1
|
Chen C, Hao Z, Chen J, Li S, Su Y, Jiang S, Ma L, Lv H, Pei X, Zhang P, Wang H, Yang G. Design, synthesis, and biological evaluation of C-12 modified ocotillol-type derivatives as novel P-glycoprotein modulators for overcoming multidrug resistance. Eur J Med Chem 2025; 294:117757. [PMID: 40382839 DOI: 10.1016/j.ejmech.2025.117757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
Ocotillol-type ginsenoside derivatives exhibit significant potential as modulators of P-glycoprotein (Pgp). To date, structural investigations of Ocotillol-type saponins have predominantly focused on modifications at the C-3 position of the A-ring, with limited exploration of the C-12 position on the C-ring. In this study, we designed and synthesized a series of C-12 modified ocotillol-type derivatives and assessed their efficacy in reversing multidrug resistance (MDR) in KBV cells. Most of the newly synthesized derivatives exhibited minimal cytotoxicity and potent MDR reversal capabilities. Notably, compound 9e emerged as the most effective agent in reversing tumor MDR in vitro, showing more than twice the potency of verapamil. Furthermore, 9e displayed high selectivity for Pgp, being 40- and 20-fold more effective than verapamil in inhibiting Rh123 efflux and enhancing doxorubicin sensitivity, respectively. Molecular docking analysis revealed that 9e possesses a unique T-shaped configuration that occupies the access channel of Pgp, obstructing the peristaltic extrusion mechanism of TM12 and TM9, thereby inhibiting the efflux function of Pgp. Overall, 9e represents a promising lead compound for the development of novel Pgp modulators to overcome MDR in cancer therapy.
Collapse
Affiliation(s)
- Cheng Chen
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Ziqian Hao
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Jiaxuan Chen
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Shuang Li
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Yongyuan Su
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Suwei Jiang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Lin Ma
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Hanqi Lv
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Xinjie Pei
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Peng Zhang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Hongbo Wang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China
| | - Gangqiang Yang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, 264005, China.
| |
Collapse
|
2
|
Ying TT, Hu HQ, Wu XW, Xu XL, Lv J, Zhang SN, Wang H, Hou W, Wei B, Rao GW. Optimized ebselen derivatives as novel potent Escherichia coli β-glucuronidase covalent allosteric inhibitors. Eur J Med Chem 2025; 290:117571. [PMID: 40168911 DOI: 10.1016/j.ejmech.2025.117571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
Gut microbial β-glucuronidase (GUS) plays a key role in metabolizing compounds and influencing disease and drug metabolism, highlighting the need for potent inhibitors to improve drug efficacy and intestinal health. To identify Escherichia coli β-glucuronidase (EcGUS) inhibitors, we designed and synthesized fifty 1,2-benzoselenazol-3-one (BSEA) derivatives using a bioisosterism strategy. Among these, twenty-five BSEA derivatives demonstrated greater inhibitory efficacy than the most potent known EcGUS inhibitor, amoxapine (AMX), with compound 49 showing the strongest activity, achieving an IC50 of 12.9 nM. Structure-inhibitory activity relationship analysis suggested that modifications such as adding benzene rings or nitrogenous heterocycles to the BSEA scaffold enhanced inhibitory activity, influenced by the type and position of substituents. The LC-MS analysis confirmed that compounds 31 and 49 covalently modify Cys197 in EcGUS, and additional covalent linkage of compound 49 was observed on Cys28 and Cys443. In addition, the jump dilution assays proved that compounds 31 was irreversible covalent inhibitors, and its kinetic parameter kinact/KI were determined to be 21292.9 M-1s-1. The compounds 49 was reversible covalent inhibitors and its apparent steady-state inhibition constant Ki∗app were determined to be 23.33 nM. Molecular docking predicted specific interactions, such as hydrogen bonds involving Se and the pyrazole NH of compound 49 with Cys28 and Cys449, which may contribute to its inhibitory action. This study reports the first discovery of covalent inhibitors for EcGUS, with optimized BSEA derivatives acting as novel allosteric covalent inhibitors, revealing structure-activity relationships and molecular determinants that establish their potential in drug development.
