanjiang Science and Technology Project (grant nos. 2020A06004, 2021A05056 and 2022A01190).

Availability of data and materials

Not applicable.

Authors' contributions

CC, WL and XS designed the study. CC completed the first draft of this manuscript. CC, YZ, ZL and ZW collected and analyzed the data and revised the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Miller KD, Nogueira L, Devasia T, Mariotto AB, Yabroff KR, Jemal A, Kramer J and Siegel RL: Cancer treatment and survivorship statistics, 2022. CA Cancer J Clin. 72:409–436. 2022. View Article : Google Scholar : PubMed/NCBI

4 

Verrill M: Metastatic disease of the breast and local recurrence. Surgery (Oxford). 37:181–185. 2019. View Article : Google Scholar

5 

Flatley MJ and Dodwell DJ: Adjuvant treatment for breast cancer. Surgery (Oxford). 34:43–46. 2016. View Article : Google Scholar

6 

Burstein HJ, Curigliano G, Thürlimann B, Weber WP, Poortmans P, Regan MM, Senn HJ, Winer EP and Gnant M; Panelists of the St Gallen Consensus Conference, : Customizing local and systemic therapies for women with early breast cancer: The St. Gallen international consensus guidelines for treatment of early breast cancer 2021. Ann Oncol. 32:1216–1235. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Harbeck N and Gnant M: Breast cancer. Lancet. 389:1134–1150. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Pondé NF, Zardavas D and Piccart M: Progress in adjuvant systemic therapy for breast cancer. Nat Rev Clin Oncol. 16:27–44. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Poland A, Glover E and Kende AS: Stereospecific, high affinity binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin by hepatic cytosol. Evidence that the binding species is receptor for induction of aryl hydrocarbon hydroxylase. J Biol Chem. 251:4936–4946. 1976. View Article : Google Scholar : PubMed/NCBI

10 

Kung T, Murphy KA and White LA: The aryl hydrocarbon receptor (AhR) pathway as a regulatory pathway for cell adhesion and matrix metabolism. Biochem Pharmacol. 77:536–546. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Murray IA, Patterson AD and Perdew GH: Aryl hydrocarbon receptor ligands in cancer: Friend and foe. Nat Rev Cancer. 14:801–814. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Bersten DC, Sullivan AE, Peet DJ and Whitelaw ML: bHLH-PAS proteins in cancer. Nat Rev Cancer. 13:827–841. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, Schumacher T, Jestaedt L, Schrenk D, Weller M, et al: An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 478:197–203. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Denison MS and Nagy SR: Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol. 43:309–334. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Baker JR, Sakoff JA and McCluskey A: The aryl hydrocarbon receptor (AhR) as a breast cancer drug target. Med Res Rev. 40:972–1001. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Rothhammer V and Quintana FJ: The aryl hydrocarbon receptor: An environmental sensor integrating immune responses in health and disease. Nat Rev Immunol. 19:184–197. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Stockinger B, Di Meglio P, Gialitakis M and Duarte JH: The aryl hydrocarbon receptor: Multitasking in the immune system. Annu Rev Immunol. 32:403–432. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Denison MS, Soshilov AA, He G, DeGroot DE and Zhao B: Exactly the same but different: Promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol Sci. 124:1–22. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Mimura J, Ema M, Sogawa K and Fujii-Kuriyama Y: Identification of a novel mechanism of regulation of Ah (dioxin) receptor function. Genes Dev. 13:20–25. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Whitlock JP Jr: Induction of cytochrome P4501A1. Annu Rev Pharmacol Toxicol. 39:103–125. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Brauze D, Widerak M, Cwykiel J, Szyfter K and Baer-Dubowska W: The effect of aryl hydrocarbon receptor ligands on the expression of AhR, AhRR, ARNT, Hif1alpha, CYP1A1 and NQO1 genes in rat liver. Toxicol Lett. 167:212–220. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Hu W, Sorrentino C, Denison MS, Kolaja K and Fielden MR: Induction of cyp1a1 is a nonspecific biomarker of aryl hydrocarbon receptor activation: Results of large scale screening of pharmaceuticals and toxicants in vivo and in vitro. Mol Pharmacol. 71:1475–1486. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Gao X, Xie C, Wang Y, Luo Y, Yagai T, Sun D, Qin X, Krausz KW and Gonzalez FJ: The antiandrogen flutamide is a novel aryl hydrocarbon receptor ligand that disrupts bile acid homeostasis in mice through induction of Abcc4. Biochem Pharmacol. 119:93–104. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Swedenborg E and Pongratz I: AhR and ARNT modulate ER signaling. Toxicology. 268:132–138. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Tarnow P, Tralau T and Luch A: Chemical activation of estrogen and aryl hydrocarbon receptor signaling pathways and their interaction in toxicology and metabolism. Expert Opin Drug Metab Toxicol. 15:219–229. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Göttel M, Le Corre L, Dumont C, Schrenk D and Chagnon MC: Estrogen receptor α and aryl hydrocarbon receptor cross-talk in a transfected hepatoma cell line (HepG2) exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Rep. 1:1029–1036. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Kang HJ, Kim HJ, Kim SK, Barouki R, Cho CH, Khanna KK, Rosen EM and Bae I: BRCA1 modulates xenobiotic stress-inducible gene expression by interacting with ARNT in human breast cancer cells. J Biol Chem. 281:14654–14662. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Tian Y, Rabson AB and Gallo MA: Ah receptor and NF-kappaB interactions: Mechanisms and physiological implications. Chem Biol Interact. 141:97–115. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Marshall NB and Kerkvliet NI: Dioxin and immune regulation: Emerging role of aryl hydrocarbon receptor in the generation of regulatory T cells. Ann N Y Acad Sci. 1183:25–37. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Gutiérrez-Vázquez C and Quintana FJ: Regulation of the immune response by the Aryl hydrocarbon receptor. Immunity. 48:19–33. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Esser C and Rannug A: The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol Rev. 67:259–279. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Lee JH, Wada T, Febbraio M, He J, Matsubara T, Lee MJ, Gonzalez FJ and Xie W: A novel role for the dioxin receptor in fatty acid metabolism and hepatic steatosis. Gastroenterology. 139:653–663. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Lahvis GP, Lindell SL, Thomas RS, McCuskey RS, Murphy C, Glover E, Bentz M, Southard J and Bradfield CA: Portosystemic shunting and persistent fetal vascular structures in aryl hydrocarbon receptor-deficient mice. Proc Natl Acad Sci USA. 97:10442–10447. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Huang S, Shui X, He Y, Xue Y, Li J, Li G, Lei W and Chen C: AhR expression and polymorphisms are associated with risk of coronary arterial disease in Chinese population. Sci Rep. 5:80222015. View Article : Google Scholar : PubMed/NCBI

