Overview
ABC subfamily G, isoform 2 (ABCG2) is a gene encoding the Breast Cancer Resistance Protein (BCRP), which is an ATP-binding cassette (ABC) efflux transporter [1]. The transporter has a broad substrate profile, including much overlap with P-glycoprotein (P-gp; encoded by the gene ABCB1) and with MRP1 (encoded by the gene ABCC1). Together these 3 transporters are among the most studied members of the ABC efflux transporter family [2].
BCRP substrates include endogenous and naturally occurring polar molecules, particularly conjugated organic ions, in addition to chemotherapeutics [1]. These substrates include dietary flavonoids, porphyrins such as heme, estrogen precursors, and the carcinogenic heterocyclic amines (PhIP) found in cooked meat and fish [1,3]. Over-activity and overexpression of the BCRP protein, such as caused by genetic variation, have been associated with resistance to chemotherapy, including tyrosine kinase inhibitors. The International Transporter Consortium has identified ABCG2 as a pharmacogene with clinically important polymorphisms [4]. Here we describe the role of ABCG2 in efflux transport and highlight its pharmacogenetic relationships.
Molecular and Protein Structure
The ABCG2 gene was first discovered in 1998 in a breast cancer cell line, MCF-7/AdrVp, which was resistant to chemotherapy, but did not express other efflux transporters that had already been associated with resistance at that time [3]. As a result the protein was named Breast Cancer Resistance Protein (BCRP). Subsequently, the cDNA was cloned, and the gene, ABCG2, was identified on chromosome 4q22 [2]. ABCG2 consists of 16 exons and 15 introns that span 66 kb [5] (Entrez Gene ID: 9429, GenBank reference sequence NG_032067.2). The BCRP protein is 72 kDa in size and contains 655 amino acids, with the translation start site located in exon 2 [6,7]. There have been three distinct upstream promoters identified that affect the size of the 5′ UTR, but all three isoforms retain the ATG start site in exon 2 [8]. The ABCG2 promoter is TATA-less and contains multiple SP-1 sites downstream of a CpG island [5]. BCRP expression is significantly decreased by methylation of the CpG island and increased upon the inhibition of histone deacetylase (HDAC) [9–11].
Despite much overlap in substrates, the protein orientation is the reverse configuration compared to other ABC transporters, with the ATP-binding domain at the N terminus and the transmembrane domains at the C terminus [7,12]. The consequences of this reversed orientation are unknown. However, transport direction does not seem to be affected, as binding pockets flank the nucleotide binding domain as they do for P-gp [13]. G family transporters like BCRP are considered “half-transporters” because they have only one of each of the nucleotide binding and membrane spanning domains compared to other ABC transporters, which have two [14,15]. A 3-D structure of BCRP, analyzed by electron crystallography from 2-D crystals, revealed that BCRP exists as a symmetric homodimer, but recent studies have suggested the possibility that BCRP may also function as a higher order oligomer such as a tetramer [16,17]. The protein is thought to homodimerize via intermolecular disulfide bridges to create the active transporter [18,19]. This dimerization is mediated by Pim-1 phosphorylation at Thr362 in the endoplasmic reticulum before the protein is transferred to the golgi for post-translational processing, after which it is transported to the apical cell membrane [20,21]. Although BCRP is typically glycosylated at the Arg596 residue, this glycosylation does not appear to be important for expression, trafficking, or function [22].
The ABCG2 gene is found in all vertebrates that have been sequenced [23]. While fish have 3 or more copies of the ABCG2 gene, most species have only one copy [23,24].
Tissue Distribution and Physiological Function
The BCRP protein functions as an efflux transporter that protects tissues from xenobiotics and harmful metabolites. Expressed on apical cell membranes of the placenta, brain, prostate, gastrointestinal tract, testes, ovaries, hepatocytes, renal tubules, stem cells, adrenal gland, uterus, bile ducts, gallbladder, central nervous system, and endothelium of veins and capillaries, BCRP mediates absorption, distribution, and elimination of its substrates [1,25–29]. BCRP is most highly expressed in brain tissue, the cervix, the small intestine, and the uterus [30]. Efflux by BCRP requires ATP hydrolysis to shuttle molecules against the concentration gradient [2,3,31]. Activity is altered by the lipid membrane environment, specifically cholesterol content, and may vary between tissues [3,32,33].
BCRP-mediated efflux protects tissues against mutagens and carcinogens found in the diet, such as polycyclic aromatic hydrocarbons and heterocyclic amines [7]. Mice deficient in BCRP1 were found to have higher area under the plasma concentration-time curve (AUC) exposure to the carcinogen PhIP [7]. BCRP is also highly expressed in a side population of stem cells derived from the bone marrow. These cells have long-term repopulating capacity and BCRP acts as a protective barrier from oxidative stress products that could cause genetic damage. It is also thought that BCRP, along with P-gp, may play an important role in organ regeneration [34].
Efflux via BCRP acts as a protective barrier in tissues central to substrate absorption and elimination. Within the intestine, BCRP expression decreases along the length of the intestine, where it limits absorption of xenobiotics from the gut [35]. BCRP is also highly expressed in the kidney and liver, where it mediates the elimination of its substrates from the blood. A vital part of the blood-brain barrier, BCRP limits the penetration of compounds into the brain. In the kidney, BCRP is a high-capacity transporter for uric acid and actively secretes excess uric acid into the urine [36].
BCRP protein expression is upregulated in the ducts and lobules of lactating breast tissue, and may thereby concentrate vitamins such as ribaflavin and minerals into milk [2,28], while simultaneously concentrating xenobiotics present in the systemic circulation into milk. It is also highly expressed in the placental syncytiotrophoblasts, protecting the fetus from harmful metabolites and compounds [28,37].
Based on studies in humans and mice, reports conflict as to whether women or men might have increased hepatic BCRP expression [6,38,39], though gender-related differences in the pharmacokinetics of the BCRP substrates topotecan [40] and doxorubin [41] suggest that men may have higher BCRP activity [38].
Compounds that Interact with BCRP
Many factors influence BCRP expression and activity. As mentioned previously, epigenetic factors such as methylation and histone deacetylation can affect BCRP expression as well as binding to hypoxia, estrogen, and progesterone response elements within the promoter [23]. As a result, estrogen and progesterone upregulate ABCG2 expression [42–44], while estradiol and the microRNAs hsa-miR-519c, hsa-miR520h, and hsa-miR328 downregulate ABCG2 expression [45–47]. Stressors and exogenous compounds can also affect expression via activation of AhR, CAR, PXR, GR, ER-β, PPAR-γand Nrf2 [17,48]. Finally, BCRP expression is mediated via the PI3K-Akt pathway and can be down-regulated using tyrosine kinase inhibitors [49,50].
Although the exact topology of BCRP’s binding sites is unknown, kinetic and equilibrium data suggest that there are two symmetric substrate binding sites and multiple allosteric sites allowing for non-competitive inhibition [3,51,52]. The list of compounds that interact with BCRP continues to grow (Table 1). BCRP inhibition is complex because any substrate for BCRP may also serve as a competitive inhibitor at the right concentration. However, since BCRP has multiple binding sites, a lack of competitive inhibition does not imply that the compound is not a BCRP substrate. Further studies are needed to elucidate the exact properties of these binding sites and the effects of competitive and allosteric inhibition. Some compounds currently listed as inhibitors may later be discovered also to be BCRP substrates. Fumitremorgin C and its more potent derivative Ko143, however, appear to inhibit BCRP transport through an allosteric mechanism and offer an effective way to inhibit BCRP transport no matter the substrate or binding pocket. In fact, Ko143 is one of the most potent yet least toxic BCRP inhibitors known to date [51,53–56].
Table 1.
