검색
검색 팝업 닫기

Ex) Article Title, Author, Keywords

Article

Split Viewer

Review

DTT 2023; 2(2): 111-123

Published online September 30, 2023

https://doi.org/10.58502/DTT.23.0021

Copyright © The Pharmaceutical Society of Korea.

Toward a Better Understanding of NRF2/NFE2L2 and BCRP/ABCG2 in Therapy Resistance in Cancer

Steffanus Pranoto Hallis1 , Beam Ju Go2, Jun Min Yoo2, Geun Hee Cho2, Mi-Kyoung Kwak1,2

1Department of Pharmacy, Graduate School of The Catholic University of Korea, Bucheon, Korea
2College of Pharmacy, The Catholic University of Korea, Bucheon, Korea

Correspondence to:Mi-Kyoung Kwak, mkwak@catholic.ac.kr

Received: May 9, 2023; Revised: June 7, 2023; Accepted: June 7, 2023

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

The development of therapy resistance in cancer is a complex mechanism with multifaceted and catastrophic consequences. BCRP (Breast Cancer Resistance Protein/ATP-Binding Cassette Subfamily G member 2; ABCG2), a drug efflux transporter, is recognized as a key regulator in obtaining multidrug resistance properties of cancer cells. NRF2 (Nuclear Factor Erythroid 2-Like 2; NFE2L2) plays an essential role in maintaining cellular redox homeostasis by regulating the expression of an array of antioxidant and detoxifying genes. In contrast to its protective roles in normal cells, a substantial body of evidence has demonstrated the negative aspects of NRF2 accumulation in cancer cells, including facilitated tumor growth, stemness, and therapy resistance. This review focuses on the linkage between BCRP and NRF2 in cancer and its consequences in acquiring therapy resistance. In particular, we highlight the role of the NRF2/BCRP axis in photodynamic therapy resistance and cancer stem cell maintenance, demonstrating that inhibition of this axis can be a beneficial strategy to overcome resistance to cancer therapy. A comprehensive understanding of the NRF2/BCRP regulating pathway will lead to the development of new therapeutic approaches and improve the prognosis of cancer patients.

KeywordsBCRP/ABCG2, NRF2/NFE2L2, multidrug resistance, chemoresistance, photodynamic therapy, cancer stem cells

The development of resistance in cancer cells remains a major obstacle to successful chemotherapy, contributing to treatment failure and disease progression. One key mechanism of chemoresistance is the overexpression of ATP-binding cassette (ABC) transporters, which are responsible for the efflux of cytotoxic anticancer agents from cancer cells (Robey et al. 2018). Among 48 human ABC transporters, which are classified into seven subfamilies, multidrug resistance protein 1 (MDR1 or P-glycoprotein/P-gp) encoded by ABC subfamily B member 1 (ABCB1) was initially described in Chinese hamster ovary (CHO) cells and later in sublines of human KB carcinoma cells bearing multidrug-resistance to either colchicine, vinblastine, or doxorubicin (Roninson et al. 1986). The multidrug resistance-associated protein 1 (MRP1, encoded by ABCC1) was identified in 1992 as the second member of multidrug ABC transporters from lung cancer cell line H69AR, a cell line model of doxorubicin, etoposide, and vincristine resistance (Cole et al. 1992). The third member of the multidrug ABC transporter was encoded by ABCG2 and concurrently reported by three groups. The first group identified ABCG2 in human MCF7 breast cancer cells, where it was named breast cancer resistance protein (BCRP) (Doyle et al. 1998). Additionally, ABCG2 was found in mitoxantrone-resistant S1-M1-80 human colon carcinoma cells (Miyake et al. 1999), as well as in placental tissue (Allikmets et al. 1998). The overexpression of multidrug ABC transporters in cancer has been linked to chemotherapy failure in clinical settings. As a result, numerous efforts have been made to overcome multidrug-resistant cancer through the development of transporter inhibitors (Gottesman et al. 2016).

Nuclear factor erythroid 2-like 2 (NFE2L2/NRF2) is a transcription factor that regulates the expression of gene arrays involved in protection against oxidative and electrophilic stress and maintenance of cellular homeostasis (Yamamoto et al. 2018). Despite its protective roles in normal tissues, constitutive activation of NRF2 confers cancer cells survival by multiple mechanisms: increasing drug elimination through phase II metabolizing enzymes, preventing intracellular drug accumulation, and suppressing apoptotic cell death (Bai et al. 2016; Choi and Kwak 2016). In particular, the antioxidant response element (ARE) has been identified as a binding site for NRF2 in the promoter region of MRP1 and BCRP, confirming the critical role of NRF2 signaling in the regulation of multidrug-resistant ABC transporters (Kurz et al. 2001; Singh et al. 2010; Ji et al. 2013). The review article focuses on providing an overview of BCRP and NRF2, and their association with therapy resistance in cancer. In particular, we highlight the role of the NRF2/BCRP axis in cancer stem cells (CSCs) and photodynamic therapy (PDT) resistance. We also suggest that inhibiting this axis can be an effective strategy to overcome resistance to cancer therapy.

Function and regulation of BCRP

BCRP, which is also known as mitoxantrone resistance-associated protein (MXR) or placenta-specific ABC transporter (ABCP), is a type of ABC transporter that belongs to the G subfamily of ABC transporters, also known as ABCG2. BCRP is responsible for translocating its substrates across cell membranes, against a concentration gradient, and this process is achieved through the binding and hydrolysis of ATP (Dean et al. 2001). BCRP is a 72-kDa protein consisting of 665 amino acids with six transmembrane segments and one extracellular loop. Unlike other subfamilies of ABC transporters, which contain two transmembrane domains (TMD) and two nucleotide (ATP)-binding domain (NBD), BCRP is a “half-transporter” with only one TMD and one NBD fused to a single polypeptide chain (Ozvegy et al. 2001; Taylor et al. 2017). Therefore, for BCRP to function properly, it needs to homodimerize, possibly through the formation of intra- and intermolecular disulfide bonds, or oligomerize (Bhatia et al. 2005; Henriksen et al. 2005).

BCRP has diverse physiological roles and is localized in various tissues and organs, including the placenta, mammary glands, gastrointestinal tract, kidney, blood-brain barrier, and hematopoietic stem cells (Robey et al. 2009). BCRP expression in the placenta is essential for protecting the fetus from topotecan, which has been administered in maternal animals. Treatment of pregnant mice with GF120918, a BCRP inhibitor, increased the relative fetal penetration of topotecan (Jonker et al. 2000). In Bcrp1(−/−) knockout mouse model, BCRP absence increased topotecan penetration to the brain by 1.5 folds, while the penetration increased by 12 folds in Mdr1a/b(−/−) Bcrp1(−/−) double knockout model, suggesting the critical role of these transporters in the blood-brain barrier (de Vries et al. 2007). Expression analysis of BCRP mRNA has revealed that its levels decrease along the gastrointestinal tract. The highest expression levels of BCRP mRNA were observed in the duodenum, while levels continuously decreased towards the rectum (Gutmann et al. 2005). The intestinal efflux of quercetin glucuronide, a flavonoid known substrate of BCRP, is impaired in Mrp2-deficient rats treated with BCRP inhibitors. This suggests that BCRP plays a role in facilitating the intestinal uptake of quercetin, potentially by transporting them across the intestinal epithelium. Interestingly, this impairment in quercetin glucuronide efflux was not observed in Mrp2-deficient rats that were not treated with BCRP inhibitors. This suggests that BCRP may compensate for the loss of Mrp2 function in the transport of quercetin glucuronide (Sesink et al. 2005). BCRP is also found to be localized in the apical membrane of human kidney proximal tubules, where it is expressed alongside other ABC transporters, contributing to the excretion of drugs and xenobiotics in the urine (Huls et al. 2008). BCRP has also been identified as a marker of hematopoietic stem cells, due to its high expression in primitive murine stem cells and sharp decrease with differentiation (Zhou et al. 2001).

The human ABCG2 gene is located on chromosome 4, specifically in the 4q21-q22 region. It spans over 66 kb and consists of 16 exons and 15 introns (Bailey-Dell et al. 2001). Transcriptional regulation studies of ABCG2 have identified several functional cis-elements within its promoter region that are recognized by several transcription factors, including estrogen (Ee et al. 2004), progesterone (Wang et al. 2008), peroxisome proliferator-activated receptor γ (Szatmari et al. 2006), hypoxia-inducible factor (HIF)-1α (Krishnamurthy et al. 2004), HIF-2α (He et al. 2019), and NRF2 (Singh et al. 2010) (Fig. 1). At the post-transcriptional modification level, alternative splicing of the ABCG2 gene has been reported to generate three splice variants of the untranslated exon 1 in the 5’ untranslated region (UTR) of the mRNA. These splice variants can serve as alternative promoters and result in the expression of isoforms of the protein with altered function and localization (Nakanishi et al. 2006). In addition, microRNAs (miRNAs) have been suggested to be involved in BCRP regulation. Rapid amplification of cDNA ends (RACE) analysis of the 3’ UTR of ABCG2 mRNA from drug-resistant S1 colon cancer cells identified several putative miRNA binding sites (To et al. 2008). MiR-328 has been reported to directly regulate ABCG2 in MCF-7 breast cancer cells and re-sensitize the cells to mitoxantrone (Pan et al. 2009). Additionally, expression of miR-520 h inhibited migration, invasion, and side populations of pancreatic cancer cells (Wang et al. 2010).

Figure 1.BCRP regulation and its association with chemoresistance. Overactivation of NRF2 in cancer increases BCRP (ABCG2) as one of its targets, as well as other targets in antioxidant defense, metabolism, and drug detoxification, thereby promoting cell survival and resistance to therapy. The transcriptional regulation of BCRP can be controlled by additional transcription factors, including HIF-1α, HIF-2α, PPARγ, and estrogen, which can bind to their enhancer elements in the promoter sequence of BCRP genes. At the post-transcriptional level, miR-328 and miR-520h can directly suppress BCRP transcription, while NRF2-regulated miR-206 targets BCRP via the c-MET/EGFR signaling pathway. sMAFs, small MAF proteins; BCRP, breast cancer resistance protein; PPARγ, peroxisome proliferator-activated receptor-γ; HIF, hypoxia-inducible factor; ARE, antioxidant response element; PPRE, peroxisome proliferator-response element; HRE, hypoxia response element; ERE, estrogen response element; EGFR, epidermal growth factor receptor; c-MET, mesenchymal-epithelial transition factor.

BCRP and chemoresistance of cancer

ABC transporters work together to mediate the efflux of various substrates, including anticancer agents such as anthracyclines, camptothecins, taxanes, and tyrosine kinase inhibitors (Robey et al. 2018). It is also well-established that BCRP plays a major role in multidrug resistance of cancer because of its distinct action. While MRP1 selectively transports products of phase II drug metabolism, BCRP transports anticancer drugs, such as irinotecan, methotrexate, mitoxantrone, and topotecan (Mao and Unadkat 2015). Histology data indicates that BCRP is uniformly expressed in all subsets of tumors, with frequent expression observed in adenocarcinomas of the gastrointestinal tract, endometrium, lung, and melanoma (Diestra et al. 2002). The Cancer Genome Atlas (TCGA) database analysis of RNA sequencing data showed that hepatocellular carcinoma and kidney cancer, both of which are frequently resistant to vincristine and doxorubicin, expressed the highest levels of BCRP compared to other cancer types (Robey et al. 2018).

In vitro studies using cancer cell lines from various solid tumors, including breast, colon, gastric, glioma, lung, and ovarian cancer, have identified high expression of BCRP in multidrug resistance models (Maliepaard et al. 1999; Ross et al. 1999; Martin et al. 2009). An earlier study using drug-selected cancer cell lines showed that mitoxantrone, doxorubicin, and verapamil increased the expression of BCRP mRNA in human breast carcinoma (MCF-7), colon carcinoma (HT29), gastric carcinoma (EPG85-257), fibrosarcoma (EPF86-079), and myeloma (8226) (Ross et al. 1999). The human IGROV1 ovarian cancer cell line, which has developed resistance to the chemotherapy drugs topotecan and mitoxantrone following continuous exposure, expresses high levels of BCRP (Maliepaard et al. 1999). In a panel of glioma cell lines and brain tumor stem cells, high expression levels of BCRP were linked to the overexpression of the Tie2 receptor, which is associated with tumor malignancy, and BCRP downregulation decreased Tie2 expression (Martin et al. 2009). In SN-38-resistant PC-6 human small-lung cancer cell lines, which had acquired resistance to the active metabolites of irinotecan and topotecan, there was a reduction in the intracellular accumulation of SN-38. This reduction was found to be associated with the overexpression of BCRP, and suppression of BCRP enhanced the sensitivity of these cells to SN-38 (Kawabata et al. 2001). A pharmacogenetic study with 299 renal carcinoma patients in the Japanese population found that differences in the BCRP genotype (the ABCG2 C421A [Q141K] polymorphism) were associated with reduced adverse effects of sunitinib, without affecting its efficacy (Low et al. 2016).

There has been consistent clinical evidence for the relationship between BCRP overexpression and drug resistance in hematologic malignancies. A gene profiling study of older acute myeloid leukemia (AML) patients found that 77% of 170 AML samples were chemoresistant and characterized by high expressions of BCRP and MDR1 (Wilson et al. 2006). A comparison between sensitive and resistant AML patients showed a significant difference in BCRP expression, and that this difference was linked to overall patients’ survival (Marzac et al. 2011). Moreover, high levels of BCRP were identified in a population of putative leukemic stem cells (CD34+CD38) from AML patients. BCRP inhibition in these leukemic stem cells using Ko143 resulted in increased cellular levels of mitoxantrone accumulation (Raaijmakers et al. 2005). Thus, these preclinical and clinical datasets suggest that the inhibition of BCRP in multidrug-resistant AML may potentially lead to better treatment outcomes and patient survival.

BCRP inhibitors

Given the critical role of BCRP in the acquisition of chemoresistance, extensive efforts have been dedicated to discovering and developing BCRP inhibitors (Doyle and Ross 2003). The first BCRP inhibitor to be reported was Fumitremorgin C (FTC), a secondary metabolite from Aspergillus fumigatus. FTC has been shown to effectively reverse the multidrug resistance of colon carcinoma to mitoxantrone, doxorubicin, bisantrene, and topotecan through the specific inhibition of BCRP (Rabindran et al. 1998). Subsequent studies have confirmed the inhibitory effect of FTC on BCRP in various cancer cell lines, including SF295 human glioblastoma, KM12 colon cancer, and A549 non-small-cell lung cancer cell lines (Robey et al. 2001a). FTC has also been found to inhibit the transport of methotrexate in human embryonic kidney cells (Chen et al. 2003). At a concentration of 5 µM, FTC has been shown to effectively re-sensitize the multi-drug resistant MCF-7 FLV1000 cell line to mitoxantrone, SN-38, and topotecan by inhibiting the efflux of these drugs via BCRP (Robey et al. 2001b). Later, a tetracycline analog of FTC called Ko143 was identified as a more selective inhibitor of BCRP. In an experiment using mouse intestine, Ko143 selectively inhibited BCRP without affecting the function of other ABC transporters such as P-gp or MRP-1 at a concentration of 100 nM (Allen et al. 2002).

Elacridar (GF120918), which was originally developed as a P-gp inhibitor, is an acridone carboxamide and identified as a potent inhibitor of BCRP. Elacridar has been shown to effectively inhibit the efflux of mitoxantrone and topotecan (de Bruin et al. 1999). Additionally, flavonoid metabolites 6-prenylchrysin and tectochrysin have a specific inhibitory effect on BCRP. In a study conducted on BCRP-expressing cancer cells, the application of 0.5 µmol/L of 6-prenylchrysin or 1 µmol/L of tectochrysin was sufficient to sensitize the cells to mitoxantrone, with less toxicity compared to elacridar (Ahmed-Belkacem et al. 2005).

Several anti-retroviral drugs, including lopinavir, nelfinavir, and delavirdine have been shown to inhibit BCRP and increase the cellular accumulation of pheophorbide A (Pha), a photosensitizer substrate of BCRP, in canine kidney epithelial cell lines (Weiss et al. 2007). Additionally, statins such as atorvastatin and rosuvastatin were shown to inhibit BCRP in the biliary excretion system using Bcrp(−/−) mice, although some statins are substrates of BCRP (Hirano et al. 2005). Moreover, since estrogen can regulate BCRP expression and therefore, estrogen agonists and antagonists have been found to modulate BCRP activity. Diethylstilbestrol, an estrogen agonist, increased the accumulation of topotecan and reversed the chemoresistance of the BCRP-transduced K562 cell line. In contrast, tamoxifen, an estrogen antagonist, has been found to only enhance the uptake of topotecan (Sugimoto et al. 2003).

The NRF2-KEAP1 system

NRF2 is a cap‘n’collar (CNC) basic leucine zipper (bZIP) transcription factor that regulates cytoprotective genes and maintains cellular redox homeostasis. Although NRF2 is expressed in all cell types, its basal level is mainly low due to negative regulation by KEAP1-mediated proteasomal degradation (Yamamoto et al. 2018). The NRF2 protein is composed of six functional domains, known as NRF2-ECH Homology (Neh) 1-6, each of which plays a specific role in NRF2 activity. Neh1 primarily contains the CNC-bZIP domains that facilitate DNA binding and dimerization, while Neh3-5 serve as transactivation domains. In particular, Neh4 and Neh5 recruit the histone acetyl-transferase cAMP-responsive element binding protein (CBP) and mediator complex to facilitate the transcriptional activity of NRF2 (Yamamoto et al. 2018). Meanwhile, Neh2 and Neh6 primarily regulate NRF2 degradation. Neh2 contains DLG and ETGE motifs that serve as distinct and prominent binding sites for the KEAP1 protein, while Neh6 functions as a KEAP1-independent degron site of NRF2. Neh6 facilitates NRF2 degradation through the binding of β-TRCP (Tong et al. 2006; Chowdhry et al. 2013).

Under normal conditions, two KEAP1 molecules and one NRF2 molecule form a structure that accelerates the ubiquitination of NRF2. KEAP1 acts as an adaptor, forming cullin3 (CUL3)-based E3 ubiquitin ligases and RBX1 to create a functional E3 ubiquitin ligase (Kobayashi et al. 2004; McMahon et al. 2006). The KEAP1-CUL3 complex recognizes the DLG and ETGE motifs in the Neh2 domain of NRF2 and ubiquitinates it, ensuring a low level of NRF2 by subjecting it to proteasomal degradation. However, following exposure to electrophiles or reactive oxygen species (ROS), the cysteine residues of KEAP1 undergo modification, forming disulfide bonds or covalent linkages due to oxidation, which results in the alteration of the conformational structure of the KEAP1 (Sekhar et al. 2010). The disruption of the DLG-KEAP1 binding allows for the accumulation of newly synthesized NRF2, which can be translocated into the nucleus. Then, NRF2 can dimerize with small Maf proteins and activate transcription by recognizing and binding to the ARE in the promoter of its target genes.

The direct correlation between NRF2 and target genes through ARE was first reported by Itoh et al. (1997). They observed a marked decrease in the expression of phase II enzymes, including glutathione S-transferases (GSTs) and NAD(P)H quinone oxidoreductase 1 (NQO1), in butylated hydroxyanisole (BHA)-treated Nrf2-null mice. Since then, NRF2 has been identified as a master regulator of genes involved in phase I, II, and III metabolism. These include phase I enzymes such as Cyp2a5 and Cyp2b10 (Ashino et al. 2014), phase II enzymes such as GSTs (GSTA3, GSTA4, GSTM1, GSTM2, GSTM3, GSTM4, GSTM6), NQO1 (Hirotsu et al. 2012), and aldo-keto reductase family 1 member C1 (AKR1C1) (Jung et al. 2013), and phase III transporters, including MRPs (ABCC1, ABCC2, ABCC3, ABCC4) (Stöckel et al. 2000; Mahaffey et al. 2009; Xu et al. 2010; Ji et al. 2013) and BCRP (ABCG2) (Singh et al. 2010).