35 

Neavin DR, Liu D, Ray B and Weinshilboum RM: The role of the Aryl hydrocarbon receptor (AHR) in immune and inflammatory diseases. Int J Mol Sci. 19:38512018. View Article : Google Scholar : PubMed/NCBI

36 

Stockinger B, Shah K and Wincent E: AHR in the intestinal microenvironment: Safeguarding barrier function. Nat Rev Gastroenterol Hepatol. 18:559–570. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Bock KW: Aryl hydrocarbon or dioxin receptor: Biologic and toxic responses. Rev Physiol Biochem Pharmacol. 125:1–42. 1994.PubMed/NCBI

38 

Bradshaw TD and Bell DR: Relevance of the aryl hydrocarbon receptor (AhR) for clinical toxicology. Clin Toxicol (Phila). 47:632–642. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Knerr S and Schrenk D: Carcinogenicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in experimental models. Mol Nutr Food Res. 50:897–907. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Roman ÁC, Carvajal-Gonzalez JM, Merino JM, Mulero-Navarro S and Fernández-Salguero PM: The aryl hydrocarbon receptor in the crossroad of signalling networks with therapeutic value. Pharmacol Ther. 185:50–63. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Stanford EA, Wang Z, Novikov O, Mulas F, Landesman-Bollag E, Monti S, Smith BW, Seldin DC, Murphy GJ and Sherr DH: The role of the aryl hydrocarbon receptor in the development of cells with the molecular and functional characteristics of cancer stem-like cells. BMC Biol. 14:202016. View Article : Google Scholar : PubMed/NCBI

42 

Stanford EA, Ramirez-Cardenas A, Wang Z, Novikov O, Alamoud K, Koutrakis P, Mizgerd JP, Genco CA, Kukuruzinska M, Monti S, et al: Role for the Aryl hydrocarbon receptor and diverse ligands in oral squamous cell carcinoma migration and tumorigenesis. Mol Cancer Res. 14:696–706. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Liu Z, Wu X, Zhang F, Han L, Bao G, He X and Xu Z: AhR expression is increased in hepatocellular carcinoma. J Mol Histol. 44:455–461. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Chang JT, Chang H, Chen PH, Lin SL and Lin P: Requirement of aryl hydrocarbon receptor overexpression for CYP1B1 up-regulation and cell growth in human lung adenocarcinomas. Clin Cancer Res. 13:38–45. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Koliopanos A, Kleeff J, Xiao Y, Safe S, Zimmermann A, Büchler MW and Friess H: Increased arylhydrocarbon receptor expression offers a potential therapeutic target for pancreatic cancer. Oncogene. 21:6059–6070. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Guarnieri T: Aryl hydrocarbon receptor connects inflammation to breast cancer. Int J Mol Sci. 21:52642020. View Article : Google Scholar : PubMed/NCBI

47 

Benoit L, Jornod F, Zgheib E, Tomkiewicz C, Koual M, Coustillet T, Barouki R, Audouze K, Vinken M and Coumoul X: Adverse outcome pathway from activation of the AhR to breast cancer-related death. Environ Int. 165:1073232022. View Article : Google Scholar : PubMed/NCBI

48 

Narasimhan S, Stanford Zulick E, Novikov O, Parks AJ, Schlezinger JJ, Wang Z, Laroche F, Feng H, Mulas F, Monti S and Sherr DH: Towards resolving the pro- and anti-tumor effects of the Aryl hydrocarbon receptor. Int J Mol Sci. 19:13882018. View Article : Google Scholar : PubMed/NCBI

49 

Waks AG and Winer EP: Breast cancer treatment: A review. JAMA. 321:288–300. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Loibl S, Poortmans P, Morrow M, Denkert C and Curigliano G: Breast cancer. Lancet. 397:1750–1769. 2021. View Article : Google Scholar : PubMed/NCBI

51 

Yu F, Quan F, Xu J, Zhang Y, Xie Y, Zhang J, Lan Y, Yuan H, Zhang H, Cheng S, et al: Breast cancer prognosis signature: Linking risk stratification to disease subtypes. Brief Bioinform. 20:2130–2140. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Eroles P, Bosch A, Pérez-Fidalgo JA and Lluch A: Molecular biology in breast cancer: Intrinsic subtypes and signaling pathways. Cancer Treat Rev. 38:698–707. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Loibl S and Gianni L: HER2-positive breast cancer. Lancet. 389:2415–2429. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Patel HK and Bihani T: Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment. Pharmacol Ther. 186:1–24. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Goodwin PJ: Extended aromatase inhibitors in hormone-receptor-positive breast cancer. N Engl J Med. 385:462–463. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Goel S, Bergholz JS and Zhao JJ: Targeting CDK4 and CDK6 in cancer. Nat Rev Cancer. 22:356–372. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Hushka LJ, Williams JS and Greenlee WF: Characterization of 2,3,7,8-tetrachlorodibenzofuran-dependent suppression and AH receptor pathway gene expression in the developing mouse mammary gland. Toxicol Appl Pharmacol. 152:200–210. 1998. View Article : Google Scholar : PubMed/NCBI

58 

O'Donnell EF, Koch DC, Bisson WH, Jang HS and Kolluri SK: The aryl hydrocarbon receptor mediates raloxifene-induced apoptosis in estrogen receptor-negative hepatoma and breast cancer cells. Cell Death Dis. 5:e10382014. View Article : Google Scholar : PubMed/NCBI

59 

Romagnolo DF, Papoutsis AJ, Laukaitis C and Selmin OI: Constitutive expression of AhR and BRCA-1 promoter CpG hypermethylation as biomarkers of ERα-negative breast tumorigenesis. BMC Cancer. 15:10262015. View Article : Google Scholar : PubMed/NCBI

60 

Mohamed HT, Gadalla R, El-Husseiny N, Hassan H, Wang Z, Ibrahim SA, El-Shinawi M, Sherr DH and Mohamed MM: Inflammatory breast cancer: Activation of the aryl hydrocarbon receptor and its target CYP1B1 correlates closely with Wnt5a/b-β-catenin signalling, the stem cell phenotype and disease progression. J Adv Res. 16:75–86. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Jeschke U, Zhang X, Kuhn C, Jalaguier S, Colinge J, Pfender K, Mayr D, Ditsch N, Harbeck N, Mahner S, et al: The prognostic impact of the Aryl hydrocarbon receptor (AhR) in primary breast cancer depends on the lymph node status. Int J Mol Sci. 20:10162019. View Article : Google Scholar : PubMed/NCBI

62 

Vacher S, Castagnet P, Chemlali W, Lallemand F, Meseure D, Pocard M, Bieche I and Perrot-Applanat M: High AHR expression in breast tumors correlates with expression of genes from several signaling pathways namely inflammation and endogenous tryptophan metabolism. PLoS One. 13:e01906192018. View Article : Google Scholar : PubMed/NCBI