Substrates | Inhibitors |
---|---|
| |
5-fluorouracil | atazanavir |
abacavir | chromanone derivatives |
atorvastatin | curcumin |
azidopine | cyclosporin A |
ciprofloxacin | delavirdine |
daunorubicin | fumitremorgin C (FTC) Ko143 |
doxorubicin | itraconazole |
efavirenz | ketoconazole |
epirubicin | lopinavir |
erlotinib | nelfinavir |
erythromycin | poloxamines |
etoposide | reserpine |
flavopiridol | ritonavir |
fluvastatin | saquinavir |
folic acid | sirolimus |
ganciclovir | tacrolimus |
gefitinib | |
glutathione | |
glyburide | |
homocamptothecins | |
imatinib | |
irinotecan | |
lamivudine | |
methotrexate | |
mitoxantrone | |
nilotinib | |
pantoprazole | |
pheophorbide A | |
PhIP | |
pitavastatin | |
porphyrins | |
pravastatin | |
riboflavin | |
rosuvastatin | |
SN-38 | |
sorafenib | |
sulfasalazine | |
sulfated estrogens | |
sunitinib | |
tamoxifen | |
tariquidar | |
topotecan | |
urate | |
vitamin K3 | |
zidovudine |
Many substrates that undergo efflux by BCRP also undergo efflux by P-gp and MRP1 [2]. In addition to compounds listed in Table 1, more comprehensive lists of BCRP substrates can be found in reviews [1,3,7,17,23,57].
Drug Interactions/Multi-drug Resistance
BCRP-mediated drug-drug interactions (DDIs) can severely affect the safety and efficacy of BCRP substrates, particularly those with a narrow therapeutic index. Because of this, the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) recommend pre-clinical testing to determine whether a new molecular entity is a BCRP substrate, and thus a potential victim of DDIs, or a BCRP inhibitor, and thus a perpetrator of DDIs. The FDA has published complete guidance on whether a compound should be tested in a clinical DDI study based on potency and ADME characteristics [58–60].
Safety is a consideration for BCRP substrates when dosed with an inhibitor because of increased oral bioavailability and decreased hepatic clearance. Plasma concentrations of BCRP substrates increase when dosed with a BCRP inhibitor, such as the interaction between topotecan and elacridar, and between pitavastatin and cyclosporin A [1,2,7]. Conversely, an increase in BCRP expression has been linked to lower plasma drug levels, although there is little and conflicting data on compounds that might induce BCRP expression [61]. Clinical DDI studies should be designed thoughtfully, because many BCRP substrates and inhibitors interact with other membrane transporters. For example, rosuvastatin is recommended as a probe for identifying BCRP inhibition, but rosuvastatin pharmacokinetics are also highly dependent on OATP1B1 activity [19,62]. BCRP activity can also be inhibited by natural compounds in the diet such as curcumin, so dietary restrictions should also be considered for DDI studies [19].
BCRP overexpression is prevalent in many cancers, especially stomach, colon, and esophageal cancer. It has been shown that cancer cells can induce BCRP expression through gene amplification and chromosomal translocation [63]. Overexpression is typically associated with a worse prognosis, as many chemotherapeutics are BCRP substrates. In clinical studies, patients with higher expression of BCRP responded more poorly to the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib, and to photosensitizer chemotherapeutics [64–67]. ABC inhibitors, antisense oligonucleotides, siRNA, transcriptional regulation, antibodies, and drug delivery systems have all been examined as potential therapies to resensitize these resistant cancer cells, although few have shown clinical promise. Many BCRP inhibitors caused serious neurotoxicity, perhaps due to off-target BCRP inhibition at the blood-brain barrier. Although some BCRP inhibitors are still being examined for their efficacy and safety, the field has moved away from developing chemotherapeutics that undergo efflux by ABC transporters [66,68].
ABCG2 variants
Many single nucleotide polymorphisms, resulting in both synonymous and nonsynonymous changes, have been identified in the ABCG2 gene [69–71]. Additionally, sequence diversity in the cis-regulatory regions has been reported to affect BCRP expression, and over 100 non-coding variants of ABCG2 have been associated with altered mRNA expression [30,72,73]. Both germline and somatic mutations have important implications for chemotherapeutic resistance. Table 2 summarizes the most common germline BCRP missense variants.
Table 2.
Variant rs number | cDNA (NM_004827.2) |
Protein coding effect (NP_004818.2) |
Protein function effect | Global Minor Allele Frequencya |
---|---|---|---|---|
rs2231137 | c. 34G>A | p. Val12Met | no function [107, 120, 121] | 0.1076 |
rs2231142 | c. 421C>A | p. Gln141Lys | reduced function [107, 120, 121] | 0.1180 |
rs34783571 | c. 1858G>A | p. Asp620Asn | reduced function [122] | 0.0056 |
rs138606116 | c. 1060G>A | p. Gly354Arg | unknown | 0.0026 |
rs45605536 | c. 1582G>A | p. Ala528Thr | unknown | 0.0014 |
rs192169063 | c. 1465T>C | p. Phe489Leu | reduced function [121] | 0.0009 |
rs148475733 | c. 211A>G | p. Met71Val | unknown | 0.0009 |
rs147547385 | c. 1787A>G | p. Asn596Ser | unknown | 0.0005 |
rs72552713 | c. 376C>T | p. Gln126Stop | no function [107, 123, 124] | 0.0005 |
rs199473672 | c. 335C>A | p. Pro112Gln | unknown | 0.0004 |
rs34264773 | c. 1768A>T | p. Asn590Tyr | reduced function [122] | 0.0003 |
rs140207606 | c. 706 C>A | p. Arg236Stop | no function* [123] | 0.0003 |
rs149106245 | c. 383A>T | p. Asp128Val | unknown | 0.0002 |
rs35965584 | c. 1624 A>G | p. Thr542Ala | unknown | 0.0002 |
rs547742211 | c. 1312T>C | p. Cys438Arg | unknown | 0.0002 |
From the ExAC server, accessed December 15, 2016 [125]
not validated in vitro
The two most common and extensively studied variants are rs2231137 (NM_004827.2: c.34G>A), which results in a V12M replacement (NP_004818.2: p.Val12Met), and rs2231142 (NM_004827.2:c.421C>A), which results in a Q141K substitution (NP_004818.2:p.Gln141Lys) [31,74]. The c.34G>A SNP (rs2231137) variant is located in exon 2 and reportedly does not affect expression, localization, or function of BCRP, or affect the pharmacokinetics of BCRP substrates [1,6,75]. However, in in vitro studies in transfected K562 cells, the c.34G>A variant was associated with reduced activity of the BCRP transporter, resulting in decreased resistance to tyrosine kinase inhibitors [76].
