Next Article in Journal
Brain Plasticity Modulator p75 Neurotrophin Receptor in Human Urine after Different Acute Brain Injuries—A Prospective Cohort Study
Next Article in Special Issue
Drug Resistance and Novel Targets for Cancer Therapy: An Overview of Recent Findings
Previous Article in Journal
Evaluating the Impact of Benign Prostatic Hyperplasia Surgical Treatments on Sexual Health
Previous Article in Special Issue
Exploiting TLK1 and Cisplatin Synergy for Synthetic Lethality in Androgen-Insensitive Prostate Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

ATP-Binding Cassette Subfamily G Member 2 in Acute Myeloid Leukemia: A New Molecular Target?

by
Daniela Damiani
1,2,* and
Mario Tiribelli
1,2
1
Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy
2
Department of Medicine, Udine University, 33100 Udine, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(1), 111; https://doi.org/10.3390/biomedicines12010111
Submission received: 20 November 2023 / Revised: 21 December 2023 / Accepted: 3 January 2024 / Published: 5 January 2024
(This article belongs to the Special Issue Drug Resistance and Novel Targets for Cancer Therapy)

Abstract

:
Despite the progress in the knowledge of disease pathogenesis and the identification of many molecular markers as potential targets of new therapies, the cure of acute myeloid leukemia remains challenging. Disease recurrence after an initial response and the development of resistance to old and new therapies account for the poor survival rate and still make allogeneic stem cell transplantation the only curative option. Multidrug resistance (MDR) is a multifactorial phenomenon resulting from host-related characteristics and leukemia factors. Among these, the overexpression of membrane drug transporter proteins belonging to the ABC (ATP-Binding Cassette)-protein superfamily, which diverts drugs from their cellular targets, plays an important role. Moreover, a better understanding of leukemia biology has highlighted that, at least in cancer, ABC protein’s role goes beyond simple drug transport and affects many other cell functions. In this paper, we summarized the current knowledge of ABCG2 (formerly Breast Cancer Resistance Protein, BCRP) in acute myeloid leukemia and discuss the potential ways to overcome its efflux function and to revert its ability to confer stemness to leukemia cells, favoring the persistence of leukemia progenitors in the bone marrow niche and justifying relapse also after therapy intensification with allogeneic stem cell transplantation.

1. Introduction

Acute myeloid leukemia is an aggressive disease arising from immature myeloid hematopoietic progenitors that acquire proliferative and survival advantages and defective differentiation capacity, accounting for increased infective and hemorrhagic risk [1]. Standard intensive chemotherapy-based regimens remain central in inducing complete response (CR) in AMLs, with CR rates ranging from 60–85% in younger and 40–60% in older people [2]. Furthermore, relapse occurs early in a high proportion of patients, explaining the disappointing survival rates in adult AML patients and justifying the use of stem cell transplantation as the only curative option for this disease [3]. The recent advances in diagnostic tools significantly increased the knowledge of leukemia pathogenesis and permitted the development of prognostic risk scores based on the different genetic/karyotypic alterations and the identification of many molecular markers as potential targets for new therapies [4]. Moreover, the genetic-based risk classification highlighted, once again, the prognostic importance of the initial response to chemotherapy and of the persistence of minimal residual disease [5]. In acute myeloid leukemia, as well as in many other cancer types, a recognized role in affecting the initial response to chemotherapy has been attributed to the efflux pump activity of the tree most studied members of the ABC protein family, ABCB1, ABCC1, and ABCG2. By diverting transported drugs from their intracellular target, they reduce therapy efficacy and induce the Multidrug Resistance (MDR) phenomenon, by which neoplastic cells acquire cross-resistance to different “conventional” structurally unrelated anticancer molecules, still representing the backbone of cancer treatment [6,7,8,9], but also restrict the activity of many new molecular target drugs [10]. However, more recent studies suggest that ABC transporters may have many other important roles in cancer cell biology [11], despite a mechanistic understanding of how these functions contribute to tumor biology that is still lacking. In experimental models, there is a growing body of evidence that the loss or inhibition of various ABC transporters can influence tumor cell phenotype, proliferation, differentiation, migration, and survival [12,13,14], as well as tumorigenesis and tumor progression [12,15,16]. On this basis, counteracting ABC transporter activity may have additional benefits beyond efflux inhibition and could be effective regardless of the type of drugs employed. In this paper, we focused on ABCG2, one of the most recently identified components of the ABC superfamily, revising the current knowledge on its expression and prognostic role in acute myeloid leukemia and on the possible methods for reverting its activity.

2. ABCG2: Nomenclature, Structure, and Physiological Function

The human genome contains 49 genes that encode ABC proteins, organized into seven subfamilies (ABCA–ABCG) [17]. They have many physiological functions by exporting xenobiotics, proteins, and metabolic products in tissues [18], and they can export a myriad of compounds with unrelated chemical structures, decreasing their intracellular accumulation and, as a consequence, their therapeutic efficacy [19]. Among them, ABCG2 was discovered in the late 1990s by Doyle et al. in the MCF-7 drug-resistant breast cancer cell subline and, for this reason, initially named Breast Cancer Resistance Protein (BCRP) [20]. In the same year, Allikmets et al. cloned the DNA of an ABC protein involved in multidrug resistance specific to the human placenta (human placenta-specific ATP binding cassette protein, ABCP) [21]. In 1999, Miyake et al. first cloned the DNA of a protein with high homology to ABC proteins from mitoxantrone-resistant colon carcinoma cells, justifying the name of the mitoxantrone resistance protein (MXR protein) [22]. As the second discovered member of the ABCG subfamily, the protein was further designed as ABCG2 by the Human Genome Organization Committee and assigned to the 338 cluster of differentiation (CD338) of the antigen nomenclature system by the Human Cell Differentiation Molecules organization. The human gene encoding for ABCG2 is located on chromosome 4q22 and spans over 60 kilobases, containing 16 exons and 15 introns. The translational start site is in exon 2, the ATP binding sites are in exons 3 and 6, and the ABC signature motif is in exon 6 [23,24]. The mechanisms controlling ABCG2 expression are not well clarified, but in cell lines with high ABCG2 levels, multiple rearrangements on chromosome 4, including balanced translocations between chromosomes 4 and 7, have been found [25]. Moreover, many factors are known to regulate ABCG2 at the transcriptional, splicing, and epigenetic levels [24]. Transforming growth factor beta, interleukin 1, and tumor necrosis factor-alpha seem to decrease mRNA and ABCG2 protein [26,27], and insulin-like growth factor 1 increases protein expression [27]. ABCG2 is a 72-kDa protein composed of 665 amino acids. Unlike ABCB1 and ABCC1, ABCG2 is considered a half-transporter with only one nucleotide-binding domain (NBD) and one transmembrane domain (TMD). The minimal functional unit of ABCG2 is a homodimer, but the presence of higher-order oligomers is also possible [28,29], in contrast to other ABCG members, such as ABCG5 and ABCG8, whose functional form results from heterodimerization [30]. ABCG2 dimerization occurs in the endoplasmic reticulum (ER), then ABCG2 moves to the Golgi apparatus, where it undergoes further post-translational quality control before being transferred to the apical membrane of the cell [31]. ABCG2 undergoes complex post-transcriptional controls in the ER that have a crucial role in membrane protein topogenesis, facilitating its insertion, orientation, and folding and, ultimately, attaining its quaternary structure. In these mechanisms, many chaperon proteins, such as the binding immunoglobulin protein (BiP) or calnexin/calreticulin (CALN/CALR), are involved. Moreover, ER is involved in quality control of the newly synthetized proteins, which can be transferred to the Golgi apparatus only in correctly folded conformation. Two additional quality control steps are undergone in the Golgi apparatus and in the cell periphery, including internalization, recycling, and lysosomal degradation, all of which are potential targets for ABCG2 therapeutic modulation. For a detailed description of ABCG2 synthesis, see Sarkadi et al. [32].
Compared to most other ABC transporters on the cell membrane, ABCG2 is in a reverse configuration, with an N-terminal ATP binding site and a C-terminal transmembrane domain [24]. The transmembrane domain of ABCG2 (residues 361 to 655) has six transmembrane segments and an extracellular loop between segments five and six. Glycosylation is present only at asparaginase 596 in the extracellular loop, but this does not impact proper protein localization or function [33]. The GXXXG sequence located in transmembrane domain 1 plays a role in dimerization and may also be important in the formation of higher-order complexes [34]. Cysteine 603 in the extracellular loop is involved in the formation of intramolecular disulfide bonds and can contribute to dimerization. The intramolecular disulfide bonds between cysteine 592 and cysteine 608 affect its function and stability [35]. A schematic representation of the membrane organization and residues or mutations affecting the transport function is shown in Figure 1.
Although ABCG2 was first discovered in multidrug-resistant cell lines, it was soon demonstrated in normal tissues, where it is believed to be involved in protecting tissues from xenobiotic accumulation and toxicity. ABCG2 is located on the apical surface of epithelial cells [37], and a physiologically high level of proteins has been demonstrated in strategic sites such as the central nervous system, liver, adrenal gland, placenta, hematopoietic stem cells, breast, small intestine, and colon [38,39], supporting its role in tissue defense. Like ABCB1 and ABCC1, ABCG2 confers resistance to many amphipathic natural anticancer drugs and methotrexate. In addition to natural product-type drugs, ABCG2 reduces the accumulation of porphyrins, precursors of the heme biosynthetic pathway within cells [40], and it has been suggested that ABCG2 protects stem cell survival by reducing heme accumulation under hypoxic conditions [40]. Besides the efflux of natural anticancer drugs, ABCG2 is involved in the excretion of physiological metabolites, in preventing or limiting intestinal absorption, and in enhancing biliary efflux of dietary carcinogens, such as 2-amino-1-methyl-6-pheylimidazo[4,5-b] pyridine (PhIP), the most important heterocyclic amine formed from frying and cooking meat [41].

3. ABCG2 Substrates

As ABCG2 was demonstrated in multidrug-resistant cancer cell lines, the first identified protein substrates were chemotherapeutic agents such as mitoxantrone, flavopiridol, methotrexate, irinotecan, and its active metabolite SN-38. Since then, many other classes of substrates have been identified, and the list is still rapidly expanding, including antivirals, antibiotics, antiHMG-CoA reductase inhibitors, calcium channel blockers, carcinogens, and tyrosine kinase inhibitors. In general, the compounds transported by ABCG2 are large, hydrophobic molecules that are positively or negatively charged. A list of selected ABCG2 substrates is shown in Table 1.
The precise mechanism of substrate transport is still not well understood. In all ABC proteins, ATP is bound by the Walker A and B motifs of one NBD and by the signature sequence of the other NBD, resulting in a so-called “sandwich dimer” due to a close interaction of the two NBDs [79]. Recent publications have elucidated the molecular mechanism of action of ABCG2. According to data provided by cryogenic electron microscopy, the transport is linked to conformational changes of the TMDs, which are regulated by the ATP-dependent formation and separation of the NBD “sandwich dimer.” Compared to ABCB1, conformational changes in ABCG2 during the transport cycle are less large, as the NBDs are already in close proximity in the nucleotide-free state [80]. In the absence of ATP, ABCG2 adopts an inward-facing (IF) conformation, with the transmembrane cavity open toward the cytosolic side of the cell membrane and the central four transmembrane helices (TH2, TH5, TH2′, TH5′) arranged to form a large hydrophobic slit-like drug-binding pocket (cavity1). When ATP is bound, ABCG2 is set in the outward-facing (OF) state, and the substrate is forced into the smaller and less hydrophobic cavity 2, allowing the release of the substrate to the extracellular side. The change in substrate affinity in the IF and OF conformations favors substrate uptake on the cytosolic side and its release on the extracellular side [81,82].

4. ABCG2 in Acute Myeloid Leukemia: Expression and Prognostic Role

Despite the heterogeneity of employed methods (m-RNA quantification, protein expression by flow cytometry or immune-cytochemistry, protein function by efflux of naturally fluorescent substrates) and the lack of standardization that makes data comparison difficult, many studies in the past decades have reported a relationship between ABCG2 overexpression and poor clinical outcomes in AML.
A comprehensive review of ABCG2 expression and clinical impact in AML has been recently published [83].
In summary, most data support the negative role of ABCG2 on AML outcomes, both in adults and in children. Interestingly, its negative impact also emerges when evaluating ABCG2 transcription despite the known high post-transcriptional manipulation of the protein. The frequent ABCG2 and ABCB1 co-expression on leukemia cells, with synergistic effects on drug efflux, may have a role in reducing the complete remission rate and in favoring early relapse. However, it must be considered that ABCG2 overexpression may also contribute to the negative prognosis by conferring stem cell-like properties to leukemia cells, thus favoring the persistence of disease in the hypoxic bone marrow niche and accounting for “late” relapses occurring after chemotherapy intensification and stem cell transplantation.