63 

Tryggvadottir H, Sandén E, Björner S, Bressan A, Ygland Rödström M, Khazaei S, Edwards DP, Nodin B, Jirström K, Isaksson K, et al: The prognostic impact of intratumoral Aryl hydrocarbon receptor in primary breast cancer depends on the type of endocrine therapy: A population-based cohort study. Front Oncol. 11:6427682021. View Article : Google Scholar : PubMed/NCBI

64 

Goode G, Pratap S and Eltom SE: Depletion of the aryl hydrocarbon receptor in MDA-MB-231 human breast cancer cells altered the expression of genes in key regulatory pathways of cancer. PLoS One. 9:e1001032014. View Article : Google Scholar : PubMed/NCBI

65 

Goode GD, Ballard BR, Manning HC, Freeman ML, Kang Y and Eltom SE: Knockdown of aberrantly upregulated aryl hydrocarbon receptor reduces tumor growth and metastasis of MDA-MB-231 human breast cancer cell line. Int J Cancer. 133:2769–2780. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Koual M, Tomkiewicz C, Cano-Sancho G, Antignac JP, Bats AS and Coumoul X: Environmental chemicals, breast cancer progression and drug resistance. Environ Health. 19:1172020. View Article : Google Scholar : PubMed/NCBI

67 

Li Y, Qin HZ, Song Q, Wu XD and Zhu JH: Lack of association between the aryl hydrocarbon receptor rs2066853 polymorphism and breast cancer: A meta-analysis on Ahr polymorphism and breast cancer. Genet Mol Res. 14:16162–16168. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Ali S and Coombes RC: Endocrine-responsive breast cancer and strategies for combating resistance. Nat Rev Cancer. 2:101–112. 2002. View Article : Google Scholar : PubMed/NCBI

69 

Xu Y, Huangyang P, Wang Y, Xue L, Devericks E, Nguyen HG, Yu X, Oses-Prieto JA, Burlingame AL, Miglani S, et al: ERα is an RNA-binding protein sustaining tumor cell survival and drug resistance. Cell. 184:5215–5229.e17. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Shiau AK, Barstad D, Loria PM, Cheng L, Kushner PJ, Agard DA and Greene GL: The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell. 95:927–937. 1998. View Article : Google Scholar : PubMed/NCBI

71 

Metcalfe C, Friedman LS and Hager JH: Hormone-targeted therapy and resistance. Annu Rev Cancer Biol. 2:291–312. 2018. View Article : Google Scholar

72 

Safe S and Wormke M: Inhibitory aryl hydrocarbon receptor-estrogen receptor alpha cross-talk and mechanisms of action. Chem Res Toxicol. 16:807–816. 2003. View Article : Google Scholar : PubMed/NCBI

73 

Niwa A, Kumaki K, Nebert DW and Poland AP: Genetic expression of aryl hydrocarbon hydroxylase activity in the mouse. Distinction between the ‘responsive’ homozygote and heterozygote at the Ah locus. Arch Biochem Biophys. 166:559–564. 1975. View Article : Google Scholar : PubMed/NCBI

74 

Hankinson O: Single-step selection of clones of a mouse hepatoma line deficient in aryl hydrocarbon hydroxylase. Proc Natl Acad Sci USA. 76:373–376. 1979. View Article : Google Scholar : PubMed/NCBI

75 

Stark K, Burger A, Wu J, Shelton P, Polin L and Li J: Reactivation of estrogen receptor α by vorinostat sensitizes mesenchymal-like triple-negative breast cancer to aminoflavone, a ligand of the aryl hydrocarbon receptor. PLoS One. 8:e745252013. View Article : Google Scholar : PubMed/NCBI

76 

Go RE, Hwang KA and Choi KC: Cytochrome P450 1 family and cancers. J Steroid Biochem Mol Biol. 147:24–30. 2015. View Article : Google Scholar : PubMed/NCBI

77 

Luzzani GA, Callero MA, Kuruppu AI, Trapani V, Flumian C, Todaro L, Bradshaw TD and Loaiza Perez AI: In vitro antitumor effects of AHR ligands aminoflavone (AFP 464) and benzothiazole (5F 203) in human renal carcinoma cells. J Cell Biochem. 118:4526–4535. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Brunnberg S, Pettersson K, Rydin E, Matthews J, Hanberg A and Pongratz I: The basic helix-loop-helix-PAS protein ARNT functions as a potent coactivator of estrogen receptor-dependent transcription. Proc Natl Acad Sci USA. 100:6517–6522. 2003. View Article : Google Scholar : PubMed/NCBI

79 

Ohtake F, Takeyama K, Matsumoto T, Kitagawa H, Yamamoto Y, Nohara K, Tohyama C, Krust A, Mimura J, Chambon P, et al: Modulation of oestrogen receptor signalling by association with the activated dioxin receptor. Nature. 423:545–550. 2003. View Article : Google Scholar : PubMed/NCBI

80 

Kociba RJ, Keyes DG, Beyer JE, Carreon RM, Wade CE, Dittenber DA, Kalnins RP, Frauson LE, Park CN, Barnard SD, et al: Results of a two-year chronic toxicity and oncogenicity study of 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats. Toxicol Appl Pharmacol. 46:279–303. 1978. View Article : Google Scholar : PubMed/NCBI

81 

Low Dog T: Menopause: A review of botanical dietary supplements. Am J Med. 118 (Suppl 12B):S98–S108. 2005. View Article : Google Scholar : PubMed/NCBI

82 

Gong P, Madak-Erdogan Z, Flaws JA, Shapiro DJ, Katzenellenbogen JA and Katzenellenbogen BS: Estrogen receptor-alpha and aryl hydrocarbon receptor involvement in the actions of botanical estrogens in target cells. Mol Cell Endocrinol. 437:190–200. 2016. View Article : Google Scholar : PubMed/NCBI

83 

Moynahan ME: The cancer connection: BRCA1 and BRCA2 tumor suppression in mice and humans. Oncogene. 21:8994–9007. 2002. View Article : Google Scholar : PubMed/NCBI

84 

Baek HJ, Kim SE, Choi EK, Kim JK, Shin DH, Park EJ, Kim TH, Kim JY, Kim KG, Deng CX and Kim SS: Inhibition of estrogen signaling reduces the incidence of BRCA1-associated mammary tumor formation. Int J Biol Sci. 14:1755–1768. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Wang L and Di LJ: BRCA1 and estrogen/estrogen receptor in breast cancer: Where they interact? Int J Biol Sci. 10:566–575. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Kang HJ, Kim HJ, Cho CH, Hu Y, Li R and Bae I: BRCA1 transcriptional activity is enhanced by interactions between its AD1 domain and AhR. Cancer Chemother Pharmacol. 62:965–975. 2008. View Article : Google Scholar : PubMed/NCBI

