The multidrug resistance transporter P-glycoprotein confers resistance to ferroptosis inducers

William J. E. Frye , Lyn M. Huff , José M. González Dalmasy , Paula Salazar , Rachel M. Carter , Ryan T. Gensler , Dominic Esposito , Robert W. Robey , Suresh V. Ambudkar , Michael M. Gottesman

Cancer Drug Resistance ›› 2023, Vol. 6 ›› Issue (3) : 468 -80.

PDF
Cancer Drug Resistance ›› 2023, Vol. 6 ›› Issue (3) :468 -80. DOI: 10.20517/cdr.2023.29
review-article

The multidrug resistance transporter P-glycoprotein confers resistance to ferroptosis inducers

Author information +
History +
PDF

Abstract

Aim: Ferroptosis is a non-apoptotic form of cell death caused by lethal lipid peroxidation. Several small molecule ferroptosis inducers (FINs) have been reported, yet little information is available regarding their interaction with the ATP-binding cassette (ABC) transporters P-glycoprotein (P-gp, ABCB1) and ABCG2. We thus sought to characterize the interactions of FINs with P-gp and ABCG2, which may provide information regarding oral bioavailability and brain penetration and predict drug-drug interactions.

Methods: Cytotoxicity assays with ferroptosis-sensitive A673 cells transfected to express P-gp or ABCG2 were used to determine the ability of the transporters to confer resistance to FINs; confirmatory studies were performed in OVCAR8 and NCI/ADR-RES cells. The ability of FINs to inhibit P-gp or ABCG2 was determined using the fluorescent substrates rhodamine 123 or purpuin-18, respectively.

Results: P-gp overexpression conferred resistance to FIN56 and the erastin derivatives imidazole ketone erastin and piperazine erastin. P-gp-mediated resistance to imidazole ketone erastin and piperazine erastin was also reversed in UO-31 renal cancer cells by CRISPR-mediated knockout of ABCB1. The FINs ML-162, GPX inhibitor 26a, and PACMA31 at 10 µM were able to increase intracellular rhodamine 123 fluorescence over 10-fold in P-gp-expressing MDR-19 cells. GPX inhibitor 26a was able to increase intracellular purpurin-18 fluorescence over 4-fold in ABCG2-expressing R-5 cells.

Conclusion: Expression of P-gp may reduce the efficacy of these FINs in cancers that express the transporter and may prevent access to sanctuary sites such as the brain. The ability of some FINs to inhibit P-gp and ABCG2 suggests potential drug-drug interactions.

Keywords

Ferroptosis / drug resistance / P-glycoprotein / ABCG2

Cite this article

Download citation ▾
William J. E. Frye, Lyn M. Huff, José M. González Dalmasy, Paula Salazar, Rachel M. Carter, Ryan T. Gensler, Dominic Esposito, Robert W. Robey, Suresh V. Ambudkar, Michael M. Gottesman. The multidrug resistance transporter P-glycoprotein confers resistance to ferroptosis inducers. Cancer Drug Resistance, 2023, 6(3): 468-80 DOI:10.20517/cdr.2023.29

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Stockwell BR,Bayir H.Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease.Cell2017;171:273-85 PMCID:PMC5685180

[2]

Dolma S,Hahn WC.Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells.Cancer Cell2003;3:285-96

[3]

Dixon SJ,Lamprecht MR.Ferroptosis: an iron-dependent form of nonapoptotic cell death.Cell2012;149:1060-72 PMCID:PMC3367386

[4]

Yang WS,Welsch ME.Regulation of ferroptotic cancer cell death by GPX4.Cell2014;156:317-31 PMCID:PMC4076414

[5]

Dixon SJ,Welsch M.Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis.Elife2014;3:e02523 PMCID:PMC4054777

[6]

Zhang Y,Daniels JD.Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model.Cell Chem Biol2019;26:623-633.e9 PMCID:PMC6525071

[7]

Shimada K,Kaplan A.Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis.Nat Chem Biol2016;12:497-503 PMCID:PMC4920070

[8]

Yang WS.Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells.Chem Biol2008;15:234-45 PMCID:PMC2683762

[9]

Gaschler MM,Liu H.FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation.Nat Chem Biol2018;14:507-15 PMCID:PMC5899674

[10]

Chen YC,Pope LE,Dixon SJ.Reactivity-based probe of the iron(II)-dependent interactome identifies new cellular modulators of ferroptosis.J Am Chem Soc2020;142:19085-93 PMCID:PMC8297516

[11]

Xu C,Wang J.Discovery of a potent glutathione peroxidase 4 inhibitor as a selective ferroptosis inducer.J Med Chem2021;64:13312-26

[12]

Weïwer M,Lewis TA.Development of small-molecule probes that selectively kill cells induced to express mutant RAS.Bioorg Med Chem Lett2012;22:1822-6 PMCID:PMC3528973

[13]

Kanamaru H,Yoshida O,Pastan I.MDR1 RNA levels in human renal cell carcinomas: correlation with grade and prediction of reversal of doxorubicin resistance by quinidine in tumor explants.J Natl Cancer Inst1989;81:844-9

