Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Sep 18.
Published in final edited form as: Biomed Pharmacother. 2022 Apr 5;149:112922. doi: 10.1016/j.biopha.2022.112922

The multi-targeted tyrosine kinase inhibitor SKLB610 resensitizes ABCG2-overexpressing multidrug-resistant cancer cells to chemotherapeutic drugs

Chung-Pu Wu a,b,c,*, Megumi Murakami d, Yu-Shan Wu e, Chun-Ling Lin a, Yan-Qing Li a, Yang-Hui Huang a,b, Tai-Ho Hung c,f,g, Suresh V Ambudkar d
PMCID: PMC10506422  NIHMSID: NIHMS1926012  PMID: 36068781

Abstract

The overexpression of ATP-binding cassette (ABC) transporter ABCB1 (P-glycoprotein) or ABCG2 (BCRP/MXR/ABCP) in cancer cells is frequently associated with the development of multidrug resistance (MDR) in cancer patients, which remains a major obstacle to effective cancer treatment. By utilizing energy derived from ATP hydrolysis, both transporters have been shown to reduce the chemosensitivity of cancer cells by actively effluxing cytotoxic anticancer drugs out of cancer cells. Knowing that there are presently no approved drugs or other therapeutics for the treatment of multidrug-resistant cancers, in recent years, studies have investigated the repurposing of tyrosine kinase inhibitors (TKIs) to act as agents against MDR mediated by ABCB1 and/or ABCG2. SKLB610 is a multi-targeted TKI with potent activity against vascular endothelial growth factor receptor 2 (VEGFR2), platelet-derived growth factor receptor (PDGFR), and fibroblast growth factor receptor 2 (FGFR2). In this study, we investigate the interaction of SKLB610 with ABCB1 and ABCG2. We discovered that neither ABCB1 nor ABCG2 confers resistance to SKLB610, but SKLB610 selectively sensitizes ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic anticancer agents in a concentration-dependent manner. Our data indicate that SKLB610 reverses ABCG2-mediated MDR by attenuating the drug-efflux function of ABCG2 without affecting its total cell expression. These findings are further supported by results of SKLB610-stimulated ABCG2 ATPase activity and in silico docking of SKLB610 in the drug-binding pocket of ABCG2. In summary, we reveal the potential of SKLB610 to overcome resistance to cytotoxic anticancer drugs, which offers an additional treatment option for patients with multidrug-resistant cancers and warrants further investigation.

Keywords: Drug repurposing, ABCG2, Multidrug resistance, SKLB610, VEGFR

1. Introduction

ABCB1 (MDR1; P-glycoprotein) and ABCG2 (BCRP; MXR; ABCP) are two of the most well-characterized members of the human ATP-binding cassette (ABC) proteins known to utilize the chemical energy produced by ATP hydrolysis to translocate a wide range of structurally unrelated xenobiotics and chemotherapeutic drugs across biological membranes [13]. Many of the most commonly prescribed cytotoxic anticancer drugs, such as Vinca alkaloids, mitoxantrone, topotecan, taxanes and anthracyclines, as well as molecularly targeted agents, are known substrate drugs of ABCB1 and/or ABCG2 [310]. These transporters are recognized to play an important role in modulating the oral availability and tissue distribution of substrate drugs because of the protective barriers they form at the intestinal epithelium, blood–brain barrier (BBB), and the blood–placenta barrier (BPB) [3,11,12]. Moreover, the development of multidrug-resistance (MDR) associated with the overexpression of ABCB1 or ABCG2 in cancer cells has been linked to poor prognosis [1318] and failure of cancer chemotherapy [13,19], especially in patients with solid tumors such as lung carcinoma [20], esophageal squamous cell carcinoma (ESCC)[21,22] and metastatic breast cancer (MBC) [18], and in individuals with blood cancers such as acute myelogenous leukemia (AML), acute lymphocytic leukemia (ALL), chronic lymphocytic leukemia (CLL) [1316], and multiple myeloma (MM) [17,23,24]. Therefore, it is of great significance to discover selective and effective therapeutic agents against the activity of ABCB1 or ABCG2 for clinical application [3,4].

Despite the tremendous efforts that have been invested in the preclinical development of synthetic inhibitors of ABCB1 and ABCG2, the U.S. Food and Drug Administration (FDA) has yet to approve any inhibitor to overcome MDR in cancer patients; the progress has been slow due to unforeseen adverse drug effects [3,2528]. For instance, the ABCB1 inhibitor tariquidar (XR9576) failed in two phase III clinical trials (ClinicalTrials.gov Identifier: NCT00042315 and NCT00042302) due to increased toxicity in patients. As a result, a drug repositioning (also known as drug repurposing or drug retasking) approach has been considered by many as an alternative approach to the traditional drug discovery process. SKLB610 is a multi-targeted inhibitor of angiogenesis that is highly effective against the activity of angiogenesis vascular endothelial growth factor receptor 2 (VEGFR2), platelet-derived growth factor receptor (PDGFR) and fibroblast growth factor receptor 2 (FGFR2) [29]. SKLB610 was found to inhibit the proliferation of a variety of human cancer cell lines with IC50 values in the range of 5–26 μM [29]. Additional pharmacokinetic studies of SKLB610 were also performed [30,31]. In the present study, we explore the prospects of repurposing SKLB610 to sensitize multidrug-resistant cancer cells overexpressing ABCB1 or ABCG2 to conventional cytotoxic anticancer drugs. We found that neither ABCB1 nor ABCG2 mediates resistance to SKLB610 in cancer cells. More importantly, we discovered an additional pharmacological activity of SKLB610 as an inhibitor of ABCG2-mediated drug transport and its ability to sensitize ABCG2-overexpressing multidrug-resistant cancer cells to apoptosis and cytotoxicity induced by cytotoxic anticancer drugs. Collectively, in addition to its anti-proliferative activity, the additional pharmacological activity of SKLB610 against ABCG2-mediated MDR warrants further studies for its use in combination therapies in patients with multidrug-resistant cancers.

2. Materials and methods

2.1. Cell culture

The human epidermal cancer cell line KB-3–1 and its ABCB1-overexpressing variant KB-V1 [32], the human embryonic kidney 293 cell line (HEK 293) transfected with either empty pcDNA 3.1 vector, or human ABCB1 (MDR19-HEK293) [33] or human ABCG2 (R482-HEK293) [34] were maintained in Dulbecco’s Modified Eagle’s Medium (DMEM) (Gibco, Invitrogen, Carlsbad, CA, USA). The human ovarian cancer cell line OVCAR-8 and its ABCB1-overexpressing variant NCI-ADR-RES [35], the human colon cancer cell line S1 and its ABCG2-overexpressing variant S1-MI-80 [36], the human non-small cell lung cancer (NSCLC) cell line H460 and its ABCG2-overexpressing variant H460-MX20 [37] were maintained in Roswell Park Memorial Institute (RPMI-1640) medium (Gibco, Invitrogen, Carlsbad, CA, USA). The HEK293 transfectants were maintained in the presence of 2 mg/mL of G418 [38]; KB-V1 cells were cultured in the presence of 1 μg/mL of vinblastine [39] and NCI-ADR-RES cells were cultured in the presence of 0.85 μM doxorubicin [35], whereas H460-MX20 cells were cultured in the presence of 20 nM of mitoxantrone [40] and S1-MI-80 cells were cultured in the presence of 80 μM mitoxantrone [36]. All cells were cultured at 37 °C in 5% CO2 humidified air and grown in media supplemented with 10% FCS, L-glutamine and 100 units/mL of penicillin and streptomycin. Cell lines were generous gifts from Drs. Michael Gottesman and Susan Bates (NCI, NIH, Bethesda, MD, USA). Cells were screened periodically for mycoplasma contamination using a TOOLS Mycoplasma Detection Kit and maintained in drug-free medium for 7 days before assay.

2.2. Chemicals

The TOOLS Cell Counting (CCK-8) kit was purchased from Biotools Co., Ltd. (Taipei, Taiwan). The annexin V FITC-apoptosis detection kit was purchased from BD Pharmingen (San Diego, CA, USA). SKLB610 was obtained from Selleckchem (Houston, TX, USA). Ko143, tariquidar, and all other chemicals were purchased from Sigma (St. Louis, MO, USA), unless stated otherwise.

2.3. Cytotoxicity assays

To determine the sensitivity of cell lines to SKLB610 or a combination of SKLB610 with another cytotoxic drug, cytotoxic MTT and Cell Counting Kit-8 (CCK-8) assays were performed based on the method described by Ishiyama et al. [41] and as described previously [42]. Briefly, cells were seeded in 96-well flat-bottom plates and allowed to attach at 37 °C in 5% CO2 humidified air. After 24 h, different concentrations of SKLB610 or drug combinations were added to each well with 0.5% (v/v) final concentration of DMSO in all wells and incubated for an additional 72 h before being processed with either MTT or CCK-8 reagent as described previously [38]. The IC50 value of each drug regimen was calculated using a fitted concentration-response curve from at least three independent experiments. The extent of drug resistance was presented as a resistance-factor (R.F.) value as described previously [43]. For the drug resistance reversal assays, the extent of chemosensitization by SKLB610 was presented as a fold-reversal (F.R.) value, determined by adding SKLB610 or a reference inhibitor of ABCB1 or ABCG2, at nontoxic concentrations, to the cytotoxicity assays as described previously [44].

2.4. Flow cytometry

A BD FACScan flow cytometer (BD Biosciences) was used to determine the intracellular accumulation of pheophorbide A (PhA) (395 nm excitation and 670 nm emission), a known fluorescent substrate of ABCG2 [45], as described previously [46]. Briefly, cells were first trypsinized and resuspended in Iscove’s modified Dulbecco’s medium (IMDM) containing 5% FBS before PhA (1 μM) was added to 3 × 105 cells in 4 mL of IMDM in the presence of DMSO (control), 10 μM SKLB610 or 5 μM Ko143, an ABCG2 reference inhibitor. The relative fluorescence intensity of PhA was analyzed using CellQuest software (Becton-Dickinson Biosciences, San Jose, CA) and FlowJo software (Tree Star, Inc., Ashland, OR, USA), as described previously [43,45].