NRF2 and cancer

While NRF2 activity is crucial in normal unstressed conditions, persistent activation of NRF2 has been found to contribute to impairing the efficacy of chemotherapy in cancer (Rojo de la Vega et al. 2018). The hyperactivation of NRF2 in cancer can be resulted from somatic mutations or genomic alterations in NRF2 and KEAP1 genes or the expression of KEAP1-competing proteins such as p62/SQSTM1 (Kitamura and Motohashi 2018). In non-small-cell lung cancer patients and cell lines, genomic domain mutation and high percentage loss of heterozygosity at 19p13.2 of KEAP1 results in the specific abrogation of the NRF2 repressor system (Singh et al. 2006). Thus, NRF2 is accumulated and enhances the transcriptional activation of genes involved in xenobiotic detoxification and drug efflux pumps. Moreover, mutations in either the IVR or DGR region of KEAP1, or in the DLG or ETGE motifs of NRF2, have consequences for the constitutive expression of cytoprotective enzymes induced by NRF2 and can facilitate tumor malignancies (Ohta et al. 2008; Shibata et al. 2008). The genomic alteration of exon 2 deletions in NRF2 results in persistent NRF2 activation in lung cancers and head and neck carcinomas (Goldstein et al. 2016). KEAP1 gene methylation leads to reduced expression of KEAP1 and activation of NRF2 and its corresponding targets, such as NQO1 and AKR1C1, during the carcinogenesis and cancer progression (Hanada et al. 2012; Barbano et al. 2013). One well-established KEAP1-competing protein that has been identified as responsible for NRF2 accumulation in cancer cells is the autophagy adaptor protein p62/SQSTM1. Its phosphorylation at the S349 residue by the mammalian target of rapamycin complex 1 (mTORC1) increases the binding affinity to KEAP1, subsequently disrupting the DLG-mediated KEAP1-NRF2 interaction (Ichimura and Komatsu 2018). Pharmacological inhibition of phosphorylated p62/SQSTM1 and KEAP1 interaction by K67 resulted in the suppression of NRF2 activity, which inhibited growth and re-sensitized the tumors to anti-cancer agents (Saito et al. 2016).

The high expression of NRF2, which has been observed in multiple types of cancers, contributes to chemoresistance by inducing downstream genes involved in electrophiles/ROS detoxification and the efflux of cytotoxic anticancer drugs (Srivastava et al. 2022). A recent report demonstrated that the activation of NRF2 by FAM117B-mediated KEAP1 competitive binding in gastric cancer facilitated tumor growth and chemoresistance to 5-fluorouracil and oxaliplatin (Zhou et al. 2023). In pancreatic cancer, oncogenic KRAS upregulated NRF2-dependent metabolic rewiring of glutamine metabolism, which leads to gemcitabine resistance (Mukhopadhyay et al. 2020). Keap1-deficient KrasG12D mice model developed lung tumors with hyperactivation of NRF2 and subsequent activation of the pentose phosphate pathway (PPP) (Best et al. 2019). Similarly, in KrasG12D–induced lung cancer, constitutive KRAS activation increased NRF2 transcription and resulted in cisplatin resistance. In this context, treatment with the NRF2 inhibitor brusatol was found to enhance the anti-tumor efficacy of cisplatin in KrasG12D-induced lung cancer (Tao et al. 2014).

BCRP regulation by NRF2

Several studies have shown a direct correlation between NRF2 and BCRP in cancer. An early report demonstrated that treatment with oltipraz, an activator of NRF2, increased the mRNA level of BCRP in cultured human hepatocytes (Jigorel et al. 2006). This observation was further confirmed by a study that used tert-butylhydroquinone (tBHQ), another potent activator of NRF2, in HepG2 cells (Adachi et al. 2007). In this study, treatment of HepG2 cells with tBHQ induced translocation of NRF2 to the nucleus, and concomitantly increased the mRNA level of BCRP, as well as other ABC transporters and NRF2 targets, such as the heavy and light chains of γ-glutamylcysteine synthase. The decrease and increase in expression of BCRP mRNA mediated by NRF2- and KEAP1-silencing, respectively, suggest the involvement of the NRF2/KEAP1 system in the transcriptional regulation of BCRP (Adachi et al. 2007).

An advanced study by Singh et al. identified the ARE region located in the 5’-flanking region of the BCRP gene at -431 to -420 bp, which is responsible for NRF2-mediated expression in lung cancer cells in vitro and in vivo (Singh et al. 2010) (Fig. 1). This observation confirms previous findings of ARE located in the promoter regions of other multidrug ABC transporters, such as MRP1-3 (Stöckel et al. 2000; Kurz et al. 2001; Mahaffey et al. 2009). Similarly, in normal kidney tubular cells, knockdown of KEAP1 was found to increase BCRP expression. However, unlike in the previous studies where NRF2 activation increased BCRP expression, pharmacological activation of NRF2 did not elevate BCRP mRNA in this case (Jeong et al. 2015). In addition to transcriptional regulation, NRF2 has been reported to regulate BCRP through a post-transcriptional mechanism as well. For example, in ovarian and renal carcinoma cell lines, silencing of NRF2 led to the activation of miR-206, which targeted c-MET/EGFR and downregulated BCRP expression, resulting in increased intracellular accumulation of the anticancer drug doxorubicin (Singh et al. 2013; Choi et al. 2017).

NRF2/BCRP axis and PDT

PDT is an emerging chemotherapy strategy that utilizes photosensitizing drugs to selectively kill cancer cells by generating cytotoxic ROS upon light activation (Agostinis et al. 2011). PDT has gained increasing attention as a minimally invasive and more target-specific alternative to conventional chemotherapy (Dolmans et al. 2003; Correia et al. 2021). PDT has been approved by the FDA for the treatment of several types of cancer, including bladder, esophageal, head and neck, lung, and skin cancer (Correia et al. 2021). The expression of BCRP in cancer cells can limit the effectiveness of PDT, and therefore there has been interest in investigating the relationship between BCRP and PDT. One study used imatinib mesylate, a tyrosine kinase inhibitor (TKI) known to reduce BCRP levels, and found that it enhanced the accumulation of various photosensitizers, including 2-(1-hexyloxethyl)-2-devinyl pyropheophorbide-a (Photochlor), protoporphyrin IX, and the benzoporphyrin derivative monoacid ring A (Verteporfin), in ABCG+ human and murine cell lines both in vitro and in vivo (Liu et al. 2007). Another study showed that BCRP-silencing using siRNA or inhibition of its activity using a chemical inhibitor, resulted in increased intracellular accumulation of chlorin e6 and improved PDT efficacy in cancer cells. This suggests that targeting BCRP may be a promising strategy to enhance the efficacy of PDT in cancer therapy (Roh et al. 2017). In the doxorubicin-resistant ovarian cancer A2780DR cells, the increase of BCRP-mediated drug resistance is associated with c-MET elevation. Treating the A2780DR cells with BCRP inhibitor Ko143 or c-MET inhibitor resulted in the accumulation of the photosensitizer Pba within cancer cells, which in turn sensitized the cells to PDT (Jung et al. 2015). Another study showed that A431/BCRP lung cancer cells, which overexpress BCRP, are more resistant to Photofrin-mediated PDT. However, inhibition of BCRP with FTC reversed this resistance and increased the efficacy of PDT (Usuda et al. 2010). In line with this, NRF2-knockdown in several types of cancer cells, including triple-negative breast cancer, renal carcinoma, and colon cancer, led to an increase in the effectiveness of PDT by lowering the levels of BCRP (Choi et al. 2014). Furthermore, a recent report has demonstrated that nanoparticle-mediated NRF2 silencing can increase the sensitivity of tumor cells to oxidative stress during PDT. These findings further support the importance of the NRF2/BCRP pathway in the response of cancer cells to PDT (Sun et al. 2023).

NRF2/BCRP axis and CSC properties

Ever since the initial discovery of a subpopulation of leukemic stem cells characterized by the surface markers CD34+ and CD38, CSCs have been recognized as promising targets for cancer therapy (Lapidot et al. 1994). The unique characteristics of CSCs, including their ability of self-renewal and differentiation potential, have been shown to play a critical role in cancer malignancies such as metastasis, therapy resistance, and recurrence (Batlle and Clevers 2017). CSCs and normal stem cells share many common transcription factors and signaling pathways, including several pluripotent transcription factors such as octamer-binding transcription factor 4 (OCT4), krupple-like factor 4 (KLF4), MYC, homeobox protein Nanog (NANOG), and SRY (sex determining region Y)-box 2 (SOX2) (Yang et al. 2020). Furthermore, a subpopulation of cancer cells known as the side population has been identified as a chemo- and radio-therapy-resistant population, which is thought to be due to its high expression of BCRP. As a result, BCRP has become recognized as one of the CSC markers (Zhou et al. 2001).

Recent studies have suggested that NRF2 may contribute to the development or maintenance of CSC properties, including chemoresistance (Kahroba et al. 2019; Choi et al. 2021; Hallis et al. 2023a). In the colonosphere model of in vitro CSCs, increased levels of P-gp, BCRP, and concomitant NRF2 signaling were observed, which exhibited resistance to doxorubicin (Ryoo et al. 2016). Subsequently, NRF2 knockdown in the colonosphere model sensitized these cells to drug treatment by decreasing efflux transporter activities. Consistent with these findings, the CD133high population from HCT116 colon cancer cells exhibited high levels of NRF2 and BCRP, along with elevated CSC markers, indicating that they possess CSC phenotypes such as enhanced proliferation, migration, colony formation, chemoresistance, and sphere-forming capacity (Park et al. 2022). In this experimental setting, colonospheres derived from NRF2-silenced HCT116 expressed low levels of BCRP along with reduced KLF4 expression, which resulted in the suppression of sphere-forming capacity. A similar study involving NRF2 knockdown in pancreatic ductal adenocarcinoma cell lines BxPC-3 and CFPAC-1, which had high expression of NRF2, found that it decreased the expression of CSC transcription factors NANOG, OCT4, and CD133, as well as chemoresistance properties, by downregulating BCRP (Kim et al. 2020). Silencing of NRF2 in sorafenib-resistant Huh7 hepatocellular cell line also limited cancer stemness and migration, as well as expression of BCRP (Gao et al. 2021). In another drug resistance model, A2780DR ovarian cancer cells that exhibit doxorubicin resistance showed higher NRF2 signaling activity concomitantly with elevated expression of aldehyde dehydrogenase 1 (ALDH1) and BCRP, which supports to maintain CSC properties (Shim et al. 2009; Kim et al. 2018). Isolated CSCs from the side population of cervical cancer patients showed aberrant activation of NRF2, BCRP, and Bcl-2, which resulted in chemoresistance and apoptosis resistance (Jia et al. 2015). Meanwhile, our recent study demonstrated a positive association between NRF2 and HIF-2α-induced CSC phenotypes under chronic hypoxic conditions (Hallis et al. 2023b). As previous studies have reported that HIF-2α directly regulates BCRP and confers CSC properties in ovarian CSCs, leading to resistance to doxorubicin and promoting tumor growth, this may suggest the potential association of NRF2 with HIF-2α/BCRP in hypoxic tumor environment (He et al. 2019). Collectively, these studies indicate that NRF2 and BCRP are intricately intertwined in maintaining resistance to chemotherapeutic agents and photodynamic therapy, and the maintenance of CSC properties in cancer (Fig. 2).

Figure 2.Role of NRF2 and BCRP in CSC maintenance and PDT resistance. (A) NRF2 signaling is activated in CSCs models, contributing to CSC properties, including tumor initiation and growth, invasion, metastasis, and therapy resistance. NRF2 upregulates the antioxidants gene to maintain low ROS levels and increases ABC transporters such as BCRP to protect CSCs against external harmful chemicals, which eventually leads to therapy resistance. High levels of NRF2 are also associated with the upregulation of other transcription factors, including OCT4, NANOG, KLF4, MYC, and SOX2, which help to maintain CSC phenotypes. (B) Enhanced BCRP in cancers facilitates the efflux of photosensitizers such as Pba, photochlor, and photofirin, thus leading to PDT resistance. BCRP inhibition by chemical inhibitors increases intracellular accumulation of photosensitizers and consequent ROS generation, leading to cancer cell apoptosis. OCT4, octamer-binding transcription factor 4; KLF4, krupple-like factor 4; NANOG, homeobox protein nanog; SOX2, SRY (sex determining region Y)-box 2; FTC, fumitremorgin C.

In this review, we have highlighted the critical role of NRF2 and BCRP as potential targets to counteract the therapy resistance of cancers. Elevated expression of BCRP in most cancer cases results in a poor prognosis and is associated with multidrug resistance, thereby impeding the efficacy of cancer therapy (Robey et al. 2018). Many studies have demonstrated the potential effect of BCRP inhibitors in multiple models of resistant cancers, and current efforts are focusing on investigating BCRP inhibitors for clinical trials. Thus, the regulation of BCRP by transcription factors or other signaling pathways is of great interest. Although several transcription factors have been implicated, the identification of ARE sequences in the promoter region of BCRP has highlighted the direct regulation by NRF2 (Singh et al. 2010). This is important since NRF2 overexpression in cancer clinical settings is correlated with poor prognosis and chemoresistance (Solis et al. 2010). Moreover, recent insights suggest that the suppression of BCRP and its regulator NRF2 may have an impact on PDT resistance and the maintenance of CSC properties. Thus, ongoing efforts are focused on strategies aimed at suppressing NRF2 activity to increase the sensitivity of cancer patients to chemotherapy (Dinkova-Kostova and Copple 2023). In particular, compared to the inhibition of BCRP, the inhibition of NRF2 is expected to be a more effective strategy for cancer treatment in that it can suppress the expression of other target genes that contribute to cancer growth and development as well as control of BCRP-mediated anticancer drug resistance. Currently, extensive efforts are being made to develop NRF2 inhibitors. Several natural compounds, including luteolin, procyanidins, and wedelolactone, have demonstrated NRF2 inhibitory effects in various experimental settings involving NRF2-overactive cancers (Sharifi-Rad et al. 2023). Brusatol from Brucea javanica has shown potent anti-tumor and anti-metastatic activities by decreasing NRF2 protein levels, which is attributed to multiple mechanisms, including protein translation inhibition (He et al. 2023). In a high-throughput chemical library screening, halofuginone has been identified as an inhibitor of NRF2 protein synthesis (Tsuchida et al. 2017). ML385, derived from a high-throughput chemical library screening and subsequent chemical optimization, has been discovered to interfere with the interaction between NRF2 and sMAF by directly binding to the NRF2 protein. Consequently, ML385 demonstrated the ability to sensitize KEAP1 mutant lung cancer cells to chemotherapy (Singh et al. 2016). While NRF2 inhibitors hold strong potential for enhancing chemotherapy sensitivity and suppressing tumorigenesis in NRF2-high cancers, the dual functionality of NRF2 in normal cells presents a limitation for its clinical application. In this context, achieving cancer-selective inhibition of NRF2 becomes a crucial strategy. In a recent study employing a synthetic lethal strategy, mitomycin C has emerged as a selectively cytotoxic agent for NRF2-overactive cancer cells, primarily through enhanced bioactivation via NRF2-target genes, such as NQO1 (Baird and Yamamoto 2021). Geldanamycin-derived HSP90 inhibitors have demonstrated selective lethality towards NRF2-high cancer cells, and their mechanism of action has been attributed to the enhancement of NQO1-mediated bioactivation (Baird et al. 2020). In summary, although a strategy for NRF2 inhibitors that selectively act on NRF2-overexpressing cancers has not been developed, with the progression of more advanced studies on the NRF2-KEAP1 system, there is an optimistic expectation for the future discovery of highly selective and effective NRF2/BCRP inhibitors.

This work was supported by National Research Foundation of Korea (NRF) grants funded by the Korean government (MSIT) (2022R1A2C2011866).

The authors declare that they have no conflict of interest.