The c.421C>A (rs2231142) variant is located in exon 5 and is in the nucleotide binding domain of the transporter. It has been found to decrease the expression of BCRP due to increased degradation of the variant protein in the endoplasmic reticulum [1,6,7,38,77]. Expression of the BCRP protein was reported to be 30-40% that of protein with the reference sequence, and expression in the placenta, specifically, also was reportedly lower [74,75]. The c.421C>A variant has been found to affect pharmacokinetics of, response to, and toxicity of compounds that are BCRP substrates, including chemotherapeutics and endogenous compounds [70,78–81]. This variant was identified by the International Transporter Consortium as a clinically important transporter pharmacogene based on three criteria: 1) genome wide significance of an association between the variant and one or more drugs from genome wide association studies, 2) significant association of the variant and drug outcome from candidate gene studies, and 3) functional changes resulting from the polymorphism found in in vitro studies [4]. The in vivo intestinal BCRP transport activity in people homozygous for the A allele is reported to be approximately 23% of that in the c.421CC subjects [80]. Furthermore, the AUC of sulfasalazine, a drug used as a BCRP probe in vivo, is reportedly 2.5 times greater in patients carrying the A allele compared to patients without it [82]. A study in people of Japanese descent reported a 1.9 - 3.5 times greater AUC of sulfasalazine in patients with the c.421C>A variant, but this finding was not replicated in a study of people of Chinese descent [83]. Other studies found that patients of Japanese descent with the c.421C>A variant who were being treated with sunitinib for advanced renal cell carcinoma were more likely to develop grade 3 or grade 4 thrombocytopenia [81,84,85], and that the variant was significantly associated with increased sunitinib exposure [86]. Furthermore, patients homozygous for the reference allele may have lower plasma concentrations of atorvastatin, simvastatin, and fluvastatin [1,87,88] and may respond more poorly to FOLFOX/XELOX therapy [89] compared to patients homozygous for the A allele. Additionally, patients with the AA genotype at c.421 may have reduced response to allopurinol when treated for gout as compared to patients with the AC or CC genotypes [90,91], and a greater incidence of adverse events with gefitinib treatment was reported with the c.421C>A variant [2,38]. The A allele is found more commonly in East Asian populations (27-35% in Japanese and Chinese study participants) than in Caucasian populations (6-14%) and African American populations (1-4%) [4,38,75,92]. This difference in allele frequency at c.421 may be responsible for the different recommended initial dosing of rosuvastatin for patients of Asian descent (5 mg/day) compared to patients not of Asian descent (10-20 mg/day) [93]. Compared to Caucasian patients, Asian patients were found to have a 2-fold increase in drug exposure, resulting in increased risk for myopathy due to higher drug concentrations [93].
Besides what is known about c.34 and c.421, there are nonsense and missense variants that have been characterized in vitro but are rarely found in germline variation. The variants rs1061018 (NM_004827.2:c.623T>C; NP_004818.2:p.Phe208Ser), rs3116448 (NM_004827.2:c.742T>C; NP_004818.2:p.Ser248Pro), rs3201997 (NM_004827.2:c.1000G>T; NP_004818.2:p.Glu334Stop), rs773828630 (NM_004827.2:c.1321_1331delAGTGTTTCAGC; NP_004818.2:p.Ser441Argfs) and rs192169063 (NM_004827.2:c.1465T>C; NP_004818.2:p.Phe489Leu) are all reported to result in null BCRP protein activity based on the impact on the transport of methotrexate and haematoporphyrin [71]. Furthermore, the variants c.623T>C, and c.1321_1331delAGTGTTTCAGC were reported to have severely reduced protein levels, perhaps due to instability or degradation [71,75]. BCRP variants c.742T>C and c.1465T>C were all reported to have altered protein expression and to affect drug resistance [71]. In addition to what was found in K562 cells transfected with c.34G>A, in vitro studies in K562 cells showed reduced sensitivity to tyrosine kinase inhibitors in cells transfected with c.421C>A, c.623T>C, c.886G>C, or c.1574T>G [76].
Acquired somatic cell variants that have been implicated in chemotherapeutic resistance include protein sequence changes Arg482Gly and Arg482Thr, which are thought to increase BCRP activity [2,94,95]. Due to increased efflux activity, patients with these variants exhibit decreased response to the anthracyclines, doxorubicin, daunorubicin, and to rhodamine 123 [2,94,95]. The Arg482 site is likely involved in substrate recognition, as it is found in a large drug binding pocket at the cytoplasmic interface [1]. On the other hand, the variants Thr402Ala and Thr402Arg reportedly decrease BCRP activity, reducing mitoxantrone and the probes BODIPY-prazosin and Hoechst33342 efflux by 50-90% [1]. The Pro485Ala variant is also thought to decrease BCRP function, and has been found to reduce efflux of the probe BODIPY-prazosin by 70% [96]. However, this variant did not affect the efflux of mitoxantrone or Hoechst33342, suggesting that the variant lies in the drug binding pocket and thereby determines substrate specificity [1].
Fluorescent probes, including BODIPY-prazosin, Hoechst33342, and pheophorbide A, can be used to assess BCRP expression and function [1]. Probes specific for particular variants of the ABCG2 gene include Rhodamine 123 for R482G and Lyso-Tracker Green for R482T, as these probes are not substrates of the protein produced from the ABCG2 reference sequence [97].
Genetic Associations
Variants in ABCG2 have been associated with a number of phenotypes via genome-wide association studies (GWAS), both as a risk for developing disease and altered drug response. One of the most well-characterized associations is that of the reduced-function BCRP Q141K variant and the risk for developing gout and hyperuricemia [36,98–100]. Since BCRP is a high-capacity urate transporter, reduced-function variants are linked to a decrease in urate excretion and higher uric acid levels. This in turn increases the risk for developing gout. Since the time BCRP Q141K was linked to urate excretion via GWAS, other reduced-function variants have also been linked to uric acid levels, such as Q126X [100,101].
The Clinical Pharmacogenetics Implementation Consortium (CPIC) evaluates the strength of evidence for gene-drug associations and publishes peer-reviewed guidelines for changing drug dose or drug choice based on the genotype of a patient. While most CPIC dosing guidelines have focused on genetic variation in enzymes causing an increase in drug toxicity [102–104], two large GWAS have linked ABCG2 c.421 variants with altered drug efficacy. The JUPITER trial in over 4,000 patients with cardiovascular disease demonstrated a strong association (at genome-wide level significance) of the C genotype with improved LDL cholesterol response to rosuvastatin [105]. These results were further studied in a cohort of 291 Chinese patients to reveal that this altered efficacy was due to an increased bioavailability in these patients [106]. In a recent GWAS in over 1500 multi-ethnic gout patients, the C genotype was associated at genome-wide level significance with worse uric-acid lowering response to allopurinol [90]. One follow up study in 264 patients confirmed this association, even when controlling for adherence to allopurinol, a prevalent factor of non-response in gout treatment [91].
Conclusion
ABCG2, encoding the efflux pump BCRP, plays an important role in drug response and disposition. It is widely expressed and has a broad substrate specificity to efflux xenobiotics and harmful metabolites from systemic circulation. BCRP is also an important mediator of multi-drug resistance in cancers and protection of vital yet vulnerable tissues such as the brain, stem cells, and placenta.
The regulation of ABCG2 is complex. To date, studies have shown that BCRP expression and function can be controlled via genetic response elements, post-translational modifications, epigenetics, DDIs, and genetic variation. Variation in BCRP activity affects the pharmacokinetic profiles of many drugs, and has been associated with risks of toxicity and efficacy of therapy. One of the most common reduced-function variants, BCRP p.Q141K, has been implicated in improved response to statins, increased side effects of chemotherapeutics, altered response to allopurinol, and reduced excretion of endogenous substrates such as uric acid. This variant reduces the expression of BCRP on the plasma membrane due to an increased breakdown in the endoplasmic reticulum. In vitro site-directed mutagenesis studies have helped identify other residues important for BCRP transport, trafficking, and expression, but identification of a BCRP crystal structure is needed to definitively describe the multiple binding pockets and allosteric sites of the efflux protein.
Acknowledgments
Sources of funding: NIH/NIGMS R24 GM61374
Footnotes
Disclaimers: none
Conflicts of Interest: RBA is a stockholder in Personalis Inc. and a paid advisor for Pfizer and Karius. TEK is a paid scientific advisor to Rxight Pharmacogenetics. Remaining authors have no conflicts of interest.