2.5. Immunoblot analysis

Cancer cells were treated with either DMSO (control) or SKLB610 at 0.5–3.0 μM for 72 h before being harvested and subjected to SDS-polyacrylamide electrophoresis and Western blotting as described previously [38]. Primary antibodies BXP-21 (1:1000 dilution) and anti-alpha tubulin (1:100,000 dilution) (Sigma-Aldrich, St. Louis, MO, USA) were used to detect human ABCG2 and the positive loading control tubulin, respectively. Horseradish peroxidase-conjugated goat anti-mouse immunoglobulin G (IgG) (1:100,000 dilution) (Abcam, Cambridge, MA, USA) was used as a secondary antibody. Signals were detected using the enhanced chemiluminescence (ECL) kit (Merck Millipore, Billerica, MA, USA) as described previously [38].

2.6. Apoptosis assays

The concurrent annexin V–FITC and propidium iodide (PI) staining method was used to determine the pro-apoptotic effect of drug regimens in cancer cells as previously described [47]. Briefly, parental drug-sensitive and ABCG2-overexpressing multidrug-resistant cancer cells were treated with either DMSO, 10 μM of SKLB610 alone, or 5 μM of the known apoptotic inducer topotecan, or a combination of 5 μM topotecan and 10 μM SKLB610 for 48 h. Cells were subsequently processed using the FITC Annexin V Apoptosis Detection Kit (BD Pharmingen, San Diego, CA, USA) according to the manufacturer’s protocol and analyzed (10,000 cells per sample) using a FACSCalibur flow cytometer equipped with CellQuest software (Becton-Dickinson Biosciences, San Jose, CA) as described previously [42].

2.7. ATPase assay

The vanadate (Vi)-sensitive ATPase activity of ABCG2 was measured using membrane vesicles prepared from ABCG2 baculovirus-infected High-Five insect cells based on the endpoint Pi assay as described previously [48].

2.8. Docking analysis

The energy was minimized for both the structures of ABCG2 protein (PDB: 6VXH) [49] and SKLB610 with CHARMM force field at pH 7.4 using Accelrys Discovery Studio 4.0. Ligand preparation and docking of SKLB610 in ABCG2 was performed using the CDOCKER module of the same software. The respective interaction energy was calculated and the conformation with the lowest CDOCKER interaction energy was selected as described previously [50].

2.9. Quantification and statistical analysis

Experimental values are presented as mean ± standard deviation (S. D.) or as mean ± standard error of the mean (S.E.M) calculated from at least three independent experiments. GraphPad Prism software (GraphPad Software, La Jolla, CA, USA) and KaleidaGraph software (Synergy Software, Reading, PA, USA) were used for curve plotting and statistical analysis, respectively. Two-tailed Student’s t-tests were performed to analyze the difference between mean values of experimental and control or improvement in fit and labeled with asterisks as “statistically significant” if the probability, p, was less than 0.05.

3. Results

3.1. SKLB610 is equally cytotoxic to drug-sensitive and multidrug-resistant cells overexpressing ABCB1 or ABCG2

Previous studies have reported that ABCB1 and ABCG2 are capable of transporting and/or conferring resistance to some tyrosine kinase inhibitors (TKIs) [5156]. Therefore, the intrinsic cytotoxicity of SKLB610, a multi-targeted inhibitor of tyrosine kinases, was examined in multiple pairs of drug-sensitive cell lines and the corresponding ABCB1- or ABCG2-overexpressing multidrug-resistant sublines. The extent of cellular resistance to SKLB610 mediated by ABCB1 or ABCG2 was represented as the resistance factor (R.F.), which was calculated by dividing the IC50 value of SKLB610 in the multidrug-resistant subline by the IC50 value of SKLB610 in the respective parental line. As summarized in Table 1, the ABCB1-overexpressing human epidermal cancer cell line KB-V1 and human ovarian cancer cell line NCI-ADR-RES, as well as the ABCG2-overexpressing human lung cancer cell line H460-MX20 and the human colon cancer cell line S1-M180, and their corresponding parental cell lines were equally sensitive to SKLB610. Similarly, SKLB610 was cytotoxic to the same level for HEK293 cells transfected with human ABCB1 (referred to as MDR19-HEK293) or human ABCG2 (referred to as R482-HEK293), and parental HEK293 transfected with empty pcDNA 3.1 vector (referred to as pcDNA-HEK293).

Table 1.

Cytotoxicity of SKLB610 in human cell lines overexpressing ABCB1 or ABCG2.

Cell line Type Transporter
overexpressed
IC50 (μM)a RFb

KB-3–1 Epidermal 15.75 ± 2.48 1.0
KB-V1 Epidermal ABCB1 10.75 ± 2.32 0.7
OVCAR-8 Ovarian 20.92 ± 1.31 1.0
NCI-ADR-RES Ovarian ABCB1 25.07 ± 2.64 1.2
H460 Lung 31.07 ± 3.22 1.0
H460-MX20 Lung ABCG2 25.10 ± 2.76 0.8
S1 Colon 16.06 ± 4.15 1.0
S1-MI-80 Colon ABCG2 17.06 ± 2.26 1.1
pcDNA3.1-HEK293 54.72 ± 7.46 1.0
MDR19-HEK293 ABCB1 68.61 ± 12.58 1.3
R482-HEK293 ABCG2 77.76 ± 12.89 1.4

Abbreviations: RF, resistance factor.

a

IC50 values were calculated from dose-response curves obtained from at least three independent experiments as described in Section 2.

b

RF values were obtained by dividing the IC50 value of SKLB610 in the ABCB1- or ABCG2-overexpressing multidrug-resistant cell lines by the IC50 value of SKLB610 in the corresponding drug-sensitive parental cell lines.

3.2. SKLB610 re-sensitizes ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic anticancer drugs

Given that some TKIs are known to reverse ABCB1- and ABCG2-mediated multidrug resistance in cancer cells [42,44,50,5761], SKLB610 was assessed for its ability to re-sensitize multidrug-resistant human cancer cells overexpressing ABCB1 or ABCG2, as well as in HEK293 cells transfected with human ABCB1 or ABCG2 to cytotoxic drugs. As summarized in Table 2, SKLB610 at sub-toxic concentrations (0.5–3.0 μM) had no significant effect on ABCB1-mediated resistance to colchicine, vincristine or doxorubicin [62] in KB-V1, NCI-ADR-RES cancer cells or MDR19-HEK293 cells. In contrast, we found that SKLB610 could reverse ABCG2-mediated resistance to its substrates mitoxantrone (Fig. 1AC), SN-38 (Fig. 1DF) and topotecan (Fig. 1GI) [6365] in S1-MI-80 and H460-MX20 cancer cells, as well as in R482-HEK293 cells in a concentration-dependent manner. As shown in Table 3, the extent of re-sensitization by SKLB610 was represented as the fold-reversal (F.R.) value, which was calculated by dividing the IC50 value of a particular cytotoxic drug in the absence of SKLB610 by the IC50 value of the same cytotoxic drug in the presence of SKLB610 in the same cell line [44]. Tariquidar and Ko143 were used here as positive controls to demonstrate the reversal of drug resistance mediated by ABCB1 and ABCG2, respectively. Of note, other than the H460 cell line that expresses a noticeable amount of ABCG2 intrinsically, SKLB610 had no significant effect on the chemosensitivity of drug-sensitive parental cells. Our data revealed that SKLB610 selectively reversed ABCG2-mediated MDR in cancer cells.

Table 2.

The effect of SKLB610 on ABCB1-mediated multidrug resistance.