  1. Adachi T, Nakagawa H, Chung I, Hagiya Y, Hoshijima K, Noguchi N, Kuo MT, Ishikawa T (2007) Nrf2-dependent and -independent induction of ABC transporters ABCC1, ABCC2, and ABCG2 in HepG2 cells under oxidative stress. J Exp Ther Oncol 6:335-348.
    Pubmed
  2. Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, Hahn SM, Hamblin MR, Juzeniene A, Kessel D, Korbelik M, Moan J, Mroz P, Nowis D, Piette J, Wilson BC, Golab J (2011) Photodynamic therapy of cancer: an update. CA Cancer J Clin 61:250-281. doi: 10.3322/caac.20114
    Pubmed KoreaMed CrossRef
  3. Ahmed-Belkacem A, Pozza A, Muñoz-Martínez F, Bates SE, Castanys S, Gamarro F, Di Pietro A, Pérez-Victoria JM (2005) Flavonoid structure-activity studies identify 6-prenylchrysin and tectochrysin as potent and specific inhibitors of breast cancer resistance protein ABCG2. Cancer Res 65:4852-4860. doi: 10.1158/0008-5472.CAN-04-1817
    Pubmed CrossRef
  4. Allen JD, van Loevezijn A, Lakhai JM, van der Valk M, van Tellingen O, Reid G, Schellens JH, Koomen GJ, Schinkel AH (2002) Potent and specific inhibition of the breast cancer resistance protein multidrug transporter in vitro and in mouse intestine by a novel analogue of fumitremorgin C. Mol Cancer Ther 1:417-425.
    Pubmed
  5. Allikmets R, Schriml LM, Hutchinson A, Romano-Spica V, Dean M (1998) A human placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res 58:5337-5339.
    Pubmed
  6. Ashino T, Ohkubo-Morita H, Yamamoto M, Yoshida T, Numazawa S (2014) Possible involvement of nuclear factor erythroid 2-related factor 2 in the gene expression of Cyp2b10 and Cyp2a5. Redox Biol 2:284-288. doi: 10.1016/j.redox.2013.12.025
    Pubmed KoreaMed CrossRef
  7. Bai X, Chen Y, Hou X, Huang M, Jin J (2016) Emerging role of NRF2 in chemoresistance by regulating drug-metabolizing enzymes and efflux transporters. Drug Metab Rev 48:541-567. doi: 10.1080/03602532.2016.1197239
    Pubmed CrossRef
  8. Bailey-Dell KJ, Hassel B, Doyle LA, Ross DD (2001) Promoter characterization and genomic organization of the human breast cancer resistance protein (ATP-binding cassette transporter G2) gene. Biochim Biophys Acta 1520:234-241. doi: 10.1016/s0167-4781(01)00270-6
    Pubmed CrossRef
  9. Baird L, Suzuki T, Takahashi Y, Hishinuma E, Saigusa D, Yamamoto M (2020) Geldanamycin-derived HSP90 inhibitors are synthetic lethal with NRF2. Mol Cell Biol 40:e00377-e00320. doi: 10.1128/MCB.00377-20
    Pubmed KoreaMed CrossRef
  10. Baird L, Yamamoto M (2021) NRF2-dependent bioactivation of mitomycin C as a novel strategy to target Keap1-NRF2 pathway activation in human cancer. Mol Cell Biol 41:e00473-e00420. doi: 10.1128/MCB.00473-20
    Pubmed KoreaMed CrossRef
  11. Barbano R, Muscarella LA, Pasculli B, Valori VM, Fontana A, Coco M, la Torre A, Balsamo T, Poeta ML, Marangi GF, Maiello E, Castelvetere M, Pellegrini F, Murgo R, Fazio VM, Parrella P (2013) Aberrant Keap1 methylation in breast cancer and association with clinicopathological features. Epigenetics 8:105-112. doi: 10.4161/epi.23319
    Pubmed KoreaMed CrossRef
  12. Batlle E, Clevers H (2017) Cancer stem cells revisited. Nat Med 23:1124-1134. doi: 10.1038/nm.4409
    Pubmed CrossRef
  13. Best SA, Ding S, Kersbergen A, Dong X, Song JY, Xie Y, Reljic B, Li K, Vince JE, Rathi V, Wright GM, Ritchie ME, Sutherland KD (2019) Distinct initiating events underpin the immune and metabolic heterogeneity of KRAS-mutant lung adenocarcinoma. Nat Commun 10:4190. doi: 10.1038/s41467-019-12164-y
    Pubmed KoreaMed CrossRef
  14. Bhatia A, Schäfer HJ, Hrycyna CA (2005) Oligomerization of the human ABC transporter ABCG2: evaluation of the native protein and chimeric dimers. Biochemistry 44:10893-10904. doi: 10.1021/bi0503807
    Pubmed CrossRef
  15. Chen ZS, Robey RW, Belinsky MG, Shchaveleva I, Ren XQ, Sugimoto Y, Ross DD, Bates SE, Kruh GD (2003) Transport of methotrexate, methotrexate polyglutamates, and 17beta-estradiol 17-(beta-D-glucuronide) by ABCG2: effects of acquired mutations at R482 on methotrexate transport. Cancer Res 63:4048-4054.
    Pubmed
  16. Choi BH, Kim JM, Kwak MK (2021) The multifaceted role of NRF2 in cancer progression and cancer stem cells maintenance. Arch Pharm Res 44:263-280. doi: 10.1007/s12272-021-01316-8
    Pubmed CrossRef
  17. Choi B, Kwak MK (2016) Shadows of NRF2 in cancer: resistance to chemotherapy. Curr Opin Toxicol 1:20-28. doi: 10.1016/j.cotox.2016.08.003
    CrossRef
  18. Choi BH, Ryoo IG, Kang HC, Kwak MK (2014) The sensitivity of cancer cells to pheophorbide a-based photodynamic therapy is enhanced by Nrf2 silencing. PLoS One 9:e107158. doi: 10.1371/journal.pone.0107158
    Pubmed KoreaMed CrossRef
  19. Choi BH, Ryu DY, Ryoo IG, Kwak MK (2017) NFE2L2/NRF2 silencing-inducible miR-206 targets c-MET/EGFR and suppresses BCRP/ABCG2 in cancer cells. Oncotarget 8:107188-107205. doi: 10.18632/oncotarget.22513
    Pubmed KoreaMed CrossRef
  20. Chowdhry S, Zhang Y, McMahon M, Sutherland C, Cuadrado A, Hayes JD (2013) Nrf2 is controlled by two distinct β-TrCP recognition motifs in its Neh6 domain, one of which can be modulated by GSK-3 activity. Oncogene 2:3765-3781. doi: 10.1038/onc.2012.388
    Pubmed KoreaMed CrossRef
  21. Cole SP, Bhardwaj G, Gerlach JH, Mackie JE, Grant CE, Almquist KC, Stewart AJ, Kurz EU, Duncan AM, Deeley RG (1992) Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science 258:1650-1654. doi: 10.1126/science.1360704
    Pubmed CrossRef
  22. Correia JH, Rodrigues JA, Pimenta S, Dong T, Yang Z (2021) Photodynamic therapy review: principles, photosensitizers, applications, and future directions. Pharmaceutics 13:1332. doi: 10.3390/pharmaceutics13091332
    Pubmed KoreaMed CrossRef
  23. de Bruin M, Miyake K, Litman T, Robey R, Bates SE (1999) Reversal of resistance by GF120918 in cell lines expressing the ABC half-transporter, MXR. Cancer Lett 146:117-126. doi: 10.1016/s0304-3835(99)00182-2
    Pubmed CrossRef
  24. de Vries NA, Zhao J, Kroon E, Buckle T, Beijnen JH, van Tellingen O (2007) P-glycoprotein and breast cancer resistance protein: two dominant transporters working together in limiting the brain penetration of topotecan. Clin Cancer Res 13:6440-6449. doi: 10.1158/1078-0432.CCR-07-1335
    Pubmed CrossRef
  25. Dean M, Rzhetsky A, Allikmets R (2001) The human ATP-binding cassette (ABC) transporter superfamily. Genome Res 11:1156-1166. doi: 10.1101/gr.184901
    Pubmed CrossRef
  26. Diestra JE, Scheffer GL, Català I, Maliepaard M, Schellens JH, Scheper RJ, Germà-Lluch JR, Izquierdo MA (2002) Frequent expression of the multi-drug resistance-associated protein BCRP/MXR/ABCP/ABCG2 in human tumours detected by the BXP-21 monoclonal antibody in paraffin-embedded material. J Pathol 198:213-219. doi: 10.1002/path.1203
    Pubmed CrossRef
  27. Dinkova-Kostova AT, Copple IM (2023) Advances and challenges in therapeutic targeting of NRF2. Trends Pharmacol Sci 44:137-149. doi: 10.1016/j.tips.2022.12.003
    Pubmed CrossRef
  28. Dolmans DE, Fukumura D, Jain RK (2003) Photodynamic therapy for cancer. Nat Rev Cancer 3:380-387. doi: 10.1038/nrc1071
    Pubmed CrossRef
  29. Doyle L, Ross DD (2003) Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2). Oncogene 22:7340-7358. doi: 10.1038/sj.onc.1206938
    Pubmed CrossRef
  30. Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, Ross DD (1998) A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A 95:15665-15670. doi: 10.1073/pnas.95.26.15665
    Pubmed KoreaMed CrossRef
  31. Ee PL, Kamalakaran S, Tonetti D, He X, Ross DD, Beck WT (2004) Identification of a novel estrogen response element in the breast cancer resistance protein (ABCG2) gene. Cancer Res 64:1247-1251. doi: 10.1158/0008-5472.can-03-3583
    Pubmed CrossRef
  32. Gao L, Morine Y, Yamada S, Saito Y, Ikemoto T, Tokuda K, Takasu C, Miyazaki K, Shimada M (2021) Nrf2 signaling promotes cancer stemness, migration, and expression of ABC transporter genes in sorafenib-resistant hepatocellular carcinoma cells. PLoS One 16:e0256755. doi: 10.1371/journal.pone.0256755
    Pubmed KoreaMed CrossRef
  33. Goldstein LD, Lee J, Gnad F, Klijn C, Schaub A, Reeder J, Daemen A, Bakalarski CE, Holcomb T, Shames DS, Hartmaier RJ, Chmielecki J, Seshagiri S, Gentleman R, Stokoe D (2016) Recurrent loss of NFE2L2 exon 2 is a mechanism for Nrf2 pathway activation in human cancers. Cell Rep 16:2605-2617. doi: 10.1016/j.celrep.2016.08.010
    Pubmed CrossRef
  34. Gottesman MM, Lavi O, Hall MD, Gillet JP (2016) Toward a better understanding of the complexity of cancer drug resistance. Annu Rev Pharmacol Toxicol 56:85-102. doi: 10.1146/annurev-pharmtox-010715-103111
    Pubmed CrossRef
  35. Gutmann H, Hruz P, Zimmermann C, Beglinger C, Drewe J (2005) Distribution of breast cancer resistance protein (BCRP/ABCG2) mRNA expression along the human GI tract. Biochem Pharmacol 70:695-699. doi: 10.1016/j.bcp.2005.05.031
    Pubmed CrossRef
  36. Hallis SP, Kim JM, Kwak MK (2023a) Emerging role of NRF2 signaling in cancer stem cell phenotype. Mol Cells 46:153-164. doi: 10.14348/molcells.2023.2196
    Pubmed KoreaMed CrossRef
  37. Hallis SP, Kim SK, Lee JH, Kwak MK (2023b) Association of NRF2 with HIF-2α-induced cancer stem cell phenotypes in chronic hypoxic condition. Redox Biol 60:102632. doi: 10.1016/j.redox.2023.102632
    Pubmed KoreaMed CrossRef
  38. Hanada N, Takahata T, Zhou Q, Ye X, Sun R, Itoh J, Ishiguro A, Kijima H, Mimura J, Itoh K, Fukuda S, Saijo Y (2012) Methylation of the Keap1 gene promoter region in human colorectal cancer. BMC Cancer 12:66. doi: 10.1186/1471-2407-12-66
    Pubmed KoreaMed CrossRef
  39. He M, Wu H, Jiang Q, Liu Y, Han L, Yan Y, Wei B, Liu F, Deng X, Chen H, Zhao L, Wang M, Wu X, Yao W, Zhao H, Chen J, Wei M (2019) Hypoxia-inducible factor-2α directly promotes BCRP expression and mediates the resistance of ovarian cancer stem cells to adriamycin. Mol Oncol 13:403-421. doi: 10.1002/1878-0261.12419
    Pubmed KoreaMed CrossRef
  40. He T, Zhou F, Su A, Zhang Y, Xing Z, Mi L, Li Z, Wu W (2023) Brusatol: a potential sensitizing agent for cancer therapy from Brucea javanica. Biomed Pharmacother 158:114134. doi: 10.1016/j.biopha.2022.114134
    Pubmed CrossRef
  41. Henriksen U, Fog JU, Litman T, Gether U (2005) Identification of intra- and intermolecular disulfide bridges in the multidrug resistance transporter ABCG2. J Biol Chem 280:36926-36934. doi: 10.1074/jbc.M502937200
    Pubmed CrossRef
  42. Hirano M, Maeda K, Matsushima S, Nozaki Y, Kusuhara H, Sugiyama Y (2005) Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin. Mol Pharmacol 68:800-807. doi: 10.1124/mol.105.014019
    Pubmed CrossRef
  43. Hirotsu Y, Katsuoka F, Funayama R, Nagashima T, Nishida Y, Nakayama K, Engel JD, Yamamoto M (2012) Nrf2-MafG heterodimers contribute globally to antioxidant and metabolic networks. Nucleic Acids Res 40:10228-10239. doi: 10.1093/nar/gks827
    Pubmed KoreaMed CrossRef
  44. Huls M, Brown CD, Windass AS, Sayer R, van den Heuvel JJ, Heemskerk S, Russel FG, Masereeuw R (2008) The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane. Kidney Int 73:220-225. doi: 10.1038/sj.ki.5002645
    Pubmed CrossRef
  45. Ichimura Y, Komatsu M (2018) Activation of p62/SQSTM1-Keap1-nuclear factor erythroid 2-related factor 2 pathway in cancer. Front Oncol 8:210. doi: 10.3389/fonc.2018.00210
    Pubmed KoreaMed CrossRef
  46. Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, Yamamoto M, Nabeshima Y (1997) An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun 236:313-322. doi: 10.1006/bbrc.1997.6943
    Pubmed CrossRef
  47. Jeong HS, Ryoo IG, Kwak MK (2015) Regulation of the expression of renal drug transporters in Keap1-knockdown human tubular cells. Toxicol In Vitro 9:884-892. doi: 10.1016/j.tiv.2015.03.013
    Pubmed CrossRef
  48. Ji L, Li H, Gao P, Shang G, Zhang DD, Zhang N, Jiang T (2013) Nrf2 pathway regulates multidrug-resistance-associated protein 1 in small cell lung cancer. PLoS One 8:e63404. doi: 10.1371/journal.pone.0063404
    Pubmed KoreaMed CrossRef
  49. Jia Y, Chen J, Zhu H, Jia ZH, Cui MH (2015) Aberrantly elevated redox sensing factor Nrf2 promotes cancer stem cell survival via enhanced transcriptional regulation of ABCG2 and Bcl-2/Bmi-1 genes. Oncol Rep 34:2296-2304. doi: 10.3892/or.2015.4214
    Pubmed CrossRef
  50. Jigorel E, Le Vee M, Boursier-Neyret C, Parmentier Y, Fardel O (2006) Differential regulation of sinusoidal and canalicular hepatic drug transporter expression by xenobiotics activating drug-sensing receptors in primary human hepatocytes. Drug Metab Dispos 34:1756-1763. doi: 10.1124/dmd.106.010033
    Pubmed CrossRef
  51. Jonker JW, Smit JW, Brinkhuis RF, Maliepaard M, Beijnen JH, Schellens JH, Schinkel AH (2000) Role of breast cancer resistance protein in the bioavailability and fetal penetration of topotecan. J Natl Cancer Inst 92:1651-1656. doi: 10.1093/jnci/92.20.1651
    Pubmed CrossRef
  52. Jung KA, Choi BH, Kwak MK (2015) The c-MET/PI3K signaling is associated with cancer resistance to doxorubicin and photodynamic therapy by elevating BCRP/ABCG2 expression. Mol Pharmacol 87:465-476. doi: 10.1124/mol.114.096065
    Pubmed CrossRef
  53. Jung KA, Choi BH, Nam CW, Song M, Kim ST, Lee JY, Kwak MK (2013) Identification of aldo-keto reductases as NRF2-target marker genes in human cells. Toxicol Lett 218:39-49. doi: 10.1016/j.toxlet.2012.12.026
    Pubmed CrossRef
  54. Kahroba H, Shirmohamadi M, Hejazi MS, Samadi N (2019) The role of Nrf2 signaling in cancer stem cells: from stemness and self-renewal to tumorigenesis and chemoresistance. Life Sci 239:116986. doi: 10.1016/j.lfs.2019.116986
    Pubmed CrossRef
  55. Kawabata S, Oka M, Shiozawa K, Tsukamoto K, Nakatomi K, Soda H, Fukuda M, Ikegami Y, Sugahara K, Yamada Y, Kamihira S, Doyle LA, Ross DD, Kohno S (2001) Breast cancer resistance protein directly confers SN-38 resistance of lung cancer cells. Biochem Biophys Res Commun 280:1216-1223. doi: 10.1006/bbrc.2001.4267
    Pubmed CrossRef
  56. Kim D, Choi BH, Ryoo IG, Kwak MK (2018) High NRF2 level mediates cancer stem cell-like properties of aldehyde dehydrogenase (ALDH)-high ovarian cancer cells: inhibitory role of all-trans retinoic acid in ALDH/NRF2 signaling. Cell Death Dis 9:896. doi: 10.1038/s41419-018-0903-4
    Pubmed KoreaMed CrossRef
  57. Kim EJ, Kim YJ, Lee HI, Jeong SH, Nam HJ, Cho JH (2020) NRF2 knockdown resensitizes 5-fluorouracil-resistant pancreatic cancer cells by suppressing HO-1 and ABCG2 expression. Int J Mol Sci 21:4646. doi: 10.3390/ijms21134646
    Pubmed KoreaMed CrossRef
  58. Kitamura H, Motohashi H (2018) NRF2 addiction in cancer cells. Cancer Sci 109:900-911. doi: 10.1111/cas.13537
    Pubmed KoreaMed CrossRef
  59. Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, Igarashi K, Yamamoto M (2004) Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol Cell Biol 24:7130-7139. doi: 10.1128/MCB.24.16.7130-7139.2004
    Pubmed KoreaMed CrossRef
  60. Krishnamurthy P, Ross DD, Nakanishi T, Bailey-Dell K, Zhou S, Mercer KE, Sarkadi B, Sorrentino BP, Schuetz JD (2004) The stem cell marker Bcrp/ABCG2 enhances hypoxic cell survival through interactions with heme. J Biol Chem 279:24218-24225. doi: 10.1074/jbc.M313599200
    Pubmed CrossRef
  61. Kurz EU, Cole SP, Deeley RG (2001) Identification of DNA-protein interactions in the 5' flanking and 5' untranslated regions of the human multidrug resistance protein (MRP1) gene: evaluation of a putative antioxidant response element/AP-1 binding site. Biochem Biophys Res Commun 285:981-990. doi: 10.1006/bbrc.2001.5262
    Pubmed CrossRef
  62. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367:645-648. doi: 10.1038/367645a0
    Pubmed CrossRef
  63. Liu W, Baer MR, Bowman MJ, Pera P, Zheng X, Morgan J, Pandey RA, Oseroff AR (2007) The tyrosine kinase inhibitor imatinib mesylate enhances the efficacy of photodynamic therapy by inhibiting ABCG2. Clin Cancer Res 13:2463-2470. doi: 10.1158/1078-0432.CCR-06-1599
    Pubmed CrossRef
  64. Low SK, Fukunaga K, Takahashi A, Matsuda K, Hongo F, Nakanishi H, Kitamura H, Inoue T, Kato Y, Tomita Y, Fukasawa S, Tanaka T, Nishimura K, Uemura H, Hara I, Fujisawa M, Matsuyama H, Hashine K, Tatsugami K, Enokida H, Kubo M, Miki T, Mushiroda T (2016) Association study of a functional variant on ABCG2 gene with sunitinib-induced severe adverse drug reaction. PLoS One 11:e0148177. doi: 10.1371/journal.pone.0148177
    Pubmed KoreaMed CrossRef
  65. Mahaffey CM, Zhang H, Rinna A, Holland W, Mack PC, Forman HJ (2009) Multidrug-resistant protein-3 gene regulation by the transcription factor Nrf2 in human bronchial epithelial and non-small-cell lung carcinoma. Free Radic Biol Med 46:1650-1657. doi: 10.1016/j.freeradbiomed.2009.03.023
    Pubmed KoreaMed CrossRef
  66. Maliepaard M, van Gastelen MA, de Jong LA, Pluim D, van Waardenburg RC, Ruevekamp-Helmers MC, Floot BG, Schellens JH (1999) Overexpression of the BCRP/MXR/ABCP gene in a topotecan-selected ovarian tumor cell line. Cancer Res 59:4559-4563.
  67. Mao Q, Unadkat JD (2015) Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport--an update. AAPS J 17:65-82. doi: 10.1208/s12248-014-9668-6
    Pubmed KoreaMed CrossRef
  68. Martin V, Xu J, Pabbisetty SK, Alonso MM, Liu D, Lee OH, Gumin J, Bhat KP, Colman H, Lang FF, Fueyo J, Gomez-Manzano C (2009) Tie2-mediated multidrug resistance in malignant gliomas is associated with upregulation of ABC transporters. Oncogene 28:2358-2363. doi: 10.1038/onc.2009.103
    Pubmed KoreaMed CrossRef
  69. Marzac C, Garrido E, Tang R, Fava F, Hirsch P, De Benedictis C, Corre E, Lapusan S, Lallemand JY, Marie JP, Jacquet E, Legrand O (2011) ATP Binding Cassette transporters associated with chemoresistance: transcriptional profiling in extreme cohorts and their prognostic impact in a cohort of 281 acute myeloid leukemia patients. Haematologica 96:1293-1301. doi: 10.3324/haematol.2010.031823
    Pubmed KoreaMed CrossRef
  70. McMahon M, Thomas N, Itoh K, Yamamoto M, Hayes JD (2006) Dimerization of substrate adaptors can facilitate cullin-mediated ubiquitylation of proteins by a "tethering" mechanism: a two-site interaction model for the Nrf2-Keap1 complex. J Biol Chem 281:24756-24768. doi: 10.1074/jbc.M601119200
    Pubmed CrossRef
  71. Miyake K, Mickley L, Litman T, Zhan Z, Robey R, Cristensen B, Brangi M, Greenberger L, Dean M, Fojo T, Bates SE (1999) Molecular cloning of cDNAs which are highly overexpressed in mitoxantrone-resistant cells: demonstration of homology to ABC transport genes. Cancer Res 59:8-13.
    Pubmed
  72. Mukhopadhyay S, Goswami D, Adiseshaiah PP, Burgan W, Yi M, Guerin TM, Kozlov SV, Nissley DV, McCormick F (2020) Undermining glutaminolysis bolsters chemotherapy while NRF2 promotes chemoresistance in KRAS-driven pancreatic cancers. Cancer Res 80:1630-1643. doi: 10.1158/0008-5472.CAN-19-1363
    Pubmed KoreaMed CrossRef
  73. Nakanishi T, Bailey-Dell KJ, Hassel BA, Shiozawa K, Sullivan DM, Turner J, Ross DD (2006) Novel 5' untranslated region variants of BCRP mRNA are differentially expressed in drug-selected cancer cells and in normal human tissues: implications for drug resistance, tissue-specific expression, and alternative promoter usage. Cancer Res 66:5007-5011. doi: 10.1158/0008-5472.CAN-05-4572
    Pubmed CrossRef
  74. Ohta T, Iijima K, Miyamoto M, Nakahara I, Tanaka H, Ohtsuji M, Suzuki T, Kobayashi A, Yokota J, Sakiyama T, Shibata T, Yamamoto M, Hirohashi S (2008) Loss of Keap1 function activates Nrf2 and provides advantages for lung cancer cell growth. Cancer Res 68:1303-1309. doi: 10.1158/0008-5472.CAN-07-5003
    Pubmed CrossRef
  75. Ozvegy C, Litman T, Szakács G, Nagy Z, Bates S, Váradi A, Sarkadi B (2001) Functional characterization of the human multidrug transporter, ABCG2, expressed in insect cells. Biochem Biophys Res Commun 285:111-117. doi: 10.1006/bbrc.2001.5130
    Pubmed CrossRef
  76. Pan YZ, Morris ME, Yu AM (2009) MicroRNA-328 negatively regulates the expression of breast cancer resistance protein (BCRP/ABCG2) in human cancer cells. Mol Pharmacol 75:1374-1379. doi: 10.1124/mol.108.054163
    Pubmed KoreaMed CrossRef
  77. Park J, Kim SK, Hallis SP, Choi BH, Kwak MK (2022) Role of CD133/NRF2 axis in the development of colon cancer stem cell-like properties. Front Oncol 11:808300. doi: 10.3389/fonc.2021.808300
    Pubmed KoreaMed CrossRef
  78. Raaijmakers MH, de Grouw EP, Heuver LH, van der Reijden BA, Jansen JH, Scheper RJ, Scheffer GL, de Witte TJ, Raymakers RA (2005) Breast cancer resistance protein in drug resistance of primitive CD34+38- cells in acute myeloid leukemia. Clin Cancer Res 11:2436-2444. doi: 10.1158/1078-0432.CCR-04-0212
    Pubmed CrossRef
  79. Rabindran SK, He H, Singh M, Brown E, Collins KI, Annable T, Greenberger LM (1998) Reversal of a novel multidrug resistance mechanism in human colon carcinoma cells by fumitremorgin C. Cancer Res 58:5850-5858.
    Pubmed
  80. Robey RW, Honjo Y, van de Laar A, Miyake K, Regis JT, Litman T, Bates SE (2001a) A functional assay for detection of the mitoxantrone resistance protein, MXR (ABCG2). Biochim Biophys Acta 1512:171-182. doi: 10.1016/s0005-2736(01)00308-x
    CrossRef
  81. Robey RW, Medina-Pérez WY, Nishiyama K, Lahusen T, Miyake K, Litman T, Senderowicz AM, Ross DD, Bates SE (2001b) Overexpression of the ATP-binding cassette half-transporter, ABCG2 (Mxr/BCrp/ABCP1), in flavopiridol-resistant human breast cancer cells. Clin Cancer Res 7:145-152.
  82. Robey RW, Pluchino KM, Hall MD, Fojo AT, Bates SE, Gottesman MM (2018) Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat Rev Cancer 18:452-464. doi: 10.1038/s41568-018-0005-8
    Pubmed KoreaMed CrossRef
  83. Robey RW, To KK, Polgar O, Dohse M, Fetsch P, Dean M, Bates SE (2009) ABCG2: a perspective. Adv Drug Deliv Rev 61:3-13. doi: 10.1016/j.addr.2008.11.003
    Pubmed KoreaMed CrossRef
  84. Roh YJ, Kim JH, Kim IW, Na K, Park JM, Choi MG (2017) Photodynamic therapy using photosensitizer-encapsulated polymeric nanoparticle to overcome ATP-binding cassette transporter subfamily G2 function in pancreatic cancer. Mol Cancer Ther 16:1487-1496. doi: 10.1158/1535-7163.MCT-16-0642
    Pubmed CrossRef
  85. Rojo de la Vega M, Chapman E, Zhang DD (2018) NRF2 and the hallmarks of cancer. Cancer Cell 34:21-43. doi: 10.1016/j.ccell.2018.03.022
    Pubmed KoreaMed CrossRef
  86. Roninson IB, Chin JE, Choi KG, Gros P, Housman DE, Fojo A, Shen DW, Gottesman MM, Pastan I (1986) Isolation of human mdr DNA sequences amplified in multidrug-resistant KB carcinoma cells. Proc Natl Acad Sci U S A 83:4538-4542. doi: 10.1073/pnas.83.12.4538
    Pubmed KoreaMed CrossRef
  87. Ross DD, Yang W, Abruzzo LV, Dalton WS, Schneider E, Lage H, Dietel M, Greenberger L, Cole SP, Doyle LA (1999) Atypical multidrug resistance: breast cancer resistance protein messenger RNA expression in mitoxantrone-selected cell lines. J Natl Cancer Inst 91:429-433. doi: 10.1093/jnci/91.5.429
    Pubmed CrossRef
  88. Ryoo IG, Kim G, Choi BH, Lee SH, Kwak MK (2016) Involvement of NRF2 signaling in doxorubicin resistance of cancer stem cell-enriched colonospheres. Biomol Ther (Seoul) 24:482-488. doi: 10.4062/biomolther.2016.145
    Pubmed KoreaMed CrossRef
  89. Saito T, Ichimura Y, Taguchi K, Suzuki T, Mizushima T, Takagi K, Hirose Y, Nagahashi M, Iso T, Fukutomi T, Ohishi M, Endo K, Uemura T, Nishito Y, Okuda S, Obata M, Kouno T, Imamura R, Tada Y, Obata R, Yasuda D, Takahashi K, Fujimura T, Pi J, Lee MS, Ueno T, Ohe T, Mashino T, Wakai T, Kojima H, Okabe T, Nagano T, Motohashi H, Waguri S, Soga T, Yamamoto M, Tanaka K, Komatsu M (2016) p62/Sqstm1 promotes malignancy of HCV-positive hepatocellular carcinoma through Nrf2-dependent metabolic reprogramming. Nat Commun 7:12030. doi: 10.1038/ncomms12030
    Pubmed KoreaMed CrossRef
  90. Sekhar KR, Rachakonda G, Freeman ML (2010) Cysteine-based regulation of the CUL3 adaptor protein Keap1. Toxicol Appl Pharmacol 244:21-26. doi: 10.1016/j.taap.2009.06.016
    Pubmed KoreaMed CrossRef
  91. Sesink AL, Arts IC, de Boer VC, Breedveld P, Schellens JH, Hollman PC, Russel FG (2005) Breast cancer resistance protein (Bcrp1/Abcg2) limits net intestinal uptake of quercetin in rats by facilitating apical efflux of glucuronides. Mol Pharmacol 67:1999-2006. doi: 10.1124/mol.104.009753
    Pubmed CrossRef
  92. Sharifi-Rad J, Seidel V, Izabela M, Monserrat-Mequida M, Sureda A, Ormazabal V, Zuniga FA, Mangalpady SS, Pezzani R, Ydyrys A, Tussupbekova G, Martorell M, Calina D, Cho WC (2023) Phenolic compounds as Nrf2 inhibitors: potential applications in cancer therapy. Cell Commun Signal 21:89. doi: 10.1186/s12964-023-01109-0
    Pubmed KoreaMed CrossRef
  93. Shibata T, Ohta T, Tong KI, Kokubu A, Odogawa R, Tsuta K, Asamura H, Yamamoto M, Hirohashi S (2008) Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Proc Natl Acad Sci U S A 105:13568-13573. doi: 10.1073/pnas.0806268105
    Pubmed KoreaMed CrossRef
  94. Shim GS, Manandhar S, Shin DH, Kim TH, Kwak MK (2009) Acquisition of doxorubicin resistance in ovarian carcinoma cells accompanies activation of the NRF2 pathway. Free Radic Biol Med 47:1619-1631. doi: 10.1016/j.freeradbiomed.2009.09.006
    Pubmed CrossRef
  95. Singh A, Happel C, Manna SK, Acquaah-Mensah G, Carrerero J, Kumar S, Nasipuri P, Krausz KW, Wakabayashi N, Dewi R, Boros LG, Gonzalez FJ, Gabrielson E, Wong KK, Girnun G, Biswal S (2013) Transcription factor NRF2 regulates miR-1 and miR-206 to drive tumorigenesis. J Clin Invest 123:2921-2934. doi: 10.1172/JCI66353
    Pubmed KoreaMed CrossRef
  96. Singh A, Misra V, Thimmulappa RK, Lee H, Ames S, Hoque MO, Herman JG, Baylin SB, Sidransky D, Gabrielson E, Brock MV, Biswal S (2006) Dysfunctional Keap1-NRF2 interaction in non-small-cell lung cancer. PLoS Med 3:e420. doi: 10.1371/journal.pmed.0030420
    Pubmed KoreaMed CrossRef
  97. Singh A, Venkannagari S, Oh KH, Zhang YQ, Rohde JM, Liu L, Nimmagadda S, Sudini K, Brimacombe KR, Gajghate S, Ma J, Wang A, Xu X, Shahane SA, Xia M, Woo J, Mensah GA, Wang Z, Ferrer M, Gabrielson E, Li Z, Rastinejad F, Shen M, Boxer MB, Biswal S (2016) Small molecule inhibitor of NRF2 selectively intervenes therapeutic resistance in Keap1-deficient NSCLC tumors. ACS Chem Biol 11:3214-3225. doi: 10.1021/acschembio.6b00651
    Pubmed KoreaMed CrossRef
  98. Singh A, Wu H, Zhang P, Happel C, Ma J, Biswal S (2010) Expression of ABCG2 (BCRP) is regulated by Nrf2 in cancer cells that confers side population and chemoresistance phenotype. Mol Cancer Ther 9:2365-2376. doi: 10.1158/1535-7163.MCT-10-0108
    Pubmed KoreaMed CrossRef
  99. Solis LM, Behrens C, Dong W, Suraokar M, Ozburn NC, Moran CA, Corvalan AH, Biswal S, Swisher SG, Bekele BN, Minna JD, Stewart DJ, Wistuba II (2010) Nrf2 and Keap1 abnormalities in non-small cell lung carcinoma and association with clinicopathologic features. Clin Cancer Res 16:3743-3753. doi: 10.1158/1078-0432.CCR-09-3352
    Pubmed KoreaMed CrossRef
  100. ivastava R Sr, Fernández-Ginés R, Encinar JA, Cuadrado A, Wells G (2022) The current status and future prospects for therapeutic targeting of Keap1-NRF2 and β-TrCP-NRF2 interactions in cancer chemoresistance. Free Radic Biol Med 192:246-260. doi: 10.1016/j.freeradbiomed.2022.09.023
    Pubmed CrossRef
  101. Stöckel B, König J, Nies AT, Cui Y, Brom M, Keppler D (2000) Characterization of the 5'-flanking region of the human multidrug resistance protein 2 (MRP2) gene and its regulation in comparison withthe multidrug resistance protein 3 (MRP3) gene. Eur J Biochem 267:1347-1358. doi: 10.1046/j.1432-1327.2000.01106.x
    Pubmed CrossRef
  102. Sugimoto Y, Tsukahara S, Imai Y, Sugimoto Y, Ueda K, Tsuruo T (2003) Reversal of breast cancer resistance protein-mediated drug resistance by estrogen antagonists and agonists. Mol Cancer Ther 2:105-112.
    Pubmed CrossRef
  103. Sun S, Yu M, Yu L, Huang W, Zhu M, Fu Y, Yan L, Wang Q, Ji X, Zhao J, Wu M (2023) Nrf2 silencing amplifies DNA photooxidative damage to activate the STING pathway for synergistic tumor immunotherapy. Biomaterials 296:122068. doi: 10.1016/j.biomaterials.2023.122068
    Pubmed CrossRef
  104. Szatmari I, Vámosi G, Brazda P, Balint BL, Benko S, Széles L, Jeney V, Ozvegy-Laczka C, Szántó A, Barta E, Balla J, Sarkadi B, Nagy L (2006) Peroxisome proliferator-activated receptor gamma-regulated ABCG2 expression confers cytoprotection to human dendritic cells. J Biol Chem 281:23812-23823. doi: 10.1074/jbc.M604890200
    Pubmed CrossRef
  105. Tao S, Wang S, Moghaddam SJ, Ooi A, Chapman E, Wong PK, Zhang DD (2014) Oncogenic KRAS confers chemoresistance by upregulating NRF2. Cancer Res 74:7430-7441. doi: 10.1158/0008-5472.CAN-14-1439
    Pubmed KoreaMed CrossRef
  106. Taylor NMI, Manolaridis I, Jackson SM, Kowal J, Stahlberg H, Locher KP (2017) Structure of the human multidrug transporter ABCG2. Nature 546:504-509. doi: 10.1038/nature22345
    Pubmed CrossRef
  107. To KK, Zhan Z, Litman T, Bates SE (2008) Regulation of ABCG2 expression at the 3' untranslated region of its mRNA through modulation of transcript stability and protein translation by a putative microRNA in the S1 colon cancer cell line. Mol Cell Biol 28:5147-5161. doi: 10.1128/MCB.00331-08
    Pubmed KoreaMed CrossRef
  108. Tong KI, Katoh Y, Kusunoki H, Itoh K, Tanaka T, Yamamoto M (2006) Keap1 recruits Neh2 through binding to ETGE and DLG motifs: characterization of the two-site molecular recognition model. Mol Cell Biol 26:2887-2900. doi: 10.1128/MCB.26.8.2887-2900.2006
    Pubmed KoreaMed CrossRef
  109. Tsuchida K, Tsujita T, Hayashi M, Ojima A, Keleku-Lukwete N, Katsuoka F, Otsuki A, Kikuchi H, Oshima Y, Suzuki M, Yamamoto M (2017) Halofuginone enhances the chemo-sensitivity of cancer cells by suppressing NRF2 accumulation. Free Radic Biol Med 103:236-247. doi: 10.1016/j.freeradbiomed.2016.12.041
    Pubmed CrossRef
  110. Usuda J, Tsunoda Y, Ichinose S, Ishizumi T, Ohtani K, Maehara S, Ono S, Tsutsui H, Ohira T, Okunaka T, Furukawa K, Sugimoto Y, Kato H, Ikeda N (2010) Breast cancer resistant protein (BCRP) is a molecular determinant of the outcome of photodynamic therapy (PDT) for centrally located early lung cancer. Lung Cancer 67:198-204. doi: 10.1016/j.lungcan.2009.04.002
    Pubmed CrossRef
  111. Wang F, Xue X, Wei J, An Y, Yao J, Cai H, Wu J, Dai C, Qian Z, Xu Z, Miao Y (2010) hsa-miR-520h downregulates ABCG2 in pancreatic cancer cells to inhibit migration, invasion, and side populations. Br J Cancer 103:567-574. doi: 10.1038/sj.bjc.6605724
    Pubmed KoreaMed CrossRef
  112. Wang H, Lee EW, Zhou L, Leung PC, Ross DD, Unadkat JD, Mao Q (2008) Progesterone receptor (PR) isoforms PRA and PRB differentially regulate expression of the breast cancer resistance protein in human placental choriocarcinoma BeWo cells. Mol Pharmacol 73:845-854. doi: 10.1124/mol.107.041087
    Pubmed CrossRef
  113. Weiss J, Rose J, Storch CH, Ketabi-Kiyanvash N, Sauer A, Haefeli WE, Efferth T (2007) Modulation of human BCRP (ABCG2) activity by anti-HIV drugs. J Antimicrob Chemother 59:238-245. doi: 10.1093/jac/dkl474
    Pubmed CrossRef
  114. Wilson CS, Davidson GS, Martin SB, Andries E, Potter J, Harvey R, Ar K, Xu Y, Kopecky KJ, Ankerst DP, Gundacker H, Slovak ML, Mosquera-Caro M, Chen IM, Stirewalt DL, Murphy M, Schultz FA, Kang H, Wang X, Radich JP, Appelbaum FR, Atlas SR, Godwin J, Willman CL (2006) Gene expression profiling of adult acute myeloid leukemia identifies novel biologic clusters for risk classification and outcome prediction. Blood 108:685-696. doi: 10.1182/blood-2004-12-4633
    Pubmed KoreaMed CrossRef
  115. Xu S, Weerachayaphorn J, Cai SY, Soroka CJ, Boyer JL (2010) Aryl hydrocarbon receptor and NF-E2-related factor 2 are key regulators of human MRP4 expression. Am J Physiol Gastrointest Liver Physiol 299:G126-G135. doi: 10.1152/ajpgi.00522.2010
    Pubmed KoreaMed CrossRef
  116. Yamamoto M, Kensler TW, Motohashi H (2018) The Keap1-NRF2 system: a thiol-based sensor-effector apparatus for maintaining redox homeostasis. Physiol Rev 98:1169-1203. doi: 10.1152/physrev.00023.2017
    Pubmed KoreaMed CrossRef
  117. Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, Zhang G, Wang X, Dong Z, Chen F, Cui H (2020) Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther 5:8. doi: 10.1038/s41392-020-0110-5
    Pubmed KoreaMed CrossRef
  118. Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J, Morris JJ, Lagutina I, Grosveld GC, Osawa M, Nakauchi H, Sorrentino BP (2001) The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med 7:1028-1034. doi: 10.1038/nm0901-1028
    Pubmed CrossRef
  119. Zhou Y, Chen Y, Shi Y, Wu L, Tan Y, Li T, Chen Y, Xia J, Hu R (2023) FAM117B promotes gastric cancer growth and drug resistance by targeting the Keap1/NRF2 signaling pathway. J Clin Invest 133:e158705. doi: 10.1172/JCI158705
    Pubmed KoreaMed CrossRef