87 

Tapia T, Smalley SV, Kohen P, Muñoz A, Solis LM, Corvalan A, Faundez P, Devoto L, Camus M, Alvarez M and Carvallo P: Promoter hypermethylation of BRCA1 correlates with absence of expression in hereditary breast cancer tumors. Epigenetics. 3:157–163. 2008. View Article : Google Scholar : PubMed/NCBI

88 

Papoutsis AJ, Selmin OI, Borg JL and Romagnolo DF: Gestational exposure to the AhR agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin induces BRCA-1 promoter hypermethylation and reduces BRCA-1 expression in mammary tissue of rat offspring: Preventive effects of resveratrol. Mol Carcinog. 54:261–269. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Wormke M, Stoner M, Saville B, Walker K, Abdelrahim M, Burghardt R and Safe S: The aryl hydrocarbon receptor mediates degradation of estrogen receptor alpha through activation of proteasomes. Mol Cell Biol. 23:1843–1855. 2003. View Article : Google Scholar : PubMed/NCBI

90 

Crimini E, Repetto M, Aftimos P, Botticelli A, Marchetti P and Curigliano G: Precision medicine in breast cancer: From clinical trials to clinical practice. Cancer Treat Rev. 98:1022232021. View Article : Google Scholar : PubMed/NCBI

91 

Zhang S, Lei P, Liu X, Li X, Walker K, Kotha L, Rowlands C and Safe S: The aryl hydrocarbon receptor as a target for estrogen receptor-negative breast cancer chemotherapy. Endocr Relat Cancer. 16:835–844. 2009. View Article : Google Scholar : PubMed/NCBI

92 

Zhang S, Kim K, Jin UH, Pfent C, Cao H, Amendt B, Liu X, Wilson-Robles H and Safe S: Aryl hydrocarbon receptor agonists induce microRNA-335 expression and inhibit lung metastasis of estrogen receptor negative breast cancer cells. Mol Cancer Ther. 11:108–118. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Hanieh H: Aryl hydrocarbon receptor-microRNA-212/132 axis in human breast cancer suppresses metastasis by targeting SOX4. Mol Cancer. 14:1722015. View Article : Google Scholar : PubMed/NCBI

94 

Hall JM, Barhoover MA, Kazmin D, McDonnell DP, Greenlee WF and Thomas RS: Activation of the aryl-hydrocarbon receptor inhibits invasive and metastatic features of human breast cancer cells and promotes breast cancer cell differentiation. Mol Endocrinol. 24:359–369. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Barhoover MA, Hall JM, Greenlee WF and Thomas RS: Aryl hydrocarbon receptor regulates cell cycle progression in human breast cancer cells via a functional interaction with cyclin-dependent kinase 4. Mol Pharmacol. 77:195–201. 2010. View Article : Google Scholar : PubMed/NCBI

96 

Wang T, Wyrick KL, Meadows GG, Wills TB and Vorderstrasse BA: Activation of the aryl hydrocarbon receptor by TCDD inhibits mammary tumor metastasis in a syngeneic mouse model of breast cancer. Toxicol Sci. 124:291–298. 2011. View Article : Google Scholar : PubMed/NCBI

97 

Safe S and Zhang L: The role of the Aryl hydrocarbon receptor (AhR) and its ligands in breast cancer. Cancers (Basel). 14:55742022. View Article : Google Scholar : PubMed/NCBI

98 

Ho JN, Jun W, Choue R and Lee J: I3C and ICZ inhibit migration by suppressing the EMT process and FAK expression in breast cancer cells. Mol Med Rep. 7:384–388. 2013. View Article : Google Scholar : PubMed/NCBI

99 

Nguyen CH, Brenner S, Huttary N, Atanasov AG, Dirsch VM, Chatuphonprasert W, Holzner S, Stadler S, Riha J, Krieger S, et al: AHR/CYP1A1 interplay triggers lymphatic barrier breaching in breast cancer spheroids by inducing 12(S)-HETE synthesis. Hum Mol Genet. 25:5006–5016. 2016.PubMed/NCBI

100 

Yerushalmi R, Bargil S, Ber Y, Ozlavo R, Sivan T, Rapson Y, Pomerantz A, Tsoref D, Sharon E, Caspi O, et al: 3,3-Diindolylmethane (DIM): A nutritional intervention and its impact on breast density in healthy BRCA carriers. A prospective clinical trial. Carcinogenesis. 41:1395–1401. 2020. View Article : Google Scholar : PubMed/NCBI

101 

Mobini K, Tamaddon G, Fardid R, Keshavarzi M and Mohammadi-Bardbori A: Aryl hydrocarbon-estrogen alpha receptor-dependent expression of miR-206, miR-27b, and miR-133a suppress cell proliferation and migration in MCF-7 cells. J Biochem Mol Toxicol. 33:e223042019. View Article : Google Scholar : PubMed/NCBI

102 

Piwarski SA, Thompson C, Chaudhry AR, Denvir J, Primerano DA, Fan J and Salisbury TB: The putative endogenous AHR ligand ITE reduces JAG1 and associated NOTCH1 signaling in triple negative breast cancer cells. Biochem Pharmacol. 174:1138452020. View Article : Google Scholar : PubMed/NCBI

103 

Sári Z, Mikó E, Kovács T, Boratkó A, Ujlaki G, Jankó L, Kiss B, Uray K and Bai P: Indoxylsulfate, a metabolite of the microbiome, has cytostatic effects in breast cancer via activation of AHR and PXR receptors and induction of oxidative stress. Cancers (Basel). 12:29152020. View Article : Google Scholar : PubMed/NCBI

104 

Sári Z, Mikó E, Kovács T, Jankó L, Csonka T, Lente G, Sebő É, Tóth J, Tóth D, Árkosy P, et al: Indolepropionic acid, a metabolite of the microbiome, has cytostatic properties in breast cancer by activating AHR and PXR receptors and inducing oxidative stress. Cancers (Basel). 12:24112020. View Article : Google Scholar : PubMed/NCBI

105 

Safe S, Jin UH, Park H, Chapkin RS and Jayaraman A: Aryl hydrocarbon receptor (AHR) ligands as selective AHR modulators (SAhRMs). Int J Mol Sci. 21:66542020. View Article : Google Scholar : PubMed/NCBI

106 

Akama T, Ishida H, Shida Y, Kimura U, Gomi K, Saito H, Fuse E, Kobayashi S, Yoda N and Kasai M: Design and synthesis of potent antitumor 5,4′-diaminoflavone derivatives based on metabolic considerations. J Med Chem. 40:1894–1900. 1997. View Article : Google Scholar : PubMed/NCBI