Mean IC50a ± SD and (FRb)
Treatment Concentration (μM) OVCAR-8 (parental) [nM] NCI-ADR-RES (resistant) [μM]
Colchicine 23.96 ± 6.25 (1.0) 2.75 ± 0.52 (1.0)
+ SKLB610 0.5 25.78 ± 7.41 (0.9) 3.07 ± 0.55 (0.9)
+ SKLB610 1.0 26.72 ± 8.02 (0.9) 3.58 ± 0.66 (0.8)
+ SKLB610 2.0 26.08 ± 7.30 (0.9) 2.74 ± 0.55 (1.0)
+ SKLB610 3.0 25.91 ± 7.31 (0.9) 2.70 ± 0.52 (1.0)
+ Tariquidar 1.0 24.44 ± 6.74 (1.0) 46.79 ± 15.01 [nM]*** (59)
[nM] [μM]
Vincristine 15.47 ± 1.80 (1.0) 6.09 ± 1.32 (1.0)
+ SKLB610 0.5 16.83 ± 2.42 (0.9) 6.06 ± 1.47 (1.0)
+ SKLB610 1.0 16.37 ± 2.45 (0.9) 5.33 ± 1.08 (1.1)
+ SKLB610 2.0 15.35 ± 1.82 (1.0) 4.12 ± 0.88 (1.5)
+ SKLB610 3.0 16.05 ± 2.66 (1.0) 3.13 ± 0.64** (1.9)
+ Tariquidar 1.0 13.34 ± 1.56 (1.2) 29.96 ± 5.25 [nM]** (203)
[nM] [μM]
Doxorubicin 296.98 ± 35.69 (1.0) 7.17 ± 0.77 (1.0)
+ SKLB610 0.5 276.25 ± 22.13 (1.1) 8.32 ± 0.80 (0.9)
+ SKLB610 1.0 267.15 ± 30.99 (1.1) 8.81 ± 0.93 (0.8)
+ SKLB610 2.0 253.97 ± 28.85 (1.2) 7.12 ± 0.62 (1.0)
+ SKLB610 3.0 227.27 ± 28.82 (1.3) 6.25 ± 0.62 (1.1)
+ Tariquidar 1.0 261.99 ± 33.88 (1.1) 0.33 ± 0.05*** (21.7)
Treatment Concentration (μM) KB-3–1 (parental) [nM] KB-V1 (resistant) [μM]
Colchicine 11.35 ± 4.35 (1.0) 1.29 ± 0.16 (1.0)
+ SKLB610 0.5 11.72 ± 4.58 (1.0) 1.59 ± 0.18 (0.8)
+ SKLB610 1.0 11.78 ± 4.68 (1.0) 1.71 ± 0.23 (0.8)
+ SKLB610 2.0 12.00 ± 4.78 (0.9) 1.59 ± 0.25 (0.8)
+ SKLB610 3.0 11.82 ± 4.75 (1.0) 1.34 ± 0.23 (1.0)
+ Tariquidar 1.0 11.73 ± 4.62 (1.0) 14.18 ± 5.29 [nM]*** (91)
[nM] [nM]
Vincristine 1.46 ± 0.41 (1.0) 1609.50 ± 381.83 (1.0)
+ SKLB610 0.5 1.75 ± 0.54 (0.8) 1385.80 ± 308.97 (1.2)
+ SKLB610 1.0 1.90 ± 0.62 (0.8) 1289.69 ± 299.71 (1.2)
+ SKLB610 2.0 1.82 ± 0.58 (0.8) 1005.78 ± 230.61 (1.6)
+ SKLB610 3.0 1.96 ± 0.66 (0.7) 912.63 ± 214.78 (1.8)
+ Tariquidar 1.0 1.49 ± 0.41 (1.0) 2.79 ± 0.51** (577)
[nM] [μM]
Doxorubicin 47.03 ± 8.60 (1.0) 2.10 ± 0.22 (1.0)
+ SKLB610 0.5 42.56 ± 6.70 (1.1) 2.27 ± 0.22 (0.9)
+ SKLB610 1.0 40.90 ± 7.40 (1.1) 2.18 ± 0.22 (1.0)
+ SKLB610 2.0 41.54 ± 7.72 (1.1) 2.34 ± 0.28 (0.9)
+ SKLB610 3.0 42.95 ± 7.76 (1.1) 1.69 ± 0.21 (1.2)
+ Tariquidar 1.0 47.09 ± 9.26 (1.0) 0.15 ± 0.02*** (14.0)
Treatment Concentration (μM) pcDNA3.1-HEK293 (parental) [nM] MDR19-HEK293 (resistant) [nM]
Colchicine 14.57 ± 4.88 (1.0) 147.25 ± 29.93 (1.0)
+ SKLB610 0.5 14.75 ± 5.02 (1.0) 181.71 ± 38.28 (0.8)
+ SKLB610 1.0 13.77 ± 4.57 (1.1) 169.78 ± 46.56 (0.9)
+ SKLB610 2.0 13.96 ± 4.54 (1.0) 198.09 ± 49.60 (0.7)
+ SKLB610 3.0 13.69 ± 4.56 (1.1) 175.21 ± 49.45 (0.8)
+ Tariquidar 1.0 15.00 ± 5.05 (1.0) 11.53 ± 4.10** (12.8)
[nM] [nM]
Vincristine 3.76 ± 0.60 (1.0) 547.08 ± 110.31 (1.0)
+ SKLB610 0.5 3.25 ± 0.44 (1.2) 655.39 ± 104.48 (0.8)
+ SKLB610 1.0 3.26 ± 0.46 (1.2) 572.03 ± 138.76 (1.0)
+ SKLB610 2.0 3.36 ± 0.56 (1.1) 523.91 ± 111.69 (1.0)
+ SKLB610 3.0 3.43 ± 0.62 (1.1) 418.40 ± 65.50 (1.3)
+ Tariquidar 1.0 2.10 ± 0.32* (1.8) 1.75 ± 0.37** (312.6)
[nM] [nM]
Doxorubicin 20.71 ± 3.84 (1.0) 413.62 ± 56.02 (1.0)
+ SKLB610 0.5 24.43 ± 5.45 (0.8) 462.24 ± 43.34 (0.9)
+ SKLB610 1.0 27.63 ± 6.51 (0.7) 496.15 ± 76.15 (0.8)
+ SKLB610 2.0 24.55 ± 4.28 (0.8) 501.70 ± 76.17 (0.8)
+ SKLB610 3.0 23.72 ± 4.25 (0.9) 461.56 ± 64.79 (0.9)
+ Tariquidar 1.0 21.05 ± 3.76 (1.0) 63.71 ± 6.71*** (6.5)

Abbreviations: FR, fold-reversal.

**

p < 0.01

***

p < 0.001.

a

IC50 values were calculated as described in the legend to Table 1.

b

FR values were calculated by dividing the IC50 value of a known ABCB1 substrate drug by the IC50 value of the same substrate drug in the presence of SKLB610 or tariquidar.

Fig. 1.

Fig. 1.

Fig. 1.

Fig. 1.

SKLB610 resensitizes ABCG2-overexpressing multidrug-resistant cells to anticancer drugs in a concentration-dependent manner. The effect of SKLB610 on ABCG2-mediated resistance to mitoxantrone (A–C), SN-38 (D–F), and topotecan (G–I) was evaluated in the ABCG2-overexpressing human colon cancer cell line S1-MI-80 (A, D and E, left panels) and its drug-sensitive parental line S1 (A, D and E, right panels), ABCG2-overexpressing human NSCLC cell line H460-MX20 (B, E and H, left panels) and its drug-sensitive parental line H460 (B, E and H, right panels), ABCG2-transfected R482-HEK293 cell line (C, F and I, left panels) and the parental line HEK293 (C, F and I, right panels). Cells were treated with increasing concentrations of mitoxantrone, SN-38 or topotecan in the presence of DMSO (empty circles) or SKLB610 at 500 nM (empty squares), 1 μM (filled squares), 2 μM (empty triangles), or 3 μM (filled triangles) for 72 h before analysis as described in Section 2. Points, mean values from at least three independent experiments; bars; S.E.M.

Table 3.

The effect of SKLB610 on ABCG2-mediated multidrug resistance.

Mean IC50a ± SD and (FRb)
Treatment Concentration S1 (parental) [nM] S1-MI-80 (resistant)
(μM) [μM]
Mitoxantrone 8.20 ± 1.15 (1.0) 63.99 ± 4.49 (1.0)
+ SKLB610 0.5 6.41 ± 1.02 (1.3) 25.27 ± 2.83*** (2.5)
+ SKLB610 1.0 7.26 ± 1.31 (1.1) 14.05 ± 1.94*** (4.6)
+ SKLB610 2.0 7.22 ± 1.51 (1.1) 8.06 ± 1.06*** (7.9)
+ SKLB610 3.0 5.52 ± 1.91 (15) 4.22 ± 0.58*** (15.2)
+ Ko143 1.0 6.86 ± 1.04 (1.2) 0.73 ± 0.07*** (57.4)
[nM] [μM]
SN-38 5.69 ± 0.82 (1.0) 7.59 ± 2.16 (1.0)
+ SKLB610 0.5 6.61 ± 0.92 (0.9) 4.72 ± 1.69 (1.6)
+ SKLB610 1.0 6.62 ± 0.86 (0.9) 3.17 ± 1.23* (2.4)
+ SKLB610 2.0 6.52 ± 0.90 (0.9) 1.88 ± 0.49* (4.0)
+ SKLB610 3.0 6.63 ± 1.00 (0.9) 1.41 ± 0.39** (5.4)
+ Ko143 1.0 6.78 ± 0.96 (0.8) 110.32 ± 32.73 [nM]*** (69)
[nM] [μM]
Topotecan 47.97 ± 3.39 (1.0) 15.55 ± 2.67 (1.0)
+ SKLB610 0.5 49.86 ± 5.31 (1.0) 4.76 ± 1.01** (3.3)
+ SKLB610 1.0 48.74 ± 5.25 (1.0) 3.98 ± 0.86** (3.9)
+ SKLB610 2.0 44.91 ± 4.74 (1.1) 3.09 ± 0.72** (5.0)
+ SKLB610 3.0 43.41 ± 4.33 (1.1) 1.97 ± 0.43*** (7.9)
+ Ko143 1.0 44.36 ± 5.08 (1.1) 0.33 ± 0.07*** (47.1)
Treatment Concentration H460 (parental) [nM] H460-MX20
μM) (resistant) [nM]
Mitoxantrone 35.04 ± 5.26 (1.0) 841.13 ± 99.22 (1.0)
+ SKLB610 0.5 24.57 ± 3.96 (1.4) 337.64 ± 65.21** (2.5)
+ SKLB610 1.0 20.13 ± 3.67* (1.7) 228.04 ± 51.99*** (3.7)
+ SKLB610 2.0 17.96 ± 3.15** (2.0) 186.38 ± 44.86*** (4.5)
+ SKLB610 3.0 17.87 ± 3.31** (2.0) 175.70 ± 48.63*** (4.8)
+ Ko143 1.0 17.83 ± 3.31** (2.0) 86.46 ± 25.36*** (9.7)
[nM] [μM]
SN-38 16.62 ± 1.98 (1.0) 417.04 ± 77.66 (1.0)
+ SKLB610 0.5 12.46 ± 1.76 (1.3) 214.29 ± 46.00* (1.9)
+ SKLB610 1.0 10.06 ± 1.63* (1.7) 139.10 ± 31.03** (3.0)
+ SKLB610 2.0 7.69 ± 1.24** (2.2) 103.37 ± 25.14** (4.0)
+ SKLB610 3.0 7.56 ± 1.30** (2.2) 86.54 ± 22.74** (4.8)
+ Ko143 1.0 4.90 ± 1.09*** (3.4) 9.56 ± 2.62*** (43.6)
[nM] [μM]
Topotecan 139.14 ± 18.39 (1.0) 1329.22 ± 308.64 (1.0)
+ SKLB610 0.5 93.70 ± 13.58* (1.5) 666.77 ± 149.84* (2.0)
+ SKLB610 1.0 80.22 ± 11.64** (1.7) 591.98 ± 126.24* (2.2)
+ SKLB610 2.0 66.32 ± 10.30** (2.1) 437.45 ± 96.69** (3.0)
+ SKLB610 3.0 56.40 ± 9.16** (2.5) 326.33 ± 75.55** (4.1)
+ Ko143 1.0 48.96 ± 8.50** (2.8) 85.75 ± 20.07** (15.5)
Treatment Concentration pcDNA3.1-HEK293 R482-HEK293
(μM) (parental) [nM] (resistant) [nM]
Mitoxantrone 4.76 ± 1.15 (1.0)
+ SKLB610 0.5 4.51 ± 0.83 (1.1) 125.97 ± 17.05 (1.0)
63.54 ± 7.62** (2.0)
+ SKLB610 1.0 4.47 ± 0.92 (1.1) 55.34 ± 5.17** (2.3)
+ SKLB610 2.0 4.48 ± 0.99 (1.1) 38.78 ± 5.29** (3.2)
+ SKLB610 3.0 4.38 ± 0.95 (1.1) 31.27 ± 4.65*** (4.0)
+ Ko143 1.0 4.29 ± 0.84 (1.1) 12.20 ± 1.02*** (10.3)
SN-38 [nM]
4.26 ± 0.97 (1.0)
[nM]
335.81 ± 36.28 (1.0)
+ SKLB610 0.5 4.27 ± 1.00 (1.0) 131.70 ± 18.69*** (2.5)
+ SKLB610 1.0 4.18 ± 1.03 (1.0) 93.22 ± 17.39*** (3.6)
+ SKLB610 2.0 3.97 ± 0.90 (1.1) 49.99 ± 11.55*** (6.7)
+ SKLB610 3.0 3.61 ± 0.75 (1.2) 38.76 ± 7.92*** (8.7)
+ Ko143 1.0 3.75 ± 0.87 (1.1) 12.22 ± 2.43*** (27.4)
Topotecan [nM]
37.24 ± 9.83 (1.0)
[nM]
731.78 ± 89.04 (1.0)
+ SKLB610 0.5 36.16 ± 9.12 (1.0) 423.60 ± 53.13** (1.7)
+ SKLB610 1.0 32.29 ± 7.10 (1.2) 301.48 ± 44.27** (2.4)
+ SKLB610 2.0 31.15 ± 7.84 (1.2) 203.06 ± 29.44*** (3.6)
+ SKLB610 3.0 28.80 ± 6.25 (1.3) 170.08 ± 30.54*** (4.3)
+ Ko143 1.0 34.71 ± 8.07 (1.1) 138.25 ± 25.38*** (5.3)