Article

Review

DTT 2023; 2(2): 111-123

Published online September 30, 2023 https://doi.org/10.58502/DTT.23.0021

Copyright © The Pharmaceutical Society of Korea.

Toward a Better Understanding of NRF2/NFE2L2 and BCRP/ABCG2 in Therapy Resistance in Cancer

Steffanus Pranoto Hallis1 , Beam Ju Go2, Jun Min Yoo2, Geun Hee Cho2, Mi-Kyoung Kwak1,2

1Department of Pharmacy, Graduate School of The Catholic University of Korea, Bucheon, Korea
2College of Pharmacy, The Catholic University of Korea, Bucheon, Korea

Correspondence to:Mi-Kyoung Kwak, mkwak@catholic.ac.kr

Received: May 9, 2023; Revised: June 7, 2023; Accepted: June 7, 2023

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

The development of therapy resistance in cancer is a complex mechanism with multifaceted and catastrophic consequences. BCRP (Breast Cancer Resistance Protein/ATP-Binding Cassette Subfamily G member 2; ABCG2), a drug efflux transporter, is recognized as a key regulator in obtaining multidrug resistance properties of cancer cells. NRF2 (Nuclear Factor Erythroid 2-Like 2; NFE2L2) plays an essential role in maintaining cellular redox homeostasis by regulating the expression of an array of antioxidant and detoxifying genes. In contrast to its protective roles in normal cells, a substantial body of evidence has demonstrated the negative aspects of NRF2 accumulation in cancer cells, including facilitated tumor growth, stemness, and therapy resistance. This review focuses on the linkage between BCRP and NRF2 in cancer and its consequences in acquiring therapy resistance. In particular, we highlight the role of the NRF2/BCRP axis in photodynamic therapy resistance and cancer stem cell maintenance, demonstrating that inhibition of this axis can be a beneficial strategy to overcome resistance to cancer therapy. A comprehensive understanding of the NRF2/BCRP regulating pathway will lead to the development of new therapeutic approaches and improve the prognosis of cancer patients.