References
- 1.Mao Q, Unadkat JD. Role of the breast cancer resistance protein (BCRP/ABCG2) in drug transport–an update. AAPS J. 2015;17(1):65–82. doi: 10.1208/s12248-014-9668-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Stacy AE, Jansson PJ, Richardson DR. Molecular pharmacology of ABCG2 and its role in chemoresistance. Mol Pharmacol. 2013;84(5):655–669. doi: 10.1124/mol.113.088609. [DOI] [PubMed] [Google Scholar]
- 3.Ni Z, Bikadi Z, Rosenberg MF, Mao Q. Structure and function of the human breast cancer resistance protein (BCRP/ABCG2) Current Drug Metabolism. 2010;11(7) doi: 10.2174/138920010792927325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.International Transoporter Consortium. Giacomini KM, Balimane PV, Cho SK, Eadon M, Edeki T, et al. International Transporter Consortium commentary on clinically important transporter polymorphisms. Clin Pharmacol Ther. 2013;94(1):23–26. doi: 10.1038/clpt.2013.12. [DOI] [PubMed] [Google Scholar]
- 5.Bailey-Dell K, Hassel B, Doyle L, Ross D. Promoter characterization and genomic organization of the human breast cancer resistance protein (ATP-binding cassette transporter G2) gene. Biochim Biophys Acta. 2001;1520(3):234–241. doi: 10.1016/s0167-4781(01)00270-6. [DOI] [PubMed] [Google Scholar]
- 6.Hu M, To KK, Mak VW, Tomlinson B. The ABCG2 transporter and its relations with the pharmacokinetics, drug interaction and lipid-lowering effects of statins. Expert Opin Drug Metab Toxicol. 2011;7(1):49–62. doi: 10.1517/17425255.2011.538383. [DOI] [PubMed] [Google Scholar]
- 7.Nakanishi T, Ross DD. Breast cancer resistance protein (BCRP/ABCG2): its role in multidrug resistance and regulation of its gene expression. Chinese Journal of Cancer. 2012;31(2):73–99. doi: 10.5732/cjc.011.10320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Campbell PK, Zong Y, Yang S, Zhou S, Rubnitz JE, Sorrentino BP. Identification of a novel, tissue-specific ABCG2 promoter expressed in pediatric acute megakaryoblastic leukemia. Leuk Res. 2011;35(10):1321–1329. doi: 10.1016/j.leukres.2011.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.To KK, Polgar O, Huff LM, Morisaki K, Bates SE. Histone modifications at the ABCG2 promoter following treatment with histone deacetylase inhibitor mirror those in multidrug-resistant cells. Mol Cancer Res. 2008;6(1):151–164. doi: 10.1158/1541-7786.MCR-07-0175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Turner JG, Gump JL, Zhang C, Cook JM, Marchion D, Hazlehurst L, et al. ABCG2 expression, function, and promoter methylation in human multiple myeloma. Blood. 2006;108(12):3881–3889. doi: 10.1182/blood-2005-10-009084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.To KK, Zhan Z, Bates SE. Aberrant promoter methylation of the ABCG2 gene in renal carcinoma. Mol Cell Biol. 2006;26(22):8572–8585. doi: 10.1128/MCB.00650-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Polgar O, Robey RW, Bates SE. ABCG2: structure, function and role in drug response. Expert Opin Drug Metab Toxicol. 2008;4(1):1–15. doi: 10.1517/17425255.4.1.1. [DOI] [PubMed] [Google Scholar]
- 13.McDevitt CA, Collins R, Kerr ID, Callaghan R. Purification and structural analyses of ABCG2. Adv Drug Deliv Rev. 2009;61(1):57–65. doi: 10.1016/j.addr.2008.07.004. [DOI] [PubMed] [Google Scholar]
- 14.Volk EL, Schneider E. Wild-type breast cancer resistance protein (BCRP/ABCG2) is a methotrexate polyglutamate transporter. Cancer research. 2003;63(17):5538–5543. [PubMed] [Google Scholar]
- 15.Hazai E, Bikadi Z. Homology modeling of breast cancer resistance protein (ABCG2) J Struct Biol. 2008;162(1):63–74. doi: 10.1016/j.jsb.2007.12.001. [DOI] [PubMed] [Google Scholar]
- 16.Rosenberg MF, Bikadi Z, Hazai E, Starborg T, Kelley L, Chayen NE, et al. Three-dimensional structure of the human breast cancer resistance protein (BCRP/ABCG2) in an inward-facing conformation. Acta Crystallogr D Biol Crystallogr. 2015;71(Pt 8):1725–1735. doi: 10.1107/S1399004715010676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mo W, Zhang JT. Human ABCG2: structure, function, and its role in multidrug resistance. International Journal of Biochemistry and Molecular Biology. 2012;3(1):1–27. [PMC free article] [PubMed] [Google Scholar]
- 18.Kage K, Tsukahara S, Sugiyama T, Asada S, Ishikawa E, Tsuruo T, et al. Dominant-negative inhibition of breast cancer resistance protein as drug efflux pump through the inhibition of S-S dependent homodimerization. Int J Cancer. 2002;97(5):626–630. doi: 10.1002/ijc.10100. [DOI] [PubMed] [Google Scholar]
- 19.Lee CA, O’Connor MA, Ritchie TK, Galetin A, Cook JA, Ragueneau-Majlessi I, et al. Breast cancer resistance protein (ABCG2) in clinical pharmacokinetics and drug interactions: practical recommendations for clinical victim and perpetrator drug-drug interaction study design. Drug Metab Dispos. 2015;43(4):490–509. doi: 10.1124/dmd.114.062174. [DOI] [PubMed] [Google Scholar]
- 20.Graf GA, Yu L, Li WP, Gerard R, Tuma PL, Cohen JC, et al. ABCG5 and ABCG8 are obligate heterodimers for protein trafficking and biliary cholesterol excretion. J Biol Chem. 2003;278(48):48275–48282. doi: 10.1074/jbc.M310223200. [DOI] [PubMed] [Google Scholar]
- 21.Xie Y, Xu K, Linn DE, Yang X, Guo Z, Shimelis H, et al. The 44-kDa Pim-1 kinase phosphorylates BCRP/ABCG2 and thereby promotes its multimerization and drug-resistant activity in human prostate cancer cells. J Biol Chem. 2008;283(6):3349–3356. doi: 10.1074/jbc.M707773200. [DOI] [PubMed] [Google Scholar]
- 22.Diop NK, Hrycyna CA. 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. doi: 10.1021/bi0479858. [DOI] [PubMed] [Google Scholar]
- 23.Robey RW, To KK, Polgar O, Dohse M, Fetsch P, Dean M, et al. ABCG2: a perspective. Adv Drug Deliv Rev. 2009;61(1):3–13. doi: 10.1016/j.addr.2008.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Annilo T, Chen ZQ, Shulenin S, Costantino J, Thomas L, Lou H, et al. Evolution of the vertebrate ABC gene family: analysis of gene birth and death. Genomics. 2006;88(1):1–11. doi: 10.1016/j.ygeno.2006.03.001. [DOI] [PubMed] [Google Scholar]
- 25.Mao Q, Unadkat JD. Role of the Breast Cancer Resistance Protein (ABCG2) in Drug Transport. The AAPS Journal. 2005;7(1) doi: 10.1208/aapsj070112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Doyle L, Ross DD. Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2) Oncogene. 2003;22(47):7340–7358. doi: 10.1038/sj.onc.1206938. [DOI] [PubMed] [Google Scholar]
- 27.Aust S, Obrist P, Jaeger W, Klimpfinger M, Tucek G, Wrba F, et al. Subcellular localization of the ABCG2 transporter in normal and malignant human gallbladder epithelium. Lab Invest. 2004;84(8):1024–1036. doi: 10.1038/labinvest.3700127. [DOI] [PubMed] [Google Scholar]