Abbreviations: FR, fold-reversal.

*

p < 0.05

**

p < 0.01

***

p < 0.001.

a

IC50 values calculation (see legend to Table 1).

b

FR values were calculated by dividing the IC50 value of a known ABCG2 substrate drug by the IC50 value of the same substrate drug in the presence of SKLB610 or Ko143.

3.3. SKLB610 inhibits ABCG2-mediated drug efflux

Knowing that a common way to re-sensitize ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic drugs is by directly inhibiting the drug transport function of ABCG2 [27,58,59,6668], we examined the ability of SKLB610 to inhibit efflux of Pheophorbide A (PhA), a known fluorescent substrate of ABCG2 [45], from ABCG2-overexpressing cells. Cells were incubated in IMDM containing PhA in the presence of DMSO (solid line), SKLB610 (filled solid line), or Ko143 (dotted line) for 45 min before the intracellular accumulation of PhA was determined as described in Materials and methods. We found that SKLB610 and Ko143 had no significant effect on the intracellular accumulation of PhA in drug-sensitive parental S1 (Fig. 2A, left panel), H460 (Fig. 2B, left panel) cancer cells, or pcDNA-HEK293 cells (Fig. 2C, left panel). In contrast, the intracellular fluorescence of PhA was significantly higher in ABCG2-overexpressing S1-MI-80 (Fig. 2A, right panel) and H460-MX20 (Fig. 2B, right panel) cancer cells, and ABCG2-transfected R482-HEK293 cells (Fig. 2C, right panel) in the presence of SKLB610 or Ko143 versus in the absence of any inhibitor.

Fig. 2.

Fig. 2.

SKLB610 attenuates the drug transport function of ABCG2. The intracellular accumulation of pheophorbide A (PhA), a known fluorescent substrate drug of ABCG2, was determined in (A) S1 and S1-MI-80, (B) H460 and H460-MX20, and (C) pcDNA-HEK293 and R482-HEK293 cells, in the presence of DMSO (control, solid lines), 10 μM SKLB610 (filled solid lines), or 5 μM Ko143, a benchmark inhibitor of ABCG2 (dotted lines). (D) The corresponding quantification of intracellular PhA accumulation in ABCG2-overexpressing S1-MI-80 (black bars), H460-MX20 (white bars) and R482-HEK293 (gray bars). Representative histograms and the quantification values presented as mean ± S.D. calculated from at least three independent experiments are shown. The fluorescence signal was analyzed by flow cytometry as described previously [38].

3.4. SKLB610 does not alter the protein expression of ABCG2 in multidrug-resistant cancer cells

In addition to inhibiting the drug transport function of ABCG2, studies have shown that drug-induced down-regulation of ABCG2 is also a common mechanism by which ABCG2-overexpressing multidrug-resistant cancer cells can be re-sensitized to cytotoxic drugs [69,70]. Therefore, S1-MI-80 and H460-MX20 cancer cell lines were treated with increasing concentrations (0.5–3 μM) of SKLB610 for 72 h and the protein expression of ABCG2 was examined as given in Materials and methods. As shown in Fig. 3, the expression of ABCG2 was not significantly affected by SKLB610 at the protein level in S1-MI-80 (Fig. 3A) or H460-MX20 (Fig. 3B) cancer cells over a period of 72 h. Our results suggest that SKLB610 re-sensitized ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic drugs by inhibiting the drug efflux function of ABCG2.

Fig. 3.

Fig. 3.

SKLB610 has no significant effect on ABCG2 protein expression. The ABCG2-overexpressing S1-MI-80 (A) and H460-MX20 (B) cancer cells were treated with DMSO (vehicle control) or SKLB610 at 0.5 μM, 1 μM, 2 μM, or 3 μM for 72 h and the cell lysates were processed for Western blotting with indicated antibodies as described in Section 2. Representative immunoblots (top) and the corresponding quantification (bottom) of human ABCG2 protein and the internal loading controlα-tubulin are shown. Values are presented as mean ± S.D. calculated from at least three independent experiments.

3.5. SKLB610 restores drug-induced apoptosis in ABCG2-overexpressing multidrug-resistant cancer cells

Knowing that drug-induced growth retardation could also appear to be chemosensitization of multidrug-resistant cancer cells, the effect of SKLB610 on topotecan-induced apoptosis was examined in ABCG2-overexpressing S1-MI-80 cancer cells. S1 and S1-MI-80 cancer cells were treated with DMSO (control), 10 μM SKLB610, 5 μM topotecan or a combination of 5 μM topotecan and 10 μM SKLB610 for 48 h and processed as described in Materials and methods. As shown in Fig. 4, topotecan substantially increased the apoptotic cell population from approximately 4% basal to 39% in the drug-sensitive S1 cancer cell line. In contrast, being a known substrate of ABCG2 [63], topotecan merely increased the apoptotic cell population from approximately 4% basal to 7% in the ABCG2-overexpressing S1-MI-80 subline. However, without affecting the extent of overall apoptosis in S1 cancer cells, SKLB610 significantly enhanced topotecan-induced apoptosis in S1-MI-80 cancer cells from 7% to approximately 19% total apoptosis. Our results indicate that by blocking the drug efflux function of ABCG2, SKLB610 restored drug-induced apoptosis-mediated cytotoxicity in ABCG2-overexpressing multidrug-resistant cancer cells.

Fig. 4.

Fig. 4.

SKLB610 enhances drug-induced apoptosis in ABCG2-overexpressing cancer cells. The drug-sensitive human colon cancer cell line S1 and its ABCG2-overexpressing multidrug-resistant subline S1-MI-80 were treated with DMSO (control), 10 μM SKLB610 alone (+ SKLB610), 5 μM topotecan alone (+ topotecan), or a combination of topotecan and SKLB610 (+ topotecan + SKLB610) for 48 h, processed and analyzed by flow cytometry as described in Materials and methods. Representative flow cytometric dot plots are shown (top), and the corresponding quantifications (bottom) are presented as mean ± S.D. calculated from at least three independent experiments. **p < 0.01, versus the same treatment in the absence of SKLB610.

3.6. Effect of SKLB610 on ABCG2-mediated ATP hydrolysis

The effect of SKLB610 on the ATPase activity of ABCG2 was examined to obtain additional biochemical information on the interactions between SKLB610 and ABCG2 as ATP hydrolysis is known to be associated with the transport activity of ABCG2 [71]. SKLB610 stimulated Vi-sensitive ATPase activity of ABCG2 in a concentration-dependent manner, with a half-maximal effective concentration (EC50) value of approximately 98 nM and a maximum stimulation of almost 300% higher than the basal activity of 58.1 ± 11.2 nmole Pi/min/mg protein (Fig. 5). These results suggest that SKLB610 interacts with ABCG2 at the substrate-binding site.

Fig. 5.

Fig. 5.

SKLB610 stimulates the ATPase activity of ABCG2. The effect of SKLB610 (0–5 μM) on ABCG2-mediated ATP hydrolysis was measured in membrane vesicles prepared from ABCG2 baculovirus-infected High-Five insect cells and recorded as vanadate (Vi)-sensitive ATPase activity as described previously [43]. Points, mean from at least three independent experiments; bars, S.D.

3.7. Docking of SKLB610 in the drug-binding pocket of ABCG2

To further understand the binding of SKLB610 with ABCG2, the potential site of interaction between SKLB610 and ABCG2 was determined by docking SKLB610 into the drug-binding pocket within the transmembrane domain of the human ABCG2 structure (PDB: 6VXH) [49]. The best ligand binding pose was chosen with the calculated binding energy of − 73 kcal/mol. As shown in Fig. 6, hydrophobic interactions between the aromatic rings of SKLB610 and the residues Met549 and Val546 as well as interaction of the trifluoromethyl moiety with LEU405 were predicted within the transmembrane domain of ABCG2.

Fig. 6.

Fig. 6.

Docking of SKLB610 in the drug-binding pocket of ABCG2. Binding modes of SKLB610 with the protein structure of ABCG2 protein structure (PDB: 6VXH) were predicted by Accelrys Discovery Studio 4.0 software as described in Section 2. SKLB610 (yellow color) is presented in stick representation and the atoms for interacting amino acid residues are colored carbon-gray, hydrogen-light gray, oxygen-red, nitrogen-blue and fluorine-cyan. Proposed interactions are presented as dotted lines.

4. Discussion

The development of MDR associated with the overexpression of drug transporters such as ABCB1 or ABCG2 remains a significant obstacle in cancer chemotherapy [3,19]. Patients with multidrug-resistant cancers display reduced responses to conventional cytotoxic anticancer drugs. Unfortunately, the progress of developing selective and potent synthetic inhibitors of these drug transporters has been hindered by unforeseen toxicity [3,2528]. We and others have discovered in recent years that the overexpression of ABCB1 or ABCG2 could be responsible for reduced susceptibility of cancer cells to certain TKIs [46,54,7274]. Some of the TKIs were found to reverse MDR mediated by ABCB1 and/or ABCG2 in cancer cells [50,58,61,7578]. Most recently, studies have demonstrated that branebrutinib [78], almonertinib [77], erdafitinib [61], and sitravatinib [50] inhibit drug efflux mediated by ABCB1, whereas rociletinib [79] and cabozantinib [80] inhibit drug efflux mediated by ABCG2. Notably, results of several combination therapy trials have shown the advantages of combination therapy of TKIs with conventional anticancer drugs over monotherapy in patients with advanced pancreatic cancer [81,82] or human epidermal growth factor receptor 2 (HER2)-positive advanced breast cancer [83,84]. More importantly, TKIs could potentiate the antiproliferative effect of cytotoxic anticancer drugs against multidrug-resistant cancers [85]. Collectively, these findings indicate that further studies are warranted to investigate combination therapies of TKIs and cytotoxic anticancer drugs to overcome ABC transporter-mediated MDR.

In this study, we investigated the potential interactions between the multi-targeted TKI SKLB610, ABCB1 and ABCG2 in multidrug-resistant cancer cell lines overexpressing human ABCB1 or ABCG2. First, we found that SKLB610 was cytotoxic in all tested cancer cell lines (Table 1), with IC50 values in the range of 10–32 μM, which are comparable to the previously reported IC50 values against 12 other cancer cell lines [29]. More importantly, the drug-sensitive parental cancer cell line and the multidrug-resistant sublines overexpressing ABCB1 or ABCG2 were equally sensitive to SKLB610, indicating that SKLB610 is not rapidly transported out of cancer cells by either ABCB1 or ABCG2. Our results suggest that the overexpression of ABCB1 or ABCG2 is unlikely to be one of the key contributing factors in the development of SKLB610 resistance in patients, but the clinical effects of prolonged treatment of patients with SKLB610 remain to be determined. Next, we examined the potential chemosensitizing effect of SKLB610 on drug resistance mediated by ABCB1 and ABCG2 in cells overexpressing ABCB1 or ABCG2. We found that SKLB610 had no significant effect on ABCB1-mediated resistance to colchicine, vincristine, or doxorubicin in ABCB1-overexpressing cancer cells or in HEK293 cells transfected with human ABCB1 (Table 2). In contrast, SKLB610 significantly sensitized ABCG2-overexpressing multidrug-resistant H460-MX20 and S1-MI-80 cancer cells, as well as HEK293 cells transfected with human ABCG2, to mitoxantrone, SN-38, and topotecan in a concentration-dependent manner (Fig. 1), signifying that the activity of SKLB610 is more selective to ABCG2 as compared to ABCB1. Interestingly, although studies have identified TKIs that are capable of re-sensitizing ABCG2-overexpressing multidrug-resistant cancer cells to anticancer drugs, most of these TKIs are not ABCG2-selective due to a substantial overlapping substrate specificity between ABCB1 and ABCG2 [44,50,60,66,86,87]. It should be noted that Cao et al. reported that SKLB610 at 10 μM inhibited the activity of VEGFR2, FGFR2, and PDGFR by 97%, 65%, and 55% in biochemical kinase assays [29], while at the same concentration, we demonstrated that SKLB610 inhibited ABCG2-mediated efflux of a fluorescent substrate drug (Fig. 2D) and enhanced the extent of drug-induced apoptosis (Fig. 4) in ABCG2-overexpressing multidrug-resistant cancer cells. Furthermore, SKLB610 had no significant effect on the total protein level of ABCG2 (Fig. 3) in ABCG2-overexpressing cancer cells, suggesting that SKLB610 resensitizes ABCG2-overexpressing cancer cells to drug-induced apoptosis (Fig. 4) and cytotoxicity (Table 3) by attenuating the drug transport function of ABCG2. Interaction of SKLB610 at the drug-binding pocket of ABCG2 was confirmed by its stimulatory effect on the ATPase activity in a concentration-dependent manner (Fig. 5), and the in silico docking analysis of SKLB610 binding to the inward-open conformation of human ABCG2 (Fig. 6). Our data indicate that SKLB610 interacts with multiple residues within the substrate-binding pocket of ABCG2 and competes with the binding of substrate drugs at the same site, thus inhibiting the transport function of ABCG2 (Fig. 7).

Fig. 7.

Fig. 7.

Simplified schematic illustration of SKLB610 re-sensitizing ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic anticancer drugs by blocking the drug efflux function of ABCG2. The intracellular concentration of ABCG2 substrate drugs (represented by blue circles) in ABCG2-overexpressing cells is actively reduced by ABCG2-mediated drug transport. In contrast, in the presence of SKLB610 (red triangle), ABCG2-mediated drug efflux is attenuated by SKLB610 outcompeting the binding of ABCG2 substrate drug at the same drug-binding pocket, thus restoring the intracellular accumulation of cytotoxic drugs in ABCG2-overexpressing multidrug-resistant cancer cells.

In summary, we demonstrated that SKLB610 could selectively sensitize ABCG2-overexpressing multidrug-resistant cancer cells by modulating the drug transport activity of ABCG2. While undesirable responses or altered pharmacokinetics of concomitant anticancer drugs may occur in combination therapies [3,8891], our findings revealed an additional pharmacological activity of SKLB610 that could potentially be used in combination therapy with cytotoxic anticancer drugs against multidrug-resistant cancers associated with the overexpression of ABCG2.

Supplementary Material

36068781_ambudkar_sm

Acknowledgments

We thank George Leiman for editorial help. This work was supported by grants from the Ministry of Science and Technology, Taiwan (MOST-108–2320-B-182–038-MY3 to CPW and 109–2314-B-182A-097-MY3 to THH) and Chang Gung Memorial Hospital (CMRPD1K0391, CMRPD1L0051 and BMRPC17 to CPW and CMRPG1J0073, CORPG1L0051, and CORPG2L0031 to THH). The authors are grateful to the Taipei Common Laboratory of Chang Gung Memorial Hospital for providing technical assistance. MM and SVA were supported by the Intramural Research Program of the National Institutes of Health, National Cancer Institute, Center for Cancer Research.

Abbreviations:

ABC

ATP-binding cassette

MDR

multidrug resistance

VEGFR

vascular endothelial growth factor

Vi

sodium orthovanadate

FR

fold-reversal

Footnotes

Conflict of interest

The authors have no conflict of interest to declare.

CRediT authorship contribution statement

Chung-Pu Wu: Conceptualization, Methodology, Software, Writing – original draft, Supervision. Megumi Murakami: Data curation, Visualization, Investigation, Writing – review & editing. Yu-Shan Wu: Data curation, Visualization, Investigation, Writing – review & editing. Chun-Ling Lin: Data curation, Visualization, Investigation. Yan-Qing Li: Data curation, Visualization, Investigation. Yang-Hui Huang: Data curation, Visualization, Investigation. Tai-Ho Hung: Conceptualization, Methodology, Supervision. Suresh. V. Ambudkar: Conceptualization, Methodology, Supervision, Writing – review & editing.

Appendix A. Supporting information

Supplementary data associated with this article can be found in the online version at doi:10.1016/j.biopha.2022.112922.