Keywords: BCRP/ABCG2, NRF2/NFE2L2, multidrug resistance, chemoresistance, photodynamic therapy, cancer stem cells

Introduction

The development of resistance in cancer cells remains a major obstacle to successful chemotherapy, contributing to treatment failure and disease progression. One key mechanism of chemoresistance is the overexpression of ATP-binding cassette (ABC) transporters, which are responsible for the efflux of cytotoxic anticancer agents from cancer cells (Robey et al. 2018). Among 48 human ABC transporters, which are classified into seven subfamilies, multidrug resistance protein 1 (MDR1 or P-glycoprotein/P-gp) encoded by ABC subfamily B member 1 (ABCB1) was initially described in Chinese hamster ovary (CHO) cells and later in sublines of human KB carcinoma cells bearing multidrug-resistance to either colchicine, vinblastine, or doxorubicin (Roninson et al. 1986). The multidrug resistance-associated protein 1 (MRP1, encoded by ABCC1) was identified in 1992 as the second member of multidrug ABC transporters from lung cancer cell line H69AR, a cell line model of doxorubicin, etoposide, and vincristine resistance (Cole et al. 1992). The third member of the multidrug ABC transporter was encoded by ABCG2 and concurrently reported by three groups. The first group identified ABCG2 in human MCF7 breast cancer cells, where it was named breast cancer resistance protein (BCRP) (Doyle et al. 1998). Additionally, ABCG2 was found in mitoxantrone-resistant S1-M1-80 human colon carcinoma cells (Miyake et al. 1999), as well as in placental tissue (Allikmets et al. 1998). The overexpression of multidrug ABC transporters in cancer has been linked to chemotherapy failure in clinical settings. As a result, numerous efforts have been made to overcome multidrug-resistant cancer through the development of transporter inhibitors (Gottesman et al. 2016).

Nuclear factor erythroid 2-like 2 (NFE2L2/NRF2) is a transcription factor that regulates the expression of gene arrays involved in protection against oxidative and electrophilic stress and maintenance of cellular homeostasis (Yamamoto et al. 2018). Despite its protective roles in normal tissues, constitutive activation of NRF2 confers cancer cells survival by multiple mechanisms: increasing drug elimination through phase II metabolizing enzymes, preventing intracellular drug accumulation, and suppressing apoptotic cell death (Bai et al. 2016; Choi and Kwak 2016). In particular, the antioxidant response element (ARE) has been identified as a binding site for NRF2 in the promoter region of MRP1 and BCRP, confirming the critical role of NRF2 signaling in the regulation of multidrug-resistant ABC transporters (Kurz et al. 2001; Singh et al. 2010; Ji et al. 2013). The review article focuses on providing an overview of BCRP and NRF2, and their association with therapy resistance in cancer. In particular, we highlight the role of the NRF2/BCRP axis in cancer stem cells (CSCs) and photodynamic therapy (PDT) resistance. We also suggest that inhibiting this axis can be an effective strategy to overcome resistance to cancer therapy.

BCRP

Function and regulation of BCRP

BCRP, which is also known as mitoxantrone resistance-associated protein (MXR) or placenta-specific ABC transporter (ABCP), is a type of ABC transporter that belongs to the G subfamily of ABC transporters, also known as ABCG2. BCRP is responsible for translocating its substrates across cell membranes, against a concentration gradient, and this process is achieved through the binding and hydrolysis of ATP (Dean et al. 2001). BCRP is a 72-kDa protein consisting of 665 amino acids with six transmembrane segments and one extracellular loop. Unlike other subfamilies of ABC transporters, which contain two transmembrane domains (TMD) and two nucleotide (ATP)-binding domain (NBD), BCRP is a “half-transporter” with only one TMD and one NBD fused to a single polypeptide chain (Ozvegy et al. 2001; Taylor et al. 2017). Therefore, for BCRP to function properly, it needs to homodimerize, possibly through the formation of intra- and intermolecular disulfide bonds, or oligomerize (Bhatia et al. 2005; Henriksen et al. 2005).

BCRP has diverse physiological roles and is localized in various tissues and organs, including the placenta, mammary glands, gastrointestinal tract, kidney, blood-brain barrier, and hematopoietic stem cells (Robey et al. 2009). BCRP expression in the placenta is essential for protecting the fetus from topotecan, which has been administered in maternal animals. Treatment of pregnant mice with GF120918, a BCRP inhibitor, increased the relative fetal penetration of topotecan (Jonker et al. 2000). In Bcrp1(−/−) knockout mouse model, BCRP absence increased topotecan penetration to the brain by 1.5 folds, while the penetration increased by 12 folds in Mdr1a/b(−/−) Bcrp1(−/−) double knockout model, suggesting the critical role of these transporters in the blood-brain barrier (de Vries et al. 2007). Expression analysis of BCRP mRNA has revealed that its levels decrease along the gastrointestinal tract. The highest expression levels of BCRP mRNA were observed in the duodenum, while levels continuously decreased towards the rectum (Gutmann et al. 2005). The intestinal efflux of quercetin glucuronide, a flavonoid known substrate of BCRP, is impaired in Mrp2-deficient rats treated with BCRP inhibitors. This suggests that BCRP plays a role in facilitating the intestinal uptake of quercetin, potentially by transporting them across the intestinal epithelium. Interestingly, this impairment in quercetin glucuronide efflux was not observed in Mrp2-deficient rats that were not treated with BCRP inhibitors. This suggests that BCRP may compensate for the loss of Mrp2 function in the transport of quercetin glucuronide (Sesink et al. 2005). BCRP is also found to be localized in the apical membrane of human kidney proximal tubules, where it is expressed alongside other ABC transporters, contributing to the excretion of drugs and xenobiotics in the urine (Huls et al. 2008). BCRP has also been identified as a marker of hematopoietic stem cells, due to its high expression in primitive murine stem cells and sharp decrease with differentiation (Zhou et al. 2001).

The human ABCG2 gene is located on chromosome 4, specifically in the 4q21-q22 region. It spans over 66 kb and consists of 16 exons and 15 introns (Bailey-Dell et al. 2001). Transcriptional regulation studies of ABCG2 have identified several functional cis-elements within its promoter region that are recognized by several transcription factors, including estrogen (Ee et al. 2004), progesterone (Wang et al. 2008), peroxisome proliferator-activated receptor γ (Szatmari et al. 2006), hypoxia-inducible factor (HIF)-1α (Krishnamurthy et al. 2004), HIF-2α (He et al. 2019), and NRF2 (Singh et al. 2010) (Fig. 1). At the post-transcriptional modification level, alternative splicing of the ABCG2 gene has been reported to generate three splice variants of the untranslated exon 1 in the 5’ untranslated region (UTR) of the mRNA. These splice variants can serve as alternative promoters and result in the expression of isoforms of the protein with altered function and localization (Nakanishi et al. 2006). In addition, microRNAs (miRNAs) have been suggested to be involved in BCRP regulation. Rapid amplification of cDNA ends (RACE) analysis of the 3’ UTR of ABCG2 mRNA from drug-resistant S1 colon cancer cells identified several putative miRNA binding sites (To et al. 2008). MiR-328 has been reported to directly regulate ABCG2 in MCF-7 breast cancer cells and re-sensitize the cells to mitoxantrone (Pan et al. 2009). Additionally, expression of miR-520 h inhibited migration, invasion, and side populations of pancreatic cancer cells (Wang et al. 2010).

Figure 1. BCRP regulation and its association with chemoresistance. Overactivation of NRF2 in cancer increases BCRP (ABCG2) as one of its targets, as well as other targets in antioxidant defense, metabolism, and drug detoxification, thereby promoting cell survival and resistance to therapy. The transcriptional regulation of BCRP can be controlled by additional transcription factors, including HIF-1α, HIF-2α, PPARγ, and estrogen, which can bind to their enhancer elements in the promoter sequence of BCRP genes. At the post-transcriptional level, miR-328 and miR-520h can directly suppress BCRP transcription, while NRF2-regulated miR-206 targets BCRP via the c-MET/EGFR signaling pathway. sMAFs, small MAF proteins; BCRP, breast cancer resistance protein; PPARγ, peroxisome proliferator-activated receptor-γ; HIF, hypoxia-inducible factor; ARE, antioxidant response element; PPRE, peroxisome proliferator-response element; HRE, hypoxia response element; ERE, estrogen response element; EGFR, epidermal growth factor receptor; c-MET, mesenchymal-epithelial transition factor.

BCRP and chemoresistance of cancer

ABC transporters work together to mediate the efflux of various substrates, including anticancer agents such as anthracyclines, camptothecins, taxanes, and tyrosine kinase inhibitors (Robey et al. 2018). It is also well-established that BCRP plays a major role in multidrug resistance of cancer because of its distinct action. While MRP1 selectively transports products of phase II drug metabolism, BCRP transports anticancer drugs, such as irinotecan, methotrexate, mitoxantrone, and topotecan (Mao and Unadkat 2015). Histology data indicates that BCRP is uniformly expressed in all subsets of tumors, with frequent expression observed in adenocarcinomas of the gastrointestinal tract, endometrium, lung, and melanoma (Diestra et al. 2002). The Cancer Genome Atlas (TCGA) database analysis of RNA sequencing data showed that hepatocellular carcinoma and kidney cancer, both of which are frequently resistant to vincristine and doxorubicin, expressed the highest levels of BCRP compared to other cancer types (Robey et al. 2018).

In vitro studies using cancer cell lines from various solid tumors, including breast, colon, gastric, glioma, lung, and ovarian cancer, have identified high expression of BCRP in multidrug resistance models (Maliepaard et al. 1999; Ross et al. 1999; Martin et al. 2009). An earlier study using drug-selected cancer cell lines showed that mitoxantrone, doxorubicin, and verapamil increased the expression of BCRP mRNA in human breast carcinoma (MCF-7), colon carcinoma (HT29), gastric carcinoma (EPG85-257), fibrosarcoma (EPF86-079), and myeloma (8226) (Ross et al. 1999). The human IGROV1 ovarian cancer cell line, which has developed resistance to the chemotherapy drugs topotecan and mitoxantrone following continuous exposure, expresses high levels of BCRP (Maliepaard et al. 1999). In a panel of glioma cell lines and brain tumor stem cells, high expression levels of BCRP were linked to the overexpression of the Tie2 receptor, which is associated with tumor malignancy, and BCRP downregulation decreased Tie2 expression (Martin et al. 2009). In SN-38-resistant PC-6 human small-lung cancer cell lines, which had acquired resistance to the active metabolites of irinotecan and topotecan, there was a reduction in the intracellular accumulation of SN-38. This reduction was found to be associated with the overexpression of BCRP, and suppression of BCRP enhanced the sensitivity of these cells to SN-38 (Kawabata et al. 2001). A pharmacogenetic study with 299 renal carcinoma patients in the Japanese population found that differences in the BCRP genotype (the ABCG2 C421A [Q141K] polymorphism) were associated with reduced adverse effects of sunitinib, without affecting its efficacy (Low et al. 2016).

There has been consistent clinical evidence for the relationship between BCRP overexpression and drug resistance in hematologic malignancies. A gene profiling study of older acute myeloid leukemia (AML) patients found that 77% of 170 AML samples were chemoresistant and characterized by high expressions of BCRP and MDR1 (Wilson et al. 2006). A comparison between sensitive and resistant AML patients showed a significant difference in BCRP expression, and that this difference was linked to overall patients’ survival (Marzac et al. 2011). Moreover, high levels of BCRP were identified in a population of putative leukemic stem cells (CD34+CD38) from AML patients. BCRP inhibition in these leukemic stem cells using Ko143 resulted in increased cellular levels of mitoxantrone accumulation (Raaijmakers et al. 2005). Thus, these preclinical and clinical datasets suggest that the inhibition of BCRP in multidrug-resistant AML may potentially lead to better treatment outcomes and patient survival.

BCRP inhibitors

Given the critical role of BCRP in the acquisition of chemoresistance, extensive efforts have been dedicated to discovering and developing BCRP inhibitors (Doyle and Ross 2003). The first BCRP inhibitor to be reported was Fumitremorgin C (FTC), a secondary metabolite from Aspergillus fumigatus. FTC has been shown to effectively reverse the multidrug resistance of colon carcinoma to mitoxantrone, doxorubicin, bisantrene, and topotecan through the specific inhibition of BCRP (Rabindran et al. 1998). Subsequent studies have confirmed the inhibitory effect of FTC on BCRP in various cancer cell lines, including SF295 human glioblastoma, KM12 colon cancer, and A549 non-small-cell lung cancer cell lines (Robey et al. 2001a). FTC has also been found to inhibit the transport of methotrexate in human embryonic kidney cells (Chen et al. 2003). At a concentration of 5 µM, FTC has been shown to effectively re-sensitize the multi-drug resistant MCF-7 FLV1000 cell line to mitoxantrone, SN-38, and topotecan by inhibiting the efflux of these drugs via BCRP (Robey et al. 2001b). Later, a tetracycline analog of FTC called Ko143 was identified as a more selective inhibitor of BCRP. In an experiment using mouse intestine, Ko143 selectively inhibited BCRP without affecting the function of other ABC transporters such as P-gp or MRP-1 at a concentration of 100 nM (Allen et al. 2002).

Elacridar (GF120918), which was originally developed as a P-gp inhibitor, is an acridone carboxamide and identified as a potent inhibitor of BCRP. Elacridar has been shown to effectively inhibit the efflux of mitoxantrone and topotecan (de Bruin et al. 1999). Additionally, flavonoid metabolites 6-prenylchrysin and tectochrysin have a specific inhibitory effect on BCRP. In a study conducted on BCRP-expressing cancer cells, the application of 0.5 µmol/L of 6-prenylchrysin or 1 µmol/L of tectochrysin was sufficient to sensitize the cells to mitoxantrone, with less toxicity compared to elacridar (Ahmed-Belkacem et al. 2005).

Several anti-retroviral drugs, including lopinavir, nelfinavir, and delavirdine have been shown to inhibit BCRP and increase the cellular accumulation of pheophorbide A (Pha), a photosensitizer substrate of BCRP, in canine kidney epithelial cell lines (Weiss et al. 2007). Additionally, statins such as atorvastatin and rosuvastatin were shown to inhibit BCRP in the biliary excretion system using Bcrp(−/−) mice, although some statins are substrates of BCRP (Hirano et al. 2005). Moreover, since estrogen can regulate BCRP expression and therefore, estrogen agonists and antagonists have been found to modulate BCRP activity. Diethylstilbestrol, an estrogen agonist, increased the accumulation of topotecan and reversed the chemoresistance of the BCRP-transduced K562 cell line. In contrast, tamoxifen, an estrogen antagonist, has been found to only enhance the uptake of topotecan (Sugimoto et al. 2003).

NRF2

The NRF2-KEAP1 system

NRF2 is a cap‘n’collar (CNC) basic leucine zipper (bZIP) transcription factor that regulates cytoprotective genes and maintains cellular redox homeostasis. Although NRF2 is expressed in all cell types, its basal level is mainly low due to negative regulation by KEAP1-mediated proteasomal degradation (Yamamoto et al. 2018). The NRF2 protein is composed of six functional domains, known as NRF2-ECH Homology (Neh) 1-6, each of which plays a specific role in NRF2 activity. Neh1 primarily contains the CNC-bZIP domains that facilitate DNA binding and dimerization, while Neh3-5 serve as transactivation domains. In particular, Neh4 and Neh5 recruit the histone acetyl-transferase cAMP-responsive element binding protein (CBP) and mediator complex to facilitate the transcriptional activity of NRF2 (Yamamoto et al. 2018). Meanwhile, Neh2 and Neh6 primarily regulate NRF2 degradation. Neh2 contains DLG and ETGE motifs that serve as distinct and prominent binding sites for the KEAP1 protein, while Neh6 functions as a KEAP1-independent degron site of NRF2. Neh6 facilitates NRF2 degradation through the binding of β-TRCP (Tong et al. 2006; Chowdhry et al. 2013).

Under normal conditions, two KEAP1 molecules and one NRF2 molecule form a structure that accelerates the ubiquitination of NRF2. KEAP1 acts as an adaptor, forming cullin3 (CUL3)-based E3 ubiquitin ligases and RBX1 to create a functional E3 ubiquitin ligase (Kobayashi et al. 2004; McMahon et al. 2006). The KEAP1-CUL3 complex recognizes the DLG and ETGE motifs in the Neh2 domain of NRF2 and ubiquitinates it, ensuring a low level of NRF2 by subjecting it to proteasomal degradation. However, following exposure to electrophiles or reactive oxygen species (ROS), the cysteine residues of KEAP1 undergo modification, forming disulfide bonds or covalent linkages due to oxidation, which results in the alteration of the conformational structure of the KEAP1 (Sekhar et al. 2010). The disruption of the DLG-KEAP1 binding allows for the accumulation of newly synthesized NRF2, which can be translocated into the nucleus. Then, NRF2 can dimerize with small Maf proteins and activate transcription by recognizing and binding to the ARE in the promoter of its target genes.

The direct correlation between NRF2 and target genes through ARE was first reported by Itoh et al. (1997). They observed a marked decrease in the expression of phase II enzymes, including glutathione S-transferases (GSTs) and NAD(P)H quinone oxidoreductase 1 (NQO1), in butylated hydroxyanisole (BHA)-treated Nrf2-null mice. Since then, NRF2 has been identified as a master regulator of genes involved in phase I, II, and III metabolism. These include phase I enzymes such as Cyp2a5 and Cyp2b10 (Ashino et al. 2014), phase II enzymes such as GSTs (GSTA3, GSTA4, GSTM1, GSTM2, GSTM3, GSTM4, GSTM6), NQO1 (Hirotsu et al. 2012), and aldo-keto reductase family 1 member C1 (AKR1C1) (Jung et al. 2013), and phase III transporters, including MRPs (ABCC1, ABCC2, ABCC3, ABCC4) (Stöckel et al. 2000; Mahaffey et al. 2009; Xu et al. 2010; Ji et al. 2013) and BCRP (ABCG2) (Singh et al. 2010).