5. ABCG2 Reversal: Counteracting Efflux Activity

With this premise, the identification of strategies able to overcome multidrug resistance appears crucial to improving the leukemia cure and patients’ survival. Over the past three decades, many strategies, e.g., the synthesis of drugs eluding ABC transporters or the use of drug combinations to take advantage of the “collateral sensitivity” phenomenon, have been proposed with disappointing results. The most pursued way was to prevent multidrug resistance by inhibiting ABC protein efflux activity and re-sensitizing resistant cells to conventional chemotherapy. Most of the studies tried a combination therapy aimed at inhibiting transporter function by inhibitor molecules and then induced leukemia cell death by using chemotherapeutic drugs. These combined therapies, initially used to inhibit ABCB1, demonstrated the best results in vitro [84,85] and displayed some encouraging results in vivo, limited by off-target toxicity. So far, there are no approved sensitizers for clinical use. Among molecules able to inhibit ABCG2-mediated efflux, the first identified was fumitremorgin C (FTC), a mycotoxin produced by Aspergillus fumigatus. Since then, many other potent ABCG2 inhibitors have been identified by random screening of different classes of drugs sharing structural characteristics known to favor ABCG2 inhibition [86] or by drug repurposing, which has the advantage of shortening the drug approval process [87] and can take advantage of known pharmacokinetic indexes [87]. Among them, the most interesting are tivozanib, fostamatinib, ponatinib, and febuxostat, all active at nanomolar IC50 and compatible with clinical use [88,89,90]. Some inhibitors selectively inhibit ABCG2 efflux. Others were first selected as ABCB1 blockers and also demonstrated anti-ABCG2 activity [91,92]. A few compounds, such as vandetanib, showed dual ABCG2 and ABCC1 inhibition [93], but at present, only a major curcumin metabolite, tetrahydrocurcumin, seems to act as a pan-inhibitor (anti-ABCB1,-ABCC1,-ABCG2) [94]. The advantage of multi-inhibitors on leukemic cells is evident, considering the frequent co-expression of ABC proteins, but the cumulative off-target toxicity remains an unsolved problem. At present, over one hundred molecules distributed in 45 classes of compounds have shown in vitro inhibitory activity toward ABCG2; many of them demonstrated inhibitory effects only at high concentrations, not reachable in vivo. However, it must be underlined that in vitro tests were performed in systems with very high ABCG2 expression and that, in wild leukemia samples, ABCG2 levels are significantly lower than in cell lines, and ABCG2 inhibition may be reached with lower concentrations. Table 2 lists some ABCG2 inhibitors by class of compound selected through in vitro tests, with IC50 attainable in clinical settings.
Recently, other ABCG2 inhibitors have been identified by drug repurposing. Ribociclib, a CDK4/6 inhibitor approved for the treatment of locally advanced/ metastatic breast cancer, revealed ABCB1 and ABCG2-mediated daunorubicin and mitoxantrone efflux inhibition in AML cell lines and in CD4+/Flt3-WT leukemia cells [117]. In vitro dual inhibition of ABCC1 and ABCG2-mediated daunorubicin and mitoxantrone efflux was demonstrated by talazoparib, a TK inhibitor used in metastatic BCRA1/2-mutated breast cancer [118]. Tucatinib, a selective HER-2 inhibitor, approved in 2020 for the treatment of HER-2+ breast cancer, showed high ABCG2 inhibition activity in primary leukemia blast cells and in the HL60/ABCG2 cell line, increasing cell death, Hoechst 3342 cell accumulation, and significantly reducing the leukemia stem cell population [119]. ABCG2-mediated efflux block was also demonstrated by venetoclax, bcl-2 inhibitors originally approved for lymphoproliferative diseases and recently employed in elderly acute myeloid leukemia in association with hypomethylating agents [120]. In vitro and modeling studies have demonstrated high ABCG2 inhibition by bexarotene and by vemurafenib, a BRAF inhibitor approved for melanoma and used in the treatment of hairy cell leukemia [121]. Cermáková et al. reported that zanubrutinib, a second-generation Bruton tyrosine kinase inhibitor approved for relapsed/refractory marginal zone lymphoma (MZL), mantle cell lymphoma (MCL), and Waldenström’s macroglobulinemia (WM), counteracts anthracycline resistance by targeting aldo-keto reductase 1C3 (AKR13) and inhibits daunorubicin efflux mediated by ABCB1, ABCC1, and ABCG2, suggesting a possible use in acute myeloid leukemia [122].
The recent advances in the knowledge of ABCG2 structure and transport mechanism will be helpful in discovering new potential inhibitors, modifying known molecules, or starting from new scaffolds. It is well recognized that inhibition potency is closely related to drug physicochemical properties, like hydrophilic/lipophilic balance, which reflect solubility and permeability on biological membranes [123]. Lipinski et al. proposed “the rule of 5” regarding solubility and permeability to guide new drug development, considering that for ABCG2 inhibition, solubility is necessary for membrane penetration, and only hydrophobic moieties can bind TMDs [124]. Moreover, the “ideal” ABCG2 inhibitor should combine high potency of inhibition, low/null toxicity, and high ABCG2 selectivity. Moinul et al. investigated the structure-inhibition relationship of many molecular scaffolds, analyzing their different pharmacological activities, and proposed the minimum possible structural features required for the design of new ABCG2 inhibitors. They identified a benzene ring fused with a heterocyclic aromatic ring as essential for activity and suggested many additional groups to modulate polarity to improve TMD binding and selectivity [125].

6. ABCG2 Reversal: Affecting Protein Expression

In the past years, the study of ABCG2 activity highlighted that beyond drug efflux, it contributes to changing leukemia cell biology, favoring the acquisition of stem cell properties, resulting in protection from drug toxicity and immune surveillance, and eventually preventing disease eradication also after stem cell transplantation. Moreover, the study of complex molecular pathways involved in ABCG2 expression has underlined that many of them are also involved in cancer/leukemia pathogenesis.

6.1. Regulation by Transcription Factors

To date, several cis-regulatory elements and transcription factor binding sites in the ABCG2 gene region have been identified, many of them expressed in a tissue-specific manner, many of them overexpressed in acute myeloid leukemia, and often already targets of new anticancer molecules.
Krishnamurthy et al. found that ABCG2 transcription is activated by the binding of hypoxia-inducible factor 1α (HIF1α)/ARNT heterodimer to a hypoxia response element (HRE) under low oxygen conditions [40], enhancing hypoxic cell survival, protecting hematopoietic stem cells in the bone marrow niche, and also leukemia stem cells. High levels of HIF1α were reported in acute myeloid leukemia in de novo AML patients by Jabari et al. [126]. Xu et al. observed a positive correlation between HIF1α and ABCG2 expression [127], suggesting that HIF1α could be considered a prognostic indicator in AMLs and making HIF1α a potential therapeutic target to down-modulate ABCG2 expression and eliminate leukemia stem cells [128].
C-Myc is a proto-oncogene and encodes an HLH-leucine zipper transcription factor to regulate gene expression [129]. Overexpression of c-MYC in human mammary epithelial cells results in enhanced ABCG2 expression and activity. In the leukemia setting, high expression of c-MYC-driven ABCG2 was observed in CD34 cells of chronic myeloid leukemia [130]. However, Zhang et al. demonstrated that c-MYC can access the ABCG2 promoter only if its binding sites are unmethylated [131]. In FLT3-ITD acute myeloid leukemia, the combination of homoharringtonine and quizartinib AKT and its downstream targets, including c-MYC, reduces the ABCG2 overexpressing side population [132].
As c-MYC, also one other HLH transcription factor, E2F1 is often overexpressed in many cancers and acts by directly bonding the ABCG2 promoter, activating protein transcription, and inducing chemotherapy resistance [133]. In vitro studies in leukemia cell lines K562 and K562/A02 transiently transfected with miR-98 show that miR-98 upregulation results in reduced E2F1 levels, a reduction of ABCG2 levels, and increased chemosensitivity [134].
Nuclear factor kappa B (NF-kB) consists of five subunits that form homo or hetero dimers to bind a gene promoter [135]. In acute leukemia, the activation of the NF-kB pathway affects cell survival, promotes immune evasion, favors proliferation, induces drug resistance, and enhances the stem cell population [136]. Studies in cancer cell lines prove that NF-kB directly binds the ABCG2 promoter, enhancing BCG2 expression and membrane translocation of the protein [137].
Spalt-like transcription factor 4 (SALL4) is one of the transcription factors involved in maintaining the pluripotency of stem cells. In acute myeloid leukemia, Jeong et al. demonstrated its aberrant overexpression in myeloid leukemia and a correlation between SALL4 and ABCG2 levels [138]. Moreover, they showed that SALL4 modulates ABCG2, not the binding promoter but some distal region, or represses PTEN expression [139].
Szatmari et al. demonstrated ABCG2 upregulation via activation of peroxisome proliferator-activated receptor (PPAR)-γ in human myeloid dendritic cells [140]. Recently, Kim et al. demonstrated high ABCG2 expression levels in M2 macrophages [141]. They reported that IL-4 and IL-13, responsible for macrophage polarization to M2 type, induce PPARγ transcription, PPARγ bound to the ABCG2 promoter, and ultimately ABCG2 overexpression in M2 macrophages, limiting the intracellular accumulation of macrophage-targeting antibiotics [141]. In acute myeloid leukemia, PPARγ stimulation exerts an opposite regulatory effect. It results in phosphatase and tensin homolog (PTEN) upregulation and consequent ABCG2 internalization through reduced Akt phosphorylation [142,143].
Nrf2, a nuclear factor-erythroid 2-related transcription factor, plays a critical role in the transcriptional regulation of many metabolizing enzymes that rescue cells from oxidative stress and is found to be a transcription factor for ABCG2 [144]. In acute myeloid leukemia, overexpression of Nrf2 was observed at diagnosis and relapse [145] and has been associated with disease progression and increased resistance to cytarabine in cases with poor prognostic gene mutations [146]. Hu et al. hypothesized that Nrf2-mediated drug resistance occurs via JNK/NF-kB signaling pathway activation, both involved in the regulation of ABCG2, which may contribute to chemotherapy resistance [147]. In pancreatic cancer cell lines, knockout Nrf2 re-sensitized cancer cells to 5-Fluorouracil by suppressing ABCG2 expression [148]. Moreover, Choi et al. reported that Nrf2 increased ABCG2 expression by modulating the microRNA miR-206 level [149].

6.2. Regulation by Kinase Signaling

Many kinases, such as PIM1, PI3K/AKT, ERK, and JNK, are involved in ABCG2 expression, activity, and trafficking [150], either by direct phosphorylation of ABCG2 or indirectly by phosphorylation of downstream effector kinases/regulatory proteins. Among them, PIM1 kinase phosphorylates ABCG2 at threonine 362, promotes protein membrane translocation, stabilization, and multimerization, and enhances its activity. PIM1 inhibitors display a dual-mode effect on ABCG2-expressing cells. They impair efflux function and reduce protein levels on the membrane [151]. The PI3K/AKT pathway also mediates ABCG2 sequestration in extracellular micro-vesicles (ESs), contributing to the dissemination of drug resistance [152]. Pharmacologically inhibition of the PI3K/AKT pathway transfers ABCG2 from ESs to the cell compartment, reversing drug resistance [152]. In addition, PI3K/AKT mediates ABCG2 at the transcriptional level [153]. JNK pathways and consequent upregulation of ABCG2 are activated by high levels of CXCR4, a chemokine receptor frequently expressed in AML [154]. Finally, Wnt and Hedgehog pathways, involved in the maintenance of leukemia stem cell survival, bind the ABCG2 promoter and increase protein expression [155].

6.3. Epigenetic Regulation

Epigenetic changes commonly involve DNA methylation and/or post-translational modification of histone proteins, with permissive or repressive effects on gene expression. Hypermethylation of the CpG island of the ABCG2 promoter by DNMT1 and DNMT3a and recruitment to the methylated site of methyl CpG binding domain proteins (MBDs) along with co-repressors mSIN3a and HDAC1 lead to gene silencing [156]. This is associated with HDAC1-mediated deacetylation at H3K9 and with histone methyltransferases’ (HMTs) addition of the repressive histone mark H3K9me3 [156]. ABCG2 upregulation has been observed in cell lines after treatment with 5-aza-2′ deoxycytidine (5-AZA) [157,158]. Given the large clinical use of 5AZA in older patients with AML, the possible impact on clinical outcomes deserves further investigation. Histone modification by acetylation, methylation, phosphorylation, citrullination, or ubiquitination of lysine and arginine can result in enhanced gene expression or suppression. ABCG2 transcriptional activation results from the replacement of the repressive histone mark H3K9me3 with permissive changes, including H3K4me and H3S10p, the release of HDCA1s, and the recruitment of chromatin remodeler Brg-1 and RNA polymerase II to the promoter. Loss of function mutant of enhancer of zeste homologue 2 (EZH2), an enzymatic component of polycomb repressive complex 2 (PRC2), increases ABCG2 mRNA expression and contributes to myelodysplatic syndrome pathogenesis [159]. Histone deacetylase inhibitors, such as romidepsin, vorinostat, and panobinostat, have demonstrated the ability to increase ABCG2 synthesis and function [160]. Developing drugs able to inhibit histone acetyltransferase or activate histone deacetylase may be a new strategy for decreasing ABCG2 expression. Moreover, post-transcriptional regulation of ABCG2, with increased mRNA stability, is the product of the bound to ABCG2 mRNA of the hypermethylated RNA recognition motif (RRM) of heterogeneous nuclear ribonucleoprotein A1 (HnRNPA1) [161].
MicroRNAs (miRNAs) are small, non-coding RNAs able to cause mRNA degradation and/or decreased translational efficiency, affecting protein expression and functions [162]. Several studies have shown that miRNAs regulate ABCG2 mRNA or protein expression. MiR-487a, mi-R-328, mi-R-181a, and mi-R-302 were demonstrated to increase mitoxantrone sensitivity in cell lines and reduce ABCG2 expression in cell lines [163,164,165,166]. Mi-R-519c may be involved in ABCG2 overexpression, affecting mRNA stability [156].