References

  • [1].Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM, Targeting multidrug resistance in cancer, Nat. Rev. 5 (3) (2006) 219–234. [DOI] [PubMed] [Google Scholar]
  • [2].Wu C-P, Hsieh C-H, Wu Y-S, The emergence of drug transporter-mediated multidrug resistance to cancer chemotherapy, Mol. Pharmaceut. 8 (6) (2011) 1996–2011. [DOI] [PubMed] [Google Scholar]
  • [3].Robey RW, Pluchino KM, Hall MD, Fojo AT, Bates SE, Gottesman MM, Revisiting the role of ABC transporters in multidrug-resistant cancer, Nat. Rev. Cancer 18 (7) (2018) 452–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Gottesman M, Ambudkar SV, Overview: ABC transporters and human disease, J. Bioenerget. Biomembr. 33 (6) (2001) 453–458. [DOI] [PubMed] [Google Scholar]
  • [5].Dohse M, Scharenberg C, Shukla S, Robey RW, Volkmann T, Deeken JF, Brendel C, Ambudkar SV, Neubauer A, Bates SE, Comparison of ATP-binding cassette transporter interactions with the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib, Drug Metab. Dispos.: Biol. Fate Chem. 38 (8) (2010) 1371–1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Ozvegy-Laczka C, Hegedus T, Varady G, Ujhelly O, Schuetz JD, Varadi A, Keri G, Orfi L, Nemet K, Sarkadi B, High-affinity interaction of tyrosine kinase inhibitors with the ABCG2 multidrug transporter, Mol. Pharm. 65 (6) (2004) 1485–1495. [DOI] [PubMed] [Google Scholar]
  • [7].Agarwal S, Sane R, Gallardo JL, Ohlfest JR, Elmquist WF, Distribution of gefitinib to the brain is limited by P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2)-mediated active efflux, J. Pharm. Exp. Ther. 334 (1) (2010) 147–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Noguchi K, Katayama K, Sugimoto Y, Human ABC transporter ABCG2/BCRP expression in chemoresistance: basic and clinical perspectives for molecular cancer therapeutics, Pharmacogenom. Pers. Med. 7 (2014) 53–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Wu CP, Hsieh YJ, Hsiao SH, Su CY, Li YQ, Huang YH, Huang CW, Hsieh CH, Yu JS, Wu YS, Human ATP-binding cassette transporter ABCG2 confers resistance to CUDC-907, a dual inhibitor of histone deacetylase and phosphatidylinositol 3-kinase, Mol. Pharm. 13 (3) (2016) 784–794. [DOI] [PubMed] [Google Scholar]
  • [10].Wu CP, Hsieh YJ, Murakami M, Vahedi S, Hsiao SH, Yeh N, Chou AW, Li YQ, Wu YS, Yu JS, Ambudkar SV, Human ATP-binding cassette transporters ABCB1 and ABCG2 confer resistance to histone deacetylase 6 inhibitor ricolinostat (ACY-1215) in cancer cell lines, Biochem. Pharm. 155 (2018) 316–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Breedveld P, Beijnen JH, Schellens JH, Use of P-glycoprotein and BCRP inhibitors to improve oral bioavailability and CNS penetration of anticancer drugs, Trends Pharmacol. Sci. 27 (1) (2006) 17–24. [DOI] [PubMed] [Google Scholar]
  • [12].Sarkadi B, Homolya L, Szakacs G, Varadi A, Human multidrug resistance ABCB and ABCG transporters: participation in a chemoimmunity defense system, Physiol. Rev. 86 (4) (2006) 1179–1236. [DOI] [PubMed] [Google Scholar]
  • [13].Ross DD, Karp JE, Chen TT, Doyle LA, Expression of breast cancer resistance protein in blast cells from patients with acute leukemia, Blood 96 (1) (2000) 365–368. [PubMed] [Google Scholar]
  • [14].Steinbach D, Sell W, Voigt A, Hermann J, Zintl F, Sauerbrey A, BCRP gene expression is associated with a poor response to remission induction therapy in childhood acute myeloid leukemia, Leukemia 16 (8) (2002) 1443–1447. [DOI] [PubMed] [Google Scholar]
  • [15].Uggla B, Stahl E, Wagsater D, Paul C, Karlsson MG, Sirsjo A, Tidefelt U, BCRP mRNA expression v. clinical outcome in 40 adult AML patients, Leuk. Res. 29 (2) (2005) 141–146. [DOI] [PubMed] [Google Scholar]
  • [16].Matthews C, Catherwood MA, Larkin AM, Clynes M, Morris TC, Alexander HD, MDR-1, but not MDR-3 gene expression, is associated with unmutated IgVH genes and poor prognosis chromosomal aberrations in chronic lymphocytic leukemia, Leuk. Lymphoma 47 (11) (2006) 2308–2313. [DOI] [PubMed] [Google Scholar]
  • [17].Turner JG, Gump JL, Zhang C, Cook JM, Marchion D, Hazlehurst L, Munster P, Schell MJ, Dalton WS, Sullivan DM, ABCG2 expression, function, and promoter methylation in human multiple myeloma, Blood 108 (12) (2006) 3881–3889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Kovalev AA, Tsvetaeva DA, Grudinskaja TV, Role of ABC-cassette transporters (MDR1, MRP1, BCRP) in the development of primary and acquired multiple drug resistance in patients with early and metastatic breast cancer, Exp. Oncol. 35 (4) (2013) 287–290. [PubMed] [Google Scholar]
  • [19].Gillet JP, Gottesman MM, Mechanisms of multidrug resistance in cancer, Methods Mol. Biol. 596 (2010) 47–76. [DOI] [PubMed] [Google Scholar]
  • [20].Li F, Zeng H, Ying K, The combination of stem cell markers CD133 and ABCG2 predicts relapse in stage I non-small cell lung carcinomas, Med. Oncol. 28 (4) (2011) 1458–1462. [DOI] [PubMed] [Google Scholar]
  • [21].Hang D, Dong HC, Ning T, Dong B, Hou DL, Xu WG, Prognostic value of the stem cell markers CD133 and ABCG2 expression in esophageal squamous cell carcinoma, Dis. Esophagus 25 (7) (2012) 638–644. [DOI] [PubMed] [Google Scholar]
  • [22].Tsunoda S, Okumura T, Ito T, Kondo K, Ortiz C, Tanaka E, Watanabe G, Itami A, Sakai Y, Shimada Y, ABCG2 expression is an independent unfavorable prognostic factor in esophageal squamous cell carcinoma, Oncology 71 (3–4) (2006) 251–258. [DOI] [PubMed] [Google Scholar]
  • [23].Schwarzenbach H, Expression of MDR1/P-glycoprotein, the multidrug resistance protein MRP, and the lung-resistance protein LRP in multiple myeloma, Med. Oncol. 19 (2) (2002) 87–104. [DOI] [PubMed] [Google Scholar]
  • [24].Tsubaki M, Satou T, Itoh T, Imano M, Komai M, Nishinobo M, Yamashita M, Yanae M, Yamazoe Y, Nishida S, Overexpression of MDR1 and survivin, and decreased Bim expression mediate multidrug-resistance in multiple myeloma cells, Leuk. Res. 36 (10) (2012) 1315–1322. [DOI] [PubMed] [Google Scholar]
  • [25].Kannan P, Telu S, Shukla S, Ambudkar SV, Pike VW, Halldin C, Gottesman MM, Innis RB, Hall MD, The “specific” P-glycoprotein inhibitor Tariquidar is also a substrate and an inhibitor for breast cancer resistance protein (BCRP/ABCG2), ACS Chem. Neurosci. 2 (2) (2011) 82–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Weidner LD, Zoghbi SS, Lu S, Shukla S, Ambudkar SV, Pike VW, Mulder J, Gottesman MM, Innis RB, Hall MD, The inhibitor Ko143 is not specific for ABCG2, J. Pharm. Exp. Ther. 354 (3) (2015) 384–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Toyoda Y, Takada T, Suzuki H, Inhibitors of human ABCG2: from technical background to recent updates with clinical implications, Front. Pharm. 10 (2019) 208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Dong J, Qin Z, Zhang WD, Cheng G, Yehuda AG, Ashby CR Jr., Chen ZS, Cheng XD, Qin JJ, Medicinal chemistry strategies to discover P-glycoprotein inhibitors: An update, Drug Resist. Updates 49 (2020), 100681. [DOI] [PubMed] [Google Scholar]
  • [29].Cao ZX, Zheng RL, Lin HJ, Luo SD, Zhou Y, Xu YZ, Zeng XX, Wang Z, Zhou LN, Mao YQ, Yang L, Wei YQ, Yu LT, Yang SY, Zhao YL, SKLB610: a novel potential inhibitor of vascular endothelial growth factor receptor tyrosine kinases inhibits angiogenesis and tumor growth in vivo, Cell. Physiol. Biochem. 27 (5) (2011) 565–574. [DOI] [PubMed] [Google Scholar]
  • [30].Luo X, Li S, Xie Y, He J, Li J, Lin H, Wang N, Yang S, Zhao Y, Yu L, Song X, Pharmacokinetic studies of a novel multikinase inhibitor for treating cancer by HPLC-UV, J. Chromatogr. Sci. 51 (1) (2013) 17–20. [DOI] [PubMed] [Google Scholar]
  • [31].Huang Y, Luo X, You X, Xia Y, Song X, Yu L, The preparation and evaluation of water-soluble SKLB610 nanosuspensions with improved bioavailability, AAPS PharmSciTech 14 (3) (2013) 1236–1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Shen DW, Cardarelli C, Hwang J, Cornwell M, Richert N, Ishii S, Pastan I, Gottesman MM, Multiple drug-resistant human KB carcinoma cells independently selected for high-level resistance to colchicine, adriamycin, or vinblastine show changes in expression of specific proteins, J. Biol. Chem. 261 (17) (1986) 7762–7770. [PubMed] [Google Scholar]