NRF2 and cancer

While NRF2 activity is crucial in normal unstressed conditions, persistent activation of NRF2 has been found to contribute to impairing the efficacy of chemotherapy in cancer (Rojo de la Vega et al. 2018). The hyperactivation of NRF2 in cancer can be resulted from somatic mutations or genomic alterations in NRF2 and KEAP1 genes or the expression of KEAP1-competing proteins such as p62/SQSTM1 (Kitamura and Motohashi 2018). In non-small-cell lung cancer patients and cell lines, genomic domain mutation and high percentage loss of heterozygosity at 19p13.2 of KEAP1 results in the specific abrogation of the NRF2 repressor system (Singh et al. 2006). Thus, NRF2 is accumulated and enhances the transcriptional activation of genes involved in xenobiotic detoxification and drug efflux pumps. Moreover, mutations in either the IVR or DGR region of KEAP1, or in the DLG or ETGE motifs of NRF2, have consequences for the constitutive expression of cytoprotective enzymes induced by NRF2 and can facilitate tumor malignancies (Ohta et al. 2008; Shibata et al. 2008). The genomic alteration of exon 2 deletions in NRF2 results in persistent NRF2 activation in lung cancers and head and neck carcinomas (Goldstein et al. 2016). KEAP1 gene methylation leads to reduced expression of KEAP1 and activation of NRF2 and its corresponding targets, such as NQO1 and AKR1C1, during the carcinogenesis and cancer progression (Hanada et al. 2012; Barbano et al. 2013). One well-established KEAP1-competing protein that has been identified as responsible for NRF2 accumulation in cancer cells is the autophagy adaptor protein p62/SQSTM1. Its phosphorylation at the S349 residue by the mammalian target of rapamycin complex 1 (mTORC1) increases the binding affinity to KEAP1, subsequently disrupting the DLG-mediated KEAP1-NRF2 interaction (Ichimura and Komatsu 2018). Pharmacological inhibition of phosphorylated p62/SQSTM1 and KEAP1 interaction by K67 resulted in the suppression of NRF2 activity, which inhibited growth and re-sensitized the tumors to anti-cancer agents (Saito et al. 2016).

The high expression of NRF2, which has been observed in multiple types of cancers, contributes to chemoresistance by inducing downstream genes involved in electrophiles/ROS detoxification and the efflux of cytotoxic anticancer drugs (Srivastava et al. 2022). A recent report demonstrated that the activation of NRF2 by FAM117B-mediated KEAP1 competitive binding in gastric cancer facilitated tumor growth and chemoresistance to 5-fluorouracil and oxaliplatin (Zhou et al. 2023). In pancreatic cancer, oncogenic KRAS upregulated NRF2-dependent metabolic rewiring of glutamine metabolism, which leads to gemcitabine resistance (Mukhopadhyay et al. 2020). Keap1-deficient KrasG12D mice model developed lung tumors with hyperactivation of NRF2 and subsequent activation of the pentose phosphate pathway (PPP) (Best et al. 2019). Similarly, in KrasG12D–induced lung cancer, constitutive KRAS activation increased NRF2 transcription and resulted in cisplatin resistance. In this context, treatment with the NRF2 inhibitor brusatol was found to enhance the anti-tumor efficacy of cisplatin in KrasG12D-induced lung cancer (Tao et al. 2014).

NRF2 and BCRP

BCRP regulation by NRF2

Several studies have shown a direct correlation between NRF2 and BCRP in cancer. An early report demonstrated that treatment with oltipraz, an activator of NRF2, increased the mRNA level of BCRP in cultured human hepatocytes (Jigorel et al. 2006). This observation was further confirmed by a study that used tert-butylhydroquinone (tBHQ), another potent activator of NRF2, in HepG2 cells (Adachi et al. 2007). In this study, treatment of HepG2 cells with tBHQ induced translocation of NRF2 to the nucleus, and concomitantly increased the mRNA level of BCRP, as well as other ABC transporters and NRF2 targets, such as the heavy and light chains of γ-glutamylcysteine synthase. The decrease and increase in expression of BCRP mRNA mediated by NRF2- and KEAP1-silencing, respectively, suggest the involvement of the NRF2/KEAP1 system in the transcriptional regulation of BCRP (Adachi et al. 2007).

An advanced study by Singh et al. identified the ARE region located in the 5’-flanking region of the BCRP gene at -431 to -420 bp, which is responsible for NRF2-mediated expression in lung cancer cells in vitro and in vivo (Singh et al. 2010) (Fig. 1). This observation confirms previous findings of ARE located in the promoter regions of other multidrug ABC transporters, such as MRP1-3 (Stöckel et al. 2000; Kurz et al. 2001; Mahaffey et al. 2009). Similarly, in normal kidney tubular cells, knockdown of KEAP1 was found to increase BCRP expression. However, unlike in the previous studies where NRF2 activation increased BCRP expression, pharmacological activation of NRF2 did not elevate BCRP mRNA in this case (Jeong et al. 2015). In addition to transcriptional regulation, NRF2 has been reported to regulate BCRP through a post-transcriptional mechanism as well. For example, in ovarian and renal carcinoma cell lines, silencing of NRF2 led to the activation of miR-206, which targeted c-MET/EGFR and downregulated BCRP expression, resulting in increased intracellular accumulation of the anticancer drug doxorubicin (Singh et al. 2013; Choi et al. 2017).

NRF2/BCRP axis and PDT

PDT is an emerging chemotherapy strategy that utilizes photosensitizing drugs to selectively kill cancer cells by generating cytotoxic ROS upon light activation (Agostinis et al. 2011). PDT has gained increasing attention as a minimally invasive and more target-specific alternative to conventional chemotherapy (Dolmans et al. 2003; Correia et al. 2021). PDT has been approved by the FDA for the treatment of several types of cancer, including bladder, esophageal, head and neck, lung, and skin cancer (Correia et al. 2021). The expression of BCRP in cancer cells can limit the effectiveness of PDT, and therefore there has been interest in investigating the relationship between BCRP and PDT. One study used imatinib mesylate, a tyrosine kinase inhibitor (TKI) known to reduce BCRP levels, and found that it enhanced the accumulation of various photosensitizers, including 2-(1-hexyloxethyl)-2-devinyl pyropheophorbide-a (Photochlor), protoporphyrin IX, and the benzoporphyrin derivative monoacid ring A (Verteporfin), in ABCG+ human and murine cell lines both in vitro and in vivo (Liu et al. 2007). Another study showed that BCRP-silencing using siRNA or inhibition of its activity using a chemical inhibitor, resulted in increased intracellular accumulation of chlorin e6 and improved PDT efficacy in cancer cells. This suggests that targeting BCRP may be a promising strategy to enhance the efficacy of PDT in cancer therapy (Roh et al. 2017). In the doxorubicin-resistant ovarian cancer A2780DR cells, the increase of BCRP-mediated drug resistance is associated with c-MET elevation. Treating the A2780DR cells with BCRP inhibitor Ko143 or c-MET inhibitor resulted in the accumulation of the photosensitizer Pba within cancer cells, which in turn sensitized the cells to PDT (Jung et al. 2015). Another study showed that A431/BCRP lung cancer cells, which overexpress BCRP, are more resistant to Photofrin-mediated PDT. However, inhibition of BCRP with FTC reversed this resistance and increased the efficacy of PDT (Usuda et al. 2010). In line with this, NRF2-knockdown in several types of cancer cells, including triple-negative breast cancer, renal carcinoma, and colon cancer, led to an increase in the effectiveness of PDT by lowering the levels of BCRP (Choi et al. 2014). Furthermore, a recent report has demonstrated that nanoparticle-mediated NRF2 silencing can increase the sensitivity of tumor cells to oxidative stress during PDT. These findings further support the importance of the NRF2/BCRP pathway in the response of cancer cells to PDT (Sun et al. 2023).

NRF2/BCRP axis and CSC properties

Ever since the initial discovery of a subpopulation of leukemic stem cells characterized by the surface markers CD34+ and CD38, CSCs have been recognized as promising targets for cancer therapy (Lapidot et al. 1994). The unique characteristics of CSCs, including their ability of self-renewal and differentiation potential, have been shown to play a critical role in cancer malignancies such as metastasis, therapy resistance, and recurrence (Batlle and Clevers 2017). CSCs and normal stem cells share many common transcription factors and signaling pathways, including several pluripotent transcription factors such as octamer-binding transcription factor 4 (OCT4), krupple-like factor 4 (KLF4), MYC, homeobox protein Nanog (NANOG), and SRY (sex determining region Y)-box 2 (SOX2) (Yang et al. 2020). Furthermore, a subpopulation of cancer cells known as the side population has been identified as a chemo- and radio-therapy-resistant population, which is thought to be due to its high expression of BCRP. As a result, BCRP has become recognized as one of the CSC markers (Zhou et al. 2001).

Recent studies have suggested that NRF2 may contribute to the development or maintenance of CSC properties, including chemoresistance (Kahroba et al. 2019; Choi et al. 2021; Hallis et al. 2023a). In the colonosphere model of in vitro CSCs, increased levels of P-gp, BCRP, and concomitant NRF2 signaling were observed, which exhibited resistance to doxorubicin (Ryoo et al. 2016). Subsequently, NRF2 knockdown in the colonosphere model sensitized these cells to drug treatment by decreasing efflux transporter activities. Consistent with these findings, the CD133high population from HCT116 colon cancer cells exhibited high levels of NRF2 and BCRP, along with elevated CSC markers, indicating that they possess CSC phenotypes such as enhanced proliferation, migration, colony formation, chemoresistance, and sphere-forming capacity (Park et al. 2022). In this experimental setting, colonospheres derived from NRF2-silenced HCT116 expressed low levels of BCRP along with reduced KLF4 expression, which resulted in the suppression of sphere-forming capacity. A similar study involving NRF2 knockdown in pancreatic ductal adenocarcinoma cell lines BxPC-3 and CFPAC-1, which had high expression of NRF2, found that it decreased the expression of CSC transcription factors NANOG, OCT4, and CD133, as well as chemoresistance properties, by downregulating BCRP (Kim et al. 2020). Silencing of NRF2 in sorafenib-resistant Huh7 hepatocellular cell line also limited cancer stemness and migration, as well as expression of BCRP (Gao et al. 2021). In another drug resistance model, A2780DR ovarian cancer cells that exhibit doxorubicin resistance showed higher NRF2 signaling activity concomitantly with elevated expression of aldehyde dehydrogenase 1 (ALDH1) and BCRP, which supports to maintain CSC properties (Shim et al. 2009; Kim et al. 2018). Isolated CSCs from the side population of cervical cancer patients showed aberrant activation of NRF2, BCRP, and Bcl-2, which resulted in chemoresistance and apoptosis resistance (Jia et al. 2015). Meanwhile, our recent study demonstrated a positive association between NRF2 and HIF-2α-induced CSC phenotypes under chronic hypoxic conditions (Hallis et al. 2023b). As previous studies have reported that HIF-2α directly regulates BCRP and confers CSC properties in ovarian CSCs, leading to resistance to doxorubicin and promoting tumor growth, this may suggest the potential association of NRF2 with HIF-2α/BCRP in hypoxic tumor environment (He et al. 2019). Collectively, these studies indicate that NRF2 and BCRP are intricately intertwined in maintaining resistance to chemotherapeutic agents and photodynamic therapy, and the maintenance of CSC properties in cancer (Fig. 2).

Figure 2. Role of NRF2 and BCRP in CSC maintenance and PDT resistance. (A) NRF2 signaling is activated in CSCs models, contributing to CSC properties, including tumor initiation and growth, invasion, metastasis, and therapy resistance. NRF2 upregulates the antioxidants gene to maintain low ROS levels and increases ABC transporters such as BCRP to protect CSCs against external harmful chemicals, which eventually leads to therapy resistance. High levels of NRF2 are also associated with the upregulation of other transcription factors, including OCT4, NANOG, KLF4, MYC, and SOX2, which help to maintain CSC phenotypes. (B) Enhanced BCRP in cancers facilitates the efflux of photosensitizers such as Pba, photochlor, and photofirin, thus leading to PDT resistance. BCRP inhibition by chemical inhibitors increases intracellular accumulation of photosensitizers and consequent ROS generation, leading to cancer cell apoptosis. OCT4, octamer-binding transcription factor 4; KLF4, krupple-like factor 4; NANOG, homeobox protein nanog; SOX2, SRY (sex determining region Y)-box 2; FTC, fumitremorgin C.

Conclusion

In this review, we have highlighted the critical role of NRF2 and BCRP as potential targets to counteract the therapy resistance of cancers. Elevated expression of BCRP in most cancer cases results in a poor prognosis and is associated with multidrug resistance, thereby impeding the efficacy of cancer therapy (Robey et al. 2018). Many studies have demonstrated the potential effect of BCRP inhibitors in multiple models of resistant cancers, and current efforts are focusing on investigating BCRP inhibitors for clinical trials. Thus, the regulation of BCRP by transcription factors or other signaling pathways is of great interest. Although several transcription factors have been implicated, the identification of ARE sequences in the promoter region of BCRP has highlighted the direct regulation by NRF2 (Singh et al. 2010). This is important since NRF2 overexpression in cancer clinical settings is correlated with poor prognosis and chemoresistance (Solis et al. 2010). Moreover, recent insights suggest that the suppression of BCRP and its regulator NRF2 may have an impact on PDT resistance and the maintenance of CSC properties. Thus, ongoing efforts are focused on strategies aimed at suppressing NRF2 activity to increase the sensitivity of cancer patients to chemotherapy (Dinkova-Kostova and Copple 2023). In particular, compared to the inhibition of BCRP, the inhibition of NRF2 is expected to be a more effective strategy for cancer treatment in that it can suppress the expression of other target genes that contribute to cancer growth and development as well as control of BCRP-mediated anticancer drug resistance. Currently, extensive efforts are being made to develop NRF2 inhibitors. Several natural compounds, including luteolin, procyanidins, and wedelolactone, have demonstrated NRF2 inhibitory effects in various experimental settings involving NRF2-overactive cancers (Sharifi-Rad et al. 2023). Brusatol from Brucea javanica has shown potent anti-tumor and anti-metastatic activities by decreasing NRF2 protein levels, which is attributed to multiple mechanisms, including protein translation inhibition (He et al. 2023). In a high-throughput chemical library screening, halofuginone has been identified as an inhibitor of NRF2 protein synthesis (Tsuchida et al. 2017). ML385, derived from a high-throughput chemical library screening and subsequent chemical optimization, has been discovered to interfere with the interaction between NRF2 and sMAF by directly binding to the NRF2 protein. Consequently, ML385 demonstrated the ability to sensitize KEAP1 mutant lung cancer cells to chemotherapy (Singh et al. 2016). While NRF2 inhibitors hold strong potential for enhancing chemotherapy sensitivity and suppressing tumorigenesis in NRF2-high cancers, the dual functionality of NRF2 in normal cells presents a limitation for its clinical application. In this context, achieving cancer-selective inhibition of NRF2 becomes a crucial strategy. In a recent study employing a synthetic lethal strategy, mitomycin C has emerged as a selectively cytotoxic agent for NRF2-overactive cancer cells, primarily through enhanced bioactivation via NRF2-target genes, such as NQO1 (Baird and Yamamoto 2021). Geldanamycin-derived HSP90 inhibitors have demonstrated selective lethality towards NRF2-high cancer cells, and their mechanism of action has been attributed to the enhancement of NQO1-mediated bioactivation (Baird et al. 2020). In summary, although a strategy for NRF2 inhibitors that selectively act on NRF2-overexpressing cancers has not been developed, with the progression of more advanced studies on the NRF2-KEAP1 system, there is an optimistic expectation for the future discovery of highly selective and effective NRF2/BCRP inhibitors.

Acknowledgements

This work was supported by National Research Foundation of Korea (NRF) grants funded by the Korean government (MSIT) (2022R1A2C2011866).

Conflict of interest

The authors declare that they have no conflict of interest.

Fig 1.

Figure 1.BCRP regulation and its association with chemoresistance. Overactivation of NRF2 in cancer increases BCRP (ABCG2) as one of its targets, as well as other targets in antioxidant defense, metabolism, and drug detoxification, thereby promoting cell survival and resistance to therapy. The transcriptional regulation of BCRP can be controlled by additional transcription factors, including HIF-1α, HIF-2α, PPARγ, and estrogen, which can bind to their enhancer elements in the promoter sequence of BCRP genes. At the post-transcriptional level, miR-328 and miR-520h can directly suppress BCRP transcription, while NRF2-regulated miR-206 targets BCRP via the c-MET/EGFR signaling pathway. sMAFs, small MAF proteins; BCRP, breast cancer resistance protein; PPARγ, peroxisome proliferator-activated receptor-γ; HIF, hypoxia-inducible factor; ARE, antioxidant response element; PPRE, peroxisome proliferator-response element; HRE, hypoxia response element; ERE, estrogen response element; EGFR, epidermal growth factor receptor; c-MET, mesenchymal-epithelial transition factor.
Drug Targets and Therapeutics 2023; 2: 111-123https://doi.org/10.58502/DTT.23.0021

Fig 2.

Figure 2.Role of NRF2 and BCRP in CSC maintenance and PDT resistance. (A) NRF2 signaling is activated in CSCs models, contributing to CSC properties, including tumor initiation and growth, invasion, metastasis, and therapy resistance. NRF2 upregulates the antioxidants gene to maintain low ROS levels and increases ABC transporters such as BCRP to protect CSCs against external harmful chemicals, which eventually leads to therapy resistance. High levels of NRF2 are also associated with the upregulation of other transcription factors, including OCT4, NANOG, KLF4, MYC, and SOX2, which help to maintain CSC phenotypes. (B) Enhanced BCRP in cancers facilitates the efflux of photosensitizers such as Pba, photochlor, and photofirin, thus leading to PDT resistance. BCRP inhibition by chemical inhibitors increases intracellular accumulation of photosensitizers and consequent ROS generation, leading to cancer cell apoptosis. OCT4, octamer-binding transcription factor 4; KLF4, krupple-like factor 4; NANOG, homeobox protein nanog; SOX2, SRY (sex determining region Y)-box 2; FTC, fumitremorgin C.
Drug Targets and Therapeutics 2023; 2: 111-123https://doi.org/10.58502/DTT.23.0021