6.4. Regulation by Protein Degradation

At least three degradation pathways have been identified through “in vitro” studies on cell lines. The first mechanism involves Derlin1-mediated block of endoplasmic reticulum (ER) to Golgi transport that affects protein maturation [167]. Moreover, Derlin-1 facilitates ubiquitin-mediated ER-associated degradation (ERAD) of both glycosylated and non-glycosylated forms [167]. In cancer cell lines, this mechanism was induced by low molecular weight heparin (LMWH). The second mechanism is endocytosis-mediated internalization and lysosomal degradation of ABCG2. The functional consequence of protein down-regulation has been demonstrated by the increased sensitivity of mitoxantrone in the MCF7-MX cell line [168]. Wei et al. demonstrated that sorafenib, a multi-kinase inhibitor employed in hepatocarcinoma and in relapsed FLT3-ITD acute myeloid leukemia, not only inhibits ABCG2 efflux activity but induces ABCG2 degradation in the lysosome [169]. The third mechanism is ABCG2 half-life reduction through calpain-mediated protein degradation. This mechanism was described by Hu et al., who tested the cytotoxic effect of Escallonia acid D (SAD), a toxic metabolite of Penicillium oxalicum, in the side population of a lung cancer cell line. They observed that SAD treatments reduced side population size by reducing membrane ABCG2 and demonstrated that protein down-modulation was due to the acceleration of ABCG2 degradation by activation of the calpain pathway [170].
On this basis, a new approach to counteract ABCG2 may be hypothesized, targeting the protein rather than its function. A partial selection of molecules with demonstrated activity on ABCG2 expression in cancer cell lines is reported in Table 3.

7. Conclusions

Since its first description in the late 1990s, several studies have associated ABCG2 overexpression with poor outcomes in acute myeloid leukemia. Despite this, very few efforts have been made in the attempt to counteract its function, probably as a consequence of the disappointing results in the clinical use of ABCB1 inhibitors, almost due to their off-target toxicity. However, the recent explosion of anti-cancer target drugs, often substrates of ABCG2, has led to the identification of several compounds with high ABCG2 affinity and strong efflux inhibition ability that may be “repurposed” as ABCG2 reversal agents. Moreover, knowledge of the chemical structure of molecules with proven inhibitory activity helps better understand the mechanisms of ABCG2-mediated transmembrane transport and paves the way for the development of new molecules combining high inhibition capacity with reduced toxicity. In addition, the discovery of mechanisms underlying leukemia pathogenesis has highlighted that many of them are involved in ABCG2 regulation, so identifying potential therapeutic targets to indirectly inhibit ABCG2 expression and offering a new perspective to counteract ABCG2-mediated drug resistance is able to modify leukemia cell biology. At present, data on drugs targeting ABCG2 transcription, internalization, and stability are quite scarce or preliminary but clearly warranted, along with extensive studies on cellular pathways specifically activated in leukemia cells and efforts toward the standardization of methods to evaluate protein expression and function.