  • [33].Robey RW, Shukla S, Finley EM, Oldham RK, Barnett D, Ambudkar SV, Fojo T, Bates SE, Inhibition of P-glycoprotein (ABCB1)- and multidrug resistance-associated protein 1 (ABCC1)-mediated transport by the orally administered inhibitor, CBT-1((R), Biochem. Pharm. 75 (6) (2008) 1302–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Robey RW, Honjo Y, Morisaki K, Nadjem TA, Runge S, Risbood M, Poruchynsky MS, Bates SE, Mutations at amino-acid 482 in the ABCG2 gene affect substrate and antagonist specificity, Br. J. Cancer 89 (10) (2003) 1971–1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Roschke AV, Tonon G, Gehlhaus KS, McTyre N, Bussey KJ, Lababidi S, Scudiero DA, Weinstein JN, Kirsch IR, Karyotypic complexity of the NCI-60 drug-screening panel, Cancer Res. 63 (24) (2003) 8634–8647. [PubMed] [Google Scholar]
  • [36].Miyake K, Mickley L, Litman T, Zhan Z, Robey R, Cristensen B, Brangi M, Greenberger L, Dean M, Fojo T, Bates SE, Molecular cloning of cDNAs which are highly overexpressed in mitoxantrone-resistant cells: demonstration of homology to ABC transport genes, Cancer Res. 59 (1) (1999) 8–13. [PubMed] [Google Scholar]
  • [37].Henrich CJ, Robey RW, Bokesch HR, Bates SE, Shukla S, Ambudkar SV, Dean M, McMahon JB, New inhibitors of ABCG2 identified by high-throughput screening, Mol. Cancer Ther. 6 (12 Pt 1) (2007) 3271–3278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Wu CP, Shukla S, Calcagno AM, Hall MD, Gottesman MM, Ambudkar SV, Evidence for dual mode of action of a thiosemicarbazone, NSC73306: a potent substrate of the multidrug resistance linked ABCG2 transporter, Mol. Cancer Ther. 6 (12 Pt 1) (2007) 3287–3296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Shen DW, Fojo A, Chin JE, Roninson IB, Richert N, Pastan I, Gottesman MM, Human multidrug-resistant cell lines: increased mdr1 expression can precede gene amplification, Science 232 (4750) (1986) 643–645. [DOI] [PubMed] [Google Scholar]
  • [40].Henrich CJ, Bokesch HR, Dean M, Bates SE, Robey RW, Goncharova EI, Wilson JA, McMahon JB, A high-throughput cell-based assay for inhibitors of ABCG2 activity, J. Biomol. Screen. 11 (2) (2006) 176–183. [DOI] [PubMed] [Google Scholar]
  • [41].Ishiyama M, Tominaga H, Shiga M, Sasamoto K, Ohkura Y, Ueno K, A combined assay of cell viability and in vitro cytotoxicity with a highly water-soluble tetrazolium salt, neutral red and crystal violet, Biol. Pharm. Bull. 19 (11) (1996) 1518–1520. [DOI] [PubMed] [Google Scholar]
  • [42].Hsiao SH, Lu YJ, Li YQ, Huang YH, Hsieh CH, Wu CP, Osimertinib (AZD9291) attenuates the function of multidrug resistance-linked ATP-binding cassette transporter ABCB1 in vitro, Mol. Pharm. (2016). [DOI] [PubMed] [Google Scholar]
  • [43].Wu CP, Hsiao SH, Sim HM, Luo SY, Tuo WC, Cheng HW, Li YQ, Huang YH, Ambudkar SV, Human ABCB1 (P-glycoprotein) and ABCG2 mediate resistance to BI 2536, a potent and selective inhibitor of Polo-like kinase 1, Biochem. Pharm. 86 (7) (2013) 904–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Dai CL, Tiwari AK, Wu CP, Su XD, Wang SR, Liu DG, Ashby CR Jr., Huang Y, Robey RW, Liang YJ, Chen LM, Shi CJ, Ambudkar SV, Chen ZS, Fu LW, 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. 68 (19) (2008) 7905–7914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Robey RW, Steadman K, Polgar O, Morisaki K, Blayney M, Mistry P, Bates SE, Pheophorbide a is a specific probe for ABCG2 function and inhibition, Cancer Res. 64 (4) (2004) 1242–1246. [DOI] [PubMed] [Google Scholar]
  • [46].Wu CP, Hsiao SH, Su CY, Luo SY, Li YQ, Huang YH, Hsieh CH, Huang CW, Human ATP-binding cassette transporters ABCB1 and ABCG2 confer resistance to CUDC-101, a multi-acting inhibitor of histone deacetylase, epidermal growth factor receptor and human epidermal growth factor receptor 2, Biochem. Pharm. 92 (4) (2014) 567–576. [DOI] [PubMed] [Google Scholar]
  • [47].Anderson HA, Maylock CA, Williams JA, Paweletz CP, Shu H, Shacter E, Serum-derived protein S binds to phosphatidylserine and stimulates the phagocytosis of apoptotic cells, Nat. Immunol. 4 (1) (2003) 87–91. [DOI] [PubMed] [Google Scholar]
  • [48].Ambudkar SV, Drug-stimulatable ATPase activity in crude membranes of human MDR1-transfected mammalian cells, Methods Enzym. 292 (1998) 504–514. [DOI] [PubMed] [Google Scholar]
  • [49].Orlando BJ, Liao M, ABCG2 transports anticancer drugs via a closed-to-open switch, Nat. Commun. 11 (1) (2020) 2264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Wu CP, Hsiao SH, Huang YH, Hung LC, Yu YJ, Chang YT, Hung TH, Wu YS, Sitravatinib sensitizes ABCB1- and ABCG2-overexpressing multidrug-resistant cancer cells to chemotherapeutic drugs, Cancers 12 (1) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Mahon FX, Belloc F, Lagarde V, Chollet C, Moreau-Gaudry F, Reiffers J, Goldman JM, Melo JV, MDR1 gene overexpression confers resistance to imatinib mesylate in leukemia cell line models, Blood 101 (6) (2003) 2368–2373. [DOI] [PubMed] [Google Scholar]
  • [52].Hiwase DK, Saunders V, Hewett D, Frede A, Zrim S, Dang P, Eadie L, To LB, Melo J, Kumar S, Hughes TP, White DL, Dasatinib cellular uptake and efflux in chronic myeloid leukemia cells: therapeutic implications, Clin. Cancer Res. 14 (12) (2008) 3881–3888. [DOI] [PubMed] [Google Scholar]
  • [53].Eechoute K, Sparreboom A, Burger H, Franke RM, Schiavon G, Verweij J, Loos WJ, Wiemer EA, Mathijssen RH, Drug transporters and imatinib treatment: implications for clinical practice, Clin. Cancer Res. 17 (3) (2011) 406–415. [DOI] [PubMed] [Google Scholar]
  • [54].Tang C, Schafranek L, Watkins DB, Parker WT, Moore S, Prime JA, White DL, Hughes TP, Tyrosine kinase inhibitor resistance in chronic myeloid leukemia cell lines: investigating resistance pathways, Leuk. Lymphoma 52 (11) (2011) 2139–2147. [DOI] [PubMed] [Google Scholar]
  • [55].van Hoppe S, Jamalpoor A, Rood JJM, Wagenaar E, Sparidans RW, Beijnen JH, Schinkel AH, Brain accumulation of osimertinib and its active metabolite AZ5104 is restricted by ABCB1 (P-glycoprotein) and ABCG2 (breast cancer resistance protein), Pharm. Res. 146 (2019), 104297. [DOI] [PubMed] [Google Scholar]
  • [56].Li W, Tibben M, Wang Y, Lebre MC, Rosing H, Beijnen JH, Schinkel AH, P-glycoprotein (MDR1/ABCB1) controls brain accumulation and intestinal disposition of the novel TGF-beta signaling pathway inhibitor galunisertib, Int. J. Cancer, 2019. [DOI] [PubMed] [Google Scholar]
  • [57].Shi Z, Peng XX, Kim IW, Shukla S, Si QS, Robey RW, Bates SE, Shen T, Ashby CR Jr., Fu LW, Ambudkar SV, Chen ZS, Erlotinib (Tarceva, OSI-774) antagonizes ATP-binding cassette subfamily B member 1 and ATP-binding cassette subfamily G member 2-mediated drug resistance, Cancer Res. 67 (22) (2007) 11012–11020. [DOI] [PubMed] [Google Scholar]
  • [58].Wu CP, Hsiao SH, Murakami M, Lu MJ, Li YQ, Hsieh CH, Ambudkar SV, Wu YS, Tyrphostin RG14620 selectively reverses ABCG2-mediated multidrug resistance in cancer cell lines, Cancer Lett. 409 (2017) 56–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Zhang GN, Zhang YK, Wang YJ, Gupta P, Ashby CR Jr., Alqahtani S, Deng T, Bates SE, Kaddoumi A, Wurpel JND, Lei YX, Chen ZS, Epidermal growth factor receptor (EGFR) inhibitor PD153035 reverses ABCG2-mediated multidrug resistance in non-small cell lung cancer: in vitro and in vivo, Cancer Lett. 424 (2018) 19–29. [DOI] [PubMed] [Google Scholar]
  • [60].Wu CP, Lusvarghi S, Wang JC, Hsiao SH, Huang YH, Hung TH, Ambudkar SV, Avapritinib: a selective inhibitor of KIT and PDGFRalpha that reverses ABCB1 and ABCG2-mediated multidrug resistance in cancer cell lines, Mol. Pharm. 16 (7) (2019) 3040–3052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Wu CP, Hung TH, Hsiao SH, Huang YH, Hung LC, Yu YJ, Chang YT, Wang SP, Wu YS, Erdafitinib resensitizes ABCB1-overexpressing multidrug-resistant cancer cells to cytotoxic anticancer drugs, Cancers 12 (6) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Kartner N, Riordan JR, Ling V, Cell surface P-glycoprotein associated with multidrug resistance in mammalian cell lines, Science 221 (4617) (1983) 1285–1288. [DOI] [PubMed] [Google Scholar]