107 

Akama T, Shida Y, Sugaya T, Ishida H, Gomi K and Kasai M: Novel 5-aminoflavone derivatives as specific antitumor agents in breast cancer. J Med Chem. 39:3461–3469. 1996. View Article : Google Scholar : PubMed/NCBI

108 

Kenz S, Haas CS, Werth SC, Bohnet S and Brabant G: High sensitivity to tolvaptan in paraneoplastic syndrome of inappropriate ADH secretion (SIADH). Ann Oncol. 22:26962011. View Article : Google Scholar : PubMed/NCBI

109 

Kuffel MJ, Schroeder JC, Pobst LJ, Naylor S, Reid JM, Kaufmann SH and Ames MM: Activation of the antitumor agent aminoflavone (NSC 686288) is mediated by induction of tumor cell cytochrome P450 1A1/1A2. Mol Pharmacol. 62:143–153. 2002. View Article : Google Scholar : PubMed/NCBI

110 

Loaiza-Perez AI, Kenney S, Boswell J, Hollingshead M, Alley MC, Hose C, Ciolino HP, Yeh GC, Trepel JB, Vistica DT and Sausville EA: Aryl hydrocarbon receptor activation of an antitumor aminoflavone: Basis of selective toxicity for MCF-7 breast tumor cells. Mol Cancer Ther. 3:715–725. 2004. View Article : Google Scholar : PubMed/NCBI

111 

Loaiza-Perez AI, Kenney S, Boswell J, Hollingshead M, Hose C, Linehan WM, Worrell R, Rubinstein L, Sausville EA and Vistica DT: Sensitivity of renal cell carcinoma to aminoflavone: role of CYP1A1. J Urol. 171:1688–1697. 2004. View Article : Google Scholar : PubMed/NCBI

112 

Meng LH, Shankavaram U, Chen C, Agama K, Fu HQ, Gonzalez FJ, Weinstein J and Pommier Y: Activation of aminoflavone (NSC 686288) by a sulfotransferase is required for the antiproliferative effect of the drug and for induction of histone gamma-H2AX. Cancer Res. 66:9656–9664. 2006. View Article : Google Scholar : PubMed/NCBI

113 

Mavingire N, Campbell P, Liu T, Wooten J, Khan S, Chen X, Matthews J, Wang C and Brantley E: Aminoflavone upregulates putative tumor suppressor miR-125b-2-3p to inhibit luminal A breast cancer stem cell-like properties. Precis Clin Med. 5:pbac0082022. View Article : Google Scholar : PubMed/NCBI

114 

Campbell PS, Mavingire N, Khan S, Rowland LK, Wooten JV, Opoku-Agyeman A, Guevara A, Soto U, Cavalli F, Loaiza-Pérez AI, et al: AhR ligand aminoflavone suppresses α6-integrin-Src-Akt signaling to attenuate tamoxifen resistance in breast cancer cells. J Cell Physiol. 234:108–121. 2018. View Article : Google Scholar : PubMed/NCBI

115 

Brantley E, Callero MA, Berardi DE, Campbell P, Rowland L, Zylstra D, Amis L, Yee M, Simian M, Todaro L, et al: AhR ligand Aminoflavone inhibits α6-integrin expression and breast cancer sphere-initiating capacity. Cancer Lett. 376:53–61. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Hu T, Zhou R, Zhao Y and Wu G: Integrin α6/Akt/Erk signaling is essential for human breast cancer resistance to radiotherapy. Sci Rep. 6:333762016. View Article : Google Scholar : PubMed/NCBI

117 

Zhao S, Kanno Y, Nakayama M, Makimura M, Ohara S and Inouye Y: Activation of the aryl hydrocarbon receptor represses mammosphere formation in MCF-7 cells. Cancer Lett. 317:192–198. 2012. View Article : Google Scholar : PubMed/NCBI

118 

Wang C, Xu CX, Bu Y, Bottum KM and Tischkau SA: Beta-naphthoflavone (DB06732) mediates estrogen receptor-positive breast cancer cell cycle arrest through AhR-dependent regulation of PI3K/AKT and MAPK/ERK signaling. Carcinogenesis. 35:703–713. 2014. View Article : Google Scholar : PubMed/NCBI

119 

Fukasawa K, Kagaya S, Maruyama S, Kuroiwa S, Masuda K, Kameyama Y, Satoh Y, Akatsu Y, Tomura A, Nishikawa K, et al: A novel compound, NK150460, exhibits selective antitumor activity against breast cancer cell lines through activation of aryl hydrocarbon receptor. Mol Cancer Ther. 14:343–354. 2015. View Article : Google Scholar : PubMed/NCBI

120 

Tarleton M, Gilbert J, Robertson MJ, McCluskey A and Sakoff JA: Library synthesis and cytotoxicity of a family of 2-phenylacrylonitriles and discovery of an estrogen dependent breast cancer lead compound†. Med Chem Commun. 2:31–37. 2011. View Article : Google Scholar

121 

Gilbert J, De Iuliis GN, Tarleton M, McCluskey A and Sakoff JA: (Z)-2-(3,4-Dichlorophenyl)-3-(1 H-Pyrrol-2-yl)acrylonitrile exhibits selective antitumor activity in breast cancer cell lines via the aryl hydrocarbon receptor pathway. Mol Pharmacol. 93:168–177. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Corsello SM, Bittker JA, Liu Z, Gould J, McCarren P, Hirschman JE, Johnston SE, Vrcic A, Wong B, Khan M, et al: The drug repurposing Hub: A next-generation drug library and information resource. Nat Med. 23:405–408. 2017. View Article : Google Scholar : PubMed/NCBI

123 

Quattrochi LC and Tukey RH: Nuclear uptake of the Ah (dioxin) receptor in response to omeprazole: Transcriptional activation of the human CYP1A1 gene. Mol Pharmacol. 43:504–508. 1993.PubMed/NCBI

124 

Prud'homme GJ, Glinka Y, Toulina A, Ace O, Subramaniam V and Jothy S: Breast cancer stem-like cells are inhibited by a non-toxic aryl hydrocarbon receptor agonist. PLoS One. 5:e138312010. View Article : Google Scholar : PubMed/NCBI

125 

Jin UH, Lee SO and Safe S: Aryl hydrocarbon receptor (AHR)-active pharmaceuticals are selective AHR modulators in MDA-MB-468 and BT474 breast cancer cells. J Pharmacol Exp Ther. 343:333–341. 2012. View Article : Google Scholar : PubMed/NCBI

126 

Cummings SR, Tice JA, Bauer S, Browner WS, Cuzick J, Ziv E, Vogel V, Shepherd J, Vachon C, Smith-Bindman R and Kerlikowske K: Prevention of breast cancer in postmenopausal women: Approaches to estimating and reducing risk. J Natl Cancer Inst. 101:384–398. 2009. View Article : Google Scholar : PubMed/NCBI