- 28.Vander Borght S, Libbrecht L, Katoonizadeh A, van Pelt J, Cassiman D, Nevens F, et al. Breast cancer resistance protein (BCRP/ABCG2) is expressed by progenitor cells/reactive ductules and hepatocytes and its expression pattern is influenced by disease etiology and species type: possible functional consequences. J Histochem Cytochem. 2006;54(9):1051–1059. doi: 10.1369/jhc.5A6912.2006. [DOI] [PubMed] [Google Scholar]
- 29.Han B, Zhang JT. Multidrug resistance in cancer chemotherapy and xenobiotic protection mediated by the half ATP-binding cassette transporter ABCG2. Current medicinal chemistry Anti-cancer agents. 2004;4(1):31–42. doi: 10.2174/1568011043482205. [DOI] [PubMed] [Google Scholar]
- 30.Broad Institute. GTEx Portal. Available at: http://www.gtexportal.org/home/ [Accessed Accessed December 15 2016]
- 31.Krishnamurthy P, Schuetz JD. The ABC transporter Abcg2/Bcrp: role in hypoxia mediated survival. Biometals. 2005;18(4):349–358. doi: 10.1007/s10534-005-3709-7. [DOI] [PubMed] [Google Scholar]
- 32.Pal A, Mehn D, Molnar E, Gedey S, Meszaros P, Nagy T, et al. Cholesterol potentiates ABCG2 activity in a heterologous expression system: improved in vitro model to study function of human ABCG2. J Pharmacol Exp Ther. 2007;321(3):1085–1094. doi: 10.1124/jpet.106.119289. [DOI] [PubMed] [Google Scholar]
- 33.To KK, Hu M, Tomlinson B. Expression and activity of ABCG2, but not ABCB1 or OATP1B1, are associated with cholesterol levels: evidence from in vitro and in vivo experiments. Pharmacogenomics. 2014;15(8):1091–1104. doi: 10.2217/pgs.14.58. [DOI] [PubMed] [Google Scholar]
- 34.Huls M, Russel FG, Masereeuw R. The role of ATP binding cassette transporters in tissue defense and organ regeneration. J Pharmacol Exp Ther. 2009;328(1):3–9. doi: 10.1124/jpet.107.132225. [DOI] [PubMed] [Google Scholar]
- 35.Gutmann H, Hruz P, Zimmermann C, Beglinger C, Drewe J. Distribution of breast cancer resistance protein (BCRP/ABCG2) mRNA expression along the human GI tract. Biochem Pharmacol. 2005;70(5):695–699. doi: 10.1016/j.bcp.2005.05.031. [DOI] [PubMed] [Google Scholar]
- 36.Woodward OM. ABCG2: the molecular mechanisms of urate secretion and gout. Am J Physiol Renal Physiol. 2015;309(6):F485–488. doi: 10.1152/ajprenal.00242.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Maliepaard M, Scheffer GL, Faneyte IF, van Gastelen MA, Pijnenborg AC, Schinkel AH, et al. Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer research. 2001;61(8):3458–3464. [PubMed] [Google Scholar]
- 38.Gradhand U, Kim RB. Pharmacogenomics of MRP transporters (ABCC1-5) and BCRP (ABCG2) Drug metabolism reviews. 2008;40(2):317–354. doi: 10.1080/03602530801952617. [DOI] [PubMed] [Google Scholar]
- 39.Merino G, van Herwaarden AE, Wagenaar E, Jonker JW, Schinkel AH. Sex-dependent expression and activity of the ATP-binding cassette transporter breast cancer resistance protein (BCRP/ABCG2) in liver. Mol Pharmacol. 2005;67(5):1765–1771. doi: 10.1124/mol.105.011080. [DOI] [PubMed] [Google Scholar]
- 40.Loos WJ, Gelderblom H, Verweij J, van Boven-van Zomeren DM, Nooter K, Stoter G, et al. Red blood cells: a neglected compartment in topotecan pharmacokinetic analysis. Anticancer Drugs. 2003;14(3):227–232. doi: 10.1097/00001813-200303000-00006. [DOI] [PubMed] [Google Scholar]
- 41.Dobbs NA, Twelves CJ, Gillies H, James CA, Harper PG, Rubens RD. Gender affects doxorubicin pharmacokinetics in patients with normal liver biochemistry. Cancer Chemother Pharmacol. 1995;36:473–476. doi: 10.1007/BF00685796. [DOI] [PubMed] [Google Scholar]
- 42.Ee PL, Kamalakaran S, Tonetti D, He X, Ross DD, Beck WT. Identification of a novel estrogen response element in the breast cancer resistance protein (ABCG2) gene. Cancer research. 2004;64(4):1247–1251. doi: 10.1158/0008-5472.can-03-3583. [DOI] [PubMed] [Google Scholar]
- 43.Wang H, Lee EW, Zhou L, Leung PC, Ross DD, Unadkat JD, et al. Progesterone receptor (PR) isoforms PRA and PRB differentially regulate expression of the breast cancer resistance protein in human placental choriocarcinoma BeWo cells. Mol Pharmacol. 2008;73(3):845–854. doi: 10.1124/mol.107.041087. [DOI] [PubMed] [Google Scholar]
- 44.Pradhan M, Bembinster LA, Baumgarten SC, Frasor J. Proinflammatory cytokines enhance estrogen-dependent expression of the multidrug transporter gene ABCG2 through estrogen receptor and NF{kappa}B cooperativity at adjacent response elements. J Biol Chem. 2010;285(41):31100–31106. doi: 10.1074/jbc.M110.155309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.To KK, Robey RW, Knutsen T, Zhan Z, Ried T, Bates SE. Escape from hsa-miR-519c enables drug-resistant cells to maintain high expression of ABCG2. Mol Cancer Ther. 2009;8(10):2959–2968. doi: 10.1158/1535-7163.MCT-09-0292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wang F, Xue X, Wei J, An Y, Yao J, Cai H, et al. hsa-miR-520h downregulates ABCG2 in pancreatic cancer cells to inhibit migration, invasion, and side populations. Br J Cancer. 2010;103(4):567–574. doi: 10.1038/sj.bjc.6605724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pan YZ, Morris ME, Yu AM. MicroRNA-328 negatively regulates the expression of breast cancer resistance protein (BCRP/ABCG2) in human cancer cells. Mol Pharmacol. 2009;75(6):1374–1379. doi: 10.1124/mol.108.054163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Miller DS. ABC transporter regulation by signaling at the blood-brain barrier: relevance to pharmacology. Adv Pharmacol. 2014;71:1–24. doi: 10.1016/bs.apha.2014.06.008. [DOI] [PubMed] [Google Scholar]
- 49.Pick A, Wiese M. Tyrosine kinase inhibitors influence ABCG2 expression in EGFR-positive MDCK BCRP cells via the PI3K/Akt signaling pathway. ChemMedChem. 2012;7(4):650–662. doi: 10.1002/cmdc.201100543. [DOI] [PubMed] [Google Scholar]
- 50.Nakanishi T. Complex interaction of BCRP/ABCG2 and imatinib in BCR-ABL-expressing cells: BCRP-mediated resistance to imatinib is attenuated by imatinib-induced reduction of BCRP expression. Blood. 2006;108(2):678–684. doi: 10.1182/blood-2005-10-4020. [DOI] [PubMed] [Google Scholar]
- 51.Pozza A, Perez-Victoria JM, Sardo A, Ahmed-Belkacem A, Di Pietro A. Purification of breast cancer resistance protein ABCG2 and role of arginine-482. Cell Mol Life Sci. 2006;63(16):1912–1922. doi: 10.1007/s00018-006-6159-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Clark R, Kerr ID, Callaghan R. Multiple drugbinding sites on the R482G isoform of the ABCG2 transporter. Br J Pharmacol. 2006;149(5):506–515. doi: 10.1038/sj.bjp.0706904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Rabindran SK, He H, Singh M, Brown E, Collins KI, Annable T, et al. Reversal of a novel multidrug resistance mechanism in human colon carcinoma cells by fumitremorgin C. Cancer research. 1998;58 [PubMed] [Google Scholar]
- 54.Valdameri G, Genoux-Bastide E, Peres B, Gauthier C, Guitton J, Terreux R, et al. Substituted chromones as highly potent nontoxic inhibitors, specific for the breast cancer resistance protein. J Med Chem. 2012;55(2):966–970. doi: 10.1021/jm201404w. [DOI] [PubMed] [Google Scholar]
- 55.Nakanishi T, Doyle LA, Hassel B, Wei Y, Bauer KS, Wu S, et al. Functional characterization of human breast cancer resistance protein (BCRP, ABCG2) expressed in the oocytes of Xenopus laevis. Mol Pharmacol. 2003;64(6):1452–1462. doi: 10.1124/mol.64.6.1452. [DOI] [PubMed] [Google Scholar]
- 56.Giri N, Agarwal S, Shaik N, Pan G, Chen Y, Elmquist WF. Substrate-dependent breast cancer resistance protein (Bcrp1/Abcg2)-mediated interactions: consideration of multiple binding sites in in vitro assay design. Drug Metab Dispos. 2009;37(3):560–570. doi: 10.1124/dmd.108.022046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sarkadi B, Homolya L, Szakacs G, Varadi A. Human multidrug resistance ABCB and ABCG transporters: participation in a chemoimmunity defense system. Physiol Rev. 2006;86(4):1179–1236. doi: 10.1152/physrev.00037.2005. [DOI] [PubMed] [Google Scholar]
- 58.International Transporter Consortium. Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–236. doi: 10.1038/nrd3028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Guidance for Industry: Drug Interaction Studies - Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations. United States Department of Health and Human Services Food and Drug Administration, Center for Drug Evaluation and Research editors. Clinical Pharmacology. 2012 [Google Scholar]
- 60.Prueksaritanont T, Chu X, Gibson C, Cui D, Yee KL, Ballard J, et al. Drug-drug interaction studies: regulatory guidance and an industry perspective. Aaps j. 2013;15(3):629–645. doi: 10.1208/s12248-013-9470-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Konig J, Muller F, Fromm MF. Transporters and drug-drug interactions: important determinants of drug disposition and effects. Pharmacol Rev. 2013;65(3):944–966. doi: 10.1124/pr.113.007518. [DOI] [PubMed] [Google Scholar]
- 62.Kitamura S, Maeda K, Wang Y, Sugiyama Y. Involvement of multiple transporters in the hepatobiliary transport of rosuvastatin. Drug Metab Dispos. 2008;36(10):2014–2023. doi: 10.1124/dmd.108.021410. [DOI] [PubMed] [Google Scholar]
- 63.Calcagno AM, Ambudkar SV. Molecular mechanisms of drug resistance in single-step and multi-step drug-selected cancer cells. Methods in molecular biology (Clifton, NJ) 2010;596:77–93. doi: 10.1007/978-1-60761-416-6_5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Dohse M, Scharenberg C, Shukla S, Robey RW, Volkmann T, Deeken JF, et al. Comparison of ATP-binding cassette transporter interactions with the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib. Drug Metab Dispos. 2010;38(8):1371–1380. doi: 10.1124/dmd.109.031302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Jiang X, Zhao Y, Smith C, Gasparetto M, Turhan A, Eaves A, et al. Chronic myeloid leukemia stem cells possess multiple unique features of resistance to BCR-ABL targeted therapies. Leukemia. 2007;21(5):926–935. doi: 10.1038/sj.leu.2404609. [DOI] [PubMed] [Google Scholar]
- 66.Wu CP, Calcagno AM, Ambudkar SV. Reversal of ABC drug transporter-mediated multidrug resistance in cancer cells: evaluation of current strategies. Current molecular pharmacology. 2008;1(2):93–105. doi: 10.2174/1874467210801020093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yamada M, Wilber JF. Reciprocal regulation of preprothyrotropin-releasing hormone (TRH) mRNA in the rat anterior hypothalamus by thyroid hormone: dissociation from TRH concentrations during hypothyroidism. Neuropeptides. 1990;15(1):49–53. doi: 10.1016/0143-4179(90)90160-z. [DOI] [PubMed] [Google Scholar]
- 68.Westover D, Li F. New trends for overcoming ABCG2/BCRP-mediated resistance to cancer therapies. Journal of experimental & clinical cancer research : CR. 2015;34:159. doi: 10.1186/s13046-015-0275-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Sharom F. ABC multidrug transporters: structure, function and role in chemoresistance. Pharmacogenomics. 2008;9(1):105–127. doi: 10.2217/14622416.9.1.105. [DOI] [PubMed] [Google Scholar]
- 70.Rabindran SK, Ross DD, Doyle LA, Yang W, Greenberger LM. Fumitremorgin C reverses multidrug resistance in cells transfected with the breast cancer resistance protein. Cancer research. 2000;60(1):47–50. [PubMed] [Google Scholar]
- 71.Nakagawa H, Tamura A, Wakabayashi K, Hoshijima K, Komada M, Yoshida T, et al. Ubiquitin-mediated proteasomal degradation of non-synonymous SNP variants of human ABC transporter ABCG2. Biochem J. 2008;411(3):623–631. doi: 10.1042/BJ20071229. [DOI] [PubMed] [Google Scholar]
- 72.Cusatis G, Sparreboom A. Pharmacogenomic Importance of ABCG2. Pharmacogenomics. 2008;9(8):1005–1009. doi: 10.2217/14622416.9.8.1005. [DOI] [PubMed] [Google Scholar]
- 73.GTex Consortium. The Genotype-Tissue Expression (GTEx) project. Nat Genet. 2013;45(6):580–585. doi: 10.1038/ng.2653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kobayashi D, Ieiri I, Hirota T, Takane H, Maegawa S, Kigawa J, et al. Functional assessment of ABCG2 (BCRP) gene polymorphisms to protein expression in human placenta. Drug Metab Dispos. 2005;33(1):94–101. doi: 10.1124/dmd.104.001628. [DOI] [PubMed] [Google Scholar]
- 75.Kondo C, Suzuki H, Itoda M, Ozawa S, Sawada J-I, Kobayashi D, et al. Functional Analysis of SNPs Variants of BCRP/ABCG2. Pharmaceutical Research. 2004;21(10) doi: 10.1023/b:pham.0000045245.21637.d4. [DOI] [PubMed] [Google Scholar]
- 76.Skoglund K, Boiso Moreno S, Jonsson JI, Vikingsson S, Carlsson B, Green H. Single-nucleotide polymorphisms of ABCG2 increase the efficacy of tyrosine kinase inhibitors in the K562 chronic myeloid leukemia cell line. Pharmacogenet Genomics. 2014;24(1):52–61. doi: 10.1097/FPC.0000000000000022. [DOI] [PubMed] [Google Scholar]
- 77.Furukawa T, Wakabayashi K, Tamura A, Nakagawa H, Morishima Y, Osawa Y, et al. Major SNP (Q141K) variant of human ABC transporter ABCG2 undergoes lysosomal and proteasomal degradations. Pharm Res. 2009;26(2):469–479. doi: 10.1007/s11095-008-9752-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Nakatomi K, Yoshikawa M, Oka M, Ikegami Y, Hayasaka S, Sano K, et al. Transport of 7-ethyl-10-hydroxycamptothecin (SN-38) by breast cancer resistance protein ABCG2 in human lung cancer cells. Biochemical and biophysical research communications. 2001;288(4):827–832. doi: 10.1006/bbrc.2001.5850. [DOI] [PubMed] [Google Scholar]
- 79.Maliepaard M, van Gastelen MA, Tohgo A, Hausheer FH, van Waardenburg RCAM, de Jong LA, et al. Circumvention of Breast Cancer Resistance Protein (BCRP)- mediated Resistance to Camptothecins in Vitro Using Non- Substrate Drugs or the BCRP Inhibitor GF120918. Clinical Cancer Research. 2001;7:935–941. [PubMed] [Google Scholar]
- 80.Tanaka Y, Kitamura Y, Maeda K, Sugiyama Y. Quantitative Analysis of the ABCG2 c.421C>A Polymorphism Effect on In Vivo Transport Activity of Breast Cancer Resistance Protein (BCRP) Using an Intestinal Absorption Model. J Pharm Sci. 2015;104(9):3039–3048. doi: 10.1002/jps.24366. [DOI] [PubMed] [Google Scholar]
- 81.Low SK, Fukunaga K, Takahashi A, Matsuda K, Hongo F, Nakanishi H, et al. Association Study of a Functional Variant on ABCG2 Gene with Sunitinib-Induced Severe Adverse Drug Reaction. PLoS One. 2016;11(2):e0148177. doi: 10.1371/journal.pone.0148177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Yamasaki Y, Ieiri I, Kusuhara H, Sasaki T, Kimura M, Tabuchi H, et al. Pharmacogenetic characterization of sulfasalazine disposition based on NAT2 and ABCG2 (BCRP) gene polymorphisms in humans. Clin Pharmacol Ther. 2008;84(1):95–103. doi: 10.1038/sj.clpt.6100459. [DOI] [PubMed] [Google Scholar]
- 83.Adkison KK, Vaidya SS, Lee DY, Koo SH, Li L, Mehta AA, et al. Oral sulfasalazine as a clinical BCRP probe substrate: pharmacokinetic effects of genetic variation (C421A) and pantoprazole coadministration. J Pharm Sci. 2010;99(2):1046–1062. doi: 10.1002/jps.21860. [DOI] [PubMed] [Google Scholar]
- 84.Miura Y, Imamura CK, Fukunaga K, Katsuyama Y, Suyama K, Okaneya T, et al. Sunitinib-induced severe toxicities in a Japanese patient with the ABCG2 421 AA genotype. BMC Cancer. 2014;14:964. doi: 10.1186/1471-2407-14-964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kato R, Kato Y, Matsuura T, Kanehira M, Takata R, Obara W. Characteristics of early-onset hematotoxicity of sunitinib in Japanese patients with renal cell carcinoma. BMC Cancer. 2017;17(1):214. doi: 10.1186/s12885-017-3205-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Narjoz C, Cessot A, Thomas-Schoemann A, Golmard JL, Huillard O, Boudou-Rouquette P, et al. Role of the lean body mass and of pharmacogenetic variants on the pharmacokinetics and pharmacodynamics of sunitinib in cancer patients. Invest New Drugs. 2015;33(1):257–268. doi: 10.1007/s10637-014-0178-2. [DOI] [PubMed] [Google Scholar]
- 87.Keskitalo JE, Zolk O, Fromm MF, Kurkinen KJ, Neuvonen PJ, Niemi M. ABCG2 polymorphism markedly affects the pharmacokinetics of atorvastatin and rosuvastatin. Clin Pharmacol Ther. 2009;86(2):197–203. doi: 10.1038/clpt.2009.79. [DOI] [PubMed] [Google Scholar]
- 88.Keskitalo JE, Pasanen MK, Neuvonen PJ, Niemi M. Different effects of the ABCG2 c.421C>A SNP on the pharmacokinetics of fluvastatin, pravastatin and simvastatin. Pharmacogenomics. 2009;10(10):1617–1624. doi: 10.2217/pgs.09.85. [DOI] [PubMed] [Google Scholar]
- 89.Zhao J, Li W, Zhu D, Yu Q, Zhang Z, Sun M, et al. Association of single nucleotide polymorphisms in MTHFR and ABCG2 with the different efficacy of first-line chemotherapy in metastatic colorectal cancer. Medical oncology (Northwood, London, England) 2014;31(1):802. doi: 10.1007/s12032-013-0802-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Wen CC, Yee SW, Liang X, Hoffmann TJ, Kvale MN, Banda Y, et al. Genome-wide association study identifies ABCG2 (BCRP) as an allopurinol transporter and a determinant of drug response. Clin Pharmacol Ther. 2015;97(5):518–525. doi: 10.1002/cpt.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Roberts RL, Wallace MC, Phipps-Green AJ, Topless R, Drake JM, Tan P, et al. ABCG2 loss-of-function polymorphism predicts poor response to allopurinol in patients with gout. The Pharmacogenomics Journal. 2016 doi: 10.1038/tpj.2015.101. [DOI] [PubMed] [Google Scholar]
- 92.Ono C, Kikkawa H, Suzuki A, Suzuki M, Yamamoto Y, Ichikawa K, et al. Clinical impact of genetic variants of drug transporters in different ethnic groups within and across regions. Pharmacogenomics. 2013;14(14):1745–1764. doi: 10.2217/pgs.13.171. [DOI] [PubMed] [Google Scholar]
- 93.AstraZeneca Pharmaceuticals LP. Crestor [Package Insert] Wilmington, DE: 2016. [Google Scholar]
- 94.Honjo Y, Hrycyna CA, Yan QW, Medina-Perez WY, Robey RW, van de Laar A, et al. Acquired mutations in the MXR/BCRP/ABCP gene alter substrate specificity in MXR/BCRP/ABCP-overexpressing cells. Cancer research. 2001;61(18):6635–6639. [PubMed] [Google Scholar]
- 95.Allen JD, Jackson SC, Schinkel AH. A Mutation Hot Spot in the Bcrp1 (Abcg2) Multidrug Transporter in Mouse Cell Lines Selected for Doxorubicin Resistance. Cancer research. 2002;62:2294–2299. [PubMed] [Google Scholar]
- 96.Ni Z, Bikadi Z, Shuster DL, Zhao C, Rosenberg MF, Mao Q. Identification of proline residues in or near the transmembrane helices of the human breast cancer resistance protein (BCRP/ABCG2) that are important for transport activity and substrate specificity. Biochemistry. 2011;50(37):8057–8066. doi: 10.1021/bi200573t. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Robey RW, Honjo Y, van de Laar A, Miyake K, Regis JT, Litman T, et al. A functional assay for detection of the mitoxantrone resistance protein, MXR (ABCG2) Biochimica et Biophysica Acta. 2001;1512:171–182. doi: 10.1016/s0005-2736(01)00308-x. [DOI] [PubMed] [Google Scholar]
- 98.Matsuo H, Yamamoto K, Nakaoka H, Nakayama A, Sakiyama M, Chiba T, et al. Genome-wide association study of clinically defined gout identifies multiple risk loci and its association with clinical subtypes. Ann Rheum Dis. 2016;75(4):652–659. doi: 10.1136/annrheumdis-2014-206191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kannangara DR, Phipps-Green AJ, Dalbeth N, Stamp LK, Williams KM, Graham GG, et al. Hyperuricaemia: contributions of urate transporter ABCG2 and the fractional renal clearance of urate. Ann Rheum Dis. 2016;75(7):1363–1366. doi: 10.1136/annrheumdis-2015-208111. [DOI] [PubMed] [Google Scholar]
- 100.Matsuo H, Takada T, Ichida K, Nakamura T, Nakayama A, Suzuki H, et al. ABCG2/BCRP dysfunction as a major cause of gout. Nucleosides, nucleotides & nucleic acids. 2011;30(12):1117–1128. doi: 10.1080/15257770.2011.633954. [DOI] [PubMed] [Google Scholar]
- 101.Li R, Miao L, Qin L, Xiang Y, Zhang X, Peng H, et al. A meta-analysis of the associations between the Q141K and Q126X ABCG2 gene variants and gout risk. International journal of clinical and experimental pathology. 2015;8(9):9812–9823. [PMC free article] [PubMed] [Google Scholar]
- 102.Moriyama B, Obeng AO, Barbarino J, Penzak SR, Henning SA, Scott SA, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC(R)) Guideline for CYP2C19 and Voriconazole Therapy. Clin Pharmacol Ther. 2016 doi: 10.1002/cpt.583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Relling MV, Gardner EE, Sandborn WJ, Schmiegelow K, Pui CH, Yee SW, et al. Clinical Pharmacogenetics Implementation Consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing. Clin Pharmacol Ther. 2011;89(3):387–391. doi: 10.1038/clpt.2010.320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Bell GC, Caudle KE, Whirl-Carrillo M, Gordon RJ, Hikino K, Prows CA, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6 Genotype and Use of Ondansetron and Tropisetron. Clin Pharmacol Ther. 2016 doi: 10.1002/cpt.598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Chasman DI, Giulianini F, MacFadyen J, Barratt BJ, Nyberg F, Ridker PM. Genetic determinants of statin-induced low-density lipoprotein cholesterol reduction: the Justification for the Use of Statins in Prevention: an Intervention Trial Evaluating Rosuvastatin (JUPITER) trial. Circ Cardiovasc Genet. 2012;5(2):257–264. doi: 10.1161/CIRCGENETICS.111.961144. [DOI] [PubMed] [Google Scholar]
- 106.Lee HK, Hu M, Lui S, Ho CS, Wong CK, Tomlinson B. Effects of polymorphisms in ABCG2, SLCO1B1, SLC10A1 and CYP2C9/19 on plasma concentrations of rosuvastatin and lipid response in Chinese patients. Pharmacogenomics. 2013;14(11):1283–1294. doi: 10.2217/pgs.13.115. [DOI] [PubMed] [Google Scholar]
- 107.Yanase K, Tsukahara S, Mitsuhashi J, Sugimoto Y. Functional SNPs of the breast cancer resistance protein-therapeutic effects and inhibitor development. Cancer letters. 2006;234(1):73–80. doi: 10.1016/j.canlet.2005.04.039. [DOI] [PubMed] [Google Scholar]
- 108.Natarajan K, Xie Y, Baer MR, Ross DD. Role of breast cancer resistance protein (BCRP/ABCG2) in cancer drug resistance. Biochem Pharmacol. 2012;83(8):1084–1103. doi: 10.1016/j.bcp.2012.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Wang L, Leggas M, Empey PE, McNamara PJ. Stereoselective interaction of pantoprazole with ABCG2. II. In vitro flux analysis. Drug Metab Dispos. 2012;40(5):1024–1031. doi: 10.1124/dmd.111.041616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Bankstahl JP, Bankstahl M, Romermann K, Wanek T, Stanek J, Windhorst AD, et al. Tariquidar and elacridar are dose-dependently transported by P-glycoprotein and Bcrp at the blood-brain barrier: a small-animal positron emission tomography and in vitro study. Drug Metab Dispos. 2013;41(4):754–762. doi: 10.1124/dmd.112.049148. [DOI] [PubMed] [Google Scholar]
- 111.Gupta A, Dai Y, Vethanayagam RR, Hebert MF, Thummel KE, Unadkat JD, et al. 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(3):374–383. doi: 10.1007/s00280-005-0173-6. [DOI] [PubMed] [Google Scholar]
- 112.Gupta A, Unadkat JD, Mao Q. Interactions of azole antifungal agents with the human breast cancer resistance protein (BCRP) J Pharm Sci. 2007;96(12):3226–3235. doi: 10.1002/jps.20963. [DOI] [PubMed] [Google Scholar]
- 113.Gupta A, Zhang Y, Unadkat JD, Mao Q. HIV protease inhibitors are inhibitors but not substrates of the human breast cancer resistance protein (BCRP/ABCG2) J Pharmacol Exp Ther. 2004;310(1):334–341. doi: 10.1124/jpet.104.065342. [DOI] [PubMed] [Google Scholar]
- 114.Bierman WF, Scheffer GL, Schoonderwoerd A, Jansen G, van Agtmael MA, Danner SA, et al. Protease inhibitors atazanavir, lopinavir and ritonavir are potent blockers, but poor substrates, of ABC transporters in a broad panel of ABC transporter-overexpressing cell lines. J Antimicrob Chemother. 2010;65(8):1672–1680. doi: 10.1093/jac/dkq209. [DOI] [PubMed] [Google Scholar]
- 115.Peroni RN, Di Gennaro SS, Hocht C, Chiappetta DA, Rubio MC, Sosnik A, et al. Efavirenz is a substrate and in turn modulates the expression of the efflux transporter ABCG2/BCRP in the gastrointestinal tract of the rat. Biochem Pharmacol. 2011;82(9):1227–1233. doi: 10.1016/j.bcp.2011.07.081. [DOI] [PubMed] [Google Scholar]
- 116.Matsushima S, Maeda K, Kondo C, Hirano M, Sasaki M, Suzuki H, et al. 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(3):1059–1067. doi: 10.1124/jpet.105.085589. [DOI] [PubMed] [Google Scholar]
- 117.Li J, Volpe DA, Wang Y, Zhang W, Bode C, Owen A, et al. Use of transporter knockdown Caco-2 cells to investigate the in vitro efflux of statin drugs. Drug Metab Dispos. 2011;39(7):1196–1202. doi: 10.1124/dmd.111.038075. [DOI] [PubMed] [Google Scholar]
- 118.Selever J, Gu G, Lewis MT, Beyer A, Herynk MH, Covington KR, et al. Dicer-mediated upregulation of BCRP confers tamoxifen resistance in human breast cancer cells. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17(20):6510–6521. doi: 10.1158/1078-0432.CCR-11-1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Bessho Y, Oguri T, Achiwa H, Muramatsu H, Maeda H, Niimi T, et al. Role of ABCG2 as a biomarker for predicting resistance to CPT-11/SN-38 in lung cancer. Cancer science. 2006;97(3):192–198. doi: 10.1111/j.1349-7006.2006.00164.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Imai Y, Nakane M, Kage K, Tsukahara S, Ishikawa E, Tsuruo T, et al. C421A polymorphism in the human breast cancer resistance protein gene is associated with low expression of Q141K protein and low-level drug resistance. Mol Cancer Ther. 2002;1(8):611–616. [PubMed] [Google Scholar]
- 121.Tamura A, Wakabayashi K, Onishi Y, Takeda M, Ikegami Y, Sawada S, et al. Re-evaluation and functional classification of non-synonymous single nucleotide polymorphisms of the human ATP-binding cassette transporter ABCG2. Cancer science. 2007;98(2):231–239. doi: 10.1111/j.1349-7006.2006.00371.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Vethanayagam RR, Wang H, Gupta A, Zhang Y, Lewis F, Unadkat JD, et al. Functional analysis of the human variants of breast cancer resistance protein: I206L, N590Y, and D620N. Drug Metab Dispos. 2005;33(6):697–705. doi: 10.1124/dmd.105.003657. [DOI] [PubMed] [Google Scholar]
- 123.Zelinski T, Coghlan G, Liu XQ, Reid ME. ABCG2 null alleles define the Jr(a-) blood group phenotype. Nat Genet. 2012;44(2):131–132. doi: 10.1038/ng.1075. [DOI] [PubMed] [Google Scholar]
- 124.Matsuo H, Takada T, Ichida K, Nakamura T, Nakayama A, Ikebuchi Y, et al. Common defects of ABCG2, a high-capacity urate exporter, cause gout: a function-based genetic analysis in a Japanese population. Science translational medicine. 2009;1(5):5ra11. doi: 10.1126/scitranslmed.3000237. [DOI] [PubMed] [Google Scholar]
- 125.Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536(7616):285–291. doi: 10.1038/nature19057. [DOI] [PMC free article] [PubMed] [Google Scholar]