  • [63].Maliepaard M, van Gastelen MA, de Jong LA, Pluim D, van Waardenburg RC, Ruevekamp-Helmers MC, Floot BG, Schellens JH, Overexpression of the BCRP/MXR/ABCP gene in a topotecan-selected ovarian tumor cell line, Cancer Res. 59 (18) (1999) 4559–4563. [PubMed] [Google Scholar]
  • [64].Robey RW, Medina-Perez WY, Nishiyama K, Lahusen T, Miyake K, Litman T, Senderowicz AM, Ross DD, Bates SE, Overexpression of the ATP-binding cassette half-transporter, ABCG2 (MXR/BCRP/ABCP1), in flavopiridol-resistant human breast cancer cells, Clin. Cancer Res. 7 (1) (2001) 145–152. [PubMed] [Google Scholar]
  • [65].Bates SE, Medina-Perez WY, Kohlhagen G, Antony S, Nadjem T, Robey RW, Pommier Y, ABCG2 mediates differential resistance to SN-38 (7-ethyl-10-hydroxycamptothecin) and homocamptothecins, J. Pharm. Exp. Ther. 310 (2) (2004) 836–842. [DOI] [PubMed] [Google Scholar]
  • [66].Tiwari AK, Sodani K, Dai CL, Abuznait AH, Singh S, Xiao ZJ, Patel A, Talele TT, Fu L, Kaddoumi A, Gallo JM, Chen ZS, Nilotinib potentiates anticancer drug sensitivity in murine ABCB1-, ABCG2-, and ABCC10-multidrug resistance xenograft models, Cancer Lett. 328 (2) (2013) 307–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Wu CP, Murakami M, Hsiao SH, Liu TC, Yeh N, Li YQ, Hung TH, Wu YS, Ambudkar SV, SIS3, a specific inhibitor of Smad3 reverses ABCB1- and ABCG2-mediated multidrug resistance in cancer cell lines, Cancer Lett. 433 (2018) 259–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Wu CP, Lusvarghi S, Hsiao SH, Liu TC, Li YQ, Huang YH, Hung TH, Ambudkar SV, Licochalcone A selectively resensitizes ABCG2-overexpressing multidrug-resistant cancer cells to chemotherapeutic drugs, J. Nat. Prod. 83 (5) (2020) 1461–1472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Cuestas ML, Castillo AI, Sosnik A, Mathet VL, Downregulation of mdr1 and abcg2 genes is a mechanism of inhibition of efflux pumps mediated by polymeric amphiphiles, Bioorg. Med. Chem. Lett. 22 (21) (2012) 6577–6579. [DOI] [PubMed] [Google Scholar]
  • [70].Natarajan K, Bhullar J, Shukla S, Burcu M, Chen ZS, Ambudkar SV, Baer MR, The Pim kinase inhibitor SGI-1776 decreases cell surface expression of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) and drug transport by Pim-1-dependent and -independent mechanisms, Biochem. Pharm. 85 (4) (2013) 514–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Ozvegy C, Litman T, Szakacs G, Nagy Z, Bates S, Varadi A, Sarkadi B, Functional characterization of the human multidrug transporter, ABCG2, expressed in insect cells, Biochem. Biophys. Res. Commun. 285 (1) (2001) 111–117. [DOI] [PubMed] [Google Scholar]
  • [72].Ozvegy-Laczka C, Cserepes J, Elkind NB, Sarkadi B, Tyrosine kinase inhibitor resistance in cancer: role of ABC multidrug transporters, Drug Resist. Updates 8 (1–2) (2005) 15–26. [DOI] [PubMed] [Google Scholar]
  • [73].Brozik A, Hegedus C, Erdei Z, Hegedus T, Ozvegy-Laczka C, Szakacs G, Sarkadi B, Tyrosine kinase inhibitors as modulators of ATP binding cassette multidrug transporters: substrates, chemosensitizers or inducers of acquired multidrug resistance? Expert Opin. Drug Metab. Toxicol. 7 (5) (2011) 623–642. [DOI] [PubMed] [Google Scholar]
  • [74].Maia RC, Vasconcelos FC, Souza PS, Rumjanek VM, Towards comprehension of the ABCB1/P-glycoprotein role in chronic myeloid leukemia, Molecules 23 (1) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Beretta GL, Cassinelli G, Pennati M, Zuco V, Gatti L, Overcoming ABC transporter-mediated multidrug resistance: the dual role of tyrosine kinase inhibitors as multitargeting agents, Eur. J. Med. Chem. 142 (2017) 271–289. [DOI] [PubMed] [Google Scholar]
  • [76].Wu S, Fu L, Tyrosine kinase inhibitors enhanced the efficacy of conventional chemotherapeutic agent in multidrug resistant cancer cells, Mol. Cancer 17 (1) (2018) 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Wu CP, Hung TH, Lusvarghi S, Chu YH, Hsiao SH, Huang YH, Chang YT, Ambudkar SV, The third-generation EGFR inhibitor almonertinib (HS-10296) resensitizes ABCB1-overexpressing multidrug-resistant cancer cells to chemotherapeutic drugs, Biochem. Pharm. 188 (2021), 114516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Wu CP, Murakami M, Wu YS, Chi YC, Hsiao SH, Huang YH, Hung TH, Ambudkar SV, Branebrutinib (BMS-986195), a Bruton’s tyrosine kinase inhibitor, resensitizes P-glycoprotein-overexpressing multidrug-resistant cancer cells to chemotherapeutic agents, Front. Cell Dev. Biol. 9 (2021), 699571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Zeng F, Wang F, Zheng Z, Chen Z, Wah KK, Zhang H, Han Q, Fu L, Rociletinib (CO-1686) enhanced the efficacy of chemotherapeutic agents in ABCG2-overexpressing cancer cells in vitro and in vivo, Acta Pharm. Sin. B 10 (5) (2020) 799–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Lei ZN, Teng QX, Gupta P, Zhang W, Narayanan S, Yang DH, Wurpel JND, Fan YF, Chen ZS, Cabozantinib reverses topotecan resistance in human non-small cell lung cancer NCI-H460/TPT10 cell line and tumor xenograft model, Front. Cell Dev. Biol. 9 (2021), 640957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, Campos D, Lim R, Ding K, Clark G, Voskoglou-Nomikos T, Ptasynski M, Parulekar W, National G Cancer Institute of Canada Clinical Trials, Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group, J. Clin. Oncol. 25 (15) (2007) 1960–1966. [DOI] [PubMed] [Google Scholar]
  • [82].Yang ZY, Yuan JQ, Di MY, Zheng DY, Chen JZ, Ding H, Wu XY, Huang YF, Mao C, Tang JL, Gemcitabine plus erlotinib for advanced pancreatic cancer: a systematic review with meta-analysis, PLoS One 8 (3) (2013), e57528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B, Skarlos D, Campone M, Davidson N, Berger M, Oliva C, Rubin SD, Stein S, Cameron D, Lapatinib plus capecitabine for HER2-positive advanced breast cancer, N. Engl. J. Med. 355 (26) (2006) 2733–2743. [DOI] [PubMed] [Google Scholar]
  • [84].Cetin B, Benekli M, Turker I, Koral L, Ulas A, Dane F, Oksuzoglu B, Kaplan MA, Koca D, Boruban C, Yilmaz B, Sevinc A, Berk V, Uncu D, Harputluoglu H, Coskun U, Buyukberber S, Lapatinib plus capecitabine for HER2-positive advanced breast cancer: a multicentre study of Anatolian Society of Medical Oncology (ASMO), J. Chemother. 26 (5) (2014) 300–305. [DOI] [PubMed] [Google Scholar]
  • [85].Alemany R, Moura DS, Redondo A, Martinez-Trufero J, Calabuig S, Saus C, Obrador-Hevia A, Ramos RF, Villar VH, Valverde C, Vaz MA, Medina J, Felipe-Abrio I, Hindi N, Taron M, Martin-Broto J, Nilotinib as co-adjuvant treatment with doxorubicin in patients with sarcomas: a phase I trial of the Spanish Group for Research on Sarcoma, Clin. Cancer Res, 2018. [DOI] [PubMed] [Google Scholar]
  • [86].Mi YJ, Liang YJ, Huang HB, Zhao HY, Wu CP, Wang F, Tao LY, Zhang CZ, Dai CL, Tiwari AK, Ma XX, To KK, Ambudkar SV, Chen ZS, Fu LW, Apatinib (YN968D1) reverses multidrug resistance by inhibiting the efflux function of multiple ATP-binding cassette transporters, Cancer Res. 70 (20) (2010) 7981–7991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Sodani K, Tiwari AK, Singh S, Patel A, Xiao ZJ, Chen JJ, Sun YL, Talele TT, Chen ZS, GW583340 and GW2974, human EGFR and HER-2 inhibitors, reverse ABCG2- and ABCB1-mediated drug resistance, Biochem. Pharm. 83 (12) (2012) 1613–1622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Stewart CF, Leggas M, Schuetz JD, Panetta JC, Cheshire PJ, Peterson J, Daw N, Jenkins JJ 3rd, Gilbertson R, Germain GS, Harwood FC, Houghton PJ, Gefitinib enhances the antitumor activity and oral bioavailability of irinotecan in mice, Cancer Res. 64 (20) (2004) 7491–7499. [DOI] [PubMed] [Google Scholar]
  • [89].Leggas M, Panetta JC, Zhuang Y, Schuetz JD, Johnston B, Bai F, Sorrentino B, Zhou S, Houghton PJ, Stewart CF, Gefitinib modulates the function of multiple ATP-binding cassette transporters in vivo, Cancer Res. 66 (9) (2006) 4802–4807. [DOI] [PubMed] [Google Scholar]
  • [90].Shukla S, Wu CP, Ambudkar SV, Development of inhibitors of ATP-binding cassette drug transporters: present status and challenges, Expert Opin. Drug Metab. Toxicol. 4 (2) (2008) 205–223. [DOI] [PubMed] [Google Scholar]
  • [91].Libby E, Hromas R, Dismounting the MDR horse, Blood 116 (20) (2010) 4037–4038. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

36068781_ambudkar_sm

RESOURCES