127 

Moen MD and Keating GM: Raloxifene: A review of its use in the prevention of invasive breast cancer. Drugs. 68:2059–2083. 2008. View Article : Google Scholar : PubMed/NCBI

128 

Bevers TB: Raloxifene and the prevention of breast cancer. Expert Opin Pharmacother. 7:2301–2307. 2006. View Article : Google Scholar : PubMed/NCBI

129 

Martino S, Cauley JA, Barrett-Connor E, Powles TJ, Mershon J, Disch D, Secrest RJ and Cummings SR; CORE Investigators, : Continuing outcomes relevant to Evista: Breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst. 96:1751–1761. 2004. View Article : Google Scholar : PubMed/NCBI

130 

Clemens JA, Bennett DR, Black LJ and Jones CD: Effects of a new antiestrogen, keoxifene (LY156758), on growth of carcinogen-induced mammary tumors and on LH and prolactin levels. Life Sci. 32:2869–2875. 1983. View Article : Google Scholar : PubMed/NCBI

131 

Kleinberg DL, Todd J and Babitsky G: Inhibition by estradiol of the lactogenic effect of prolactin in primate mammary tissue: Reversal by antiestrogens LY 156758 and tamoxifen. Proc Natl Acad Sci USA. 80:4144–4148. 1983. View Article : Google Scholar : PubMed/NCBI

132 

Ning M, Zhou C, Weng J, Zhang S, Chen D, Yang C, Wang H, Ren J, Zhou L, Jin C and Wang MW: Biological activities of a novel selective oestrogen receptor modulator derived from raloxifene (Y134). Br J Pharmacol. 150:19–28. 2007. View Article : Google Scholar : PubMed/NCBI

133 

Jang HS, Pearce M, O'Donnell EF, Nguyen BD, Truong L, Mueller MJ, Bisson WH, Kerkvliet NI, Tanguay RL and Kolluri SK: Identification of a raloxifene analog that promotes AhR-mediated apoptosis in cancer cells. Biology (Basel). 6:412017.PubMed/NCBI

134 

Hyder T, Marino CC, Ahmad S, Nasrazadani A and Brufsky AM: Aromatase inhibitor-associated musculoskeletal syndrome: Understanding mechanisms and management. Front Endocrinol (Lausanne). 12:7137002021. View Article : Google Scholar : PubMed/NCBI

135 

Dzeletovic N, McGuire J, Daujat M, Tholander J, Ema M, Fujii-Kuriyama Y, Bergman J, Maurel P and Poellinger L: Regulation of dioxin receptor function by omeprazole. J Biol Chem. 272:12705–12713. 1997. View Article : Google Scholar : PubMed/NCBI

136 

Jin UH, Kim SB and Safe S: Omeprazole inhibits pancreatic cancer cell invasion through a nongenomic Aryl hydrocarbon receptor pathway. Chem Res Toxicol. 28:907–918. 2015. View Article : Google Scholar : PubMed/NCBI

137 

Lesca P, Peryt B, Larrieu G, Alvinerie M, Galtier P, Daujat M, Maurel P and Hoogenboom L: Evidence for the ligand-independent activation of the AH receptor. Biochem Biophys Res Commun. 209:474–482. 1995. View Article : Google Scholar : PubMed/NCBI

138 

Jin UH, Lee SO, Pfent C and Safe S: The aryl hydrocarbon receptor ligand omeprazole inhibits breast cancer cell invasion and metastasis. BMC Cancer. 14:4982014. View Article : Google Scholar : PubMed/NCBI

139 

Koda A, Nagai H, Watanabe S, Yanagihara Y and Sakamoto K: Inhibition of hypersensitivity reactions by a new drug, N(3′,4′-dimethoxycinnamoyl) anthranilic acid (N-5′). J Allergy Clin. Immunol. 57:396–407. 1976.

140 

Azuma H, Banno K and Yoshimura T: Pharmacological properties of N-(3′,4′-dimethoxycinnamoyl) anthranilic acid (N-5′), a new anti-atopic agent. Br J Pharmacol. 58:483–488. 1976. View Article : Google Scholar : PubMed/NCBI

141 

Batlle E and Clevers H: Cancer stem cells revisited. Nat Med. 23:1124–1134. 2017. View Article : Google Scholar : PubMed/NCBI

142 

Chakrabarti R, Subramaniam V, Abdalla S, Jothy S and Prud'homme GJ: Tranilast inhibits the growth and metastasis of mammary carcinoma. Anticancer Drugs. 20:334–345. 2009. View Article : Google Scholar : PubMed/NCBI

143 

Subramaniam V, Ace O, Prud'homme GJ and Jothy S: Tranilast treatment decreases cell growth, migration and inhibits colony formation of human breast cancer cells. Exp Mol Pathol. 90:116–122. 2011. View Article : Google Scholar : PubMed/NCBI

144 

Ciolino HP, MacDonald CJ, Memon OS, Bass SE and Yeh GC: Sulindac regulates the aryl hydrocarbon receptor-mediated expression of phase 1 metabolic enzymes in vivo and in vitro. Carcinogenesis. 27:1586–1592. 2006. View Article : Google Scholar : PubMed/NCBI

145 

Hahn ME: Aryl hydrocarbon receptors: Diversity and evolution. Chem Biol Interact. 141:131–160. 2002. View Article : Google Scholar : PubMed/NCBI

146 

Schlezinger JJ, Liu D, Farago M, Seldin DC, Belguise K, Sonenshein GE and Sherr DH: A role for the aryl hydrocarbon receptor in mammary gland tumorigenesis. Biol Chem. 387:1175–1187. 2006. View Article : Google Scholar : PubMed/NCBI

147 

Vogel CFA, Lazennec G, Kado SY, Dahlem C, He Y, Castaneda A, Ishihara Y, Vogeley C, Rossi A, Haarmann-Stemmann T, et al: Targeting the Aryl hydrocarbon receptor signaling pathway in breast cancer development. Front Immunol. 12:6253462021. View Article : Google Scholar : PubMed/NCBI

148 

Kolasa E, Houlbert N, Balaguer P and Fardel O: AhR- and NF-κB-dependent induction of interleukin-6 by co-exposure to the environmental contaminant benzanthracene and the cytokine tumor necrosis factor-α in human mammary MCF-7 cells. Chem Biol Interact. 203:391–400. 2013. View Article : Google Scholar : PubMed/NCBI

149 

Shan A, Leng L, Li J, Luo XM, Fan YJ, Yang Q, Xie QH, Chen YS, Ni CS, Guo LM, et al: TCDD-induced antagonism of MEHP-mediated migration and invasion partly involves aryl hydrocarbon receptor in MCF7 breast cancer cells. J Hazard Mater. 398:1228692020. View Article : Google Scholar : PubMed/NCBI