Author Contributions

All authors equally contributed to manuscript preparation. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jan, M.; Snyder, T.M.; Corces-Zimmerman, M.R.; Vyas, P.; Weissman, I.L.; Quake, S.R.; Majeti, R. Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia. Sci. Transl. Med. 2012, 4, 149ra118. [Google Scholar] [CrossRef] [PubMed]
  2. Short, N.J.; Rytting, M.E.; Cortes, J.E. Acute myeloid leukaemia. Lancet 2018, 392, 593–606. [Google Scholar] [CrossRef] [PubMed]
  3. Li, Y.; Zhou, F. Efficacy of bone marrow transplantation in treating acute myeloid leukemia: A systematic review and meta-analysis. Am. J. Transl. Res. 2023, 15, 1–12. [Google Scholar] [PubMed]
  4. Dohner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef] [PubMed]
  5. Shimony, S.; Stahl, M.; Stone, R.M. Acute myeloid leukemia: 2023 update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2023, 98, 502–526. [Google Scholar] [CrossRef] [PubMed]
  6. Gottesman, M.M.; Ling, V. The molecular basis of multidrug resistance in cancer: The early years of P-glycoprotein research. FEBS Lett. 2006, 580, 998–1009. [Google Scholar] [CrossRef] [PubMed]
  7. Theodoulou, F.L.; Kerr, I.D. ABC transporter research: Going strong 40 years on. Biochem. Soc. Trans. 2015, 43, 1033–1040. [Google Scholar] [CrossRef] [PubMed]
  8. Patel, H.; Wu, Z.X.; Chen, Y.; Bo, L.; Chen, Z.S. Drug resistance: From bacteria to cancer. Mol. Biomed. 2021, 2, 27. [Google Scholar] [CrossRef]
  9. Dean, M.; Hamon, Y.; Chimini, G. The human ATP-binding cassette (ABC) transporter superfamily. J. Lipid Res. 2001, 42, 1007–1017. [Google Scholar] [CrossRef]
  10. Sarmento-Ribeiro, A.B.; Scorilas, A.; Goncalves, A.C.; Efferth, T.; Trougakos, I.P. The emergence of drug resistance to targeted cancer therapies: Clinical evidence. Drug Resist. Update 2019, 47, 100646. [Google Scholar] [CrossRef]
  11. Fletcher, J.I.; Haber, M.; Henderson, M.J.; Norris, M.D. ABC transporters in cancer: More than just drug efflux pumps. Nat. Rev. Cancer 2010, 10, 147–156. [Google Scholar] [CrossRef] [PubMed]
  12. Copsel, S.; Bruzzone, A.; May, M.; Beyrath, J.; Wargon, V.; Cany, J.; Russel, F.G.; Shayo, C.; Davio, C. Multidrug resistance protein 4/ATP binding cassette transporter 4: A new potential therapeutic target for acute myeloid leukemia. Oncotarget 2014, 5, 9308–9321. [Google Scholar] [CrossRef] [PubMed]
  13. Henderson, M.J.; Haber, M.; Porro, A.; Munoz, M.A.; Iraci, N.; Xue, C.; Murray, J.; Flemming, C.L.; Smith, J.; Fletcher, J.I.; et al. ABCC multidrug transporters in childhood neuroblastoma: Clinical and biological effects independent of cytotoxic drug efflux. J. Natl. Cancer Inst. 2011, 103, 1236–1251. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, Z.; Wang, J.; Shen, B.; Peng, C.; Zheng, M. The ABCC4 gene is a promising target for pancreatic cancer therapy. Gene 2012, 491, 194–199. [Google Scholar] [CrossRef]
  15. Yamada, T.; Mori, Y.; Hayashi, R.; Takada, M.; Ino, Y.; Naishiro, Y.; Kondo, T.; Hirohashi, S. Suppression of intestinal polyposis in Mdr1-deficient ApcMin/+ mice. Cancer Res. 2003, 63, 895–901. [Google Scholar]
  16. Zander, S.A.; Kersbergen, A.; Sol, W.; Gonggrijp, M.; van de Wetering, K.; Jonkers, J.; Borst, P.; Rottenberg, S. Lack of ABCG2 shortens latency of BRCA1-deficient mammary tumors and this is not affected by genistein or resveratrol. Cancer Prev. Res. 2012, 5, 1053–1060. [Google Scholar] [CrossRef]
  17. Dean, M. The genetics of ATP-binding cassette transporters. Methods Enzymol. 2005, 400, 409–429. [Google Scholar] [CrossRef]
  18. Holland, I.B. ABC transporters, mechanisms and biology: An overview. Essays Biochem. 2011, 50, 1–17. [Google Scholar] [CrossRef]
  19. Mohammad, I.S.; He, W.; Yin, L. Understanding of human ATP binding cassette superfamily and novel multidrug resistance modulators to overcome MDR. Biomed. Pharmacother. 2018, 100, 335–348. [Google Scholar] [CrossRef]
  20. Doyle, L.A.; Yang, W.; Abruzzo, L.V.; Krogmann, T.; Gao, Y.; Rishi, A.K.; Ross, D.D. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc. Natl. Acad. Sci. USA 1998, 95, 15665–15670. [Google Scholar] [CrossRef]
  21. Allikmets, R.; Schriml, L.M.; Hutchinson, A.; Romano-Spica, V.; Dean, M. A human placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res. 1998, 58, 5337–5339. [Google Scholar] [PubMed]
  22. Miyake, K.; Mickley, L.; Litman, T.; Zhan, Z.; Robey, R.; Cristensen, B.; Brangi, M.; Greenberger, L.; Dean, M.; Fojo, T.; et al. Molecular cloning of cDNAs which are highly overexpressed in mitoxantrone-resistant cells: Demonstration of homology to ABC transport genes. Cancer Res. 1999, 59, 8–13. [Google Scholar] [PubMed]
  23. Bailey-Dell, K.J.; Hassel, B.; Doyle, L.A.; Ross, D.D. Promoter characterization and genomic organization of the human breast cancer resistance protein (ATP-binding cassette transporter G2) gene. Biochim. Biophys. Acta 2001, 1520, 234–241. [Google Scholar] [CrossRef]
  24. Robey, R.W.; To, K.K.; Polgar, O.; Dohse, M.; Fetsch, P.; Dean, M.; Bates, S.E. ABCG2: A perspective. Adv. Drug Deliv. Rev. 2009, 61, 3–13. [Google Scholar] [CrossRef] [PubMed]
  25. Knutsen, T.; Rao, V.K.; Ried, T.; Mickley, L.; Schneider, E.; Miyake, K.; Ghadimi, B.M.; Padilla-Nash, H.; Pack, S.; Greenberger, L.; et al. Amplification of 4q21-q22 and the MXR gene in independently derived mitoxantrone-resistant cell lines. Genes Chromosomes Cancer 2000, 27, 110–116. [Google Scholar] [CrossRef]
  26. Yin, L.; Castagnino, P.; Assoian, R.K. ABCG2 expression and side population abundance regulated by a transforming growth factor beta-directed epithelial-mesenchymal transition. Cancer Res. 2008, 68, 800–807. [Google Scholar] [CrossRef] [PubMed]
  27. Evseenko, D.A.; Paxton, J.W.; Keelan, J.A. Independent regulation of apical and basolateral drug transporter expression and function in placental trophoblasts by cytokines, steroids, and growth factors. Drug Metab. Dispos. 2007, 35, 595–601. [Google Scholar] [CrossRef] [PubMed]
  28. McDevitt, C.A.; Collins, R.F.; Conway, M.; Modok, S.; Storm, J.; Kerr, I.D.; Ford, R.C.; Callaghan, R. Purification and 3D structural analysis of oligomeric human multidrug transporter ABCG2. Structure 2006, 14, 1623–1632. [Google Scholar] [CrossRef]
  29. Xu, J.; Liu, Y.; Yang, Y.; Bates, S.; Zhang, J.T. Characterization of oligomeric human half-ABC transporter ATP-binding cassette G2. J. Biol. Chem. 2004, 279, 19781–19789. [Google Scholar] [CrossRef]
  30. Graf, G.A.; Li, W.P.; Gerard, R.D.; Gelissen, I.; White, A.; Cohen, J.C.; Hobbs, H.H. Coexpression of ATP-binding cassette proteins ABCG5 and ABCG8 permits their transport to the apical surface. J. Clin. Investig. 2002, 110, 659–669. [Google Scholar] [CrossRef]
  31. Stacy, A.E.; Jansson, P.J.; Richardson, D.R. Molecular pharmacology of ABCG2 and its role in chemoresistance. Mol. Pharmacol. 2013, 84, 655–669. [Google Scholar] [CrossRef] [PubMed]
  32. Sarkadi, B.; Homolya, L.; Hegedus, T. The ABCG2/BCRP transporter and its variants—From structure to pathology. FEBS Lett. 2020, 594, 4012–4034. [Google Scholar] [CrossRef] [PubMed]
  33. Diop, N.K.; Hrycyna, C.A. N-Linked glycosylation of the human ABC transporter ABCG2 on asparagine 596 is not essential for expression, transport activity, or trafficking to the plasma membrane. Biochemistry 2005, 44, 5420–5429. [Google Scholar] [CrossRef] [PubMed]
  34. Polgar, O.; Robey, R.W.; Morisaki, K.; Dean, M.; Michejda, C.; Sauna, Z.E.; Ambudkar, S.V.; Tarasova, N.; Bates, S.E. Mutational analysis of ABCG2: Role of the GXXXG motif. Biochemistry 2004, 43, 9448–9456. [Google Scholar] [CrossRef]
  35. Henriksen, U.; Fog, J.U.; Litman, T.; Gether, U. Identification of intra- and intermolecular disulfide bridges in the multidrug resistance transporter ABCG2. J. Biol. Chem. 2005, 280, 36926–36934. [Google Scholar] [CrossRef] [PubMed]
  36. Noguchi, K.; Kawahara, H.; Kaji, A.; Katayama, K.; Mitsuhashi, J.; Sugimoto, Y. Substrate-dependent bidirectional modulation of P-glycoprotein-mediated drug resistance by erlotinib. Cancer Sci. 2009, 100, 1701–1707. [Google Scholar] [CrossRef]
  37. Sarkadi, B.; Ozvegy-Laczka, C.; Nemet, K.; Varadi, A. ABCG2—A transporter for all seasons. FEBS Lett. 2004, 567, 116–120. [Google Scholar] [CrossRef]
  38. Fetsch, P.A.; Abati, A.; Litman, T.; Morisaki, K.; Honjo, Y.; Mittal, K.; Bates, S.E. Localization of the ABCG2 mitoxantrone resistance-associated protein in normal tissues. Cancer Lett. 2006, 235, 84–92. [Google Scholar] [CrossRef]
  39. Maliepaard, M.; Scheffer, G.L.; Faneyte, I.F.; van Gastelen, M.A.; Pijnenborg, A.C.; Schinkel, A.H.; van De Vijver, M.J.; Scheper, R.J.; Schellens, J.H. Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer Res. 2001, 61, 3458–3464. [Google Scholar]
  40. Krishnamurthy, P.; Ross, D.D.; Nakanishi, T.; Bailey-Dell, K.; Zhou, S.; Mercer, K.E.; Sarkadi, B.; Sorrentino, B.P.; Schuetz, J.D. The stem cell marker Bcrp/ABCG2 enhances hypoxic cell survival through interactions with heme. J. Biol. Chem. 2004, 279, 24218–24225. [Google Scholar] [CrossRef]
  41. Van Herwaarden, A.E.; Jonker, J.W.; Wagenaar, E.; Brinkhuis, R.F.; Schellens, J.H.; Beijnen, J.H.; Schinkel, A.H. The breast cancer resistance protein (Bcrp1/Abcg2) restricts exposure to the dietary carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine. Cancer Res. 2003, 63, 6447–6452. [Google Scholar] [PubMed]
  42. Robey, R.W.; Honjo, Y.; Morisaki, K.; Nadjem, T.A.; Runge, S.; Risbood, M.; Poruchynsky, M.S.; Bates, S.E. Mutations at amino-acid 482 in the ABCG2 gene affect substrate and antagonist specificity. Br. J. Cancer 2003, 89, 1971–1978. [Google Scholar] [CrossRef] [PubMed]
  43. Robey, R.W.; Medina-Perez, W.Y.; Nishiyama, K.; Lahusen, T.; Miyake, K.; Litman, T.; Senderowicz, A.M.; Ross, D.D.; Bates, S.E. Overexpression of the ATP-binding cassette half-transporter, ABCG2 (Mxr/BCrp/ABCP1), in flavopiridol-resistant human breast cancer cells. Clin. Cancer Res. 2001, 7, 145–152. [Google Scholar] [PubMed]
  44. Allen, J.D.; Van Dort, S.C.; Buitelaar, M.; van Tellingen, O.; Schinkel, A.H. Mouse breast cancer resistance protein (Bcrp1/Abcg2) mediates etoposide resistance and transport, but etoposide oral availability is limited primarily by P-glycoprotein. Cancer Res. 2003, 63, 1339–1344. [Google Scholar] [PubMed]
  45. Allen, J.D.; Brinkhuis, R.F.; Wijnholds, J.; Schinkel, A.H. The mouse Bcrp1/Mxr/Abcp gene: Amplification and overexpression in cell lines selected for resistance to topotecan, mitoxantrone, or doxorubicin. Cancer Res. 1999, 59, 4237–4241. [Google Scholar] [PubMed]
  46. Schellens, J.H.; Maliepaard, M.; Scheper, R.J.; Scheffer, G.L.; Jonker, J.W.; Smit, J.W.; Beijnen, J.H.; Schinkel, A.H. Transport of topoisomerase I inhibitors by the breast cancer resistance protein. Potential clinical implications. Ann. N. Y. Acad. Sci. 2000, 922, 188–194. [Google Scholar] [CrossRef] [PubMed]