References

  1. Adachi T, Nakagawa H, Chung I, Hagiya Y, Hoshijima K, Noguchi N, Kuo MT, Ishikawa T (2007) Nrf2-dependent and -independent induction of ABC transporters ABCC1, ABCC2, and ABCG2 in HepG2 cells under oxidative stress. J Exp Ther Oncol 6:335-348.
    Pubmed
  2. Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, Hahn SM, Hamblin MR, Juzeniene A, Kessel D, Korbelik M, Moan J, Mroz P, Nowis D, Piette J, Wilson BC, Golab J (2011) Photodynamic therapy of cancer: an update. CA Cancer J Clin 61:250-281. doi: 10.3322/caac.20114
    Pubmed KoreaMed CrossRef
  3. Ahmed-Belkacem A, Pozza A, Muñoz-Martínez F, Bates SE, Castanys S, Gamarro F, Di Pietro A, Pérez-Victoria JM (2005) Flavonoid structure-activity studies identify 6-prenylchrysin and tectochrysin as potent and specific inhibitors of breast cancer resistance protein ABCG2. Cancer Res 65:4852-4860. doi: 10.1158/0008-5472.CAN-04-1817
    Pubmed CrossRef
  4. Allen JD, van Loevezijn A, Lakhai JM, van der Valk M, van Tellingen O, Reid G, Schellens JH, Koomen GJ, Schinkel AH (2002) Potent and specific inhibition of the breast cancer resistance protein multidrug transporter in vitro and in mouse intestine by a novel analogue of fumitremorgin C. Mol Cancer Ther 1:417-425.
    Pubmed
  5. Allikmets R, Schriml LM, Hutchinson A, Romano-Spica V, Dean M (1998) A human placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res 58:5337-5339.
    Pubmed
  6. Ashino T, Ohkubo-Morita H, Yamamoto M, Yoshida T, Numazawa S (2014) Possible involvement of nuclear factor erythroid 2-related factor 2 in the gene expression of Cyp2b10 and Cyp2a5. Redox Biol 2:284-288. doi: 10.1016/j.redox.2013.12.025
    Pubmed KoreaMed CrossRef
  7. Bai X, Chen Y, Hou X, Huang M, Jin J (2016) Emerging role of NRF2 in chemoresistance by regulating drug-metabolizing enzymes and efflux transporters. Drug Metab Rev 48:541-567. doi: 10.1080/03602532.2016.1197239
    Pubmed CrossRef
  8. Bailey-Dell KJ, Hassel B, Doyle LA, Ross DD (2001) Promoter characterization and genomic organization of the human breast cancer resistance protein (ATP-binding cassette transporter G2) gene. Biochim Biophys Acta 1520:234-241. doi: 10.1016/s0167-4781(01)00270-6
    Pubmed CrossRef
  9. Baird L, Suzuki T, Takahashi Y, Hishinuma E, Saigusa D, Yamamoto M (2020) Geldanamycin-derived HSP90 inhibitors are synthetic lethal with NRF2. Mol Cell Biol 40:e00377-e00320. doi: 10.1128/MCB.00377-20
    Pubmed KoreaMed CrossRef
  10. Baird L, Yamamoto M (2021) NRF2-dependent bioactivation of mitomycin C as a novel strategy to target Keap1-NRF2 pathway activation in human cancer. Mol Cell Biol 41:e00473-e00420. doi: 10.1128/MCB.00473-20
    Pubmed KoreaMed CrossRef
  11. Barbano R, Muscarella LA, Pasculli B, Valori VM, Fontana A, Coco M, la Torre A, Balsamo T, Poeta ML, Marangi GF, Maiello E, Castelvetere M, Pellegrini F, Murgo R, Fazio VM, Parrella P (2013) Aberrant Keap1 methylation in breast cancer and association with clinicopathological features. Epigenetics 8:105-112. doi: 10.4161/epi.23319
    Pubmed KoreaMed CrossRef
  12. Batlle E, Clevers H (2017) Cancer stem cells revisited. Nat Med 23:1124-1134. doi: 10.1038/nm.4409
    Pubmed CrossRef
  13. Best SA, Ding S, Kersbergen A, Dong X, Song JY, Xie Y, Reljic B, Li K, Vince JE, Rathi V, Wright GM, Ritchie ME, Sutherland KD (2019) Distinct initiating events underpin the immune and metabolic heterogeneity of KRAS-mutant lung adenocarcinoma. Nat Commun 10:4190. doi: 10.1038/s41467-019-12164-y
    Pubmed KoreaMed CrossRef
  14. Bhatia A, Schäfer HJ, Hrycyna CA (2005) Oligomerization of the human ABC transporter ABCG2: evaluation of the native protein and chimeric dimers. Biochemistry 44:10893-10904. doi: 10.1021/bi0503807
    Pubmed CrossRef
  15. Chen ZS, Robey RW, Belinsky MG, Shchaveleva I, Ren XQ, Sugimoto Y, Ross DD, Bates SE, Kruh GD (2003) Transport of methotrexate, methotrexate polyglutamates, and 17beta-estradiol 17-(beta-D-glucuronide) by ABCG2: effects of acquired mutations at R482 on methotrexate transport. Cancer Res 63:4048-4054.
    Pubmed
  16. Choi BH, Kim JM, Kwak MK (2021) The multifaceted role of NRF2 in cancer progression and cancer stem cells maintenance. Arch Pharm Res 44:263-280. doi: 10.1007/s12272-021-01316-8
    Pubmed CrossRef
  17. Choi B, Kwak MK (2016) Shadows of NRF2 in cancer: resistance to chemotherapy. Curr Opin Toxicol 1:20-28. doi: 10.1016/j.cotox.2016.08.003
    CrossRef
  18. Choi BH, Ryoo IG, Kang HC, Kwak MK (2014) The sensitivity of cancer cells to pheophorbide a-based photodynamic therapy is enhanced by Nrf2 silencing. PLoS One 9:e107158. doi: 10.1371/journal.pone.0107158
    Pubmed KoreaMed CrossRef
  19. Choi BH, Ryu DY, Ryoo IG, Kwak MK (2017) NFE2L2/NRF2 silencing-inducible miR-206 targets c-MET/EGFR and suppresses BCRP/ABCG2 in cancer cells. Oncotarget 8:107188-107205. doi: 10.18632/oncotarget.22513
    Pubmed KoreaMed CrossRef
  20. Chowdhry S, Zhang Y, McMahon M, Sutherland C, Cuadrado A, Hayes JD (2013) Nrf2 is controlled by two distinct β-TrCP recognition motifs in its Neh6 domain, one of which can be modulated by GSK-3 activity. Oncogene 2:3765-3781. doi: 10.1038/onc.2012.388
    Pubmed KoreaMed CrossRef
  21. Cole SP, Bhardwaj G, Gerlach JH, Mackie JE, Grant CE, Almquist KC, Stewart AJ, Kurz EU, Duncan AM, Deeley RG (1992) Overexpression of a transporter gene in a multidrug-resistant human lung cancer cell line. Science 258:1650-1654. doi: 10.1126/science.1360704
    Pubmed CrossRef
  22. Correia JH, Rodrigues JA, Pimenta S, Dong T, Yang Z (2021) Photodynamic therapy review: principles, photosensitizers, applications, and future directions. Pharmaceutics 13:1332. doi: 10.3390/pharmaceutics13091332
    Pubmed KoreaMed CrossRef
  23. de Bruin M, Miyake K, Litman T, Robey R, Bates SE (1999) Reversal of resistance by GF120918 in cell lines expressing the ABC half-transporter, MXR. Cancer Lett 146:117-126. doi: 10.1016/s0304-3835(99)00182-2
    Pubmed CrossRef
  24. de Vries NA, Zhao J, Kroon E, Buckle T, Beijnen JH, van Tellingen O (2007) P-glycoprotein and breast cancer resistance protein: two dominant transporters working together in limiting the brain penetration of topotecan. Clin Cancer Res 13:6440-6449. doi: 10.1158/1078-0432.CCR-07-1335
    Pubmed CrossRef
  25. Dean M, Rzhetsky A, Allikmets R (2001) The human ATP-binding cassette (ABC) transporter superfamily. Genome Res 11:1156-1166. doi: 10.1101/gr.184901
    Pubmed CrossRef
  26. Diestra JE, Scheffer GL, Català I, Maliepaard M, Schellens JH, Scheper RJ, Germà-Lluch JR, Izquierdo MA (2002) Frequent expression of the multi-drug resistance-associated protein BCRP/MXR/ABCP/ABCG2 in human tumours detected by the BXP-21 monoclonal antibody in paraffin-embedded material. J Pathol 198:213-219. doi: 10.1002/path.1203
    Pubmed CrossRef
  27. Dinkova-Kostova AT, Copple IM (2023) Advances and challenges in therapeutic targeting of NRF2. Trends Pharmacol Sci 44:137-149. doi: 10.1016/j.tips.2022.12.003
    Pubmed CrossRef
  28. Dolmans DE, Fukumura D, Jain RK (2003) Photodynamic therapy for cancer. Nat Rev Cancer 3:380-387. doi: 10.1038/nrc1071
    Pubmed CrossRef
  29. Doyle L, Ross DD (2003) Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2). Oncogene 22:7340-7358. doi: 10.1038/sj.onc.1206938
    Pubmed CrossRef
  30. Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, Ross DD (1998) A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci U S A 95:15665-15670. doi: 10.1073/pnas.95.26.15665
    Pubmed KoreaMed CrossRef
  31. Ee PL, Kamalakaran S, Tonetti D, He X, Ross DD, Beck WT (2004) Identification of a novel estrogen response element in the breast cancer resistance protein (ABCG2) gene. Cancer Res 64:1247-1251. doi: 10.1158/0008-5472.can-03-3583
    Pubmed CrossRef
  32. Gao L, Morine Y, Yamada S, Saito Y, Ikemoto T, Tokuda K, Takasu C, Miyazaki K, Shimada M (2021) Nrf2 signaling promotes cancer stemness, migration, and expression of ABC transporter genes in sorafenib-resistant hepatocellular carcinoma cells. PLoS One 16:e0256755. doi: 10.1371/journal.pone.0256755
    Pubmed KoreaMed CrossRef
  33. Goldstein LD, Lee J, Gnad F, Klijn C, Schaub A, Reeder J, Daemen A, Bakalarski CE, Holcomb T, Shames DS, Hartmaier RJ, Chmielecki J, Seshagiri S, Gentleman R, Stokoe D (2016) Recurrent loss of NFE2L2 exon 2 is a mechanism for Nrf2 pathway activation in human cancers. Cell Rep 16:2605-2617. doi: 10.1016/j.celrep.2016.08.010
    Pubmed CrossRef
  34. Gottesman MM, Lavi O, Hall MD, Gillet JP (2016) Toward a better understanding of the complexity of cancer drug resistance. Annu Rev Pharmacol Toxicol 56:85-102. doi: 10.1146/annurev-pharmtox-010715-103111
    Pubmed CrossRef
  35. Gutmann H, Hruz P, Zimmermann C, Beglinger C, Drewe J (2005) Distribution of breast cancer resistance protein (BCRP/ABCG2) mRNA expression along the human GI tract. Biochem Pharmacol 70:695-699. doi: 10.1016/j.bcp.2005.05.031
    Pubmed CrossRef
  36. Hallis SP, Kim JM, Kwak MK (2023a) Emerging role of NRF2 signaling in cancer stem cell phenotype. Mol Cells 46:153-164. doi: 10.14348/molcells.2023.2196
    Pubmed KoreaMed CrossRef
  37. Hallis SP, Kim SK, Lee JH, Kwak MK (2023b) Association of NRF2 with HIF-2α-induced cancer stem cell phenotypes in chronic hypoxic condition. Redox Biol 60:102632. doi: 10.1016/j.redox.2023.102632
    Pubmed KoreaMed CrossRef
  38. Hanada N, Takahata T, Zhou Q, Ye X, Sun R, Itoh J, Ishiguro A, Kijima H, Mimura J, Itoh K, Fukuda S, Saijo Y (2012) Methylation of the Keap1 gene promoter region in human colorectal cancer. BMC Cancer 12:66. doi: 10.1186/1471-2407-12-66
    Pubmed KoreaMed CrossRef
  39. He M, Wu H, Jiang Q, Liu Y, Han L, Yan Y, Wei B, Liu F, Deng X, Chen H, Zhao L, Wang M, Wu X, Yao W, Zhao H, Chen J, Wei M (2019) Hypoxia-inducible factor-2α directly promotes BCRP expression and mediates the resistance of ovarian cancer stem cells to adriamycin. Mol Oncol 13:403-421. doi: 10.1002/1878-0261.12419
    Pubmed KoreaMed CrossRef
  40. He T, Zhou F, Su A, Zhang Y, Xing Z, Mi L, Li Z, Wu W (2023) Brusatol: a potential sensitizing agent for cancer therapy from Brucea javanica. Biomed Pharmacother 158:114134. doi: 10.1016/j.biopha.2022.114134
    Pubmed CrossRef
  41. Henriksen U, Fog JU, Litman T, Gether U (2005) Identification of intra- and intermolecular disulfide bridges in the multidrug resistance transporter ABCG2. J Biol Chem 280:36926-36934. doi: 10.1074/jbc.M502937200
    Pubmed CrossRef
  42. Hirano M, Maeda K, Matsushima S, Nozaki Y, Kusuhara H, Sugiyama Y (2005) Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin. Mol Pharmacol 68:800-807. doi: 10.1124/mol.105.014019
    Pubmed CrossRef
  43. Hirotsu Y, Katsuoka F, Funayama R, Nagashima T, Nishida Y, Nakayama K, Engel JD, Yamamoto M (2012) Nrf2-MafG heterodimers contribute globally to antioxidant and metabolic networks. Nucleic Acids Res 40:10228-10239. doi: 10.1093/nar/gks827
    Pubmed KoreaMed CrossRef
  44. Huls M, Brown CD, Windass AS, Sayer R, van den Heuvel JJ, Heemskerk S, Russel FG, Masereeuw R (2008) The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane. Kidney Int 73:220-225. doi: 10.1038/sj.ki.5002645
    Pubmed CrossRef
  45. Ichimura Y, Komatsu M (2018) Activation of p62/SQSTM1-Keap1-nuclear factor erythroid 2-related factor 2 pathway in cancer. Front Oncol 8:210. doi: 10.3389/fonc.2018.00210
    Pubmed KoreaMed CrossRef
  46. Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, Yamamoto M, Nabeshima Y (1997) An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun 236:313-322. doi: 10.1006/bbrc.1997.6943
    Pubmed CrossRef
  47. Jeong HS, Ryoo IG, Kwak MK (2015) Regulation of the expression of renal drug transporters in Keap1-knockdown human tubular cells. Toxicol In Vitro 9:884-892. doi: 10.1016/j.tiv.2015.03.013
    Pubmed CrossRef
  48. Ji L, Li H, Gao P, Shang G, Zhang DD, Zhang N, Jiang T (2013) Nrf2 pathway regulates multidrug-resistance-associated protein 1 in small cell lung cancer. PLoS One 8:e63404. doi: 10.1371/journal.pone.0063404
    Pubmed KoreaMed CrossRef
  49. Jia Y, Chen J, Zhu H, Jia ZH, Cui MH (2015) Aberrantly elevated redox sensing factor Nrf2 promotes cancer stem cell survival via enhanced transcriptional regulation of ABCG2 and Bcl-2/Bmi-1 genes. Oncol Rep 34:2296-2304. doi: 10.3892/or.2015.4214
    Pubmed CrossRef
  50. Jigorel E, Le Vee M, Boursier-Neyret C, Parmentier Y, Fardel O (2006) Differential regulation of sinusoidal and canalicular hepatic drug transporter expression by xenobiotics activating drug-sensing receptors in primary human hepatocytes. Drug Metab Dispos 34:1756-1763. doi: 10.1124/dmd.106.010033
    Pubmed CrossRef
  51. Jonker JW, Smit JW, Brinkhuis RF, Maliepaard M, Beijnen JH, Schellens JH, Schinkel AH (2000) Role of breast cancer resistance protein in the bioavailability and fetal penetration of topotecan. J Natl Cancer Inst 92:1651-1656. doi: 10.1093/jnci/92.20.1651
    Pubmed CrossRef
  52. Jung KA, Choi BH, Kwak MK (2015) The c-MET/PI3K signaling is associated with cancer resistance to doxorubicin and photodynamic therapy by elevating BCRP/ABCG2 expression. Mol Pharmacol 87:465-476. doi: 10.1124/mol.114.096065
    Pubmed CrossRef
  53. Jung KA, Choi BH, Nam CW, Song M, Kim ST, Lee JY, Kwak MK (2013) Identification of aldo-keto reductases as NRF2-target marker genes in human cells. Toxicol Lett 218:39-49. doi: 10.1016/j.toxlet.2012.12.026
    Pubmed CrossRef
  54. Kahroba H, Shirmohamadi M, Hejazi MS, Samadi N (2019) The role of Nrf2 signaling in cancer stem cells: from stemness and self-renewal to tumorigenesis and chemoresistance. Life Sci 239:116986. doi: 10.1016/j.lfs.2019.116986
    Pubmed CrossRef
  55. Kawabata S, Oka M, Shiozawa K, Tsukamoto K, Nakatomi K, Soda H, Fukuda M, Ikegami Y, Sugahara K, Yamada Y, Kamihira S, Doyle LA, Ross DD, Kohno S (2001) Breast cancer resistance protein directly confers SN-38 resistance of lung cancer cells. Biochem Biophys Res Commun 280:1216-1223. doi: 10.1006/bbrc.2001.4267
    Pubmed CrossRef
  56. Kim D, Choi BH, Ryoo IG, Kwak MK (2018) High NRF2 level mediates cancer stem cell-like properties of aldehyde dehydrogenase (ALDH)-high ovarian cancer cells: inhibitory role of all-trans retinoic acid in ALDH/NRF2 signaling. Cell Death Dis 9:896. doi: 10.1038/s41419-018-0903-4
    Pubmed KoreaMed CrossRef
  57. Kim EJ, Kim YJ, Lee HI, Jeong SH, Nam HJ, Cho JH (2020) NRF2 knockdown resensitizes 5-fluorouracil-resistant pancreatic cancer cells by suppressing HO-1 and ABCG2 expression. Int J Mol Sci 21:4646. doi: 10.3390/ijms21134646
    Pubmed KoreaMed CrossRef
  58. Kitamura H, Motohashi H (2018) NRF2 addiction in cancer cells. Cancer Sci 109:900-911. doi: 10.1111/cas.13537
    Pubmed KoreaMed CrossRef
  59. Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, Igarashi K, Yamamoto M (2004) Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol Cell Biol 24:7130-7139. doi: 10.1128/MCB.24.16.7130-7139.2004
    Pubmed KoreaMed CrossRef
  60. Krishnamurthy P, Ross DD, Nakanishi T, Bailey-Dell K, Zhou S, Mercer KE, Sarkadi B, Sorrentino BP, Schuetz JD (2004) The stem cell marker Bcrp/ABCG2 enhances hypoxic cell survival through interactions with heme. J Biol Chem 279:24218-24225. doi: 10.1074/jbc.M313599200
    Pubmed CrossRef
  61. Kurz EU, Cole SP, Deeley RG (2001) Identification of DNA-protein interactions in the 5' flanking and 5' untranslated regions of the human multidrug resistance protein (MRP1) gene: evaluation of a putative antioxidant response element/AP-1 binding site. Biochem Biophys Res Commun 285:981-990. doi: 10.1006/bbrc.2001.5262
    Pubmed CrossRef
  62. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE (1994) A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367:645-648. doi: 10.1038/367645a0
    Pubmed CrossRef
  63. Liu W, Baer MR, Bowman MJ, Pera P, Zheng X, Morgan J, Pandey RA, Oseroff AR (2007) The tyrosine kinase inhibitor imatinib mesylate enhances the efficacy of photodynamic therapy by inhibiting ABCG2. Clin Cancer Res 13:2463-2470. doi: 10.1158/1078-0432.CCR-06-1599
    Pubmed CrossRef
  64. Low SK, Fukunaga K, Takahashi A, Matsuda K, Hongo F, Nakanishi H, Kitamura H, Inoue T, Kato Y, Tomita Y, Fukasawa S, Tanaka T, Nishimura K, Uemura H, Hara I, Fujisawa M, Matsuyama H, Hashine K, Tatsugami K, Enokida H, Kubo M, Miki T, Mushiroda T (2016) Association study of a functional variant on ABCG2 gene with sunitinib-induced severe adverse drug reaction. PLoS One 11:e0148177. doi: 10.1371/journal.pone.0148177
    Pubmed KoreaMed CrossRef
  65. Mahaffey CM, Zhang H, Rinna A, Holland W, Mack PC, Forman HJ (2009) Multidrug-resistant protein-3 gene regulation by the transcription factor Nrf2 in human bronchial epithelial and non-small-cell lung carcinoma. Free Radic Biol Med 46:1650-1657. doi: 10.1016/j.freeradbiomed.2009.03.023
    Pubmed KoreaMed CrossRef
  66. Maliepaard M, van Gastelen MA, de Jong LA, Pluim D, van Waardenburg RC, Ruevekamp-Helmers MC, Floot BG, Schellens JH (1999) Overexpression of the BCRP/MXR/ABCP gene in a topotecan-selected ovarian tumor cell line. Cancer Res 59:4559-4563.
  67. Mao Q, Unadkat JD (2015) Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport--an update. AAPS J 17:65-82. doi: 10.1208/s12248-014-9668-6
    Pubmed KoreaMed CrossRef
  68. Martin V, Xu J, Pabbisetty SK, Alonso MM, Liu D, Lee OH, Gumin J, Bhat KP, Colman H, Lang FF, Fueyo J, Gomez-Manzano C (2009) Tie2-mediated multidrug resistance in malignant gliomas is associated with upregulation of ABC transporters. Oncogene 28:2358-2363. doi: 10.1038/onc.2009.103
    Pubmed KoreaMed CrossRef
  69. Marzac C, Garrido E, Tang R, Fava F, Hirsch P, De Benedictis C, Corre E, Lapusan S, Lallemand JY, Marie JP, Jacquet E, Legrand O (2011) ATP Binding Cassette transporters associated with chemoresistance: transcriptional profiling in extreme cohorts and their prognostic impact in a cohort of 281 acute myeloid leukemia patients. Haematologica 96:1293-1301. doi: 10.3324/haematol.2010.031823
    Pubmed KoreaMed CrossRef
  70. McMahon M, Thomas N, Itoh K, Yamamoto M, Hayes JD (2006) Dimerization of substrate adaptors can facilitate cullin-mediated ubiquitylation of proteins by a "tethering" mechanism: a two-site interaction model for the Nrf2-Keap1 complex. J Biol Chem 281:24756-24768. doi: 10.1074/jbc.M601119200
    Pubmed CrossRef
  71. Miyake K, Mickley L, Litman T, Zhan Z, Robey R, Cristensen B, Brangi M, Greenberger L, Dean M, Fojo T, Bates SE (1999) Molecular cloning of cDNAs which are highly overexpressed in mitoxantrone-resistant cells: demonstration of homology to ABC transport genes. Cancer Res 59:8-13.
    Pubmed
  72. Mukhopadhyay S, Goswami D, Adiseshaiah PP, Burgan W, Yi M, Guerin TM, Kozlov SV, Nissley DV, McCormick F (2020) Undermining glutaminolysis bolsters chemotherapy while NRF2 promotes chemoresistance in KRAS-driven pancreatic cancers. Cancer Res 80:1630-1643. doi: 10.1158/0008-5472.CAN-19-1363
    Pubmed KoreaMed CrossRef
  73. Nakanishi T, Bailey-Dell KJ, Hassel BA, Shiozawa K, Sullivan DM, Turner J, Ross DD (2006) Novel 5' untranslated region variants of BCRP mRNA are differentially expressed in drug-selected cancer cells and in normal human tissues: implications for drug resistance, tissue-specific expression, and alternative promoter usage. Cancer Res 66:5007-5011. doi: 10.1158/0008-5472.CAN-05-4572
    Pubmed CrossRef
  74. Ohta T, Iijima K, Miyamoto M, Nakahara I, Tanaka H, Ohtsuji M, Suzuki T, Kobayashi A, Yokota J, Sakiyama T, Shibata T, Yamamoto M, Hirohashi S (2008) Loss of Keap1 function activates Nrf2 and provides advantages for lung cancer cell growth. Cancer Res 68:1303-1309. doi: 10.1158/0008-5472.CAN-07-5003
    Pubmed CrossRef
  75. Ozvegy C, Litman T, Szakács G, Nagy Z, Bates S, Váradi A, Sarkadi B (2001) Functional characterization of the human multidrug transporter, ABCG2, expressed in insect cells. Biochem Biophys Res Commun 285:111-117. doi: 10.1006/bbrc.2001.5130
    Pubmed CrossRef
  76. Pan YZ, Morris ME, Yu AM (2009) MicroRNA-328 negatively regulates the expression of breast cancer resistance protein (BCRP/ABCG2) in human cancer cells. Mol Pharmacol 75:1374-1379. doi: 10.1124/mol.108.054163
    Pubmed KoreaMed CrossRef
  77. Park J, Kim SK, Hallis SP, Choi BH, Kwak MK (2022) Role of CD133/NRF2 axis in the development of colon cancer stem cell-like properties. Front Oncol 11:808300. doi: 10.3389/fonc.2021.808300
    Pubmed KoreaMed CrossRef
  78. Raaijmakers MH, de Grouw EP, Heuver LH, van der Reijden BA, Jansen JH, Scheper RJ, Scheffer GL, de Witte TJ, Raymakers RA (2005) Breast cancer resistance protein in drug resistance of primitive CD34+38- cells in acute myeloid leukemia. Clin Cancer Res 11:2436-2444. doi: 10.1158/1078-0432.CCR-04-0212
    Pubmed CrossRef
  79. Rabindran SK, He H, Singh M, Brown E, Collins KI, Annable T, Greenberger LM (1998) Reversal of a novel multidrug resistance mechanism in human colon carcinoma cells by fumitremorgin C. Cancer Res 58:5850-5858.
    Pubmed
  80. Robey RW, Honjo Y, van de Laar A, Miyake K, Regis JT, Litman T, Bates SE (2001a) A functional assay for detection of the mitoxantrone resistance protein, MXR (ABCG2). Biochim Biophys Acta 1512:171-182. doi: 10.1016/s0005-2736(01)00308-x
    CrossRef
  81. Robey RW, Medina-Pérez WY, Nishiyama K, Lahusen T, Miyake K, Litman T, Senderowicz AM, Ross DD, Bates SE (2001b) Overexpression of the ATP-binding cassette half-transporter, ABCG2 (Mxr/BCrp/ABCP1), in flavopiridol-resistant human breast cancer cells. Clin Cancer Res 7:145-152.
  82. Robey RW, Pluchino KM, Hall MD, Fojo AT, Bates SE, Gottesman MM (2018) Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat Rev Cancer 18:452-464. doi: 10.1038/s41568-018-0005-8
    Pubmed KoreaMed CrossRef
  83. Robey RW, To KK, Polgar O, Dohse M, Fetsch P, Dean M, Bates SE (2009) ABCG2: a perspective. Adv Drug Deliv Rev 61:3-13. doi: 10.1016/j.addr.2008.11.003
    Pubmed KoreaMed CrossRef
  84. Roh YJ, Kim JH, Kim IW, Na K, Park JM, Choi MG (2017) Photodynamic therapy using photosensitizer-encapsulated polymeric nanoparticle to overcome ATP-binding cassette transporter subfamily G2 function in pancreatic cancer. Mol Cancer Ther 16:1487-1496. doi: 10.1158/1535-7163.MCT-16-0642
    Pubmed CrossRef
  85. Rojo de la Vega M, Chapman E, Zhang DD (2018) NRF2 and the hallmarks of cancer. Cancer Cell 34:21-43. doi: 10.1016/j.ccell.2018.03.022
    Pubmed KoreaMed CrossRef
  86. Roninson IB, Chin JE, Choi KG, Gros P, Housman DE, Fojo A, Shen DW, Gottesman MM, Pastan I (1986) Isolation of human mdr DNA sequences amplified in multidrug-resistant KB carcinoma cells. Proc Natl Acad Sci U S A 83:4538-4542. doi: 10.1073/pnas.83.12.4538
    Pubmed KoreaMed CrossRef
  87. Ross DD, Yang W, Abruzzo LV, Dalton WS, Schneider E, Lage H, Dietel M, Greenberger L, Cole SP, Doyle LA (1999) Atypical multidrug resistance: breast cancer resistance protein messenger RNA expression in mitoxantrone-selected cell lines. J Natl Cancer Inst 91:429-433. doi: 10.1093/jnci/91.5.429
    Pubmed CrossRef
  88. Ryoo IG, Kim G, Choi BH, Lee SH, Kwak MK (2016) Involvement of NRF2 signaling in doxorubicin resistance of cancer stem cell-enriched colonospheres. Biomol Ther (Seoul) 24:482-488. doi: 10.4062/biomolther.2016.145
    Pubmed KoreaMed CrossRef
  89. Saito T, Ichimura Y, Taguchi K, Suzuki T, Mizushima T, Takagi K, Hirose Y, Nagahashi M, Iso T, Fukutomi T, Ohishi M, Endo K, Uemura T, Nishito Y, Okuda S, Obata M, Kouno T, Imamura R, Tada Y, Obata R, Yasuda D, Takahashi K, Fujimura T, Pi J, Lee MS, Ueno T, Ohe T, Mashino T, Wakai T, Kojima H, Okabe T, Nagano T, Motohashi H, Waguri S, Soga T, Yamamoto M, Tanaka K, Komatsu M (2016) p62/Sqstm1 promotes malignancy of HCV-positive hepatocellular carcinoma through Nrf2-dependent metabolic reprogramming. Nat Commun 7:12030. doi: 10.1038/ncomms12030
    Pubmed KoreaMed CrossRef
  90. Sekhar KR, Rachakonda G, Freeman ML (2010) Cysteine-based regulation of the CUL3 adaptor protein Keap1. Toxicol Appl Pharmacol 244:21-26. doi: 10.1016/j.taap.2009.06.016
    Pubmed KoreaMed CrossRef
  91. Sesink AL, Arts IC, de Boer VC, Breedveld P, Schellens JH, Hollman PC, Russel FG (2005) Breast cancer resistance protein (Bcrp1/Abcg2) limits net intestinal uptake of quercetin in rats by facilitating apical efflux of glucuronides. Mol Pharmacol 67:1999-2006. doi: 10.1124/mol.104.009753
    Pubmed CrossRef
  92. Sharifi-Rad J, Seidel V, Izabela M, Monserrat-Mequida M, Sureda A, Ormazabal V, Zuniga FA, Mangalpady SS, Pezzani R, Ydyrys A, Tussupbekova G, Martorell M, Calina D, Cho WC (2023) Phenolic compounds as Nrf2 inhibitors: potential applications in cancer therapy. Cell Commun Signal 21:89. doi: 10.1186/s12964-023-01109-0
    Pubmed KoreaMed CrossRef
  93. Shibata T, Ohta T, Tong KI, Kokubu A, Odogawa R, Tsuta K, Asamura H, Yamamoto M, Hirohashi S (2008) Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Proc Natl Acad Sci U S A 105:13568-13573. doi: 10.1073/pnas.0806268105
    Pubmed KoreaMed CrossRef
  94. Shim GS, Manandhar S, Shin DH, Kim TH, Kwak MK (2009) Acquisition of doxorubicin resistance in ovarian carcinoma cells accompanies activation of the NRF2 pathway. Free Radic Biol Med 47:1619-1631. doi: 10.1016/j.freeradbiomed.2009.09.006
    Pubmed CrossRef
  95. Singh A, Happel C, Manna SK, Acquaah-Mensah G, Carrerero J, Kumar S, Nasipuri P, Krausz KW, Wakabayashi N, Dewi R, Boros LG, Gonzalez FJ, Gabrielson E, Wong KK, Girnun G, Biswal S (2013) Transcription factor NRF2 regulates miR-1 and miR-206 to drive tumorigenesis. J Clin Invest 123:2921-2934. doi: 10.1172/JCI66353
    Pubmed KoreaMed CrossRef
  96. Singh A, Misra V, Thimmulappa RK, Lee H, Ames S, Hoque MO, Herman JG, Baylin SB, Sidransky D, Gabrielson E, Brock MV, Biswal S (2006) Dysfunctional Keap1-NRF2 interaction in non-small-cell lung cancer. PLoS Med 3:e420. doi: 10.1371/journal.pmed.0030420
    Pubmed KoreaMed CrossRef
  97. Singh A, Venkannagari S, Oh KH, Zhang YQ, Rohde JM, Liu L, Nimmagadda S, Sudini K, Brimacombe KR, Gajghate S, Ma J, Wang A, Xu X, Shahane SA, Xia M, Woo J, Mensah GA, Wang Z, Ferrer M, Gabrielson E, Li Z, Rastinejad F, Shen M, Boxer MB, Biswal S (2016) Small molecule inhibitor of NRF2 selectively intervenes therapeutic resistance in Keap1-deficient NSCLC tumors. ACS Chem Biol 11:3214-3225. doi: 10.1021/acschembio.6b00651
    Pubmed KoreaMed CrossRef
  98. Singh A, Wu H, Zhang P, Happel C, Ma J, Biswal S (2010) Expression of ABCG2 (BCRP) is regulated by Nrf2 in cancer cells that confers side population and chemoresistance phenotype. Mol Cancer Ther 9:2365-2376. doi: 10.1158/1535-7163.MCT-10-0108
    Pubmed KoreaMed CrossRef
  99. Solis LM, Behrens C, Dong W, Suraokar M, Ozburn NC, Moran CA, Corvalan AH, Biswal S, Swisher SG, Bekele BN, Minna JD, Stewart DJ, Wistuba II (2010) Nrf2 and Keap1 abnormalities in non-small cell lung carcinoma and association with clinicopathologic features. Clin Cancer Res 16:3743-3753. doi: 10.1158/1078-0432.CCR-09-3352
    Pubmed KoreaMed CrossRef
  100. ivastava R Sr, Fernández-Ginés R, Encinar JA, Cuadrado A, Wells G (2022) The current status and future prospects for therapeutic targeting of Keap1-NRF2 and β-TrCP-NRF2 interactions in cancer chemoresistance. Free Radic Biol Med 192:246-260. doi: 10.1016/j.freeradbiomed.2022.09.023
    Pubmed CrossRef
  101. Stöckel B, König J, Nies AT, Cui Y, Brom M, Keppler D (2000) Characterization of the 5'-flanking region of the human multidrug resistance protein 2 (MRP2) gene and its regulation in comparison withthe multidrug resistance protein 3 (MRP3) gene. Eur J Biochem 267:1347-1358. doi: 10.1046/j.1432-1327.2000.01106.x
    Pubmed CrossRef
  102. Sugimoto Y, Tsukahara S, Imai Y, Sugimoto Y, Ueda K, Tsuruo T (2003) Reversal of breast cancer resistance protein-mediated drug resistance by estrogen antagonists and agonists. Mol Cancer Ther 2:105-112.
    Pubmed CrossRef
  103. Sun S, Yu M, Yu L, Huang W, Zhu M, Fu Y, Yan L, Wang Q, Ji X, Zhao J, Wu M (2023) Nrf2 silencing amplifies DNA photooxidative damage to activate the STING pathway for synergistic tumor immunotherapy. Biomaterials 296:122068. doi: 10.1016/j.biomaterials.2023.122068
    Pubmed CrossRef
  104. Szatmari I, Vámosi G, Brazda P, Balint BL, Benko S, Széles L, Jeney V, Ozvegy-Laczka C, Szántó A, Barta E, Balla J, Sarkadi B, Nagy L (2006) Peroxisome proliferator-activated receptor gamma-regulated ABCG2 expression confers cytoprotection to human dendritic cells. J Biol Chem 281:23812-23823. doi: 10.1074/jbc.M604890200
    Pubmed CrossRef
  105. Tao S, Wang S, Moghaddam SJ, Ooi A, Chapman E, Wong PK, Zhang DD (2014) Oncogenic KRAS confers chemoresistance by upregulating NRF2. Cancer Res 74:7430-7441. doi: 10.1158/0008-5472.CAN-14-1439
    Pubmed KoreaMed CrossRef
  106. Taylor NMI, Manolaridis I, Jackson SM, Kowal J, Stahlberg H, Locher KP (2017) Structure of the human multidrug transporter ABCG2. Nature 546:504-509. doi: 10.1038/nature22345
    Pubmed CrossRef
  107. To KK, Zhan Z, Litman T, Bates SE (2008) Regulation of ABCG2 expression at the 3' untranslated region of its mRNA through modulation of transcript stability and protein translation by a putative microRNA in the S1 colon cancer cell line. Mol Cell Biol 28:5147-5161. doi: 10.1128/MCB.00331-08
    Pubmed KoreaMed CrossRef
  108. Tong KI, Katoh Y, Kusunoki H, Itoh K, Tanaka T, Yamamoto M (2006) Keap1 recruits Neh2 through binding to ETGE and DLG motifs: characterization of the two-site molecular recognition model. Mol Cell Biol 26:2887-2900. doi: 10.1128/MCB.26.8.2887-2900.2006
    Pubmed KoreaMed CrossRef
  109. Tsuchida K, Tsujita T, Hayashi M, Ojima A, Keleku-Lukwete N, Katsuoka F, Otsuki A, Kikuchi H, Oshima Y, Suzuki M, Yamamoto M (2017) Halofuginone enhances the chemo-sensitivity of cancer cells by suppressing NRF2 accumulation. Free Radic Biol Med 103:236-247. doi: 10.1016/j.freeradbiomed.2016.12.041
    Pubmed CrossRef
  110. Usuda J, Tsunoda Y, Ichinose S, Ishizumi T, Ohtani K, Maehara S, Ono S, Tsutsui H, Ohira T, Okunaka T, Furukawa K, Sugimoto Y, Kato H, Ikeda N (2010) Breast cancer resistant protein (BCRP) is a molecular determinant of the outcome of photodynamic therapy (PDT) for centrally located early lung cancer. Lung Cancer 67:198-204. doi: 10.1016/j.lungcan.2009.04.002
    Pubmed CrossRef
  111. Wang F, Xue X, Wei J, An Y, Yao J, Cai H, Wu J, Dai C, Qian Z, Xu Z, Miao Y (2010) hsa-miR-520h downregulates ABCG2 in pancreatic cancer cells to inhibit migration, invasion, and side populations. Br J Cancer 103:567-574. doi: 10.1038/sj.bjc.6605724
    Pubmed KoreaMed CrossRef
  112. Wang H, Lee EW, Zhou L, Leung PC, Ross DD, Unadkat JD, Mao Q (2008) Progesterone receptor (PR) isoforms PRA and PRB differentially regulate expression of the breast cancer resistance protein in human placental choriocarcinoma BeWo cells. Mol Pharmacol 73:845-854. doi: 10.1124/mol.107.041087
    Pubmed CrossRef
  113. Weiss J, Rose J, Storch CH, Ketabi-Kiyanvash N, Sauer A, Haefeli WE, Efferth T (2007) Modulation of human BCRP (ABCG2) activity by anti-HIV drugs. J Antimicrob Chemother 59:238-245. doi: 10.1093/jac/dkl474
    Pubmed CrossRef
  114. Wilson CS, Davidson GS, Martin SB, Andries E, Potter J, Harvey R, Ar K, Xu Y, Kopecky KJ, Ankerst DP, Gundacker H, Slovak ML, Mosquera-Caro M, Chen IM, Stirewalt DL, Murphy M, Schultz FA, Kang H, Wang X, Radich JP, Appelbaum FR, Atlas SR, Godwin J, Willman CL (2006) Gene expression profiling of adult acute myeloid leukemia identifies novel biologic clusters for risk classification and outcome prediction. Blood 108:685-696. doi: 10.1182/blood-2004-12-4633
    Pubmed KoreaMed CrossRef
  115. Xu S, Weerachayaphorn J, Cai SY, Soroka CJ, Boyer JL (2010) Aryl hydrocarbon receptor and NF-E2-related factor 2 are key regulators of human MRP4 expression. Am J Physiol Gastrointest Liver Physiol 299:G126-G135. doi: 10.1152/ajpgi.00522.2010
    Pubmed KoreaMed CrossRef
  116. Yamamoto M, Kensler TW, Motohashi H (2018) The Keap1-NRF2 system: a thiol-based sensor-effector apparatus for maintaining redox homeostasis. Physiol Rev 98:1169-1203. doi: 10.1152/physrev.00023.2017
    Pubmed KoreaMed CrossRef
  117. Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, Zhang G, Wang X, Dong Z, Chen F, Cui H (2020) Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther 5:8. doi: 10.1038/s41392-020-0110-5
    Pubmed KoreaMed CrossRef
  118. Zhou S, Schuetz JD, Bunting KD, Colapietro AM, Sampath J, Morris JJ, Lagutina I, Grosveld GC, Osawa M, Nakauchi H, Sorrentino BP (2001) The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med 7:1028-1034. doi: 10.1038/nm0901-1028
    Pubmed CrossRef
  119. Zhou Y, Chen Y, Shi Y, Wu L, Tan Y, Li T, Chen Y, Xia J, Hu R (2023) FAM117B promotes gastric cancer growth and drug resistance by targeting the Keap1/NRF2 signaling pathway. J Clin Invest 133:e158705. doi: 10.1172/JCI158705
    Pubmed KoreaMed CrossRef

Stats or Metrics

Share this article on :

  • line