150 

Bekki K, Vogel H, Li W, Ito T, Sweeney C, Haarmann-Stemmann T, Matsumura F and Vogel CF: The aryl hydrocarbon receptor (AhR) mediates resistance to apoptosis induced in breast cancer cells. Pestic Biochem Physiol. 120:5–13. 2015. View Article : Google Scholar : PubMed/NCBI

151 

Pontillo C, Español A, Chiappini F, Miret N, Cocca C, Alvarez L, Kleiman de Pisarev D, Sales ME and Randi AS: Hexachlorobenzene promotes angiogenesis in vivo, in a breast cancer model and neovasculogenesis in vitro, in the human microvascular endothelial cell line HMEC-1. Toxicol Lett. 239:53–64. 2015. View Article : Google Scholar : PubMed/NCBI

152 

Vogel CFA, Li W, Wu D, Miller JK, Sweeney C, Lazennec G, Fujisawa Y and Matsumura F: Interaction of aryl hydrocarbon receptor and NF-κB subunit RelB in breast cancer is associated with interleukin-8 overexpression. Arch Biochem Biophys. 512:78–86. 2011. View Article : Google Scholar : PubMed/NCBI

153 

Novikov O, Wang Z, Stanford EA, Parks AJ, Ramirez-Cardenas A, Landesman E, Laklouk I, Sarita-Reyes C, Gusenleitner D, Li A, et al: An Aryl hydrocarbon receptor-mediated amplification loop that enforces cell migration in ER-/PR-/Her2-human breast cancer cells. Mol Pharmacol. 90:674–688. 2016. View Article : Google Scholar : PubMed/NCBI

154 

Hsieh TH, Tsai CF, Hsu CY, Kuo PL, Lee JN, Chai CY, Wang SC and Tsai EM: Phthalates induce proliferation and invasiveness of estrogen receptor-negative breast cancer through the AhR/HDAC6/c-Myc signaling pathway. FASEB J. 26:778–787. 2012. View Article : Google Scholar : PubMed/NCBI

155 

Miret N, Pontillo C, Ventura C, Carozzo A, Chiappini F, Kleiman de Pisarev D, Fernández N, Cocca C and Randi A: Hexachlorobenzene modulates the crosstalk between the aryl hydrocarbon receptor and transforming growth factor-β1 signaling, enhancing human breast cancer cell migration and invasion. Toxicology. 366–367. 20–31. 2016.

156 

Pontillo CA, Rojas P, Chiappini F, Sequeira G, Cocca C, Crocci M, Colombo L, Lanari C, Kleiman de Pisarev D and Randi A: Action of hexachlorobenzene on tumor growth and metastasis in different experimental models. Toxicol Appl Pharmacol. 268:331–342. 2013. View Article : Google Scholar : PubMed/NCBI

157 

Al-Dhfyan A, Alhoshani A and Korashy HM: Aryl hydrocarbon receptor/cytochrome P450 1A1 pathway mediates breast cancer stem cells expansion through PTEN inhibition and β-catenin and Akt activation. Mol Cancer. 16:142017. View Article : Google Scholar : PubMed/NCBI

158 

Jung JW, Park SB, Lee SJ, Seo MS, Trosko JE and Kang KS: Metformin represses self-renewal of the human breast carcinoma stem cells via inhibition of estrogen receptor-mediated OCT4 expression. PLoS One. 6:e280682011. View Article : Google Scholar : PubMed/NCBI

159 

Malik DE, David RM and Gooderham NJ: Interleukin-6 selectively induces drug metabolism to potentiate the genotoxicity of dietary carcinogens in mammary cells. Arch Toxicol. 93:3005–3020. 2019. View Article : Google Scholar : PubMed/NCBI

160 

Zárate LV, Pontillo CA, Español A, Miret NV, Chiappini F, Cocca C, Álvarez L, de Pisarev DK, Sales ME and Randi AS: Angiogenesis signaling in breast cancer models is induced by hexachlorobenzene and chlorpyrifos, pesticide ligands of the aryl hydrocarbon receptor. Toxicol Appl Pharmacol. 401:1150932020. View Article : Google Scholar : PubMed/NCBI

161 

Pontillo CA, Garcia MA, Peña D, Cocca C, Chiappini F, Alvarez L, Kleiman de Pisarev D and Randi AS: Activation of c-Src/HER1/STAT5b and HER1/ERK1/2 signaling pathways and cell migration by hexachlorobenzene in MDA-MB-231 human breast cancer cell line. Toxicol Sci. 120:284–296. 2011. View Article : Google Scholar : PubMed/NCBI

162 

Castillo-Sanchez R, Villegas-Comonfort S, Galindo-Hernandez O, Gomez R and Salazar EP: Benzo-[a]-pyrene induces FAK activation and cell migration in MDA-MB-231 breast cancer cells. Cell Biol Toxicol. 29:303–319. 2013. View Article : Google Scholar : PubMed/NCBI

163 

Guo J, Xu Y, Ji W, Song L, Dai C and Zhan L: Effects of exposure to benzo[a]pyrene on metastasis of breast cancer are mediated through ROS-ERK-MMP9 axis signaling. Toxicol Lett. 234:201–210. 2015. View Article : Google Scholar : PubMed/NCBI

164 

Cirillo F, Lappano R, Bruno L, Rizzuti B, Grande F, Guzzi R, Briguori S, Miglietta AM, Nakajima M, Di Martino MT and Maggiolini M: AHR and GPER mediate the stimulatory effects induced by 3-methylcholanthrene in breast cancer cells and cancer-associated fibroblasts (CAFs). J Exp Clin Cancer Res. 38:3352019. View Article : Google Scholar : PubMed/NCBI

165 

Yamashita N, Saito N, Zhao S, Terai K, Hiruta N, Park Y, Bujo H, Nemoto K and Kanno Y: Heregulin-induced cell migration is promoted by aryl hydrocarbon receptor in HER2-overexpressing breast cancer cells. Exp Cell Res. 366:34–40. 2018. View Article : Google Scholar : PubMed/NCBI

166 

D'Amato NC, Rogers TJ, Gordon MA, Greene LI, Cochrane DR, Spoelstra NS, Nemkov TG, D'Alessandro A, Hansen KC and Richer JK: A TDO2-AhR signaling axis facilitates anoikis resistance and metastasis in triple-negative breast cancer. Cancer Res. 75:4651–4664. 2015. View Article : Google Scholar : PubMed/NCBI

167 

Parks AJ, Pollastri MP, Hahn ME, Stanford EA, Novikov O, Franks DG, Haigh SE, Narasimhan S, Ashton TD, Hopper TG, et al: In silico identification of an aryl hydrocarbon receptor antagonist with biological activity in vitro and in vivo. Mol Pharmacol. 86:593–608. 2014. View Article : Google Scholar : PubMed/NCBI

168 

Nassar D and Blanpain C: Cancer stem cells: Basic concepts and therapeutic implications. Annu Rev Pathol. 11:47–76. 2016. View Article : Google Scholar : PubMed/NCBI

169 

Peitzsch C, Tyutyunnykova A, Pantel K and Dubrovska A: Cancer stem cells: The root of tumor recurrence and metastases. Semin Cancer Biol. 44:10–24. 2017. View Article : Google Scholar : PubMed/NCBI

170 

Regan Anderson TM, Ma S, Perez Kerkvliet C, Peng Y, Helle TM, Krutilina RI, Raj GV, Cidlowski JA, Ostrander JH, Schwertfeger KL, et al: Taxol induces Brk-dependent prosurvival phenotypes in TNBC cells through an AhR/GR/HIF-driven signaling axis. Mol Cancer Res. 16:1761–1772. 2018. View Article : Google Scholar : PubMed/NCBI

171 

Toomey DP, Murphy JF and Conlon KC: COX-2, VEGF and tumour angiogenesis. Surgeon. 7:174–180. 2009. View Article : Google Scholar : PubMed/NCBI

172 

Kirkpatrick K, Ogunkolade W, Elkak A, Bustin S, Jenkins P, Ghilchik M and Mokbel K: The mRNA expression of cyclo-oxygenase-2 (COX-2) and vascular endothelial growth factor (VEGF) in human breast cancer. Curr Med Res Opin. 18:237–241. 2002. View Article : Google Scholar : PubMed/NCBI

173 

Sansone P, Storci G, Tavolari S, Guarnieri T, Giovannini C, Taffurelli M, Ceccarelli C, Santini D, Paterini P, Marcu KB, et al: IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J Clin Invest. 117:3988–4002. 2007. View Article : Google Scholar : PubMed/NCBI

174 

Baumgarten SC and Frasor J: Minireview: Inflammation: An instigator of more aggressive estrogen receptor (ER) positive breast cancers. Mol Endocrinol. 26:360–371. 2012. View Article : Google Scholar : PubMed/NCBI

175 

Sasser AK, Sullivan NJ, Studebaker AW, Hendey LF, Axel AE and Hall BM: Interleukin-6 is a potent growth factor for ER-alpha-positive human breast cancer. FASEB J. 21:3763–3770. 2007. View Article : Google Scholar : PubMed/NCBI

176 

Fouad TM, Barrera AMG, Reuben JM, Lucci A, Woodward WA, Stauder MC, Lim B, DeSnyder SM, Arun B, Gildy B, et al: Inflammatory breast cancer: A proposed conceptual shift in the UICC-AJCC TNM staging system. Lancet Oncol. 18:e228–e232. 2017. View Article : Google Scholar : PubMed/NCBI

177 

Wang Z, Wang H, Ding X, Chen X and Shen K: A large-cohort retrospective study of metastatic patterns and prognostic outcomes between inflammatory and non-inflammatory breast cancer. Ther Adv Med Oncol. 12:17588359209326742020. View Article : Google Scholar : PubMed/NCBI

178 

Sovak MA, Bellas RE, Kim DW, Zanieski GJ, Rogers AE, Traish AM and Sonenshein GE: Aberrant nuclear factor-kappaB/Rel expression and the pathogenesis of breast cancer. J Clin Invest. 100:2952–2960. 1997. View Article : Google Scholar : PubMed/NCBI

179 

Kim DW, Sovak MA, Zanieski G, Nonet G, Romieu-Mourez R, Lau AW, Hafer LJ, Yaswen P, Stampfer M, Rogers AE, et al: Activation of NF-kappaB/Rel occurs early during neoplastic transformation of mammary cells. Carcinogenesis. 21:871–879. 2000. View Article : Google Scholar : PubMed/NCBI

180 

Kim DW, Gazourian L, Quadri SA, Romieu-Mourez R, Sherr DH and Sonenshein GE: The RelA NF-kappaB subunit and the aryl hydrocarbon receptor (AhR) cooperate to transactivate the c-myc promoter in mammary cells. Oncogene. 19:5498–5506. 2000. View Article : Google Scholar : PubMed/NCBI

181 

Currier N, Solomon SE, Demicco EG, Chang DL, Farago M, Ying H, Dominguez I, Sonenshein GE, Cardiff RD, Xiao ZX, et al: Oncogenic signaling pathways activated in DMBA-induced mouse mammary tumors. Toxicol Pathol. 33:726–737. 2005. View Article : Google Scholar : PubMed/NCBI

182 

Vyas D, Lopez-Hisijos N, Shah P, Deshpande KS, Basson MD, Vyas A and Chaturvedi LS: A second-generation proteasome inhibitor and doxorubicin modulates IL-6, pSTAT-3 and NF-kB activity in MDA-MB-231 breast cancer cells. J Nanosci Nanotechnol. 17:175–185. 2017. View Article : Google Scholar : PubMed/NCBI

183 

Sakamoto K, Wehde BL, Yoo KH, Kim T, Rajbhandari N, Shin HY, Triplett AA, Rädler PD, Schuler F, Villunger A, et al: Janus kinase 1 is essential for inflammatory cytokine signaling and mammary gland remodeling. Mol Cell Biol. 36:1673–1690. 2016. View Article : Google Scholar : PubMed/NCBI

184 

Poland A and Knutson JC: 2,3,7,8-tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: Examination of the mechanism of toxicity. Annu Rev Pharmacol Toxicol. 22:517–554. 1982. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2024
Volume 29 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen C, Wang Z, Liao Z, Zhang Y, Lei W and Shui X: Aryl hydrocarbon receptor: An emerging player in breast cancer pathogenesis and its potential as a drug target (Review). Mol Med Rep 29: 11, 2024
APA
Chen, C., Wang, Z., Liao, Z., Zhang, Y., Lei, W., & Shui, X. (2024). Aryl hydrocarbon receptor: An emerging player in breast cancer pathogenesis and its potential as a drug target (Review). Molecular Medicine Reports, 29, 11. https://doi.org/10.3892/mmr.2023.13134
MLA
Chen, C., Wang, Z., Liao, Z., Zhang, Y., Lei, W., Shui, X."Aryl hydrocarbon receptor: An emerging player in breast cancer pathogenesis and its potential as a drug target (Review)". Molecular Medicine Reports 29.1 (2024): 11.
Chicago
Chen, C., Wang, Z., Liao, Z., Zhang, Y., Lei, W., Shui, X."Aryl hydrocarbon receptor: An emerging player in breast cancer pathogenesis and its potential as a drug target (Review)". Molecular Medicine Reports 29, no. 1 (2024): 11. https://doi.org/10.3892/mmr.2023.13134