  47. Bates, S.E.; Medina-Perez, W.Y.; Kohlhagen, G.; Antony, S.; Nadjem, T.; Robey, R.W.; Pommier, Y. ABCG2 mediates differential resistance to SN-38 (7-ethyl-10-hydroxycamptothecin) and homocamptothecins. J. Pharmacol. Exp. Ther. 2004, 310, 836–842. [Google Scholar] [CrossRef]
  48. Volk, E.L.; Farley, K.M.; Wu, Y.; Li, F.; Robey, R.W.; Schneider, E. Overexpression of wild-type breast cancer resistance protein mediates methotrexate resistance. Cancer Res. 2002, 62, 5035–5040. [Google Scholar]
  49. Shafran, A.; Ifergan, I.; Bram, E.; Jansen, G.; Kathmann, I.; Peters, G.J.; Robey, R.W.; Bates, S.E.; Assaraf, Y.G. ABCG2 harboring the Gly482 mutation confers high-level resistance to various hydrophilic antifolates. Cancer Res. 2005, 65, 8414–8422. [Google Scholar] [CrossRef]
  50. Burger, H.; van Tol, H.; Brok, M.; Wiemer, E.A.; de Bruijn, E.A.; Guetens, G.; de Boeck, G.; Sparreboom, A.; Verweij, J.; Nooter, K. Chronic imatinib mesylate exposure leads to reduced intracellular drug accumulation by induction of the ABCG2 (BCRP) and ABCB1 (MDR1) drug transport pumps. Cancer Biol. Ther. 2005, 4, 747–752. [Google Scholar] [CrossRef]
  51. Brendel, C.; Scharenberg, C.; Dohse, M.; Robey, R.W.; Bates, S.E.; Shukla, S.; Ambudkar, S.V.; Wang, Y.; Wennemuth, G.; Burchert, A.; et al. Imatinib mesylate and nilotinib (AMN107) exhibit high-affinity interaction with ABCG2 on primitive hematopoietic stem cells. Leukemia 2007, 21, 1267–1275. [Google Scholar] [CrossRef]
  52. Elkind, N.B.; Szentpetery, Z.; Apati, A.; Ozvegy-Laczka, C.; Varady, G.; Ujhelly, O.; Szabo, K.; Homolya, L.; Varadi, A.; Buday, L.; et al. Multidrug transporter ABCG2 prevents tumor cell death induced by the epidermal growth factor receptor inhibitor Iressa (ZD1839, Gefitinib). Cancer Res. 2005, 65, 1770–1777. [Google Scholar] [CrossRef] [PubMed]
  53. Erlichman, C.; Boerner, S.A.; Hallgren, C.G.; Spieker, R.; Wang, X.Y.; James, C.D.; Scheffer, G.L.; Maliepaard, M.; Ross, D.D.; Bible, K.C.; et al. The HER tyrosine kinase inhibitor CI1033 enhances cytotoxicity of 7-ethyl-10-hydroxycamptothecin and topotecan by inhibiting breast cancer resistance protein-mediated drug efflux. Cancer Res. 2001, 61, 739–748. [Google Scholar] [PubMed]
  54. Bram, E.E.; Ifergan, I.; Grimberg, M.; Lemke, K.; Skladanowski, A.; Assaraf, Y.G. C421 allele-specific ABCG2 gene amplification confers resistance to the antitumor triazoloacridone C-1305 in human lung cancer cells. Biochem. Pharmacol. 2007, 74, 41–53. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, X.; Furukawa, T.; Nitanda, T.; Okamoto, M.; Sugimoto, Y.; Akiyama, S.; Baba, M. Breast cancer resistance protein (BCRP/ABCG2) induces cellular resistance to HIV-1 nucleoside reverse transcriptase inhibitors. Mol. Pharmacol. 2003, 63, 65–72. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, X.; Nitanda, T.; Shi, M.; Okamoto, M.; Furukawa, T.; Sugimoto, Y.; Akiyama, S.; Baba, M. Induction of cellular resistance to nucleoside reverse transcriptase inhibitors by the wild-type breast cancer resistance protein. Biochem. Pharmacol. 2004, 68, 1363–1370. [Google Scholar] [CrossRef] [PubMed]
  57. Pan, G.; Giri, N.; Elmquist, W.F. Abcg2/Bcrp1 mediates the polarized transport of antiretroviral nucleosides abacavir and zidovudine. Drug Metab. Dispos. 2007, 35, 1165–1173. [Google Scholar] [CrossRef]
  58. Kitamura, S.; Maeda, K.; Wang, Y.; Sugiyama, Y. Involvement of multiple transporters in the hepatobiliary transport of rosuvastatin. Drug Metab. Dispos. 2008, 36, 2014–2023. [Google Scholar] [CrossRef]
  59. Hirano, M.; Maeda, K.; Matsushima, S.; Nozaki, Y.; Kusuhara, H.; Sugiyama, Y. Involvement of BCRP (ABCG2) in the biliary excretion of pitavastatin. Mol. Pharmacol. 2005, 68, 800–807. [Google Scholar] [CrossRef]
  60. Matsushima, S.; Maeda, K.; Kondo, C.; Hirano, M.; Sasaki, M.; Suzuki, H.; Sugiyama, Y. Identification of the hepatic efflux transporters of organic anions using double-transfected Madin-Darby canine kidney II cells expressing human organic anion-transporting polypeptide 1B1 (OATP1B1)/multidrug resistance-associated protein 2, OATP1B1/multidrug resistance 1, and OATP1B1/breast cancer resistance protein. J. Pharmacol. Exp. Ther. 2005, 314, 1059–1067. [Google Scholar] [CrossRef]
  61. Robey, R.W.; Steadman, K.; Polgar, O.; Morisaki, K.; Blayney, M.; Mistry, P.; Bates, S.E. Pheophorbide a is a specific probe for ABCG2 function and inhibition. Cancer Res. 2004, 64, 1242–1246. [Google Scholar] [CrossRef] [PubMed]
  62. Robey, R.W.; Fetsch, P.A.; Polgar, O.; Dean, M.; Bates, S.E. The livestock photosensitizer, phytoporphyrin (phylloerythrin), is a substrate of the ATP-binding cassette transporter ABCG2. Res. Vet. Sci. 2006, 81, 345–349. [Google Scholar] [CrossRef] [PubMed]
  63. Imai, Y.; Tsukahara, S.; Asada, S.; Sugimoto, Y. Phytoestrogens/flavonoids reverse breast cancer resistance protein/ABCG2-mediated multidrug resistance. Cancer Res. 2004, 64, 4346–4352. [Google Scholar] [CrossRef] [PubMed]
  64. Youdim, K.A.; Qaiser, M.Z.; Begley, D.J.; Rice-Evans, C.A.; Abbott, N.J. Flavonoid permeability across an in situ model of the blood-brain barrier. Free Radic. Biol. Med. 2004, 36, 592–604. [Google Scholar] [CrossRef] [PubMed]
  65. Merino, G.; Alvarez, A.I.; Pulido, M.M.; Molina, A.J.; Schinkel, A.H.; Prieto, J.G. Breast cancer resistance protein (BCRP/ABCG2) transports fluoroquinolone antibiotics and affects their oral availability, pharmacokinetics, and milk secretion. Drug Metab. Dispos. 2006, 34, 690–695. [Google Scholar] [CrossRef] [PubMed]
  66. Janvilisri, T.; Shahi, S.; Venter, H.; Balakrishnan, L.; van Veen, H.W. Arginine-482 is not essential for transport of antibiotics, primary bile acids and unconjugated sterols by the human breast cancer resistance protein (ABCG2). Biochem. J. 2005, 385, 419–426. [Google Scholar] [CrossRef] [PubMed]
  67. Merino, G.; Jonker, J.W.; Wagenaar, E.; van Herwaarden, A.E.; Schinkel, A.H. The breast cancer resistance protein (BCRP/ABCG2) affects pharmacokinetics, hepatobiliary excretion, and milk secretion of the antibiotic nitrofurantoin. Mol. Pharmacol. 2005, 67, 1758–1764. [Google Scholar] [CrossRef] [PubMed]
  68. Van der Heijden, J.; de Jong, M.C.; Dijkmans, B.A.; Lems, W.F.; Oerlemans, R.; Kathmann, I.; Schalkwijk, C.G.; Scheffer, G.L.; Scheper, R.J.; Jansen, G. Development of sulfasalazine resistance in human T cells induces expression of the multidrug resistance transporter ABCG2 (BCRP) and augmented production of TNFalpha. Ann. Rheum. Dis. 2004, 63, 138–143. [Google Scholar] [CrossRef]
  69. Merino, G.; Jonker, J.W.; Wagenaar, E.; Pulido, M.M.; Molina, A.J.; Alvarez, A.I.; Schinkel, A.H. Transport of anthelmintic benzimidazole drugs by breast cancer resistance protein (BCRP/ABCG2). Drug Metab. Dispos. 2005, 33, 614–618. [Google Scholar] [CrossRef]
  70. Breedveld, P.; Zelcer, N.; Pluim, D.; Sonmezer, O.; Tibben, M.M.; Beijnen, J.H.; Schinkel, A.H.; van Tellingen, O.; Borst, P.; Schellens, J.H. Mechanism of the pharmacokinetic interaction between methotrexate and benzimidazoles: Potential role for breast cancer resistance protein in clinical drug-drug interactions. Cancer Res. 2004, 64, 5804–5811. [Google Scholar] [CrossRef]
  71. Shukla, S.; Robey, R.W.; Bates, S.E.; Ambudkar, S.V. The calcium channel blockers, 1,4-dihydropyridines, are substrates of the multidrug resistance-linked ABC drug transporter, ABCG2. Biochemistry 2006, 45, 8940–8951. [Google Scholar] [CrossRef]
  72. Zhang, Y.; Gupta, A.; Wang, H.; Zhou, L.; Vethanayagam, R.R.; Unadkat, J.D.; Mao, Q. BCRP transports dipyridamole and is inhibited by calcium channel blockers. Pharm. Res. 2005, 22, 2023–2034. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, Z.S.; Robey, R.W.; Belinsky, M.G.; Shchaveleva, I.; Ren, X.Q.; Sugimoto, Y.; Ross, D.D.; Bates, S.E.; Kruh, G.D. 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. 2003, 63, 4048–4054. [Google Scholar]
  74. Pavek, P.; Merino, G.; Wagenaar, E.; Bolscher, E.; Novotna, M.; Jonker, J.W.; Schinkel, A.H. Human breast cancer resistance protein: Interactions with steroid drugs, hormones, the dietary carcinogen 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine, and transport of cimetidine. J. Pharmacol. Exp. Ther. 2005, 312, 144–152. [Google Scholar] [CrossRef]
  75. Van Herwaarden, A.E.; Wagenaar, E.; Merino, G.; Jonker, J.W.; Rosing, H.; Beijnen, J.H.; Schinkel, A.H. Multidrug transporter ABCG2/breast cancer resistance protein secretes riboflavin (vitamin B2) into milk. Mol. Cell Biol. 2007, 27, 1247–1253. [Google Scholar] [CrossRef] [PubMed]
  76. Zhou, S.; Schuetz, J.D.; Bunting, K.D.; Colapietro, A.M.; Sampath, J.; Morris, J.J.; Lagutina, I.; Grosveld, G.C.; Osawa, M.; Nakauchi, H.; et al. 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. 2001, 7, 1028–1034. [Google Scholar] [CrossRef] [PubMed]
  77. Litman, T.; Brangi, M.; Hudson, E.; Fetsch, P.; Abati, A.; Ross, D.D.; Miyake, K.; Resau, J.H.; Bates, S.E. The multidrug-resistant phenotype associated with overexpression of the new ABC half-transporter, MXR (ABCG2). J. Cell Sci. 2000, 113 Pt 11, 2011–2021. [Google Scholar] [CrossRef]
  78. Van Herwaarden, A.E.; Wagenaar, E.; Karnekamp, B.; Merino, G.; Jonker, J.W.; Schinkel, A.H. Breast cancer resistance protein (Bcrp1/Abcg2) reduces systemic exposure of the dietary carcinogens aflatoxin B1, IQ and Trp-P-1 but also mediates their secretion into breast milk. Carcinogenesis 2006, 27, 123–130. [Google Scholar] [CrossRef]
  79. Locher, K.P. Structure and mechanism of ATP-binding cassette transporters. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2009, 364, 239–245. [Google Scholar] [CrossRef]
  80. Goebel, J.; Chmielewski, J.; Hrycyna, C.A. The roles of the human ATP-binding cassette transporters P-glycoprotein and ABCG2 in multidrug resistance in cancer and at endogenous sites: Future opportunities for structure-based drug design of inhibitors. Cancer Drug Resist. 2021, 4, 784–804. [Google Scholar] [CrossRef]
  81. Jackson, S.M.; Manolaridis, I.; Kowal, J.; Zechner, M.; Taylor, N.M.I.; Bause, M.; Bauer, S.; Bartholomaeus, R.; Bernhardt, G.; Koenig, B.; et al. Structural basis of small-molecule inhibition of human multidrug transporter ABCG2. Nat. Struct. Mol. Biol. 2018, 25, 333–340. [Google Scholar] [CrossRef] [PubMed]
  82. Gyongy, Z.; Mocsar, G.; Hegedus, E.; Stockner, T.; Ritter, Z.; Homolya, L.; Schamberger, A.; Orban, T.I.; Remenyik, J.; Szakacs, G.; et al. Nucleotide binding is the critical regulator of ABCG2 conformational transitions. Elife 2023, 12, 1–24. [Google Scholar] [CrossRef] [PubMed]
  83. Damiani, D.; Tiribelli, M. ABCG2 in Acute Myeloid Leukemia: Old and New Perspectives. Int. J. Mol. Sci. 2023, 24, 7147. [Google Scholar] [CrossRef] [PubMed]
  84. Szakacs, G.; Paterson, J.K.; Ludwig, J.A.; Booth-Genthe, C.; Gottesman, M.M. Targeting multidrug resistance in cancer. Nat. Rev. Drug Discov. 2006, 5, 219–234. [Google Scholar] [CrossRef]
  85. Qadir, M.; O’Loughlin, K.L.; Fricke, S.M.; Williamson, N.A.; Greco, W.R.; Minderman, H.; Baer, M.R. Cyclosporin A is a broad-spectrum multidrug resistance modulator. Clin. Cancer Res. 2005, 11, 2320–2326. [Google Scholar] [CrossRef]