[14]

Gottesman MM,Bates SE.Multidrug resistance in cancer: role of ATP-dependent transporters.Nature Rev Cancer2002;2:48-58

[15]

Wang H,Zhu Z.ABCG2 is a potential prognostic marker of overall survival in patients with clear cell renal cell carcinoma.BMC Cancer2017;17:222 PMCID:PMC5368932

[16]

Durmus S,Schinkel AH.Apical ABC transporters and cancer chemotherapeutic drug disposition.Adv Cancer Res2015;125:1-41

[17]

Vlaming ML,Schinkel AH.Physiological and pharmacological roles of ABCG2 (BCRP): recent findings in Abcg2 knockout mice.Adv Drug Deliv Rev2009;61:14-25

[18]

Robey RW,Qiu J,Bates SE.Rapid detection of ABC transporter interaction: potential utility in pharmacology.J Pharmacol Toxicol Methods2011;63:217-22 PMCID:PMC3086650

[19]

Robey RW,Polgar O.Pheophorbide a is a specific probe for ABCG2 function and inhibition.Cancer Res2004;64:1242-6

[20]

Ambudkar SV.Drug-stimulatable ATPase activity in crude membranes of human MDR1-transfected mammalian cells.Methods Enzymol1998;292:504-14

[21]

Lee JS,Alvarez M et al.Rhodamine efflux patterns predict P-glycoprotein substrates in the National Cancer Institute Drug Screen.Mol Pharmacol1994;46:627-38

[22]

Ambudkar SV,Pashinsky I.Relation between the turnover number for vinblastine transport and for vinblastine-stimulated ATP hydrolysis by human P-glycoprotein.J Biol Chem1997;272:21160-6

[23]

Viswanathan VS,Dhruv HD.Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway.Nature2017;547:453-7

[24]

Wang C,Wang H.Dual degradation mechanism of GPX4 degrader in induction of ferroptosis exerting anti-resistant tumor effect.Eur J Med Chem2023;247:115072

[25]

Zhou HH,Cai LY.Erastin reverses ABCB1-mediated docetaxel resistance in ovarian cancer.Front Oncol2019;9:1398 PMCID:PMC6930896

[26]

Orr GA,McDaid H.Mechanisms of Taxol resistance related to microtubules.Oncogene2003;22:7280-95 PMCID:PMC4039039

[27]

Shukla S,Ambudkar SV.Tyrosine kinase inhibitors as modulators of ABC transporter-mediated drug resistance.Drug Resist Updat2012;15:70-80 PMCID:PMC3348341

[28]

Hegedus C,Apáti A.Interaction of nilotinib, dasatinib and bosutinib with ABCB1 and ABCG2: implications for altered anti-cancer effects and pharmacological properties.Br J Pharmacol2009;158:1153-64 PMCID:PMC2785536

[29]

Dohse M,Shukla S.Comparison of ATP-binding cassette transporter interactions with the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib.Drug Metab Dispos2010;38:1371-80 PMCID:PMC2913625

[30]

Eadie LN,Hughes TP.Degree of kinase inhibition achieved in vitro by imatinib and nilotinib is decreased by high levels of ABCB1 but not ABCG2.Leuk Lymphoma2013;54:569-78

[31]

Tiwari AK,Wang SR.Nilotinib (AMN107, Tasigna) reverses multidrug resistance by inhibiting the activity of the ABCB1/Pgp and ABCG2/BCRP/MXR transporters.Biochem Pharmacol2009;78:153-61

[32]

Katayama R,Yanagitani N.P-glycoprotein Mediates ceritinib resistance in anaplastic lymphoma kinase-rearranged non-small cell lung cancer.EBioMedicine2016;3:54-66 PMCID:PMC4739423

[33]

Hu J,Wang F.Effect of ceritinib (LDK378) on enhancement of chemotherapeutic agents in ABCB1 and ABCG2 overexpressing cells in vitro and in vivo.Oncotarget2015;6:44643-59 PMCID:PMC4792582

[34]

Michaelis M,Rothweiler F,Cinatl J Jr.ABCG2 impairs the activity of the aurora kinase inhibitor tozasertib but not of alisertib.BMC Res Notes2015;8:484 PMCID:PMC4587578

[35]

Guo J,Tapang P.Identification of genes that confer tumor cell resistance to the aurora B kinase inhibitor, AZD1152.Pharmacogenomics J2009;9:90-102

[36]

Durmus S,Sparidans RW,Beijnen JH.P-glycoprotein (MDR1/ABCB1) and breast cancer resistance protein (BCRP/ABCG2) restrict brain accumulation of the JAK1/2 inhibitor, CYT387.Pharmacol Res2013;76:9-16

[37]

Kort A,Sparidans RW,Beijnen JH.Brain accumulation of ponatinib and its active metabolite, N-desmethyl ponatinib, is limited by p-glycoprotein (p-gp/abcb1) and breast cancer resistance protein (BCRP/ABCG2).Mol Pharm2017;14:3258-68

AI Summary AI Mindmap
PDF

68

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/