Collapse
Affiliation(s)
- Ti-Ti Ying
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hao-Qiang Hu
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiao-Wen Wu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xu-Liang Xu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jian Lv
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shu-Ning Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 201210, China
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China; Binjiang Institute of Artificial Intelligence, ZJUT, Hangzhou, 310051, China
| | - Wei Hou
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Guo-Wu Rao
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
3
|
Hou W, Zhou X, Yang Z, Xia H, Wang Y, Xu K, Hou S, Zhang S, Cui D, Ma P, Zhou W, Xu H. Multicomponent Reaction Integrating Selenium(II)-Nitrogen Exchange (SeNEx) Chemistry and Copper-Catalyzed Azide-Alkyne Cycloaddition (CuAAC). Angew Chem Int Ed Engl 2025; 64:e202500942. [PMID: 40000436 DOI: 10.1002/anie.202500942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 02/27/2025]
Abstract
Multicomponent reactions (MCRs) are powerful tools for rapidly constructing compound libraries with sufficient molecular diversity and complexity. Herein, to fully leverage a third aspect of molecular diversity enabled by the selenium-nitrogen exchange (SeNEx) reaction between alkynes and benzoselenazolones, a novel CuI-catalyzed three-component reaction has been successfully developed. This reaction integrates SeNEx with CuAAC click chemistry, enabling rapid and regioselective synthesis of 1,4,5-trisubstituted 5-seleno-1,2,3-triazoles with high atom economy and good to excellent yields (65 examples, 50%-95%). Notably, this MCR demonstrates excellent functional group tolerance and features modular, predictable, robust, mild reaction conditions, and operational simplicity (air and water compatibility). Extensive mechanism studies have revealed that this reaction proceeds by a unique SeNEx-CuAAC tandem reaction pathway, distinguishing it from conventional copper(I)-catalyzed interrupted click reactions. Importantly, a mononuclear σ-bound copper(I)-acetylide Cu1 was synthesized and confirmed to be an efficient catalyst for the SeNEx reaction. This discovery provides crucial mechanistic insights into the preferential reactivity of alkynyl groups toward SeNEx over CuAAC. Furthermore, preliminary biological activity screening identified compound 14 as a potent inhibitor of Escherichia coli β $\upbeta$ -glucuronidase (EcGUS), with an IC50 value of 3.16 µM. These findings underscore the significant potential of this MCR in synthetic chemistry, medical chemistry, and chemical biology.
Collapse
Affiliation(s)
- Wei Hou
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiaohui Zhou
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Zhikun Yang
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Huhui Xia
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yan Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Keren Xu
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shaoneng Hou
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shuning Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, School of Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, 201210, China
| | - Dongmei Cui
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Peixiang Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, School of Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, 201210, China
| | - Wei Zhou
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| |
Collapse
|
4
|
Moosavi F, Divar M, Khabnadideh S, Tavakkoli M, Mohabbati M, Saso L, Poustforoosh A, Firuzi O. Spiroindoline quinazolinedione derivatives as inhibitors of P-glycoprotein: potential agents for overcoming multidrug resistance in cancer therapy. Mol Divers 2025:10.1007/s11030-025-11150-5. [PMID: 40106127 DOI: 10.1007/s11030-025-11150-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
Multidrug resistance (MDR) presents a major challenge for effectiveness of chemotherapy. This study investigates the effectiveness of spiroindoline quinazolinediones in reversing MDR mediated by P-glycoprotein (P-gp) overexpression in cancer cells. A series of synthesized hybrid spiro[indoline-3,2'-quinazoline]-2,4'(3'H)-dione derivatives (compounds 5a-5l) were analyzed for their ability to enhance rhodamine 123 (Rhd123) accumulation in the MES-SA/DX5 cell line using flow cytometry. The MTT assay was also employed to evaluate the compounds' effectiveness in reversing drug resistance. Additionally, docking studies and molecular dynamics simulations were conducted to investigate the interaction of these compounds with the P-gp transporter. The Rhd123 accumulation