  86. Gandhi, Y.A.; Morris, M.E. Structure-activity relationships and quantitative structure-activity relationships for breast cancer resistance protein (ABCG2). AAPS J. 2009, 11, 541–552. [Google Scholar] [CrossRef] [PubMed]
  87. Shim, J.S.; Liu, J.O. Recent advances in drug repositioning for the discovery of new anticancer drugs. Int. J. Biol. Sci. 2014, 10, 654–663. [Google Scholar] [CrossRef]
  88. Yang, D.; Kathawala, R.J.; Chufan, E.E.; Patel, A.; Ambudkar, S.V.; Chen, Z.S.; Chen, X. Tivozanib reverses multidrug resistance mediated by ABCB1 (P-glycoprotein) and ABCG2 (BCRP). Future Oncol. 2014, 10, 1827–1841. [Google Scholar] [CrossRef]
  89. Sen, R.; Natarajan, K.; Bhullar, J.; Shukla, S.; Fang, H.B.; Cai, L.; Chen, Z.S.; Ambudkar, S.V.; Baer, M.R. The novel BCR-ABL and FLT3 inhibitor ponatinib is a potent inhibitor of the MDR-associated ATP-binding cassette transporter ABCG2. Mol. Cancer Ther. 2012, 11, 2033–2044. [Google Scholar] [CrossRef]
  90. Miyata, H.; Takada, T.; Toyoda, Y.; Matsuo, H.; Ichida, K.; Suzuki, H. Identification of Febuxostat as a New Strong ABCG2 Inhibitor: Potential Applications and Risks in Clinical Situations. Front. Pharmacol. 2016, 7, 518. [Google Scholar] [CrossRef]
  91. Weiss, J.; Theile, D.; Dvorak, Z.; Haefeli, W.E. Interaction potential of the multitargeted receptor tyrosine kinase inhibitor dovitinib with drug transporters and drug metabolising enzymes assessed in vitro. Pharmaceutics 2014, 6, 632–650. [Google Scholar] [CrossRef] [PubMed]
  92. Dai, C.L.; Tiwari, A.K.; Wu, C.P.; Su, X.D.; Wang, S.R.; Liu, D.G.; Ashby, C.R., Jr.; Huang, Y.; Robey, R.W.; Liang, Y.J.; et al. Lapatinib (Tykerb, GW572016) reverses multidrug resistance in cancer cells by inhibiting the activity of ATP-binding cassette subfamily B member 1 and G member 2. Cancer Res. 2008, 68, 7905–7914. [Google Scholar] [CrossRef] [PubMed]
  93. Zheng, L.S.; Wang, F.; Li, Y.H.; Zhang, X.; Chen, L.M.; Liang, Y.J.; Dai, C.L.; Yan, Y.Y.; Tao, L.Y.; Mi, Y.J.; et al. Vandetanib (Zactima, ZD6474) antagonizes ABCC1- and ABCG2-mediated multidrug resistance by inhibition of their transport function. PLoS ONE 2009, 4, e5172. [Google Scholar] [CrossRef] [PubMed]
  94. Limtrakul, P.; Chearwae, W.; Shukla, S.; Phisalphong, C.; Ambudkar, S.V. Modulation of function of three ABC drug transporters, P-glycoprotein (ABCB1), mitoxantrone resistance protein (ABCG2) and multidrug resistance protein 1 (ABCC1) by tetrahydrocurcumin, a major metabolite of curcumin. Mol. Cell Biochem. 2007, 296, 85–95. [Google Scholar] [CrossRef] [PubMed]
  95. Valdameri, G.; Gauthier, C.; Terreux, R.; Kachadourian, R.; Day, B.J.; Winnischofer, S.M.; Rocha, M.E.; Frachet, V.; Ronot, X.; Di Pietro, A.; et al. Investigation of chalcones as selective inhibitors of the breast cancer resistance protein: Critical role of methoxylation in both inhibition potency and cytotoxicity. J. Med. Chem. 2012, 55, 3193–3200. [Google Scholar] [CrossRef] [PubMed]
  96. Pires, A.; Lecerf-Schmidt, F.; Guragossian, N.; Pazinato, J.; Gozzi, G.J.; Winter, E.; Valdameri, G.; Veale, A.; Boumendjel, A.; Di Pietro, A.; et al. New, highly potent and non-toxic, chromone inhibitors of the human breast cancer resistance protein ABCG2. Eur. J. Med. Chem. 2016, 122, 291–301. [Google Scholar] [CrossRef]
  97. Natarajan, K.; Xie, Y.; Baer, M.R.; Ross, D.D. Role of breast cancer resistance protein (BCRP/ABCG2) in cancer drug resistance. Biochem. Pharmacol. 2012, 83, 1084–1103. [Google Scholar] [CrossRef]
  98. Weiss, J.; Rose, J.; Storch, C.H.; Ketabi-Kiyanvash, N.; Sauer, A.; Haefeli, W.E.; Efferth, T. Modulation of human BCRP (ABCG2) activity by anti-HIV drugs. J. Antimicrob. Chemother. 2007, 59, 238–245. [Google Scholar] [CrossRef]
  99. Zhu, X.; Wong, I.L.K.; Chan, K.F.; Cui, J.; Law, M.C.; Chong, T.C.; Hu, X.; Chow, L.M.C.; Chan, T.H. Triazole Bridged Flavonoid Dimers as Potent, Nontoxic, and Highly Selective Breast Cancer Resistance Protein (BCRP/ABCG2) Inhibitors. J. Med. Chem. 2019, 62, 8578–8608. [Google Scholar] [CrossRef]
  100. Ahmed-Belkacem, A.; Pozza, A.; Munoz-Martinez, F.; Bates, S.E.; Castanys, S.; Gamarro, F.; Di Pietro, A.; Perez-Victoria, J.M. Flavonoid structure-activity studies identify 6-prenylchrysin and tectochrysin as potent and specific inhibitors of breast cancer resistance protein ABCG2. Cancer Res. 2005, 65, 4852–4860. [Google Scholar] [CrossRef]
  101. Zhang, Y.; Laterra, J.; Pomper, M.G. Hedgehog pathway inhibitor HhAntag691 is a potent inhibitor of ABCG2/BCRP and ABCB1/Pgp. Neoplasia 2009, 11, 96–101. [Google Scholar] [CrossRef] [PubMed]
  102. Morell, A.; Budagaga, Y.; Vagiannis, D.; Zhang, Y.; Lastovickova, L.; Novotna, E.; Haddad, A.; Haddad, M.; Portillo, R.; Hofman, J.; et al. Isocitrate dehydrogenase 2 inhibitor enasidenib synergizes daunorubicin cytotoxicity by targeting aldo-keto reductase 1C3 and ATP-binding cassette transporters. Arch. Toxicol. 2022, 96, 3265–3277. [Google Scholar] [CrossRef] [PubMed]
  103. Gupta, A.; Dai, Y.; Vethanayagam, R.R.; Hebert, M.F.; Thummel, K.E.; Unadkat, J.D.; Ross, D.D.; Mao, Q. Cyclosporin A, tacrolimus and sirolimus are potent inhibitors of the human breast cancer resistance protein (ABCG2) and reverse resistance to mitoxantrone and topotecan. Cancer Chemother. Pharmacol. 2006, 58, 374–383. [Google Scholar] [CrossRef] [PubMed]
  104. Kuhnle, M.; Egger, M.; Muller, C.; Mahringer, A.; Bernhardt, G.; Fricker, G.; Konig, B.; Buschauer, A. Potent and selective inhibitors of breast cancer resistance protein (ABCG2) derived from the p-glycoprotein (ABCB1) modulator tariquidar. J. Med. Chem. 2009, 52, 1190–1197. [Google Scholar] [CrossRef] [PubMed]
  105. Jabor Gozzi, G.; Bouaziz, Z.; Winter, E.; Daflon-Yunes, N.; Aichele, D.; Nacereddine, A.; Marminon, C.; Valdameri, G.; Zeinyeh, W.; Bollacke, A.; et al. Converting potent indeno[1,2-b]indole inhibitors of protein kinase CK2 into selective inhibitors of the breast cancer resistance protein ABCG2. J. Med. Chem. 2015, 58, 265–277. [Google Scholar] [CrossRef]
  106. Kita, D.H.; Guragossian, N.; Zattoni, I.F.; Moure, V.R.; Rego, F.G.M.; Lusvarghi, S.; Moulenat, T.; Belhani, B.; Picheth, G.; Bouacida, S.; et al. Mechanistic basis of breast cancer resistance protein inhibition by new indeno[1,2-b]indoles. Sci. Rep. 2021, 11, 1788. [Google Scholar] [CrossRef]
  107. Guragossian, N.; Belhani, B.; Moreno, A.; Nunes, M.T.; Gonzalez-Lobato, L.; Marminon, C.; Berthier, L.; Rocio Andrade Pires, A.D.; Ozvegy-Laczka, C.; Sarkadi, B.; et al. Uncompetitive nanomolar dimeric indenoindole inhibitors of the human breast cancer resistance pump ABCG2. Eur. J. Med. Chem. 2021, 211, 113017. [Google Scholar] [CrossRef]
  108. Antoni, F.; Bause, M.; Scholler, M.; Bauer, S.; Stark, S.A.; Jackson, S.M.; Manolaridis, I.; Locher, K.P.; Konig, B.; Buschauer, A.; et al. Tariquidar-related triazoles as potent, selective and stable inhibitors of ABCG2 (BCRP). Eur. J. Med. Chem. 2020, 191, 112133. [Google Scholar] [CrossRef]
  109. Poirier, A.; Portmann, R.; Cascais, A.C.; Bader, U.; Walter, I.; Ullah, M.; Funk, C. The need for human breast cancer resistance protein substrate and inhibition evaluation in drug discovery and development: Why, when, and how? Drug Metab. Dispos. 2014, 42, 1466–1477. [Google Scholar] [CrossRef]
  110. Yang, K.; Chen, Y.; To, K.K.; Wang, F.; Li, D.; Chen, L.; Fu, L. Alectinib (CH5424802) antagonizes ABCB1- and ABCG2-mediated multidrug resistance in vitro, in vivo and ex vivo. Exp. Mol. Med. 2017, 49, e303. [Google Scholar] [CrossRef]
  111. Hegedus, C.; Ozvegy-Laczka, C.; Apati, A.; Magocsi, M.; Nemet, K.; Orfi, L.; Keri, G.; Katona, M.; Takats, Z.; Varadi, A.; et al. Interaction of nilotinib, dasatinib and bosutinib with ABCB1 and ABCG2: Implications for altered anti-cancer effects and pharmacological properties. Br. J. Pharmacol. 2009, 158, 1153–1164. [Google Scholar] [CrossRef] [PubMed]
  112. Elsby, R.; Martin, P.; Surry, D.; Sharma, P.; Fenner, K. Solitary Inhibition of the Breast Cancer Resistance Protein Efflux Transporter Results in a Clinically Significant Drug-Drug Interaction with Rosuvastatin by Causing up to a 2-Fold Increase in Statin Exposure. Drug Metab. Dispos. 2016, 44, 398–408. [Google Scholar] [CrossRef] [PubMed]
  113. Nakamura, Y.; Oka, M.; Soda, H.; Shiozawa, K.; Yoshikawa, M.; Itoh, A.; Ikegami, Y.; Tsurutani, J.; Nakatomi, K.; Kitazaki, T.; et al. Gefitinib (“Iressa”, ZD1839), an epidermal growth factor receptor tyrosine kinase inhibitor, reverses breast cancer resistance protein/ABCG2-mediated drug resistance. Cancer Res. 2005, 65, 1541–1546. [Google Scholar] [CrossRef] [PubMed]
  114. Houghton, P.J.; Germain, G.S.; Harwood, F.C.; Schuetz, J.D.; Stewart, C.F.; Buchdunger, E.; Traxler, P. Imatinib mesylate is a potent inhibitor of the ABCG2 (BCRP) transporter and reverses resistance to topotecan and SN-38 in vitro. Cancer Res. 2004, 64, 2333–2337. [Google Scholar] [CrossRef] [PubMed]
  115. Ji, N.; Yang, Y.; Lei, Z.N.; Cai, C.Y.; Wang, J.Q.; Gupta, P.; Xian, X.; Yang, D.H.; Kong, D.; Chen, Z.S. Ulixertinib (BVD-523) antagonizes ABCB1- and ABCG2-mediated chemotherapeutic drug resistance. Biochem. Pharmacol. 2018, 158, 274–285. [Google Scholar] [CrossRef]
  116. Fan, Y.F.; Zhang, W.; Zeng, L.; Lei, Z.N.; Cai, C.Y.; Gupta, P.; Yang, D.H.; Cui, Q.; Qin, Z.D.; Chen, Z.S.; et al. Dacomitinib antagonizes multidrug resistance (MDR) in cancer cells by inhibiting the efflux activity of ABCB1 and ABCG2 transporters. Cancer Lett. 2018, 421, 186–198. [Google Scholar] [CrossRef]
  117. Sorf, A.; Sucha, S.; Morell, A.; Novotna, E.; Staud, F.; Zavrelova, A.; Visek, B.; Wsol, V.; Ceckova, M. Targeting Pharmacokinetic Drug Resistance in Acute Myeloid Leukemia Cells with CDK4/6 Inhibitors. Cancers 2020, 12, 1596. [Google Scholar] [CrossRef]
  118. Sabet, Z.; Vagiannis, D.; Budagaga, Y.; Zhang, Y.; Novotna, E.; Hanke, I.; Rozkos, T.; Hofman, J. Talazoparib Does Not Interact with ABCB1 Transporter or Cytochrome P450s, but Modulates Multidrug Resistance Mediated by ABCC1 and ABCG2: An in Vitro and Ex Vivo Study. Int. J. Mol. Sci. 2022, 23, 14338. [Google Scholar] [CrossRef]
  119. Jing, W.; Zhou, M.; Chen, R.; Ye, X.; Li, W.; Su, X.; Luo, J.; Wang, Z.; Peng, S. In vitro and ex vivo anti-tumor effect and mechanism of Tucatinib in leukemia stem cells and ABCG2-overexpressing leukemia cells. Oncol. Rep. 2021, 45, 1142–1152. [Google Scholar] [CrossRef]
  120. Wang, J.Q.; Li, J.Y.; Teng, Q.X.; Lei, Z.N.; Ji, N.; Cui, Q.; Zeng, L.; Pan, Y.; Yang, D.H.; Chen, Z.S. Venetoclax, a BCL-2 Inhibitor, Enhances the Efficacy of Chemotherapeutic Agents in Wild-Type ABCG2-Overexpression-Mediated MDR Cancer Cells. Cancers 2020, 12, 466. [Google Scholar] [CrossRef]
  121. Zhang, W.; McIntyre, C.; Kuhn, M.; Forbes, H.; Kim, T.M.; Lee, J.; Demidov, L.; Colburn, D. Effect of Vemurafenib on the Pharmacokinetics of a Single Dose of Digoxin in Patients With BRAF(V600) Mutation-Positive Metastatic Malignancy. J. Clin. Pharmacol. 2018, 58, 1067–1073. [Google Scholar] [CrossRef] [PubMed]
