IJMS 您所在的位置:网站首页 administrationreversal IJMS

IJMS

2023-03-28 10:10| 来源: 网络整理| 查看: 265

Next Article in Journal Transcriptional Regulatory Network of Plant Cadmium Stress Response Previous Article in Journal Recent Progress in the Identification of Early Transition Biomarkers from Relapsing-Remitting to Progressive Multiple Sclerosis Previous Article in Special Issue Lycorine Carbamate Derivatives for Reversing P-glycoprotein-Mediated Multidrug Resistance in Human Colon Adenocarcinoma Cells Journals Active Journals Find a Journal Proceedings Series Topics Information For Authors For Reviewers For Editors For Librarians For Publishers For Societies For Conference Organizers Open Access Policy Institutional Open Access Program Special Issues Guidelines Editorial Process Research and Publication Ethics Article Processing Charges Awards Testimonials Author Services Initiatives Sciforum MDPI Books Preprints.org Scilit SciProfiles Encyclopedia JAMS Proceedings Series About Overview Contact Careers News Blog Sign In / Sign Up Notice clear Notice

You are accessing a machine-readable page. In order to be human-readable, please install an RSS reader.

Continue Cancel clear

All articles published by MDPI are made immediately available worldwide under an open access license. No special permission is required to reuse all or part of the article published by MDPI, including figures and tables. For articles published under an open access Creative Common CC BY license, any part of the article may be reused without permission provided that the original article is clearly cited. For more information, please refer to https://www.mdpi.com/openaccess.

Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications.

Feature papers are submitted upon individual invitation or recommendation by the scientific editors and must receive positive feedback from the reviewers.

Editor’s Choice articles are based on recommendations by the scientific editors of MDPI journals from around the world. Editors select a small number of articles recently published in the journal that they believe will be particularly interesting to readers, or important in the respective research area. The aim is to provide a snapshot of some of the most exciting work published in the various research areas of the journal.

Journals Active Journals Find a Journal Proceedings Series Topics Information For Authors For Reviewers For Editors For Librarians For Publishers For Societies For Conference Organizers Open Access Policy Institutional Open Access Program Special Issues Guidelines Editorial Process Research and Publication Ethics Article Processing Charges Awards Testimonials Author Services Initiatives Sciforum MDPI Books Preprints.org Scilit SciProfiles Encyclopedia JAMS Proceedings Series About Overview Contact Careers News Blog Sign In / Sign Up Submit     Journals IJMS Volume 24 Issue 5 10.3390/ijms24054377 ijms-logo Submit to this Journal Review for this Journal Edit a Special Issue ► ▼ Article Menu Article Menu Academic Editor Doriano Fabbro Subscribe SciFeed Recommended Articles Related Info Links PubMed/Medline Google Scholar More by Authors Links on DOAJ Sun, W. Wong, I. L. K. Law, H. Ka-Wai Su, X. Chan, T. C. F. Sun, G. Yang, X. Wang, X. Chan, T. Hang Wan, S. Chow, L. M. C. on Google Scholar Sun, W. Wong, I. L. K. Law, H. Ka-Wai Su, X. Chan, T. C. F. Sun, G. Yang, X. Wang, X. Chan, T. Hang Wan, S. Chow, L. M. C. on PubMed Sun, W. Wong, I. L. K. Law, H. Ka-Wai Su, X. Chan, T. C. F. Sun, G. Yang, X. Wang, X. Chan, T. Hang Wan, S. Chow, L. M. C. /ajax/scifeed/subscribe Article Views Citations - Table of Contents Altmetric share Share announcement Help format_quote Cite question_answer Discuss in SciProfiles thumb_up ... Endorse textsms ... Comment Need Help? Support

Find support for a specific problem in the support section of our website.

Get Support Feedback

Please let us know what you think of our products and services.

Give Feedback Information

Visit our dedicated information section to learn more about MDPI.

Get Information clear JSmol Viewer clear first_page settings Order Article Reprints Font Type: Arial Georgia Verdana Font Size: Aa Aa Aa Line Spacing:    Column Width:    Background: Open AccessArticle In Vivo Reversal of P-Glycoprotein-Mediated Drug Resistance in a Breast Cancer Xenograft and in Leukemia Models Using a Novel, Potent, and Nontoxic Epicatechin EC31 by Wenqin Sun 1, Iris L. K. Wong 1, Helen Ka-Wai Law 2, Xiaochun Su 1, Terry C. F. Chan 1, Gege Sun 1, Xinqing Yang 1, Xingkai Wang 3, Tak Hang Chan 1,4, Shengbiao Wan 3,* and Larry M. C. Chow 1,* 1 State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China 2 Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong SAR, China 3 Laboratory for Marine Drugs and Bioproducts of Qingdao, National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China 4 Department of Chemistry, McGill University, Montreal, QC H3A 2K6, Canada * Authors to whom correspondence should be addressed. Int. J. Mol. Sci. 2023, 24(5), 4377; https://doi.org/10.3390/ijms24054377 Received: 9 January 2023 / Revised: 10 February 2023 / Accepted: 15 February 2023 / Published: 22 February 2023 (This article belongs to the Special Issue Natural Product-Derived Compounds for Targeting Multidrug Resistance in Cancer and Microorganisms) Download Download PDF Download PDF with Cover Download XML Download Epub Browse Figures Review Reports Versions Notes

Abstract: The modulation of P-glycoprotein (P-gp, ABCB1) can reverse multidrug resistance (MDR) and potentiate the efficacy of anticancer drugs. Tea polyphenols, such as epigallocatechin gallate (EGCG), have low P-gp-modulating activity, with an EC50 over 10 μM. In this study, we optimized a series of tea polyphenol derivatives and demonstrated that epicatechin EC31 was a potent and nontoxic P-gp inhibitor. Its EC50 for reversing paclitaxel, doxorubicin, and vincristine resistance in three P-gp-overexpressing cell lines ranged from 37 to 249 nM. Mechanistic studies revealed that EC31 restored intracellular drug accumulation by inhibiting P-gp-mediated drug efflux. It did not downregulate the plasma membrane P-gp level nor inhibit P-gp ATPase. It was not a transport substrate of P-gp. A pharmacokinetic study revealed that the intraperitoneal administration of 30 mg/kg of EC31 could achieve a plasma concentration above its in vitro EC50 (94 nM) for more than 18 h. It did not affect the pharmacokinetic profile of coadministered paclitaxel. In the xenograft model of the P-gp-overexpressing LCC6MDR cell line, EC31 reversed P-gp-mediated paclitaxel resistance and inhibited tumor growth by 27.4 to 36.1% (p < 0.001). Moreover, it also increased the intratumor paclitaxel level in the LCC6MDR xenograft by 6 fold (p < 0.001). In both murine leukemia P388ADR and human leukemia K562/P-gp mice models, the cotreatment of EC31 and doxorubicin significantly prolonged the survival of the mice (p < 0.001 and p < 0.01) as compared to the doxorubicin alone group, respectively. Our results suggested that EC31 was a promising candidate for further investigation on combination therapy for treating P-gp-overexpressing cancers. Keywords: multidrug resistance; MDR; P-glycoprotein; P-gp; epicatechin; EC; modulator 1. IntroductionMultidrug resistance (MDR) is one of the major challenges in chemotherapy. The overexpression of ATP-binding cassette (ABC) transporters on the plasma membrane is one of the mechanisms for causing MDR in cancer. ABC transporters being overexpressed in naive or posttreatment tumors can limit the effectiveness of chemotherapeutic agents. They can actively efflux a broad range of structurally different cytotoxic drugs out of the cancer cells and, thus, reduce the intracellular drug concentrations [1,2]. P-glycoprotein (P-gp, MDR1 or ABCB1) is the best-characterized and most widely studied ABC transporter [3]. It is a homodimeric protein with two transmembrane domains (TMDs) and two cytosolic ATP nucleotide-binding domains (NBDs) [1]. The two TMDs form a large cavity for substrate binding and transport. The two NBDs are involved in ATP binding and hydrolysis to support the efflux [1,4]. P-gp overexpression has been correlated with poor clinical outcomes for breast cancer [5,6], sarcoma [7,8], and leukemia [9,10,11]. Many conventional anticancer drugs are P-gp substrates, including anthracyclines (doxorubicin (DOX) and daunorubicin), vinca alkaloids (vincristine (VCR) and vinblastine), colchicine, epipodophyllotoxins (etoposide and teniposide), paclitaxel (PTX), and imatinib [12]. One approach to tackling P-gp-mediated cancer MDR is to inhibit the transport activity of P-gp and to restore the anticancer drug concentration in the tumor [13,14]. First- and second-generation P-gp inhibitors are P-gp substrates, and they can compete with other substrates for binding sites, therefore acting as competitive inhibitors. These two generations of inhibitors include verapamil, cyclosporine, quinidine, reserpine, dexverapamil (R-isomer of verapamil), and PSC833 (a cyclosporine A analogue) [15,16]. They all failed in their clinical trials because of their low potency and high toxicity induced by unpredictable pharmacokinetic interactions with the anticancer drugs [17,18]. Third-generation modulators were developed through quantitative structure–activity relationship (QSAR) studies and high-throughput screening, including XR-9576 (tariquidar) [19], LY335979 (zosuquidar) [20,21], and elacridar [22]. They are noncompetitive P-gp inhibitors and have a high affinity for P-gp [23]. They bind at sites other than the substrate binding site of P-gp [24]. They are highly selective for P-gp, potent, and safe in in vitro studies, but some of them also failed in their clinical trials [20]. One factor for this failure resulted from the fact that the above-mentioned clinical trials did not stratify the patients based on the P-gp expression level in the tumor and the nonideal pharmacokinetic profiles of the modulators [18,25]. MDR can also be caused by other ABC transporters, such as breast cancer resistance protein (BCRP) [26] or multidrug-resistance-associated protein 1 (MRP1) [27], which may also result in the failure of the clinical trials of the third-generation modulators. The future development of inhibitors of ABC transporters should focus on potency, specificity, and safety. Epigallocatechin gallate (EGCG) is a natural polyphenol and is the most abundant catechin found in green tea. Its P-gp-modulating activity was first reported in 2002 [28]. EGCG at 50 µM chemosensitized the P-gp-overexpressing cell line CHRC5 cells to vinblastine and restored its IC50 to the wild-type level. EGCG was a potential P-gp modulator for reversing MDR in cancer. Its low potency, however, precluded it from further development.To potentiate the P-gp-modulating activity of EGCG, we previously designed and synthesized a catechin library containing 78 members of methylated epigallocatechin (EGC), methylated gallocatechin (GC), methylated epicatechin (EC), and methylated catechin (C) [29,30]. A structure–activity relationship (SAR) study revealed that the replacement of all the -OH groups in the A, B, and D rings with -OMe groups could significantly improve its efficacy. In addition, the presence of a longer and more rigid oxycarbonylphenylcarbamoyl linker between ring D and C3 [30] was also important (Table 1). Four methylated catechin derivatives, (2R, 3R)-EGC23, (2R, 3S)-GC51, (2R, 3R)-EC31, and (2R, 3S)-C25, were the most potent inhibitors in reversing P-gp-mediated PTX resistance in the P-gp-overexpressing LCC6MDR cells. These 4 potent modulators (1 µM) reduced the IC50 of PTX by a relative fold (RF) of 41 to 85 (Table 1), which was significantly better than the parent compound, EGCG, with an RF of 1.2 at 10 µM [29,30]. Moreover, they were strong P-gp inhibitors but weak or nonexistent BCRP and MRP1 inhibitors (Table 1). In the present study, these four potent methylated catechin derivatives were further characterized by their mechanism, pharmacokinetics, toxicity, and in vivo antitumor activity. 2. Results 2.1. Methylated Catechin Derivatives Modulated P-gp-Mediated MDR In VitroIn our previous study, EGC23, GC51, EC31, and C25 were found to be potent P-gp inhibitors in vitro [29,30]. Here, they were further characterized in vitro and in vivo. Human breast cancer cell lines LCC6 and LCC6MDR, murine leukemia cell lines P388 and P388ADR, and human leukemia cell lines K562 and K562/P-gp were used. LCC6, P388, and K562 were the sensitive parental cell lines. LCC6MDR and K562/P-gp were P-gp transfectants. P388ADR was established through a stepwise selection with Adriamycin. It was found that LCC6MDR, K562/P-gp, and P388ADR cells overexpressed more total or plasma membrane P-gp than their parental cell lines (Figure 1A–C). The IC50 of DOX, PTX, and VCR was 6.2- to 168-fold higher in the resistant cell lines (Figure 1D). In addition, P-gp (red fluorescence) was found to be localized on the cell surface of the P-gp-overexpressing cell lines but was not detected on that of the parental cell lines (Figure 1E–G).We determined the effective concentration (EC50) of the four methylated catechin derivatives and EGCG in reversing P-gp-mediated drug resistance. EC50 was defined as the concentration at which the modulator can reduce the IC50 of an anticancer drug by half in the P-gp-overexpressing cell lines. EGCG displayed no P-gp-modulating activity, with an EC50 > 10,000 nM (Table 2). In contrast, the four methylated catechin derivatives had an EC50 of 93 to 385 nM for reversing DOX resistance, 82 to 496 nM for reversing PTX resistance, and 37 to 107 nM for reversing VCR resistance (Table 2). Overall, their EC50 values were at least 20- to 270-fold lower than that of EGCG, indicating that the O-methylation of all the rings and the rigid oxycarbonylphenylcarbamoyl and oxycarbonylvinyl linkers between ring D and C3 were crucial pharmacophores for modulating P-gp activity (Table 2). The P-gp-modulating activity of methylated catechin derivatives was further studied by measuring the EC50. EC50 was defined as the concentration of modulators at which it can reduce IC50 of anticancer drug of a cell line by 50%. Three P-gp-overexpressing cell lines were employed, including breast cancer cell line LCC6MDR, mouse leukemia cell line P388ADR, and human leukemia cell line K562/P-gp. The anticancer drugs tested included DOX, PTX, and VCR. All values were presented as mean ± standard error of mean. N = 3–6 independent experiments. 2.2. Methylated Catechin Derivatives Increased DOX Accumulation by Inhibiting the Transport Activity of P-gpDOX is a fluorescent P-gp substrate and can be used to study the function of P-gp. It was found that the accumulation of DOX in P-gp-overexpressing cell lines LCC6MDR, P388ADR, and K562/P-gp was 2.8-fold (p < 0.001), 4.3-fold (p < 0.001), and 3.0-fold (p < 0.001) lower than that in their parental cell lines, respectively (Figure 2A–C). When used at 0.5 or 1 µM, EGC23, GC51, EC31, and C25 could restore DOX accumulation by 1.5 to 2.5 fold in LCC6MDR cells (Figure 2A), by 2.2 to 4.7 fold in P388ADR cells (Figure 1B), and by 1.6 to 2.8 fold in K562/P-gp cells, respectively (Figure 2C). In contrast, EGCG at 1 µM had no effect on DOX accumulation.We investigated if methylated catechin derivatives could inhibit P-gp-mediated drug efflux in P-gp-overexpressing cells. P388ADR cells were preincubated with DOX and were then resuspended in DOX-free medium with or without 1 μM EGC23, GC51, EC31, and C25. Without a modulator, intracellularly, the DOX level was maintained at 80% and 11% after 180 min for P388 and P388ADR cells, respectively (Figure 2D). When 1 μM EGC23, GC51, EC31, and C25 were included, the DOX efflux rate was significantly reduced. After 180 min of efflux, the intracellular DOX level could be maintained at 61% (p < 0.001), 59% (p < 0.01), 44 % (p < 0.001), and 37%, respectively (Figure 2D). The above results demonstrated that the reversal of DOX resistance by methylated catechin derivatives was due to an inhibition of P-gp-mediated drug efflux, leading to increased drug accumulation and, thus, restoring the drug sensitivity. 2.3. Methylated EC31 and C25 Did Not Inhibit P-gp ATPase ActivityEC31 and C25 did not affect the plasma membrane level of P-gp [29,30]. The efflux activity of P-gp is driven by ATP hydrolysis. Here, we investigated whether EC31 and C25 could affect P-gp ATPase activity. Verapamil, a well-known stimulator of P-gp ATPase activity, could increase P-gp ATPase activity in a dose-dependent manner, increasing the activity from 1.2 fold at 10 µM to 3.8 fold at 100 µM (Figure 3A). In contrast, EC31 and C25 were weak stimulators of P-gp ATPase, with up to a 2.1- to 1.6-fold increase when a 70 µM modulator was used (Figure 3A). EGCG could slightly increase P-gp ATPase activity by 1.4 fold at 10 µM and then decrease it to 0.7 fold at 70 µM (Figure 3A). These results suggested that the inhibition of the P-gp efflux function by EC31 and C25 was not due to the inhibition of ATPase activity. 2.4. Methylated EC31 Was Not a P-gp Transport SubstrateP-gp is a transporter with a wide substrate specificity. EC31 is a weak stimulator of P-gp ATPase (Figure 3A). We investigated whether EC31 was a substrate of P-gp by analyzing its intracellular retention level. It was found that EC31 was retained in a similar level in both LCC6 and LCC6MDR cells at all the concentrations tested (0.1, 1, 10, and 50 µM) (Figure 3B). For PTX, a known P-gp substrate, LCC6 accumulated 2.3- and 3.4-fold (p < 0.001) more PTX than LCC6MDR cells accumulated when 1 or 5 µM PTX was used (Figure 3B). Similar observations were found in K562 and K562/P-gp cells (Figure 3C). In the EC31 efflux assay, K562 and K562/P-gp displayed similar efflux rates. After 150 min of incubation, there was about 23% and 16% of EC31 remaining in both cell lines (Figure 3D). These results suggested that EC31 was not a transport substrate of P-gp. 2.5. Pharmacokinetic (PK) Study of Methylated Catechin Derivatives and Their Effects on the PK of PTX in MicePrior to the in vivo efficacy experiment, a PK study of EGC23, GC51, EC31, and C25 was performed using either intravenous (10 mg/kg i.v.) or intraperitoneal (30 mg/kg i.p.) injection routes (Figure 4A–D). The detailed PK profile was summarized in Figure 4E. With an i.p. injection at 30 mg/kg, the plasma levels of GC51, EC31, and C25 exceeded their corresponding in vitro EC50 values for over 18 h, whereas EGC23 lasted for 12 h, yielding a high bioavailability ranging from 63% to 82% after the dose normalization of AUCi.p. to AUCi.v. (Figure 4A–E).P-gp was widely distributed in multiple tissues and organs, including the gut, brain, kidney, liver, and placenta [31]. Therefore, it can affect the pharmacological behavior of drugs by regulating drug absorption, distribution, and elimination. Here, the plasma level of PTX with or without the coadministration of GC51, EC31, or C25 was determined up to 420 min after its administration (12 mg/kg i.v.) (Figure 4F). The AUC0–420min of PTX alone was 400,972 ng-min/mL. The coadministration of EC31 slightly increased the AUC0–420min of PTX to 478,526 ng-min/mL, but it was without statistical significance (p > 0.05) (Figure 4F). C25 or GC51 at 30 mg/kg i.p. significantly increased the AUC0–420min of PTX by 1.5 fold (** p < 0.01) and 1.6 fold (** p < 0.01), respectively (Figure 4F). EC31 was selected for an in vivo efficacy study because of its high potency and specificity in vitro [30], high bioavailability (82%), long duration of keeping its plasma level above its in vitro EC50 (>18 h), and insignificant effect on the bioavailability of PTX. 2.6. EC31 Alone and Its Combination with PTX Did Not Induce Toxicity in BALB/c MiceA toxicity study of EC31 (30 or 60 mg/kg i.p.) with or without the coadministration of PTX (12 mg/kg i.v.) was performed, and the injections were administered to BALB/c mice 12 times every other day. It was found that the repeated administration of EC31 (30 mg/kg) alone did not cause any mortality or toxicity symptoms (Figure 5A). The repeated coadministration of EC31 (either 30 or 60 mg/kg) and PTX induced a small body weight loss which was not greater than 15% for more than 3 consecutive days, and no toxicity symptoms were observed. The mice regained their body weight after the treatment was completed (Figure 5A). This result suggested that the coadministration of PTX with EC31 was well tolerated by the BALB/c mice. Therefore, the i.p. administration of EC31 at 30 or 60 mg/kg would be used in a subsequent efficacy study. 2.7. EC31 Increased the Intratumor PTX Concentration and Reversed the P-gp-Mediated PTX Resistance in the LCC6MDR Tumor Xenograft ModelAfter optimizing the administration route and dosage of EC31, we investigated whether the coadministration of EC31 could inhibit P-gp transport activity in the LCC6MDR xenograft, thereby enhancing the intratumor PTX level. When EC31 was administered alone (30 mg/kg i.p.), it could be maintained inside the LCC6MDR xenograft above its in vitro EC50 (94 nM) for 420 min (Figure 5B), suggesting that EC31 was stable in the plasma and could be continuously delivered to the tumor. When EC31 (30 mg/kg i.p.) and PTX (12 mg/kg i.v.) were coadministered, EC31 could increase the intratumor PTX level by 6 fold (*** p < 0.001) at 180 min as compared to PTX alone (Figure 5B). After 300 min, the intratumor level of EC31 was reduced to 94 nM, and the intratumor level of PTX was the same with or without EC31, suggesting that the first 300 min postadministration were an effective period for EC31 to increase the intratumor PTX concentration. This result suggested that EC31 should be administered twice a day to maintain a sufficient intratumor level of coadministered PTX. According to our established xenograft model of LCC6MDR, the injection of PTX alone (12 mg/kg i.v.) could inhibit the tumor growth of LCC6 but not that of LCC6MDR [32]. Here, we tested whether EC31 could reverse P-gp-mediated PTX resistance in the LCC6MDR xenograft model (Figure 5C). The treatment was given every other day 14 times (q2d x 14) from day 0 to day 26. There were four treatment groups, namely (1) the solvent control, (2) PTX alone (12 mg/kg i.v.), (3) EC31 (30 mg/kg i.p. at −1 h) plus PTX (12 mg/kg i.v.) plus EC31 (30 mg/kg i.p. at +5 h), and (4) EC31 (60 mg/kg i.p. at −1 h) plus PTX (12 mg/kg i.v.). The solvent control (group 1) and PTX alone (group 2) groups exhibited similar tumor growth rates (Figure 5C). Both of the combination treatment groups (groups 3 and 4) displayed promising efficacies and significantly suppressed the tumor growth of LCC6MDR. On day 30, combination treatment group 3 resulted in a 1.6-fold (*** p < 0.001) and 1.5-fold (** p < 0.01) reduction in tumor volume and tumor weight as compared to PTX alone (12 mg/kg i.v.) (Figure 5D). Combination treatment group 4 resulted in a 1.4-fold (*** p < 0.001 and ** p < 0.01) reduction in tumor volume and tumor weight as compared to PTX alone (12 mg/kg i.v.) (Figure 5D). The tumor doubling times in combination treatment groups 3 and 4 were 15.0 (* p < 0.05) and 12.4 days, which were significantly longer than that in the PTX alone group (11.1 days) (Figure 5D). No animal deaths were found in any of the four treatment groups (Figure 5D). These data suggested that EC31 could increase the intratumor PTX level by inhibiting the transport activity of P-gp in the xenograft, thereby chemosensitizing the tumor cells to PTX. 2.8. EC31 Reversed the DOX Resistance in the Murine Leukemia P388ADR and Human Leukemia K562/P-gp ModelsWe also tested the in vivo efficacy of EC31 in reversing P-gp-mediated DOX resistance in the murine leukemia P388ADR model in B6D2F1 mice (Figure 6A). It was found that the untreated group possessed the shortest median survival of 13.0 days. DOX (3 mg/kg i.p.) alone prolonged the median survival to 16.5 days. The cotreatment of EC31 (60 mg/kg i.p.) and DOX (3 mg/kg i.p.) extended the median survival to 21.5 days (Figure 6A), resulting in an increase in lifespan (ILS) of 30.3% (*** p < 0.001) compared to DOX alone (Figure 6A). The results suggested that EC31 could potentially modulate P-gp-mediated DOX resistance in the murine leukemia P388ADR model and could prolong the lifespan. To determine the in vivo efficacy of EC31 in reversing DOX resistance in the human leukemia K562/P-gp model, the survival time of leukemia-bearing NOD/SCID mice was monitored. The median survival time of the untreated group, the DOX alone (0.9 mg/kg i.v.) group, and the cotreatment (EC31 30mg/ kg i.p. + DOX 0.9 mg/kg i.v.) group was 29.0, 32.0, and 37.0 days, respectively (Figure 6B). The DOX alone group had a 10.3% ILS (p > 0.05) compared to the untreated group. The cotreatment group could prolong the animal survival by 5 days compared to the DOX alone group, representing a 15.6% ILS (** p < 0.01) (Figure 6B). This result suggested that the DOX alone treatment could not prolong the animal survival. EC31 could potentiate DOX in treating the K562/P-gp leukemia-bearing mice and could, finally, prolong the animal survival. 3. DiscussionEGCG is the most abundant polyphenol in green tea. Its P-gp-modulating activity was first reported in 2002 [28]. EGCG at 50 µM chemosensitized P-gp-overexpressing cell line CHRC5 cells to vinblastine and lowered its IC50 to the wild-type level. Despite its potential to reverse MDR in cancer, its low potency limited it from further development. The modification of the OH groups to methoxy groups at the 3′ position of the B ring (EGCG-3′OMe) can improve the P-gp-modulating activity of EGCG by 2 fold [33].We previously demonstrated that it was possible to modify EGCG to other novel catechin analogs to improve its P-gp-modulating activity. There were three important pharmacophores that were needed for the P-gp-modulating activity of EGCG, including (1) replacing all the –OH groups in the A, B, and D rings with -OMe groups, (2) conjugating the rigid oxycarbonylphenylcarbamoyl linker between ring D and C3, and (3) substituting the B ring with dimethoxylation rather than trimethoxylation [29,30]. Comparatively, stereochemistry had less of an effect on the P-gp-modulating activity of catechins [30]. EGCG displayed no P-gp-modulating activity even at 10 µM, with an RF of 1.2 (Table 1). After structural modification, (2R, 3R)-cis-methylated EC31 and (2R, 3S)-trans-methylated C25 were much more potent, with an RF of 69 to 85 at 1 µM (Table 1). They were nontoxic to the fibroblast cells, with an IC50 of >100 µM, and were highly selective for the P-gp transporter [29,30]. Here, we demonstrated that they could reverse drug resistance towards a panel of anticancer drugs, including DOX, PTX, and VCR, in three P-gp-overexpressing cell lines (EC50 = 93 to 260 nM for reversing DOX resistance, EC50 = 91 to 249 nM for reversing PTX resistance, and EC50 = 37 to 60 nM for reversing VCR resistance) (Table 2). They were at least 38- to 270-fold more potent than the parent compound EGCG. The mechanistic study demonstrated that the reversal of P-gp-mediated drug resistance by these methylated catechin derivatives was due to the inhibition of the efflux activity of P-gp (Figure 2D), restoring drug accumulation to a cytotoxic level (Figure 2A–C). They did not decrease the P-gp level at the plasma membrane [30] nor inhibit the P-gp ATPase activity (Figure 3A) to enhance drug retention. In the P-gp ATPase assay, EC31 and C25 were found to be weak stimulators of P-gp ATPase, with 1.6- to 2.0-fold stimulation at 50 µM. However, their stimulatory activities were not as strong as that of the well-known stimulator of P-gp ATPase, verapamil, which stimulated ATPase by about 3.2-fold (Figure 3A). Verapamil is a known P-gp transport substrate and worked as a competitive inhibitor to reverse MDR [34]. In the EC31 accumulation study, a similar amount of intracellular EC31 was detected in the cells with or without the overexpression of P-gp (Figure 3B,C), suggesting that EC31 was not a transport substrate of P-gp and did not work as a competitive inhibitor to reverse P-gp-mediated drug resistance. Tariquidar is known to be a nonsubstrate of P-gp, yet it can act as a noncompetitive inhibitor and stimulate P-gp ATPase activity by 10 fold [35,36]. Another example is the P-gp antibody MRK16 which can bind to the epitope at the extracellular surface, thereby simultaneously blocking drug efflux and stimulating ATPase by 2 fold [37]. The binding of tariquidar or antibody MRK16 may lock P-gp in an outward-facing conformation and hold the two NBDs in close proximity, causing continuous ATP hydrolysis [23]. With such a locked conformation, P-gp would have a reduced affinity towards the substrates. Our results suggested that EC31 was not a substrate of P-gp yet was able to stimulate P-gp ATPase activity, which is similar to tariquidar. One major drawback of competitive inhibitors is that a higher dosage is needed, which would cause unpredictable side effects or toxicity. The EC50 of EC31 for reversing PTX resistance in LCC6MDR was 94 nM, which was about 4.7-fold lower than that of verapamil (EC50 = 446 nM) [29].The pharmacokinetics of methylated catechin derivatives EGC23, GC51, EC31, and C25 were evaluated in mice (Figure 4). EC31 was selected for the in vivo efficacy study because it exhibited 82% plasma bioavailability after i.p. injection at 30 mg/kg (Figure 4C) and because its plasma level could be maintained above its in vitro EC50 (94 nM) for longer than 18 h (Figure 4C). High bioavailability in the plasma and the long residence time inside the body implied that EC31 would be sufficiently exposed to the tumor for its activity. One of the obstacles for developing the P-gp modulators in clinic was the toxicity induced by the drug–drug interactions (DDI) between anticancer drugs and the modulators [17,18,25]. Here, the cotreatment of EC31 and PTX was found to have no significant effect on the plasma PTX level in vivo (Figure 4F). This was consistent with the observation that EC31 (30 or 60 mg/kg i.p.) together with PTX (12 mg/kg i.v.) in vivo did not lead to a significant body weight reduction or significant animal deaths when compared with PTX alone (Figure 5A).The in vivo efficacy of EC31 to reverse P-gp-mediated drug resistance was investigated in three animal cancer models, including a human breast cancer tumor xenograft LCC6MDR, a human leukemia K562/P-gp model, and a murine leukemia P388ADR model. Cotreatment (EC31 30 mg/kg i.p. + PTX 12 mg/kg i.v. + EC31 30 mg/kg i.p.) exhibited potent P-gp-inhibitory activity and restored the antitumor activity of PTX in the LCC6MDR xenograft (Figure 5C). This combination significantly reduced the tumor volume by 1.6 fold (*** p < 0.001) (Figure 5C,D) and the tumor weight by 1.5 fold (** p < 0.01) (Figure 5D) compared to PTX alone. Moreover, EC31 at 30 mg/kg i.p. was demonstrated to significantly increase the intratumor PTX levels by 6 fold (*** p < 0.001) at 180 min postadministration (Figure 5B) as compared to PTX alone. In both the murine leukemia P388ADR and human leukemia K562/P-gp models, cotreatment (EC31 + DOX) significantly prolonged the survival of the mice, with an ILS of 30.3% (*** p < 0.001) and 15.6% (** p


【本文地址】

公司简介

联系我们

今日新闻

    推荐新闻

    专题文章
      CopyRight 2018-2019 实验室设备网 版权所有