assay in MDR cancer cells revealed that most compounds, in particular 5f, 5g, 5h, 5i, 5j, 5k, and 5l, exhibited significant potential as P-gp inhibitors. Among the tested derivatives, compounds 5g and 5l demonstrated the best effects, and increased Rhd123 accumulation up to 12.9 times compared to untreated cells. Additionally, compounds 5f through 5 l bearing methylbenzyl (5f), benzyl (5g), pentyl (5 ), p-bromobenzyl (5i), p-chlorobenzyl (5j), dichlorobenzyl (5k), and tert-butylbenzyl (5l) substituents on the isatin ring effectively restored sensitivity to doxorubicin at their non-toxic concentrations in resistant MES-SA/DX5 cells. Among these, compound 5l at 5 μM exhibited the highest inhibitory potential, and lowered doxorubicin's IC50 value 10.1 times compared to control. Moreover, in silico investigation identified the potential interactions of test compounds with critical residues of P-gp involved in its efflux function. Our study suggests that the synthesized spiroindoline quinazolinediones may have high potentials as agents capable of reversing MDR in cancer cells.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoumeh Divar
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soghra Khabnadideh
- School of Pharmacy, Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marjan Tavakkoli
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Mohabbati
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. Le Aldo Moro 5, 00185, Rome, Italy
| | - Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Hou W, Hou S, Gu Y, Zhang S, Ma P, Hu HY, Xu H. Selenium(II)-Nitrogen Exchange (SeNEx) Chemistry: A Good Chemistry Suitable for Nanomole-Scale Parallel Synthesis, DNA-encoded Library Synthesis and Bioconjugation. Chembiochem 2024; 25:e202400641. [PMID: 39379308 DOI: 10.1002/cbic.202400641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/10/2024]
Abstract
The continuous development of click reactions with new connecting linkage is crucial for advancing the frontiers of click chemistry. Selenium-nitrogen exchange (SeNEx) chemistry, a versatile chemistry in click chemistry, represents an all-encompassing term for nucleophilic substitution events that replace nitrogen at an electrophilic selenium(II) center, enabling the flexible and efficient assembly of linkages around a Se(II) core. Several SeNEx chemistries have been developed inspired by the biochemical reaction between Ebselen and cysteine residue, and demonstrated significant potential in on-plate nanomole-scale parallel synthesis, selenium-containing DNA-encoded library (SeDEL) synthesis, as well as peptide and protein bioconjugation. This concept aims to present the origins, advancements, and applications of selenium(II)-nitrogen exchange (SeNEx) chemistry while also outlining the potential directions for future research in this field.
Collapse
Affiliation(s)
- Wei Hou
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology Department, Hangzhou, 310014, China
| | - Shaoneng Hou
- College of Pharmaceutical Science & Green Pharmaceutical Collaborative Innovation Center of Yangtze River Delta Region, Zhejiang University of Technology Department, Hangzhou, 310014, China
| | - Yuang Gu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Shuning Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Peixiang Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Beijing Key Laboratory of Active Substances Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences, Peking UnionMedical College, Beijing, 100050, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| |
Collapse
|
6
|
Mamgain R, Mishra G, Kriti S, Singh FV. Organoselenium compounds beyond antioxidants. Future Med Chem 2024; 16:2663-2685. [PMID: 39711134 PMCID: PMC11734649 DOI: 10.1080/17568919.2024.2435254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/14/2024] [Indexed: 12/24/2024] Open
Abstract
Organoselenium chemistry has become a significant field due to its role in synthesizing numerous biologically active and therapeutic compounds. In early phase, researchers focused on designing organoselenium compounds with antioxidant properties and were quite successful. In last two decades, synthetic chemists shifted their focus toward synthesis of organoselenium compounds with biological properties, moving beyond their traditional antioxidant properties. The review includes synthesis and study of organo-selenium compounds as anticancer, antimicrobial, antiviral, antidiabetic, antithyroid, anti-inflammatory therapies, contributing to disease treatment. This review covers the synthesis and medicinal applications of synthetic organoselenium compounds over the past 10 years, thus making it a valuable resource for researchers in the field of medicinal chemistry.
Collapse
Affiliation(s)
- Ritu Mamgain
- Chemistry Division, School of Advanced Sciences (SAS), Vellore Institute of Technology - Chennai, Chennai, India
| | - Garima Mishra
- Department of Chemistry, Western Illinois University-Quad Cities, Moline, IL, USA
| | - Saumya Kriti
- Chemistry Division, School of Advanced Sciences (SAS), Vellore Institute of Technology - Chennai, Chennai, India
| | - Fateh V. Singh
- Chemistry Division, School of Advanced Sciences (SAS), Vellore Institute of Technology - Chennai, Chennai, India
| |
Collapse
|
7
|
Fu XJ, Li N, Wu J, Wang ZY, Liu RR, Niu JB, Taleb M, Yuan S, Liu HM, Song J, Zhang SY. Discovery of novel pyrazolo[1,5-a]pyrimidine derivatives as potent reversal agents against ABCB1-mediated multidrug resistance. Eur J Med Chem 2024; 277:116761. [PMID: 39151276 DOI: 10.1016/j.ejmech.2024.116761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
The P-glycoprotein (ABCB1)-mediated multidrug resistance (MDR) has emerged as a significant impediment to the efficacy of cancer chemotherapy in clinical therapy, which could promote the development of effective agents for MDR reversal. In this work, we reported the exploration of novel pyrazolo [1,5-a]pyrimidine derivatives as potent reversal agents capable of enhancing the sensitivity of ABCB1-mediated MDR MCF-7/ADR cells to paclitaxel (PTX). Among them, compound 16q remarkably increased the sensitivity of MCF-7/ADR cells to PTX at 5 μM (IC50 = 27.00 nM, RF = 247.40) and 10 μM (IC50 = 10.07 nM, RF = 663.44). Compound 16q could effectively bind and stabilize ABCB1, and does not affect the expression and subcellular localization of ABCB1 in MCF-7/ADR cells. Compound 16q inhibited the function of ABCB1, thereby increasing PTX accumulation, and interrupting the accumulation and efflux of the ABCB1-mediated Rh123, thus resulting in exhibiting good reversal effects. In addition, due to the potent reversal effects of compound 16q, the abilities of PTX to inhibit tubulin depolymerization, and induce cell cycle arrest and apoptosis in MCF-7/ADR cells under low-dose conditions were restored. These results indicate that compound 16q might be a promising potent reversal agent capable of revising ABCB1-mediated MDR, and pyrazolo [1,5-a]pyrimidine might represent a novel scaffold for the discovery of new ABCB1-mediated MDR reversal agents.
Collapse
Affiliation(s)
- Xiang-Jing Fu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Na Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Ji Wu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Zi-Yue Wang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Rui-Rui Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mohammad Taleb
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development Key, Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China; State Key Laboratory of Esophageal Cancer Prevention &Treatment, Zhengzhou, 450001, China.
| |
Collapse
|
8
|
Yu T, Zeng R, Guan Y, Pan B, Li HW, Gu J, Zheng PF, Qian Y, Ouyang Q. Discovery of new tricyclic spiroindole derivatives as potent P-glycoprotein inhibitors for reversing multidrug resistance enabled by a synthetic methodology-based library. RSC Med Chem 2024; 15:1675-1685. [PMID: 38784466 PMCID: PMC11110728 DOI: 10.1039/d4md00136b] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/23/2024] [Indexed: 05/25/2024] Open
Abstract
The discovery of novel and highly effective P-gp inhibitors is considered to be an effective strategy for overcoming tumor drug resistance. In this paper, a phenotypic screening via a self-constructed synthetic methodology-based library identified a new class of tricyclic spiroindole derivatives with excellent tumor multidrug resistance reversal activity. A stereospecific compound OY-103-B with the best reversal activity was obtained based on a detailed structure-activity relationship study, metabolic stability optimization and chiral resolution. For the VCR-resistant Eca109 cell line (Eca109/VCR), co-administration of 5.0 μM OY-103-B resulted in a reversal fold of up to 727.2, superior to the typical third-generation P-gp inhibitor tariquidar. Moreover, the compound inhibited the proliferation of Eca109/VCR cells in a concentration-dependent manner in plate cloning and flow cytometry. Furthermore, fluorescence substrate accumulation assay and chemotherapeutic drug reversal activity tests demonstrated that OY-103-B reversed tumor drug resistance via P-gp inhibition. In conclusion, this study provides a novel skeleton that inspires the design of new P-gp inhibitors, laying the foundation for the treatment of drug-resistant tumors.
Collapse
Affiliation(s)
- Tao Yu
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University Chongqing 400010 China
- Department of Medicinal Chemistry, Third Military Medical University Chongqing 400038 China
| | - Rong Zeng
- Department of Medicinal Chemistry, Third Military Medical University Chongqing 400038 China
- Department of Gastroenterology, Xinqiao Hospital, The Second Affiliated Hospital of Army Medical University (Third Military Medical University) Chongqing 400037 China
| | - Yu Guan
- College of Chemistry and Environmental Engineering, Sichuan University of Science and Engineering Zigong 643000 China
| | - Bin Pan
- Department of Medicinal Chemistry, Third Military Medical University Chongqing 400038 China
| | - Hong-Wei Li
- Department of Medicinal Chemistry, Third Military Medical University Chongqing 400038 China
| | - Jing Gu
- Department of Medicinal Chemistry, Third Military Medical University Chongqing 400038 China
| | - Peng-Fei Zheng
- Department of Medicinal Chemistry, Third Military Medical University Chongqing 400038 China
| | - Yan Qian
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University Chongqing 400010 China
| | - Qin Ouyang
- Department of Medicinal Chemistry, Third Military Medical University Chongqing 400038 China
| |
Collapse
|