  122. Cermakova, L.; Hofman, J.; Lastovickova, L.; Havlickova, L.; Springrova, I.; Novotna, E.; Wsol, V. Bruton’s Tyrosine Kinase Inhibitor Zanubrutinib Effectively Modulates Cancer Resistance by Inhibiting Anthracycline Metabolism and Efflux. Pharmaceutics 2022, 14, 1994. [Google Scholar] [CrossRef]
  123. Abi Hussein, H.; Geneix, C.; Petitjean, M.; Borrel, A.; Flatters, D.; Camproux, A.C. Global vision of druggability issues: Applications and perspectives. Drug Discov. Today 2017, 22, 404–415. [Google Scholar] [CrossRef] [PubMed]
  124. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 2001, 46, 3–26. [Google Scholar] [CrossRef] [PubMed]
  125. Moinul, M.; Amin, S.A.; Jha, T.; Gayen, S. Updated chemical scaffolds of ABCG2 inhibitors and their structure-inhibition relationships for future development. Eur. J. Med. Chem. 2022, 241, 114628. [Google Scholar] [CrossRef] [PubMed]
  126. Jabari, M.; Allahbakhshian Farsani, M.; Salari, S.; Hamidpour, M.; Amiri, V.; Mohammadi, M.H. Hypoxia-Inducible Factor1-Alpha (HIF1alpha) and Vascular Endothelial Growth Factor-A (VEGF-A) Expression in De Novo AML Patients. Asian Pac. J. Cancer Prev. 2019, 20, 705–710. [Google Scholar] [CrossRef]
  127. Xu, P.; Wang, M.; Jiang, Y.; Ouyang, J.; Chen, B. The association between expression of hypoxia inducible factor-1alpha and multi-drug resistance of acute myeloid leukemia. Transl. Cancer Res. 2017, 6, 198–205. [Google Scholar] [CrossRef]
  128. Wang, Y.; Liu, Y.; Malek, S.N.; Zheng, P.; Liu, Y. Targeting HIF1alpha eliminates cancer stem cells in hematological malignancies. Cell Stem Cell 2011, 8, 399–411. [Google Scholar] [CrossRef]
  129. Miller, D.M.; Thomas, S.D.; Islam, A.; Muench, D.; Sedoris, K. c-Myc and cancer metabolism. Clin. Cancer Res. 2012, 18, 5546–5553. [Google Scholar] [CrossRef]
  130. Porro, A.; Iraci, N.; Soverini, S.; Diolaiti, D.; Gherardi, S.; Terragna, C.; Durante, S.; Valli, E.; Kalebic, T.; Bernardoni, R.; et al. c-MYC oncoprotein dictates transcriptional profiles of ATP-binding cassette transporter genes in chronic myelogenous leukemia CD34+ hematopoietic progenitor cells. Mol. Cancer Res. 2011, 9, 1054–1066. [Google Scholar] [CrossRef]
  131. Zhang, H.L.; Wang, P.; Lu, M.Z.; Zhang, S.D. c-Myc regulation of ATP-binding cassette transporter reverses chemoresistance in CD133(+) colon cancer stem cells. Sheng Li Xue Bao 2016, 68, 171–178. [Google Scholar] [PubMed]
  132. Wang, F.; Huang, J.; Guo, T.; Zheng, Y.; Zhang, L.; Zhang, D.; Wang, F.; Naren, D.; Cui, Y.; Liu, X.; et al. Homoharringtonine synergizes with quizartinib in FLT3-ITD acute myeloid leukemia by targeting FLT3-AKT-c-Myc pathway. Biochem. Pharmacol. 2021, 188, 114538. [Google Scholar] [CrossRef] [PubMed]
  133. Rosenfeldt, M.T.; Bell, L.A.; Long, J.S.; O’Prey, J.; Nixon, C.; Roberts, F.; Dufes, C.; Ryan, K.M. E2F1 drives chemotherapeutic drug resistance via ABCG2. Oncogene 2014, 33, 4164–4172. [Google Scholar] [CrossRef] [PubMed]
  134. Huang, Y.; Hong, X.; Hu, J.; Lu, Q. Targeted regulation of MiR-98 on E2F1 increases chemosensitivity of leukemia cells K562/A02. Onco Targets Ther. 2017, 10, 3233–3239. [Google Scholar] [CrossRef] [PubMed]
  135. Giuliani, C.; Bucci, I.; Napolitano, G. The Role of the Transcription Factor Nuclear Factor-kappa B in Thyroid Autoimmunity and Cancer. Front. Endocrinol. 2018, 9, 471. [Google Scholar] [CrossRef] [PubMed]
  136. Labbozzetta, M.; Notarbartolo, M.; Poma, P. Can NF-kappaB Be Considered a Valid Drug Target in Neoplastic Diseases? Our Point of View. Int. J. Mol. Sci. 2020, 21, 3070. [Google Scholar] [CrossRef] [PubMed]
  137. Wang, J.; Xin, B.; Wang, H.; He, X.; Wei, W.; Zhang, T.; Shen, X. Gastrin regulates ABCG2 to promote the migration, invasion and side populations in pancreatic cancer cells via activation of NF-kappaB signaling. Exp. Cell Res. 2016, 346, 74–84. [Google Scholar] [CrossRef] [PubMed]
  138. Jeong, H.W.; Cui, W.; Yang, Y.; Lu, J.; He, J.; Li, A.; Song, D.; Guo, Y.; Liu, B.H.; Chai, L. SALL4, a stem cell factor, affects the side population by regulation of the ATP-binding cassette drug transport genes. PLoS ONE 2011, 6, e18372. [Google Scholar] [CrossRef]
  139. Ke, S.Z.; Ni, X.Y.; Zhang, Y.H.; Wang, Y.N.; Wu, B.; Gao, F.G. Camptothecin and cisplatin upregulate ABCG2 and MRP2 expression by activating the ATM/NF-kappaB pathway in lung cancer cells. Int. J. Oncol. 2013, 42, 1289–1296. [Google Scholar] [CrossRef]
  140. Szatmari, I.; Vamosi, G.; Brazda, P.; Balint, B.L.; Benko, S.; Szeles, L.; Jeney, V.; Ozvegy-Laczka, C.; Szanto, A.; Barta, E.; et al. Peroxisome proliferator-activated receptor gamma-regulated ABCG2 expression confers cytoprotection to human dendritic cells. J. Biol. Chem. 2006, 281, 23812–23823. [Google Scholar] [CrossRef]
  141. Kim, C.E.; Park, H.Y.; Won, H.J.; Kim, M.; Kwon, B.; Lee, S.J.; Kim, D.H.; Shin, J.G.; Seo, S.K. Repression of PPARgamma reduces the ABCG2-mediated efflux activity of M2 macrophages. Int. J. Biochem. Cell Biol. 2021, 130, 105895. [Google Scholar] [CrossRef]
  142. To, K.K.; Tomlinson, B. Targeting the ABCG2-overexpressing multidrug resistant (MDR) cancer cells by PPARgamma agonists. Br. J. Pharmacol. 2013, 170, 1137–1151. [Google Scholar] [CrossRef] [PubMed]
  143. Esmaeili, S.; Safaroghli-Azar, A.; Pourbagheri-Sigaroodi, A.; Salari, S.; Gharehbaghian, A.; Hamidpour, M.; Bashash, D. Stimulation of Peroxisome Proliferator-Activated Receptor-Gamma (PPARgamma) using Pioglitazone Decreases the Survival of Acute Promyelocytic Leukemia Cells through Up-Regulation of PTEN Expression. Anti-Cancer Agents Med. Chem. 2021, 21, 108–119. [Google Scholar] [CrossRef]
  144. Dhakshinamoorthy, S.; Long, D.J., 2nd; Jaiswal, A.K. Antioxidant regulation of genes encoding enzymes that detoxify xenobiotics and carcinogens. Curr. Top. Cell Regul. 2000, 36, 201–216. [Google Scholar] [CrossRef]
  145. Jalali, A.; Mahmoudi, S.; Larki Harchegani, A.; Mohammadiasl, J.; Ahmadzadeh, A. Evaluation of Nrf2, Keap1 and Apoptotic Pathway Genes Expression in Acute Myeloid Leukemia Patients. Iran. J. Pharm. Res. 2021, 20, 398–407. [Google Scholar] [CrossRef] [PubMed]
  146. Hu, T.; Pan, C.; Zhang, T.; Ni, M.; Wang, W.; Zhang, S.; Chen, Y.; Wang, J.; Fang, Q. Nrf2 overexpression increases the resistance of acute myeloid leukemia to cytarabine by inhibiting replication factor C4. Cancer Gene Ther. 2022, 29, 1773–1790. [Google Scholar] [CrossRef] [PubMed]
  147. Kukal, S.; Guin, D.; Rawat, C.; Bora, S.; Mishra, M.K.; Sharma, P.; Paul, P.R.; Kanojia, N.; Grewal, G.K.; Kukreti, S.; et al. Multidrug efflux transporter ABCG2: Expression and regulation. Cell Mol. Life Sci. 2021, 78, 6887–6939. [Google Scholar] [CrossRef]
  148. Kim, E.J.; Kim, Y.J.; Lee, H.I.; Jeong, S.H.; Nam, H.J.; Cho, J.H. NRF2 Knockdown Resensitizes 5-Fluorouracil-Resistant Pancreatic Cancer Cells by Suppressing HO-1 and ABCG2 Expression. Int. J. Mol. Sci. 2020, 21, 4646. [Google Scholar] [CrossRef]
  149. Choi, B.H.; Ryu, D.Y.; Ryoo, I.G.; Kwak, M.K. NFE2L2/NRF2 silencing-inducible miR-206 targets c-MET/EGFR and suppresses BCRP/ABCG2 in cancer cells. Oncotarget 2017, 8, 107188–107205. [Google Scholar] [CrossRef]
  150. Crawford, R.R.; Potukuchi, P.K.; Schuetz, E.G.; Schuetz, J.D. Beyond Competitive Inhibition: Regulation of ABC Transporters by Kinases and Protein-Protein Interactions as Potential Mechanisms of Drug-Drug Interactions. Drug Metab. Dispos. 2018, 46, 567–580. [Google Scholar] [CrossRef]
  151. Darby, R.A.; Unsworth, A.; Knapp, S.; Kerr, I.D.; Callaghan, R. Overcoming ABCG2-mediated drug resistance with imidazo-[1,2-b]-pyridazine-based Pim1 kinase inhibitors. Cancer Chemother. Pharmacol. 2015, 76, 853–864. [Google Scholar] [CrossRef] [PubMed]
  152. Goler-Baron, V.; Assaraf, Y.G. Structure and function of ABCG2-rich extracellular vesicles mediating multidrug resistance. PLoS ONE 2011, 6, e16007. [Google Scholar] [CrossRef] [PubMed]
  153. Wang, X.Q.; Ongkeko, W.M.; Chen, L.; Yang, Z.F.; Lu, P.; Chen, K.K.; Lopez, J.P.; Poon, R.T.; Fan, S.T. Octamer 4 (Oct4) mediates chemotherapeutic drug resistance in liver cancer cells through a potential Oct4-AKT-ATP-binding cassette G2 pathway. Hepatology 2010, 52, 528–539. [Google Scholar] [CrossRef] [PubMed]
  154. Tomiyasu, H.; Watanabe, M.; Sugita, K.; Goto-Koshino, Y.; Fujino, Y.; Ohno, K.; Sugano, S.; Tsujimoto, H. Regulations of ABCB1 and ABCG2 expression through MAPK pathways in acute lymphoblastic leukemia cell lines. Anticancer Res. 2013, 33, 5317–5323. [Google Scholar] [PubMed]
  155. Chau, W.K.; Ip, C.K.; Mak, A.S.; Lai, H.C.; Wong, A.S. c-Kit mediates chemoresistance and tumor-initiating capacity of ovarian cancer cells through activation of Wnt/beta-catenin-ATP-binding cassette G2 signaling. Oncogene 2013, 32, 2767–2781. [Google Scholar] [CrossRef] [PubMed]
  156. To, K.K.; Zhan, Z.; Litman, T.; Bates, S.E. 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. 2008, 28, 5147–5161. [Google Scholar] [CrossRef]
  157. To, K.K.; Zhan, Z.; Bates, S.E. Aberrant promoter methylation of the ABCG2 gene in renal carcinoma. Mol. Cell Biol. 2006, 26, 8572–8585. [Google Scholar] [CrossRef]
  158. Bram, E.E.; Stark, M.; Raz, S.; Assaraf, Y.G. Chemotherapeutic drug-induced ABCG2 promoter demethylation as a novel mechanism of acquired multidrug resistance. Neoplasia 2009, 11, 1359–1370. [Google Scholar] [CrossRef]
  159. Kawabata, K.C.; Hayashi, Y.; Inoue, D.; Meguro, H.; Sakurai, H.; Fukuyama, T.; Tanaka, Y.; Asada, S.; Fukushima, T.; Nagase, R.; et al. High expression of ABCG2 induced by EZH2 disruption has pivotal roles in MDS pathogenesis. Leukemia 2018, 32, 419–428. [Google Scholar] [CrossRef]
  160. Hauswald, S.; Duque-Afonso, J.; Wagner, M.M.; Schertl, F.M.; Lubbert, M.; Peschel, C.; Keller, U.; Licht, T. Histone deacetylase inhibitors induce a very broad, pleiotropic anticancer drug resistance phenotype in acute myeloid leukemia cells by modulation of multiple ABC transporter genes. Clin. Cancer Res. 2009, 15, 3705–3715. [Google Scholar] [CrossRef]
  161. Hsu, M.C.; Pan, M.R.; Chu, P.Y.; Tsai, Y.L.; Tsai, C.H.; Shan, Y.S.; Chen, L.T.; Hung, W.C. Protein Arginine Methyltransferase 3 Enhances Chemoresistance in Pancreatic Cancer by Methylating hnRNPA1 to Increase ABCG2 Expression. Cancers 2018, 11, 8. [Google Scholar] [CrossRef] [PubMed]
  162. Tafrihi, M.; Hasheminasab, E. MiRNAs: Biology, Biogenesis, their Web-based Tools, and Databases. Microrna 2019, 8, 4–27. [Google Scholar] [CrossRef] [PubMed]
  163. Pan, Y.Z.; Morris, M.E.; Yu, A.M. MicroRNA-328 negatively regulates the expression of breast cancer resistance protein (BCRP/ABCG2) in human cancer cells. Mol. Pharmacol. 2009, 75, 1374–1379. [Google Scholar] [CrossRef] [PubMed]
  164. Ma, M.T.; He, M.; Wang, Y.; Jiao, X.Y.; Zhao, L.; Bai, X.F.; Yu, Z.J.; Wu, H.Z.; Sun, M.L.; Song, Z.G.; et al. MiR-487a resensitizes mitoxantrone (MX)-resistant breast cancer cells (MCF-7/MX) to MX by targeting breast cancer resistance protein (BCRP/ABCG2). Cancer Lett. 2013, 339, 107–115. [Google Scholar] [CrossRef] [PubMed]
  165. Jiao, X.; Zhao, L.; Ma, M.; Bai, X.; He, M.; Yan, Y.; Wang, Y.; Chen, Q.; Zhao, X.; Zhou, M.; et al. MiR-181a enhances drug sensitivity in mitoxantone-resistant breast cancer cells by targeting breast cancer resistance protein (BCRP/ABCG2). Breast Cancer Res. Treat. 2013, 139, 717–730. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, Y.; Zhao, L.; Xiao, Q.; Jiang, L.; He, M.; Bai, X.; Ma, M.; Jiao, X.; Wei, M. miR-302a/b/c/d cooperatively inhibit BCRP expression to increase drug sensitivity in breast cancer cells. Gynecol. Oncol. 2016, 141, 592–601. [Google Scholar] [CrossRef]
  167. Sugiyama, T.; Shuto, T.; Suzuki, S.; Sato, T.; Koga, T.; Suico, M.A.; Kusuhara, H.; Sugiyama, Y.; Cyr, D.M.; Kai, H. Posttranslational negative regulation of glycosylated and non-glycosylated BCRP expression by Derlin-1. Biochem. Biophys. Res. Commun. 2011, 404, 853–858. [Google Scholar] [CrossRef]
  168. Ding, R.; Shi, J.; Pabon, K.; Scotto, K.W. Xanthines down-regulate the drug transporter ABCG2 and reverse multidrug resistance. Mol. Pharmacol. 2012, 81, 328–337. [Google Scholar] [CrossRef]
  169. Wei, Y.; Ma, Y.; Zhao, Q.; Ren, Z.; Li, Y.; Hou, T.; Peng, H. New use for an old drug: Inhibiting ABCG2 with sorafenib. Mol. Cancer Ther. 2012, 11, 1693–1702. [Google Scholar] [CrossRef]
  170. Hu, Y.P.; Tao, L.Y.; Wang, F.; Zhang, J.Y.; Liang, Y.J.; Fu, L.W. Secalonic acid D reduced the percentage of side populations by down-regulating the expression of ABCG2. Biochem. Pharmacol. 2013, 85, 1619–1625. [Google Scholar] [CrossRef]
  171. Wang, H.; Chi, C.H.; Zhang, Y.; Shi, B.; Jia, R.; Wang, B.J. Effects of histone deacetylase inhibitors on ATP-binding cassette transporters in lung cancer A549 and colorectal cancer HCT116 cells. Oncol. Lett. 2019, 18, 63–71. [Google Scholar] [CrossRef] [PubMed]
  172. Zhang, X.; Liu, X.; Zhou, W.; Yang, M.; Ding, Y.; Wang, Q.; Hu, R. Fasudil increases temozolomide sensitivity and suppresses temozolomide-resistant glioma growth via inhibiting ROCK2/ABCG2. Cell Death Dis. 2018, 9, 190. [Google Scholar] [CrossRef] [PubMed]
  173. Wen, S.H.; Su, S.C.; Liou, B.H.; Lin, C.H.; Lee, K.R. Sulbactam-enhanced cytotoxicity of doxorubicin in breast cancer cells. Cancer Cell Int. 2018, 18, 128. [Google Scholar] [CrossRef]
  174. Yang, H.; Wei, Y.C.; Li, W.C.; Chen, H.Y.; Lin, H.Y.; Chiang, C.P.; Chen, H.M. Natural Compounds Modulate Drug Transporter Mediated Oral Cancer Treatment. Biomolecules 2020, 10, 1335. [Google Scholar] [CrossRef] [PubMed]
  175. Zhu, M.M.; Tong, J.L.; Xu, Q.; Nie, F.; Xu, X.T.; Xiao, S.D.; Ran, Z.H. Increased JNK1 signaling pathway is responsible for ABCG2-mediated multidrug resistance in human colon cancer. PLoS ONE 2012, 7, e41763. [Google Scholar] [CrossRef]
  176. El-Readi, M.Z.; Eid, S.; Abdelghany, A.A.; Al-Amoudi, H.S.; Efferth, T.; Wink, M. Resveratrol mediated cancer cell apoptosis, and modulation of multidrug resistance proteins and metabolic enzymes. Phytomedicine 2019, 55, 269–281. [Google Scholar] [CrossRef]
  177. Eid, S.Y.; Althubiti, M.A.; Abdallah, M.E.; Wink, M.; El-Readi, M.Z. The carotenoid fucoxanthin can sensitize multidrug resistant cancer cells to doxorubicin via induction of apoptosis, inhibition of multidrug resistance proteins and metabolic enzymes. Phytomedicine 2020, 77, 153280. [Google Scholar] [CrossRef]
  178. Huang, W.; Wan, C.; Luo, Q.; Huang, Z.; Luo, Q. Genistein-inhibited cancer stem cell-like properties and reduced chemoresistance of gastric cancer. Int. J. Mol. Sci. 2014, 15, 3432–3443. [Google Scholar] [CrossRef]
  179. Valera, E.T.; de Freitas Cortez, M.A.; de Paula Queiroz, R.G.; de Oliveira, F.M.; Brassesco, M.S.; Jabado, N.; Faury, D.; Bobola, M.S.; Machado, H.R.; Scrideli, C.A.; et al. Pediatric glioblastoma cell line shows different patterns of expression of transmembrane ABC transporters after in vitro exposure to vinblastine. Childs Nerv. Syst. 2009, 25, 39–45. [Google Scholar] [CrossRef]
  180. Sodani, K.; Patel, A.; Anreddy, N.; Singh, S.; Yang, D.H.; Kathawala, R.J.; Kumar, P.; Talele, T.T.; Chen, Z.S. Telatinib reverses chemotherapeutic multidrug resistance mediated by ABCG2 efflux transporter in vitro and in vivo. Biochem. Pharmacol. 2014, 89, 52–61. [Google Scholar] [CrossRef]
  181. Jamieson, C.; Martinelli, G.; Papayannidis, C.; Cortes, J.E. Hedgehog Pathway Inhibitors: A New Therapeutic Class for the Treatment of Acute Myeloid Leukemia. Blood Cancer Discov. 2020, 1, 134–145. [Google Scholar] [CrossRef] [PubMed]
  182. Zhang, Y.; Wang, H.; Wei, L.; Li, G.; Yu, J.; Gao, Y.; Gao, P.; Zhang, X.; Wei, F.; Yin, D.; et al. Transcriptional modulation of BCRP gene to reverse multidrug resistance by toremifene in breast adenocarcinoma cells. Breast Cancer Res. Treat. 2010, 123, 679–689. [Google Scholar] [CrossRef]
  183. Honorat, M.; Mesnier, A.; Di Pietro, A.; Lin, V.; Cohen, P.; Dumontet, C.; Payen, L. Dexamethasone down-regulates ABCG2 expression levels in breast cancer cells. Biochem. Biophys. Res. Commun. 2008, 375, 308–314. [Google Scholar] [CrossRef] [PubMed]
  184. Imai, Y.; Ohmori, K.; Yasuda, S.; Wada, M.; Suzuki, T.; Fukuda, K.; Ueda, Y. Breast cancer resistance protein/ABCG2 is differentially regulated downstream of extracellular signal-regulated kinase. Cancer Sci. 2009, 100, 1118–1127. [Google Scholar] [CrossRef] [PubMed]
  185. Dohse, M.; Scharenberg, C.; Shukla, S.; Robey, R.W.; Volkmann, T.; Deeken, J.F.; Brendel, C.; Ambudkar, S.V.; Neubauer, A.; Bates, S.E. Comparison of ATP-binding cassette transporter interactions with the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib. Drug Metab. Dispos. 2010, 38, 1371–1380. [Google Scholar] [CrossRef] [PubMed]
  186. Hu, C.; Li, H.; Li, J.; Zhu, Z.; Yin, S.; Hao, X.; Yao, M.; Zheng, S.; Gu, J. Analysis of ABCG2 expression and side population identifies intrinsic drug efflux in the HCC cell line MHCC-97L and its modulation by Akt signaling. Carcinogenesis 2008, 29, 2289–2297. [Google Scholar] [CrossRef]
  187. Zhang, J.; Hu, J.; Li, W.; Zhang, C.; Su, P.; Wang, Y.; Sun, W.; Wang, X.; Li, L.; Wu, X. Rapamycin Antagonizes BCRP-Mediated Drug Resistance Through the PI3K/Akt/mTOR Signaling Pathway in mPRalpha-Positive Breast Cancer. Front. Oncol. 2021, 11, 608570. [Google Scholar] [CrossRef]
  188. Fischer, B.; Frei, C.; Moura, U.; Stahel, R.; Felley-Bosco, E. Inhibition of phosphoinositide-3 kinase pathway down regulates ABCG2 function and sensitizes malignant pleural mesothelioma to chemotherapy. Lung Cancer 2012, 78, 23–29. [Google Scholar] [CrossRef]
Figure 1. ABCG2 structure and variants at the N-terminus nucleotide-binding domain (NBD) in purple transmembrane domains (TMDs) [Adapted from Noguchi et al. [36]].
Figure 1. ABCG2 structure and variants at the N-terminus nucleotide-binding domain (NBD) in purple transmembrane domains (TMDs) [Adapted from Noguchi et al. [36]].
Biomedicines 12 00111 g001
Table 1. ABCG2 substrates by class of compounds.
Table 1. ABCG2 substrates by class of compounds.
Chemotherapy agents
Mitoxantrone [22], Daunorubicin [42], Doxorubicin [42], Epirubicin [42], Idarubicin [42], Bisantrene [42], Flavopiridol [43], Etoposide [44], Teniposide [44], Topotecan [45] Irinotecan [46], Diflomotecan [47], Homocamptothecan (weak) [47], Methotrexate [48], Tomudex [49], Imatinib [50], Nilotinib [51], Gefitinib [52], Canertinib [53]Triazolacridones [54]
Antivirals
Zidovudine [55], Lamivudine [56], Abacavir [57]
HMG-CoA reductase inhibitors
Rosuvastatin [58], Pitavastatin [59], Cerivastatin [60]
Porphyrins
Pheophorbide A [61], Chlorin e6 [61], Protoporphyrin IX [61], Phytoporphyrin [62]
Flavonoids
Genistein [63], quercetin [64]
Antibiotics
Ciprofloxacin [65], Ofloxacin [65], Norfloxacin [65], Erythromycin [66], Rifampicin [66], Nitrofurantoin [67], Sulfasalazine [68]
Benzimidazoles
Albedazole sulfoxide [69], Oxfendazole [69], Pantoprazole [70]
Pyridines
Nitrendipine [71], Azidopine [71], Dipyridamole [72], Folic acid [73], Cimetidine [74], Riboflavin [75]
Fluorescent compounds
Hoechst 33342 [76], Rhodamin 123 [77], Lysotracker green [77], BODIPY-prazosin [77]
Carcinogens
Aflatoxin B [78], 2-amino-3-methylimidazo[4,5-f]quinoline [78], 3-amino-1,4-dimethyl-5H-pyrido[4,3-b]indole [78], 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine [74]
Table 2. ABCG2 inhibitors are listed by structural class.
Table 2. ABCG2 inhibitors are listed by structural class.
Structural ClassCompoundIC50 (μM)Ref.
ChalconesIndolylphenylproenone0.27[95]
ChromonesChromone4a0.086[96]
Chromone310.046[97]
DiketopiperazinesKo143(FTC analog)0.01[98]
FlavonoidsFlavone2.8[99]
6-prenylchrysin0.29[100]
Flavonoid dimer1[99]
Hedgehog pathway inhibitorsVismodegib1.4[101]
Isocitrate dehydrogenase inhibitorsEnasidenib1[102]
ImmunosuppressantsSirolimus1.9[103]
Non-purine xanthine oxidase inhibitorsFebuxostat0.027[90]
Topiroxostat0.18[90]
Benzbromarone0.2[90]
ABCB1 inhibitorsTariquidar0.9[104]
Tariquidar derivative 60.06[104]
Indenoindole-type derivativesIndeno[1,2-b] indole0.21[105]
9-hydroxyindeno[1,2-b] indole0.21[106]
Indeno[1,2-b] indole homodimer0.024[107]
Tariquidar-related triazolesIR-ST10.14[108]
UR-MB1080.079[108]
Thrombopoietin receptorEltrombopag3.1[109]
Tyrosine kinase inhibitorsAlectinib1.5[110]
Bosutinib2[111]
Dasatinib2[111]
Erlotinib0.13[36]
Fostamatinib0.05[112]
Gefitinb0.5[113]
Ponatinib0.04[89]
Vandetanib0.2[93]
Tivozanib0.07[88]
Imatinib1[114]
Ulixertinib1[115]
Dacomitinib1[116]
Table 3. Modulators of ABCG2 expression by regulation site.
Table 3. Modulators of ABCG2 expression by regulation site.
Regulation LevelDrugRef.
TranscriptionVorinostat[171]
Trichostatin A[171]
Fasudil (rho kinase inhibitor)[172]
Sulbactam[173]
Ly294002 (PIK3 inhibitor)[174]
SP600125 (JNK inhibitor)[175]
Resveratrol[176]
Fucoxanthin[177]
Genistein[178]
Vinblastine[179]
Gefitinib[174]
Telatinib[180]
Glasdegib[181]
Toremifene[182]
Dexamethasone[183]
Protein degradationSorafenib[169]
PD98059 (MEK inhibitor)[184]
UO126 (MEK inhibitor)[184]
Imatinib[185]
Protein internalizationLy294002 (PIK3 inhibitor)[186]
Fasudil[172]
Rapamycin[187]
NVP-BEZ235 (dualPI3K/mTOR Inhibitor)[188]
Pioglitazone[142]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Damiani, D.; Tiribelli, M. ATP-Binding Cassette Subfamily G Member 2 in Acute Myeloid Leukemia: A New Molecular Target? Biomedicines 2024, 12, 111. https://doi.org/10.3390/biomedicines12010111

AMA Style

Damiani D, Tiribelli M. ATP-Binding Cassette Subfamily G Member 2 in Acute Myeloid Leukemia: A New Molecular Target? Biomedicines. 2024; 12(1):111. https://doi.org/10.3390/biomedicines12010111

Chicago/Turabian Style

Damiani, Daniela, and Mario Tiribelli. 2024. "ATP-Binding Cassette Subfamily G Member 2 in Acute Myeloid Leukemia: A New Molecular Target?" Biomedicines 12, no. 1: 111. https://doi.org/10.3390/biomedicines12010111

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop