1
|
Lin BH, Li YC, Murakami M, Wu YS, Huang YH, Hung TH, Ambudkar SV, Wu CP. Epertinib counteracts multidrug resistance in cancer cells by antagonizing the drug efflux function of ABCB1 and ABCG2. Biomed Pharmacother 2024; 180:117542. [PMID: 39388999 DOI: 10.1016/j.biopha.2024.117542] [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: 05/16/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
A significant hurdle in cancer treatment arises from multidrug resistance (MDR), often due to overexpression of ATP-binding cassette (ABC) transporters like ABCB1 and/or ABCG2 in cancer cells. These transporters actively diminish the efficacy of cytotoxic drugs by facilitating ATP hydrolysis-dependent drug efflux and reducing intracellular drug accumulation in cancer cells. Addressing multidrug-resistant cancers poses a significant challenge due to the lack of approved treatments, prompting the exploration of alternative avenues like drug repurposing (also referred to as drug repositioning) of molecularly targeted agents to reverse MDR-mediated by ABCB1 and/or ABCG2 in multidrug-resistant cancer cells. Epertinib, a potent inhibitor of EGFR and HER2 currently in clinical trials for solid tumors, was investigated for its potential to resensitize ABCB1- and ABCG2-overexpressing multidrug-resistant cancer cells to chemotherapeutic agents. Our findings reveal that at sub-toxic, submicromolar concentrations, epertinib restores the sensitivity of multidrug-resistant cancer cells to cytotoxic drugs in a concentration-dependent manner. The results demonstrate that epertinib enhances drug-induced apoptosis in these cancer cells by impeding the drug-efflux function of ABCB1 and ABCG2 without altering their expression. ATPase activity and molecular docking were employed to reveal potential interaction sites between epertinib and the drug-binding pockets of ABCB1 and ABCG2. In summary, our study demonstrates an additional pharmacological capability of epertinib against the activity of ABCB1 and ABCG2. These findings suggest that incorporating epertinib into combination therapy could be advantageous for a specific patient subset with tumors exhibiting high levels of ABCB1 or ABCG2, warranting further exploration.
Collapse
Affiliation(s)
- Bing-Huan Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Yen-Ching Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Megumi Murakami
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan.
| | - Yang-Hui Huang
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan.
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan; Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung 20401, Taiwan.
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | - Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan; Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
2
|
Xu H, Russell SN, Steiner K, O'Neill E, Jones KI. Targeting PI3K-gamma in myeloid driven tumour immune suppression: a systematic review and meta-analysis of the preclinical literature. Cancer Immunol Immunother 2024; 73:204. [PMID: 39105848 PMCID: PMC11303654 DOI: 10.1007/s00262-024-03779-2] [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: 12/07/2023] [Accepted: 07/11/2024] [Indexed: 08/07/2024]
Abstract
The intricate interplay between immune and stromal cells within the tumour microenvironment (TME) significantly influences tumour progression. Myeloid cells, including tumour-associated macrophages (TAMs), neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs), contribute to immune suppression in the TME (Nakamura and Smyth in Cell Mol Immunol 17(1):1-12 (2020). https://doi.org/10.1038/s41423-019-0306-1 ; DeNardo and Ruffell in Nat Rev Immunol 19(6):369-382 (2019). https://doi.org/10.1038/s41577-019-0127-6 ). This poses a significant challenge for novel immunotherapeutics that rely on host immunity to exert their effect. This systematic review explores the preclinical evidence surrounding the inhibition of phosphoinositide 3-kinase gamma (PI3Kγ) as a strategy to reverse myeloid-driven immune suppression in solid tumours. EMBASE, MEDLINE, and PubMed databases were searched on 6 October 2022 using keyword and subject heading terms to capture relevant studies. The studies, focusing on PI3Kγ inhibition in animal models, were subjected to predefined inclusion and exclusion criteria. Extracted data included tumour growth kinetics, survival endpoints, and immunological responses which were meta-analysed. PRISMA and MOOSE guidelines were followed. A total of 36 studies covering 73 animal models were included in the review and meta-analysis. Tumour models covered breast, colorectal, lung, skin, pancreas, brain, liver, prostate, head and neck, soft tissue, gastric, and oral cancer. The predominant PI3Kγ inhibitors were IPI-549 and TG100-115, demonstrating favourable specificity for the gamma isoform. Combination therapies, often involving chemotherapy, radiotherapy, immune checkpoint inhibitors, biological agents, or vaccines, were explored in 81% of studies. Analysis of tumour growth kinetics revealed a statistically significant though heterogeneous response to PI3Kγ monotherapy, whereas the tumour growth in combination treated groups were more consistently reduced. Survival analysis showed a pronounced increase in median overall survival with combination therapy. This systematic review provides a comprehensive analysis of preclinical studies investigating PI3Kγ inhibition in myeloid-driven tumour immune suppression. The identified studies underscore the potential of PI3Kγ inhibition in reshaping the TME by modulating myeloid cell functions. The combination of PI3Kγ inhibition with other therapeutic modalities demonstrated enhanced antitumour effects, suggesting a synergistic approach to overcome immune suppression. These findings support the potential of PI3Kγ-targeted therapies, particularly in combination regimens, as a promising avenue for future clinical exploration in diverse solid tumour types.
Collapse
Affiliation(s)
- Haonan Xu
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK
| | - Keaton Ian Jones
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
3
|
Zhang M, Zeng X, She M, Dong X, Chen J, Xiong Q, Qiu G, Yang S, Li X, Ren G. FRAX486, a PAK inhibitor, overcomes ABCB1-mediated multidrug resistance in breast cancer cells. Braz J Med Biol Res 2024; 57:e13357. [PMID: 38958364 PMCID: PMC11221864 DOI: 10.1590/1414-431x2024e13357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/06/2024] [Indexed: 07/04/2024] Open
Abstract
The overexpression of P-glycoprotein (P-gp/ABCB1) is a leading cause of multidrug resistance (MDR). Hence, it is crucial to discover effective pharmaceuticals that counteract ABCB1-mediated multidrug resistance. FRAX486 is a p21-activated kinase (PAK) inhibitor. The objective of this study was to investigate whether FRAX486 can reverse ABCB1-mediated multidrug resistance, while also exploring its mechanism of action. The CCK8 assay demonstrated that FRAX486 significantly reversed ABCB1-mediated multidrug resistance. Furthermore, western blotting and immunofluorescence experiments revealed that FRAX486 had no impact on expression level and intracellular localization of ABCB1. Notably, FRAX486 was found to enhance intracellular drug accumulation and reduce efflux, resulting in the reversal of multidrug resistance. Docking analysis also indicated a strong affinity between FRAX486 and ABCB1. This study highlights the ability of FRAX486 to reverse ABCB1-mediated multidrug resistance and provides valuable insights for its clinical application.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Xiaoqi Zeng
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Meiling She
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xingduo Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, USA
| | - Jun Chen
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Qingquan Xiong
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Guobin Qiu
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Shuyi Yang
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Xiangqi Li
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, China
| | - Guanghui Ren
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
4
|
Yang S, Zhang B, Zhao X, Zhang M, Zhang M, Cui L, Zhang L. Enhanced Efficacy against Drug-Resistant Tumors Enabled by Redox-Responsive Mesoporous-Silica-Nanoparticle-Supported Lipid Bilayers as Targeted Delivery Vehicles. Int J Mol Sci 2024; 25:5553. [PMID: 38791591 PMCID: PMC11122197 DOI: 10.3390/ijms25105553] [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: 04/09/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Multidrug resistance (MDR) is frequently induced after long-term exposure to reduce the therapeutic effect of chemotherapeutic drugs, which is always associated with the overexpression of efflux proteins, such as P-glycoprotein (P-gp). Nano-delivery technology can be used as an efficient strategy to overcome tumor MDR. In this study, mesoporous silica nanoparticles (MSNs) were synthesized and linked with a disulfide bond and then coated with lipid bilayers. The functionalized shell/core delivery systems (HT-LMSNs-SS@DOX) were developed by loading drugs inside the pores of MSNs and conjugating with D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) and hyaluronic acid (HA) on the outer lipid surface. HT-LMSNs-SS and other carriers were characterized and assessed in terms of various characteristics. HT-LMSNs-SS@DOX exhibited a dual pH/reduction responsive drug release. The results also showed that modified LMSNs had good dispersity, biocompatibility, and drug-loading capacity. In vitro experiment results demonstrated that HT-LMSNs-SS were internalized by cells and mainly by clathrin-mediated endocytosis, with higher uptake efficiency than other carriers. Furthermore, HT-LMSNs-SS@DOX could effectively inhibit the expression of P-gp, increase the apoptosis ratios of MCF-7/ADR cells, and arrest cell cycle at the G0/G1 phase, with enhanced ability to induce excessive reactive oxygen species (ROS) production in cells. In tumor-bearing model mice, HT-LMSNs-SS@DOX similarly exhibited the highest inhibition activity against tumor growth, with good biosafety, among all of the treatment groups. Therefore, the nano-delivery systems developed herein achieve enhanced efficacy towards resistant tumors through targeted delivery and redox-responsive drug release, with broad application prospects.
Collapse
Affiliation(s)
- Shuoye Yang
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (B.Z.); (X.Z.); (M.Z.); (M.Z.); (L.C.); (L.Z.)
- Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou 450001, China
| | - Beibei Zhang
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (B.Z.); (X.Z.); (M.Z.); (M.Z.); (L.C.); (L.Z.)
| | - Xiangguo Zhao
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (B.Z.); (X.Z.); (M.Z.); (M.Z.); (L.C.); (L.Z.)
| | - Mengwei Zhang
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (B.Z.); (X.Z.); (M.Z.); (M.Z.); (L.C.); (L.Z.)
| | - Mengna Zhang
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (B.Z.); (X.Z.); (M.Z.); (M.Z.); (L.C.); (L.Z.)
| | - Lan Cui
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (B.Z.); (X.Z.); (M.Z.); (M.Z.); (L.C.); (L.Z.)
- Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou 450001, China
| | - Lu Zhang
- School of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China; (B.Z.); (X.Z.); (M.Z.); (M.Z.); (L.C.); (L.Z.)
- Key Laboratory of Functional Molecules for Biomedical Research, Zhengzhou 450001, China
| |
Collapse
|
5
|
Albadari N, Xie Y, Li W. Deciphering treatment resistance in metastatic colorectal cancer: roles of drug transports, EGFR mutations, and HGF/c-MET signaling. Front Pharmacol 2024; 14:1340401. [PMID: 38269272 PMCID: PMC10806212 DOI: 10.3389/fphar.2023.1340401] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
In 2023, colorectal cancer (CRC) is the third most diagnosed malignancy and the third leading cause of cancer death worldwide. At the time of the initial visit, 20% of patients diagnosed with CRC have metastatic CRC (mCRC), and another 25% who present with localized disease will later develop metastases. Despite the improvement in response rates with various modulation strategies such as chemotherapy combined with targeted therapy, radiotherapy, and immunotherapy, the prognosis of mCRC is poor, with a 5-year survival rate of 14%, and the primary reason for treatment failure is believed to be the development of resistance to therapies. Herein, we provide an overview of the main mechanisms of resistance in mCRC and specifically highlight the role of drug transports, EGFR, and HGF/c-MET signaling pathway in mediating mCRC resistance, as well as discuss recent therapeutic approaches to reverse resistance caused by drug transports and resistance to anti-EGFR blockade caused by mutations in EGFR and alteration in HGF/c-MET signaling pathway.
Collapse
Affiliation(s)
| | | | - Wei Li
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
6
|
Zhang C, Huang MN, Shan JQ, Hu ZJ, Li ZW, Liu JY. Pemigatinib, a selective FGFR inhibitor overcomes ABCB1-mediated multidrug resistance in cancer cells. Biochem Biophys Res Commun 2024; 691:149314. [PMID: 38039831 DOI: 10.1016/j.bbrc.2023.149314] [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: 10/22/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023]
Abstract
P: -glycoprotein (P-gp/ABCB1) overexpression is one of the primary causes of multidrug resistance (MDR). Therefore, it is crucial to discover effective pharmaceuticals to combat multidrug resistance mediated by ABCB1. Pemigatinib is a selective the fibroblast growth factor receptor (FGFR) inhibitor that is used to treat a variety of solid tumors, Clinical Trials for Urothelial Carcinoma (NCT02872714) completed its research on Pemigatinib. This study aimed to determine whether Pemigatinib can reverse ABCB1-mediated multidrug resistance, as well as its mechanism of action. Pemigatinib substantially reversed ABCB1-mediated multidrug resistance, as determined by a CCK8 assay, and immunofluorescence experiments revealed that Pemigatinib had no effect on the intracellular localization of ABCB1. Pemigatinib was discovered to increase intracellular drug accumulation, thereby reversing multidrug resistance. In addition, Docking analysis revealed that Pemigatinib and ABCB1 have a high affinity for one another. This study concludes that Pemigatinib is capable of reversing the multidrug resistance mediated by ABCB1, offering ideas and references for the clinical application of Pemigatinib.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Urology Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440 Jiyan Road, Jinan, Shandong, 250117, PR China
| | - Min-Na Huang
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine, 134 Research Park Dr, Columbia, MO, 65211, USA
| | - Jun-Qi Shan
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No. 440 Jiyan Road, Jinan, Shandong, 250117, PR China
| | - Zun-Jie Hu
- Department of Urology Surgery, The Affiliated Taian City Central Hospital of Qingdao University, No. 29 Longtan Road, Taian, Shandong, 271000, PR China
| | - Zi-Wei Li
- Department of Experimental Center, Shandong University of Traditional Chinese Medicine, No. 4655 Daxue Road, Changqing, Jinan, Shandong, 250355, PR China.
| | - Jian-Ying Liu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Shandong First Medical University, No. 38 Wuyingshan Road, Tianqiao, Jinan, Shandong, 250031, PR China.
| |
Collapse
|
7
|
Wu CP, Murakami M, Li YC, Huang YH, Chang YT, Hung TH, Wu YS, Ambudkar SV. Imperatorin Restores Chemosensitivity of Multidrug-Resistant Cancer Cells by Antagonizing ABCG2-Mediated Drug Transport. Pharmaceuticals (Basel) 2023; 16:1595. [PMID: 38004460 PMCID: PMC10674403 DOI: 10.3390/ph16111595] [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: 10/02/2023] [Revised: 11/03/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
The high expression of the ATP-binding cassette (ABC) drug transporter ABCG2 in cancer cells contributes to the emergence of multidrug resistance (MDR) in individuals afflicted with either solid tumors or blood cancers. MDR poses a major impediment in the realm of clinical cancer chemotherapy. Recently, substantial endeavors have been dedicated to identifying bioactive compounds isolated from nature capable of counteracting ABCG2-mediated MDR in cancer cells. Imperatorin, a natural coumarin derivative renowned for its diverse pharmacological properties, has not previously been explored for its impact on cancer drug resistance. This study investigates the chemosensitizing potential of imperatorin in ABCG2-overexpressing cancer cells. Experimental results reveal that at sub-toxic concentrations, imperatorin significantly antagonizes the activity of ABCG2 and reverses ABCG2-mediated MDR in a concentration-dependent manner. Furthermore, biochemical data and in silico analysis of imperatorin docking to the inward-open conformation of human ABCG2 indicate that imperatorin directly interacts with multiple residues situated within the transmembrane substrate-binding pocket of ABCG2. Taken together, these results furnish substantiation that imperatorin holds promise for further evaluation as a potent inhibitor of ABCG2, warranting exploration in combination drug therapy to enhance the effectiveness of therapeutic agents for patients afflicted with tumors that exhibit high levels of ABCG2.
Collapse
Affiliation(s)
- Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-C.L.); (Y.-H.H.)
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan;
| | - Megumi Murakami
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Yen-Ching Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-C.L.); (Y.-H.H.)
| | - Yang-Hui Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-C.L.); (Y.-H.H.)
| | - Yu-Tzu Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (Y.-C.L.); (Y.-H.H.)
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan;
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan;
| | - Suresh V. Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
8
|
Sajid A, Rahman H, Ambudkar SV. Advances in the structure, mechanism and targeting of chemoresistance-linked ABC transporters. Nat Rev Cancer 2023; 23:762-779. [PMID: 37714963 DOI: 10.1038/s41568-023-00612-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 09/17/2023]
Abstract
Cancer cells frequently display intrinsic or acquired resistance to chemically diverse anticancer drugs, limiting therapeutic success. Among the main mechanisms of this multidrug resistance is the overexpression of ATP-binding cassette (ABC) transporters that mediate drug efflux, and, specifically, ABCB1, ABCG2 and ABCC1 are known to cause cancer chemoresistance. High-resolution structures, biophysical and in silico studies have led to tremendous progress in understanding the mechanism of drug transport by these ABC transporters, and several promising therapies, including irradiation-based immune and thermal therapies, and nanomedicine have been used to overcome ABC transporter-mediated cancer chemoresistance. In this Review, we highlight the progress achieved in the past 5 years on the three transporters, ABCB1, ABCG2 and ABCC1, that are known to be of clinical importance. We address the molecular basis of their broad substrate specificity gleaned from structural information and discuss novel approaches to block the function of ABC transporters. Furthermore, genetic modification of ABC transporters by CRISPR-Cas9 and approaches to re-engineer amino acid sequences to change the direction of transport from efflux to import are briefly discussed. We suggest that current information regarding the structure, mechanism and regulation of ABC transporters should be used in clinical trials to improve the efficiency of chemotherapeutics for patients with cancer.
Collapse
Affiliation(s)
- Andaleeb Sajid
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hadiar Rahman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Gao HL, Cui Q, Wang JQ, Ashby CR, Chen Y, Shen ZX, Chen ZS. The AKT inhibitor, MK-2206, attenuates ABCG2-mediated drug resistance in lung and colon cancer cells. Front Pharmacol 2023; 14:1235285. [PMID: 37521473 PMCID: PMC10373739 DOI: 10.3389/fphar.2023.1235285] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction: The overexpression of ATP-binding cassette (ABC) transporters, ABCB1 and ABCG2, are two of the major mediators of multidrug resistance (MDR) in cancers. Although multiple ABCB1 and ABCG2 inhibitors have been developed and some have undergone evaluation in clinical trials, none have been clinically approved. The compound, MK-2206, an inhibitor of the protein kinases AKT1/2/3, is undergoing evaluation in multiple clinical trials for the treatment of certain types of cancers, including those resistant to erlotinib. In this in vitro study, we conducted in vitro experiments to determine if MK-2206 attenuates multidrug resistance in cancer cells overexpressing the ABCB1 or ABCG2 transporter. Methodology: The efficacy of MK-2206 (0.03-1 μM), in combination with the ABCB1 transporter sub-strates doxorubicin and paclitaxel, and ABCG2 transporter substrates mitoxantrone, SN-38 and topotecan, were determined in the cancer cell lines, KB-C2 and SW620/Ad300, which overexpress the ABCB1 transporter or H460/MX20 and S1-M1-80, which overexpress the ABCG2 transporter, respectively. The expression level and the localization of ABCG2 transporter on the cancer cells membranes were determined using western blot and immunofluorescence assays, respectively, following the incubation of cells with MK-2206. Finally, the interaction between MK-2206 and human ABCG2 transporter was predicted using computer-aided molecular modeling. Results: MK-2206 significantly increased the efficacy of anticancer compounds that were substrates for the ABCG2 but not the ABCB1 transporter. MK-2206 alone (0.03-1 μM) did not significantly alter the viability of H460/MX20 and S1-M1-80 cancer cells, which overexpress the ABCG2 transporter, compared to cells incubated with vehicle. However, MK-2206 (0.3 and 1 μM) significantly increased the anticancer efficacy of mitoxantrone, SN-38 and topotecan, in H460/MX20 and S1-M1-80 cancer cells, as indicated by a significant decrease in their IC50 values, compared to cells incubated with vehicle. MK-2206 significantly increased the basal activity of the ABCG2 ATPase (EC50 = 0.46 μM) but did not significantly alter its expression level and sub-localization in the membrane. The molecular modeling results suggested that MK-2206 binds to the active pocket of the ABCG2 transporter, by a hydrogen bond, hydrophobic interactions and π-π stacking. Conclusion: These in vitro data indicated that MK-2206 surmounts resistance to mitoxantrone, SN-38 and topotecan in cancer cells overexpressing the ABCG2 transporter. If these results can be translated to humans, it is possible that MK-2206 could be used to surmount MDR in cancer cells overexpressing the ABCG2 transporter.
Collapse
Affiliation(s)
- Hai-Ling Gao
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Qingbin Cui
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Yanchun Chen
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Zhi-Xin Shen
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| |
Collapse
|
10
|
Alhaj-Suliman SO, Naguib YW, Wafa EI, Saha S, Ebeid K, Meng X, Mohammed HH, Abuo-Rahma GEDA, Yang S, Salem AK. A ciprofloxacin derivative with four mechanisms of action overcomes paclitaxel resistance in p53-mutant and MDR1 gene-expressing type II human endometrial cancer. Biomaterials 2023; 296:122093. [PMID: 36965280 PMCID: PMC10092294 DOI: 10.1016/j.biomaterials.2023.122093] [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: 11/05/2022] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/19/2023]
Abstract
Dysfunction of the p53 gene and the presence of the MDR1 gene are associated with many malignant tumors including endometrial cancer and are responsible for cancer therapeutic resistance and poor survival. Thus, there is a critical need to devise novel combinatorial therapies with multiple mechanisms of action to overcome drug resistance. Here, we report a new ciprofloxacin derivative (CIP2b) tested either alone or in combination with taxanes against four human endometrial cancer cell lines. In vitro studies revealed that a combination of paclitaxel + CIP2b had synergistic cytotoxic effects against MDR1-expressing type-II human endometrial cancer cells with loss-of-function p53 (Hec50co LOFp53). Enhanced antitumor effects were confirmed by substantial increases in caspase-3 expression, cell population shifts toward the G2/M phase, and reduction of cdc2 phosphorylation. It was found that CIP2b targets multiple pathways including the inhibition of MDR1, topoisomerase I, and topoisomerase II, as well as enhancing the effects of paclitaxel (PTX) on microtubule assembly. In vivo treatment with the combination of PTX + CIP2b also led to significantly increased accumulation of PTX in tumors (compared to CIP2b alone) and reduction in tumor growth. Enhanced in vivo cytotoxic effects were confirmed by histological and immunohistochemical examination of the tumor tissues. Complete blood count and blood biochemistry data confirmed the absence of any apparent off-target toxicity. Thus, combination therapy involving PTX and CIP2b targeted multiple pathways and represents an approach that could result in improved tolerance and efficacy in patients with type-II endometrial cancer harboring the MDR1 gene and p53 mutations.
Collapse
Affiliation(s)
- Suhaila O Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa, IA, 52242, United States
| | - Youssef W Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa, IA, 52242, United States; Departments of Pharmaceutics and Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa, IA, 52242, United States
| | - Sanjib Saha
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa, IA, 52242, United States
| | - Kareem Ebeid
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa, IA, 52242, United States; Departments of Pharmaceutics and Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Xiangbing Meng
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, United States
| | - Hamada H Mohammed
- Departments of Pharmaceutics and Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Gamal El-Din A Abuo-Rahma
- Departments of Pharmaceutics and Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Shujie Yang
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa, IA, 52242, United States.
| |
Collapse
|
11
|
Sadhukhan P, Seiwert TY. The role of macrophages in the tumor microenvironment and tumor metabolism. Semin Immunopathol 2023; 45:187-201. [PMID: 37002376 DOI: 10.1007/s00281-023-00988-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/08/2023] [Indexed: 04/03/2023]
Abstract
The complexity and plasticity of the tumor microenvironment (TME) make it difficult to fully understand the intratumoral regulation of different cell types and their activities. Macrophages play a crucial role in the signaling dynamics of the TME. Among the different subtypes of macrophages, tumor-associated macrophages (TAMs) are often associated with poor prognosis, although some subtypes of TAMs can at the same time improve treatment responsiveness and lead to favorable clinical outcomes. TAMs are key regulators of cancer cell proliferation, metastasis, angiogenesis, extracellular matrix remodeling, tumor metabolism, and importantly immunosuppression in the TME by modulating various chemokines, cytokines, and growth factors. TAMs have been identified as a key contributor to resistance to chemotherapy and cancer immunotherapy. In this review article, we aim to discuss the mechanisms by which TAMs regulate innate and adaptive immune signaling in the TME and summarize recent preclinical research on the development of therapeutics targeting TAMs and tumor metabolism.
Collapse
Affiliation(s)
- Pritam Sadhukhan
- Johns Hopkins University, Skip Viragh Outpatient Cancer Building, Baltimore, MD, 21287, USA
| | - Tanguy Y Seiwert
- Johns Hopkins University, Skip Viragh Outpatient Cancer Building, Baltimore, MD, 21287, USA.
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
12
|
Discovery of a novel highly potent and low-toxic jatrophane derivative enhancing the P-glycoprotein-mediated doxorubicin sensitivity of MCF-7/ADR cells. Eur J Med Chem 2022; 244:114822. [DOI: 10.1016/j.ejmech.2022.114822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/29/2022] [Accepted: 10/01/2022] [Indexed: 11/21/2022]
|
13
|
Wang S, Wang Z, Li Z, Xu J, Meng X, Zhao Z, Hou Y. A Catalytic Immune Activator Based on Magnetic Nanoparticles to Reprogram the Immunoecology of Breast Cancer from "Cold" to "Hot" State. Adv Healthc Mater 2022; 11:e2201240. [PMID: 36065620 DOI: 10.1002/adhm.202201240] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/03/2022] [Indexed: 01/28/2023]
Abstract
Triple-negative breast cancer (TNBC) as "cold" tumor is characterized by severe immunosuppression of the tumor microenvironment (TME). To effectively activate the immune response of TNBC, a new kind of therapy strategy called cancer catalytic immunotherapy is proposed based on magnetic nanoparticles (NPs) as immune activators. Utilizing the weak acidity and excessive hydrogen peroxide of TME, these magnetic NPs can release ferrous ions to promote Fenton reaction, leading to abundant ·OH and reactive oxygen species (ROS) for ultimately killing cancer cells. Mechanistically, these magnetic NPs activate the ROS-related signaling pathway to generate more ROS. Meanwhile, these magnetic NPs with unique immunological properties can promote the maturation of dendritic cells and the polarization of macrophages from M2 to M1, resulting in the infiltration of more T cells to reprogram the immunoecology of TNBC from "cold" to "hot" state. Besides directly affecting immune cells, these magnetic NPs can also affect the secretion of some immune-related cytokines by cancer cells, to further indirectly activate the immune response. In conclusion, these catalytic immune activators are designed to achieve the synergistic treatment of chemodynamic therapy-enhanced immunotherapy guided by computed tomography (CT)/near-infrared region-II (NIR-II) dual-mode imaging, providing a new strategy for TNBC treatment.
Collapse
Affiliation(s)
- Shuren Wang
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing, 100871, China
| | - Zhiyi Wang
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing, 100871, China
| | - Ziyuan Li
- Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing, 100191, China.,Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| | - Junjie Xu
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing, 100871, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zijing Zhao
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing, 100871, China
| | - Yanglong Hou
- Beijing Key Laboratory of Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing, 100871, China
| |
Collapse
|
14
|
Feng Y, Ma F, Wu E, Cheng Z, Wang Z, Yang L, Zhang J. Ginsenosides: Allies of gastrointestinal tumor immunotherapy. Front Pharmacol 2022; 13:922029. [PMID: 36386161 PMCID: PMC9659574 DOI: 10.3389/fphar.2022.922029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 09/26/2022] [Indexed: 09/25/2023] Open
Abstract
In the past decade, immunotherapy has been the most promising treatment for gastrointestinal tumors. But the low response rate and drug resistance remain major concerns. It is therefore imperative to develop adjuvant therapies to increase the effectiveness of immunotherapy and prevent drug resistance. Ginseng has been used in Traditional Chinese medicine as a natural immune booster for thousands of years. The active components of ginseng, ginsenosides, have played an essential role in tumor treatment for decades and are candidates for anti-tumor adjuvant therapy. They are hypothesized to cooperate with immunotherapy drugs to improve the curative effect and reduce tumor resistance and adverse reactions. This review summarizes the research into the use of ginsenosides in immunotherapy of gastrointestinal tumors and discusses potential future applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jiwei Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Zhang H, Bian S, Xu Z, Gao M, Wang H, Zhang J, Zhang M, Ke Y, Wang W, Chen ZS, Xu H. The effect and mechanistic study of encequidar on reversing the resistance of SW620/AD300 cells to doxorubicin. Biochem Pharmacol 2022; 205:115258. [PMID: 36179932 DOI: 10.1016/j.bcp.2022.115258] [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: 08/07/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022]
Abstract
Encequidar, a gut-specific P-glycoprotein (P-gp) inhibitor, makes oral paclitaxel possible, and has been used in clinical treatment of metastatic breast cancer, however, its pharmacological effect and mechanism of reversal of drug resistance in drug-resistant colon cancer cells SW620/AD300 are still unknown. Herein, we first synthesized encequidar and demonstrated that it could inhibit the transport activity of P-gp, reduced doxorubicin (DOX) efflux, enhanced DOX cytotoxicity and promoted tumor-apoptosis in SW620/AD300 cells. Metabolomic analysis of cell samples was performedusing liquid chromatography Q-Exactive mass spectrometer, the results of metabolite enrichment analysis and pathway analysis showed that the combination of encequidar and DOX could: i) significantly affect the citric acid cycle (TCA cycle) and reduce the energy supply required for P-gp to exert its transport activity; ii) affect the metabolism of glutathione, which is the main component of the anti-oxidative stress system, and reduce the ability of cells to resist oxidative stress; iii) increase the intracellular reactive oxygen species (ROS) production and enhance ROS-induced cell damage and lipid peroxidation, which in turn restore the sensitivity of drug-resistant cells to DOX. In conclusion, these results provide sufficient data support for the therapeutical application of the P-gp inhibitor encequidar to reverse MDR, and are of great significance to further understand the therapeutic advantages of encequidar in anti-tumor therapy and guide clinical rational drug use.
Collapse
Affiliation(s)
- Hang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Shaopan Bian
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhihao Xu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ming Gao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Han Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junwei Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mingkun Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yu Ke
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Weijia Wang
- Department of International Medical Services (IMS), Beijing Tiantan Hospital of Capital Medical University, Beijing, 100070, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John's University, Queens, New York, USA.
| | - Haiwei Xu
- Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Institute of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
16
|
Discovery of α-methylene-γ-lactone-δ-epoxy derivatives with anti-cancer activity: synthesis, SAR study, and biological activity. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02925-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
17
|
Narayanan S, Teng QX, Wu ZX, Nazim U, Karadkhelkar N, Acharekar N, Yoganathan S, Mansoor N, Ping FF, Chen ZS. Anticancer effect of Indanone-based thiazolyl hydrazone derivative on p53 mutant colorectal cancer cell lines: An in vitro and in vivo study. Front Oncol 2022; 12:949868. [PMID: 35992866 PMCID: PMC9386487 DOI: 10.3389/fonc.2022.949868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer is a major health problem, and it is the third most diagnosed cancer in the United States. The current treatment for colorectal cancer includes irinotecan, a topoisomerase I inhibitor, and other targeted drugs, such as bevacizumab and regorafenib. The low response rates and incidence of high toxicity caused by these drugs instigated an evaluation of the anticancer efficacy of a series of 13 thiazolyl hydrazone derivatives of 1-indanone, and four compounds among them show favorable anticancer activity against some of the tested colorectal cancer cell lines with IC50 values ranging from 0.41 ± 0.19 to 6.85 ± 1.44 μM. It is noteworthy that one of the indanone-based thiazolyl hydrazone (ITH) derivatives, N-Indan-1-ylidene-N’-(4-Biphenyl-4-yl-thiazol-2-yl)-hydrazine (ITH-6), has a better cytotoxicity profile against p53 mutant colorectal cancer cells HT-29, COLO 205, and KM 12 than a p53 wild-type colorectal cancer cell line, such as HCT 116. Mechanistic studies show that ITH-6 arrests these three cancer cell lines in the G2/M phase and induces apoptosis. It also causes a rise in the reactive oxygen species level with a remarkable decrease in the glutathione (GSH) level. Moreover, ITH-6 inhibits the expression of NF-κB p65 and Bcl-2, which proves its cytotoxic action. In addition, ITH-6 significantly decreased tumor size, growth rate, and tumor volume in mice bearing HT-29 and KM 12 tumor xenografts. Moreover, CRISPR/Cas9 was applied to establish an NF-κB p65 gene knockout HT-29 cell line model to validate the target of ITH-6. Overall, the results suggest that ITH-6 could be a potential anticancer drug candidate for p53 mutant colorectal cancers.
Collapse
Affiliation(s)
- Silpa Narayanan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Urooj Nazim
- Department of Pharmaceutical Chemistry, University of Karachi, Karachi, Pakistan
| | - Nishant Karadkhelkar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Nikita Acharekar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Sabesan Yoganathan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Najia Mansoor
- Department of Pharmaceutical Chemistry, University of Karachi, Karachi, Pakistan
| | - Feng-Feng Ping
- Department of Reproductive Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wu-xi, China
- *Correspondence: Zhe-Sheng Chen, ; Feng-Feng Ping,
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
- *Correspondence: Zhe-Sheng Chen, ; Feng-Feng Ping,
| |
Collapse
|
18
|
Karthika C, Sureshkumar R, Zehravi M, Akter R, Ali F, Ramproshad S, Mondal B, Tagde P, Ahmed Z, Khan FS, Rahman MH, Cavalu S. Multidrug Resistance of Cancer Cells and the Vital Role of P-Glycoprotein. Life (Basel) 2022; 12:897. [PMID: 35743927 PMCID: PMC9227591 DOI: 10.3390/life12060897] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
P-glycoprotein (P-gp) is a major factor in the multidrug resistance phenotype in cancer cells. P-gp is a protein that regulates the ATP-dependent efflux of a wide range of anticancer medicines and confers resistance. Due to its wide specificity, several attempts have been made to block the action of P-gp to restore the efficacy of anticancer drugs. The major goal has been to create molecules that either compete with anticancer medicines for transport or function as a direct P-gp inhibitor. Despite significant in vitro success, there are presently no drugs available in the clinic that can "block" P-gp-mediated resistance. Toxicity, unfavourable pharmacological interactions, and a variety of pharmacokinetic difficulties might all be the reason for the failure. On the other hand, P-gp has a significant effect in the body. It protects the vital organs from the entry of foreign bodies and other toxic chemicals. Hence, the inhibitors of P-gp should not hinder its action in the normal cells. To develop an effective inhibitor of P-gp, thorough background knowledge is needed in this field. The main aim of this review article was to set forth the merits and demerits of the action of P-gp on cancer cells as well as on normal cells. The influence of P-gp on cancer drug delivery and the contribution of P-gp to activating drug resistance were also mentioned.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, Tamil Nadu, India;
| | - Raman Sureshkumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, Tamil Nadu, India;
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University Alkharj, Alkharj 11942, Saudi Arabia;
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea;
| | - Faraat Ali
- Department of Licensing and Enforcement, Laboratory Services, Botswana Medicines Regulatory Authority (BoMRA), Gaborone 999106, Botswana;
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Priti Tagde
- Amity Institute of Pharmacy, Amity University, Noida 201303, Uttar Pradesh, India;
| | - Zubair Ahmed
- Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia;
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
- Mahala Campus, Community College, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Biology Department, Faculty of Sciences and Arts, King Khalid University, Dhahran Al Janoub, Abha 61413, Saudi Arabia;
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea;
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
19
|
Prognostic role of TNF alpha, LT alpha, MDR1 and codon 72 Tp53 Gene polymorphisms on Multiple Myeloma Egyptian patients. Leuk Res 2022; 117:106854. [DOI: 10.1016/j.leukres.2022.106854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 04/12/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022]
|
20
|
Yang M, Lv X, Zhan S, Lu M, Zhang X, Qiu T. Glutathione-sensitive IPI-549 nanoparticles synergized with photodynamic Chlorin e6 for the treatment of breast cancer. NANOTECHNOLOGY 2022; 33:235101. [PMID: 35193121 DOI: 10.1088/1361-6528/ac57ac] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/22/2022] [Indexed: 06/14/2023]
Abstract
We combined phosphoinositol-3-kinin inhibitor IPI-549 and photodynamic Chlorin e6 (Ce6) on carboxymethyl chitosan to develop a novel drug delivery nanoparticle (NP) system (Ce6/CMCS-DSP-IPI549) and evaluate its glutathione (GSH) sensitivity and targeting ability for breast cancer treatment. The NPs were spherical with a uniform size of 218.8 nm, a stable structure over 7 days. The maximum encapsulation efficiency was 64.42%, and NPs drug loading was 8.05%. The NPs released drugs within tumor cells due to their high GSH concentration, while they maintained structural integrity in normal cells, which have low GSH concentration. The cumulative release rates of IPI-549 and Ce6 at 108 h were 70.67% and 40.35% (at GSH 10 mM) and 8.11% and 2.71% (at GSH 2μM), respectively. The NPs showed a strong inhibitory effect on 4T1 cells yet did not affect human umbilical vein endothelial cells (HUVECs). After irradiation by a 660 nm infrared laser for 72 h, the survival rate of 4T1 cells was 15.51%. Cellular uptake studies indicated that the NPs could accurately release drugs into tumor cells. In addition, the NPs had a good photodynamic effect and promoted the release of reactive oxygen species to damage tumor cells. Overall, the combination therapy of IPI-549 and Ce6 is safe and effective, and may provide a new avenue for the treatment of breast cancer.
Collapse
Affiliation(s)
- Mengjia Yang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, People's Republic of China
- Xi'an Medical College, Xi'an 710309, People's Republic of China
| | - Xiaojun Lv
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, People's Republic of China
- Hubei Institute for Drug Control, Wuhan 430064, People's Republic of China
| | - Siwen Zhan
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Mengli Lu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Xueqiong Zhang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Tong Qiu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| |
Collapse
|
21
|
Zhang C, Zhou X, Zhang H, Han X, Li B, Yang R, Zhou X. Recent Progress of Novel Nanotechnology Challenging the Multidrug Resistance of Cancer. Front Pharmacol 2022; 13:776895. [PMID: 35237155 PMCID: PMC8883114 DOI: 10.3389/fphar.2022.776895] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistance (MDR) of tumors is one of the clinical direct reasons for chemotherapy failure. MDR directly leads to tumor recurrence and metastasis, with extremely grievous mortality. Engineering a novel nano-delivery system for the treatment of MDR tumors has become an important part of nanotechnology. Herein, this review will take those different mechanisms of MDR as the classification standards and systematically summarize the advances in nanotechnology targeting different mechanisms of MDR in recent years. However, it still needs to be seriously considered that there are still some thorny problems in the application of the nano-delivery system against MDR tumors, including the excessive utilization of carrier materials, low drug-loading capacity, relatively narrow targeting mechanism, and so on. It is hoped that through the continuous development of nanotechnology, nano-delivery systems with more universal uses and a simpler preparation process can be obtained, for achieving the goal of defeating cancer MDR and accelerating clinical transformation.
Collapse
Affiliation(s)
- Chengyuan Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Xuemei Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Hanyi Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Xuanliang Han
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Baijun Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Ran Yang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Xing Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
22
|
Xie Q, Wang J, Wu W, Zhao Y. Apatinib inhibits paclitaxel resistance of gastric carcinoma cells through VEGFR2 pathway. Am J Transl Res 2022; 14:421-431. [PMID: 35173861 PMCID: PMC8829625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/09/2021] [Indexed: 06/14/2023]
Abstract
Drug resistance of gastric carcinoma (GC) is a burning question in the medical field. This study aimed to investigating the ameliorative effect of apatinib (Apa) on paclitaxel (PTX) resistance in GC. In this research, PTX-resistant MGC803 cells were intervened by Apa. Cell proliferation was detected by cell counting kit-8 (CCK-8) assay, and cell migration and invasion was determined by Transwell assays. The levels of apoptosis-related proteins (Bcl-2, Bax), drug resistance-related proteins (MDR1, P-gp) and VEGFR2 protein were measure by Western blot, and the mRNA expression of VEGFR2 was tested by real-time quantitative polymerase chain reaction (RT-qPCR). Then VEGFR2 was overexpressed to examine the role of Apa in PTX-resistant MGC803 cells. The results identified a significantly reduced growth rate of MGC803/PTX cells after PTX induction, obviously increased invasive and migrated cells, and evidently enhanced proliferation capacity of MGC803/PTX cells as compared to MGC803 cells. In MGC803/PTX cells, VEGFR2, MDR1, P-gp and Bcl-2 were all up-regulated while Bax was down-regulated. After Apa intervention, PTX-resistant MGC803 cells showed decreased cell migration, invasion and proliferation, reduced MDR1, P-gp and VEGFR2 levels, and increased Bax protein level. Overexpression of VEGFR2 can offset the rescue effect of Apa on PTX-induced drug resistance of MGC803 cells. Taken together, Apa may inhibit PTX resistance of MGC803 cells via the VEGFR2 signaling pathway.
Collapse
Affiliation(s)
- Qian Xie
- Kindos Pharmaceuticals Co., Ltd.Chengdu 611731, Sichuan, China
| | - Jing Wang
- Kindos Pharmaceuticals Co., Ltd.Chengdu 611731, Sichuan, China
| | - Wenwen Wu
- Sichuan Institute of Food InspectionChengdu 610097, Sichuan, China
| | - Ye Zhao
- Chengdu Medical College, School of Bioscience and TechnologyChengdu 610500, Sichuan, China
| |
Collapse
|
23
|
Wang M, Zhan F, Cheng H, Li Q. Gambogenic Acid Inhibits Basal Autophagy of Drug-Resistant Hepatoma Cells and Improves Its Sensitivity to Adriamycin. Biol Pharm Bull 2022; 45:63-70. [PMID: 34980780 DOI: 10.1248/bpb.b21-00511] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gambogenic acid (GNA) is extracted from plant Gamboge, has a wide range of anti-tumor effects. In this paper, we study the inhibitory effect of GNA on the BEL-7402/ADM of hepatoma resistant cell lines and further study the mechanism of action. Cell viability test represented that GNA could improve the sensitivity of hepatoma drug-resistant cell line BEL-7402/ADM to Adriamycin (ADM), and further study by 4',6-diamidino-2-phenylindole (DAPI) staining and flow cytometry found that GNA could improve the effect of ADM on promoting apoptosis in BEL-7402/ADM cells, and the activation of apoptosis-related protein was significantly increased, and the ratio of Bax to Bcl-2 was significantly increased. Monodansylcadaverine staining and transmission electron microscopy showed that the basal autophagy level of BEL-7402/ADM cells was higher than that of BEL-7402 cells. Further detection of protein expression found that the intracellular LC3-II to LC3-I ratio and Beclin 1 protein expression increased in the combination of GNA and ADM, but the protein level of p62 increased significantly. GNA inhibit protective autophagy in BEL-7402/ADM cells and promote the role of ADM in inducing apoptosis, thereby increasing ADM sensitivity to BEL-7402/ADM cells, and the effect of GNA inhibition of autophagy may be achieved by inhibiting the degradation of autophagosomes.
Collapse
Affiliation(s)
- Meng Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Traditional Chinese Medicine, Anhui University of Chinese Medicine
| | - Fan Zhan
- Huaibei Maternity & Child Healthcare Hospital
| | - Hui Cheng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Traditional Chinese Medicine, Anhui University of Chinese Medicine
| | - Qinglin Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Traditional Chinese Medicine, Anhui University of Chinese Medicine
| |
Collapse
|
24
|
Narayanan S, Fan YF, Gujarati NA, Teng QX, Wang JQ, Cai CY, Yang Y, Chintalapati AJ, Lei Y, Korlipara VL, Chen ZS. VKNG-1 Antagonizes ABCG2-Mediated Multidrug Resistance via p-AKT and Bcl-2 Pathway in Colon Cancer: In Vitro and In Vivo Study. Cancers (Basel) 2021; 13:4675. [PMID: 34572902 PMCID: PMC8470077 DOI: 10.3390/cancers13184675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/23/2021] [Accepted: 09/07/2021] [Indexed: 11/16/2022] Open
Abstract
The emergence of multidrug resistance (MDR) to chemotherapeutic drugs is a major problem in the therapy of cancer. Knowledge of the mechanisms of drug resistance in cancer is necessary for developing efficacious therapies. ATP-binding cassette (ABC) transporters are transmembrane proteins that efflux chemotherapeutic drugs from cancer cells, thereby producing MDR. Our research efforts have led to the discovery of VKNG-1, a compound that selectively inhibits the ABCG2 transporter and reverses resistanctabe to standard anticancer drugs both in vitro and in vivo. VKNG-1, at 6 µM, selectively inhibited ABCG2 transporter and sensitized ABCG2-overexpressing drug-resistant cancer cells to the ABCG2 substrate anticancer drugs mitoxantrone, SN-38, and doxorubicin in ABCG2-overexpressing colon cancers. VKNG- 1 reverses ABCG2-mediated MDR by blocking ABCG2 efflux activity and downregulating ABCG2 expression at the mRNA and protein levels. Moreover, VKNG-1 inhibits the level of phosphorylated protein kinase B (PKB/p-AKT), and B-cell lymphoma-2 (Bcl-2) protein which may overcome resistance to anticancer drugs. However, the in vitro translocation of ABCG2 protein did not occur in the presence of 6 µM of VKNG-1. In addition, VKNG-1 enhanced the anticancer efficacy of irinotecan in ABCG2- overexpressing mouse tumor xenografts. Overall, our results suggest that VKNG-1 may, in combination with certain anticancer drugs, represent a treatment to overcome ABCG2-mediated MDR colon cancers.
Collapse
Affiliation(s)
- Silpa Narayanan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| | - Ying-Fang Fan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
- Department of Hepatobiliary Surgery, Zhu Jiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Nehaben A. Gujarati
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| | - Chao-Yun Cai
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| | - Anirudh J. Chintalapati
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| | - Yixiong Lei
- School of Public Health, Guangzhou Medical University, Guangzhou 511436, China;
| | - Vijaya L. Korlipara
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (Y.-F.F.); (N.A.G.); (Q.-X.T.); (J.-Q.W.); (C.-Y.C.); (Y.Y.); (A.J.C.)
| |
Collapse
|
25
|
Narayanan S, Wu ZX, Wang JQ, Ma H, Acharekar N, Koya J, Yoganathan S, Fang S, Chen ZS, Pan Y. The Spleen Tyrosine Kinase Inhibitor, Entospletinib (GS-9973) Restores Chemosensitivity in Lung Cancer Cells by Modulating ABCG2-mediated Multidrug Resistance. Int J Biol Sci 2021; 17:2652-2665. [PMID: 34326700 PMCID: PMC8315011 DOI: 10.7150/ijbs.61229] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are important in managing lymphoid malignancies by targeting B-cell receptor signaling pathways. Entospletinib (GS-9973) is an oral, selective inhibitor of spleen tyrosine kinase (Syk), currently in the phase II clinical trials for the treatment of chronic lymphocytic leukemia. Syk is abundantly present in the cells of hematopoietic lineage that mediates cell proliferation, differentiation, and adhesion. In this current study, we evaluated the efficacy of GS-9973 to overcome multidrug resistance (MDR) due to the overexpression of the ABCG2 transporter in the non-small cell lung cancer (NSCLC) cell line, NCI-H460/MX20. In vitro, 3 μM of GS-9973 reversed the drug resistance of NCI-H460/MX20 cell line to mitoxantrone or doxorubicin. GS-9973, at 3 μM reverses ABCG2-mediated MDR by blocking ABCG2 efflux activity and downregulating ABCG2 expression at the protein level but did not alter the ABCG2 mRNA expression and subcellular localization of the ABCG2 protein compared to drug-resistant cells incubated with the vehicle. GS-9973 produced a moderate concentration-dependent increase in the ATPase activity of ABCG2 (EC50 = 0.42 µM) and molecular docking data indicated that GS-9973 had a high affinity (-10.226 kcal/mol) for the substrate-binding site of ABCG2. Finally, HPLC analysis proved that the intracellular concentration of GS-9973 is not significantly different in both parental and resistant cell lines. In conclusion, our study suggests that in vitro, GS-9973 in combination with certain anticancer drugs, represent a strategy to overcome ABCG2-mediated MDR cancers.
Collapse
Affiliation(s)
- Silpa Narayanan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Hansu Ma
- Precision Medicine Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, PR China
| | - Nikita Acharekar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sabesan Yoganathan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Shuo Fang
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, PR China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yihang Pan
- Precision Medicine Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, PR China
| |
Collapse
|
26
|
Romero EL, Morilla MJ. Preclinical autophagy modulatory nanomedicines: big challenges, slow advances. Expert Opin Drug Deliv 2021; 18:1415-1434. [PMID: 34030559 DOI: 10.1080/17425247.2021.1933428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Autophagy is a critical housekeeping pathway to remove toxic protein aggregates, damaged organelles, providing cells with bioenergetic substrates needed to survive under adverse conditions. Since altered autophagy is associated with diverse diseases, its pharmacological modulation is considered of therapeutic interest. Nanomedicines may reduce the toxicity and improve the activity of toxic autophagy modulatory drugs (amd). AREAS COVERED The status of the most relevant anti-tumor, anti-inflammatory, and anti-infectious treatments mediated by autophagy modulatory nanomedicines (amN) published in the last 5 years is discussed. EXPERT OPINION Antitumor and anti-inflammatory treatments may be improved by administering amN for selective, massive, and targeted delivery of amd to diseased tissues. The use of amN as antimicrobial agent remains almost underexploited. Assessing the effect of amN on the complex autophagy machinery operating under different basal diseases, however, is not a trivial task. Besides structural reproducibility, nanomedicines must grant higher efficiency, and lower adverse effects than conventional medication. Simplicity of design, carefully chosen (scalable) preparation techniques, and rigorous monitoring of preclinical efficacy and nanotoxicity will improve the chances of clinical success. Currently, available data are not sufficient to envisage a fast-succeeding translation. Application of quality by design criteria would help to reach such milestones.
Collapse
Affiliation(s)
- Eder Lilia Romero
- Department of Science and Technology, Nanomedicines Research and Development Center, Quilmes National University, Bernal, Buenos Aires, Argentina
| | - Maria Jose Morilla
- Department of Science and Technology, Nanomedicines Research and Development Center, Quilmes National University, Bernal, Buenos Aires, Argentina
| |
Collapse
|
27
|
Yang H, Mamatjan A, Tang D, Aisa HA. Jatrophane diterpenoids as multidrug resistance modulators from Euphorbia sororia. Bioorg Chem 2021; 112:104989. [PMID: 34022709 DOI: 10.1016/j.bioorg.2021.104989] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
Eight new jatrophane diterpenoids, Euphosorophane F-M (1-8), as well as fourteen known jatrophane diterpenoids (9-22) were separated and purified from the fructus of Euphorbia sororia, and the chemical structures were determined based on extensive spectroscopic analysis, 1D, 2D NMR and HRESIMS data included. Their absolute configurations of compounds 1, 2, 9, and 22 were elucidated by X-ray crystallographic analysis. These jatrophane diterpenoids showed lower cytotoxicity and compounds 3, 4, 11, 12, 13, 14, and 20 revealed promising multidrug resistance (MDR) reversal ability as modulators compared to verapamil (VRP) by MTT assay. The structure-activity relationship (SAR) exhibited that the absence of keto-carbonyl at C-9 and C-14 was essential to MDR reversal activity and the acyloxies substitution at C-5, C-7, C-8, and C-14 also made the activity difference. Euphosorophane I (4) particularly unfold greater potency (EC50 = 1.82 μM) in reversing P-gp-mediated resistance to doxorubicin (DOX). As shown by fluorescence microscopy, 4 promoted intracellular accumulation of rhodamine 123 (Rh123) and DOX in a dose-dependentmanner than VRP. Flow cytometry indicated that 4 inhibitedP-glycoprotein (P-gp) -dependentRh123 efflux in drug-resistant MCF-7/ADR cells. 4 stimulated P-gp-ATPase activity in a concentration-dependent way and inhibited DOX transport activity. Western blot and real-time qPCR results further illustrated that 4 exhibited superior MDR reversal effect in MCF-7/ADR cells attributed to the activation of ATPase rather than the upregulation of P-gp expression and mRNA levels. In addition, 4 bond to the drug-binding site of P-gp predicted by the molecular docking analysis. Collectively, these results indicated that 4 efficiently reversed P-gp-mediated MDR via inhibiting the ABCB1 drug efflux function. 4 with the advantage of low toxicity and efficient could be used as an adjuvanttherapy drug for breast cancer.
Collapse
Affiliation(s)
- Hequn Yang
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, PR China; University of Chinese Academy of Sciences, Beijing 100039, PR China
| | - Aytilla Mamatjan
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, PR China; University of Chinese Academy of Sciences, Beijing 100039, PR China
| | - Dan Tang
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, PR China; The Key Laboratory of Plant Resources and Chemistry of Arid Zone, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, PR China
| | - Haji Akber Aisa
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, PR China; The Key Laboratory of Plant Resources and Chemistry of Arid Zone, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, PR China.
| |
Collapse
|
28
|
Emerging nanotaxanes for cancer therapy. Biomaterials 2021; 272:120790. [PMID: 33836293 DOI: 10.1016/j.biomaterials.2021.120790] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022]
Abstract
The clinical application of taxane (including paclitaxel, docetaxel, and cabazitaxel)-based formulations is significantly impeded by their off-target distribution, unsatisfactory release, and acquired resistance/metastasis. Recent decades have witnessed a dramatic progress in the development of high-efficiency, low-toxicity nanotaxanes via the use of novel biomaterials and nanoparticulate drug delivery systems (nano-DDSs). Thus, in this review, the achievements of nanotaxanes-targeted delivery and stimuli-responsive nano-DDSs-in preclinical or clinical trials have been outlined. Then, emerging nanotherapeutics against tumor resistance and metastasis have been overviewed, with a particular emphasis on synergistic therapy strategies (e.g., combination with surgery, chemotherapy, radiotherapy, biotherapy, immunotherapy, gas therapy, phototherapy, and multitherapy). Finally, the latest oral nanotaxanes have been briefly discussed.
Collapse
|
29
|
Mishra R, Patel H, Alanazi S, Kilroy MK, Garrett JT. PI3K Inhibitors in Cancer: Clinical Implications and Adverse Effects. Int J Mol Sci 2021; 22:3464. [PMID: 33801659 PMCID: PMC8037248 DOI: 10.3390/ijms22073464] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
The phospatidylinositol-3 kinase (PI3K) pathway is a crucial intracellular signaling pathway which is mutated or amplified in a wide variety of cancers including breast, gastric, ovarian, colorectal, prostate, glioblastoma and endometrial cancers. PI3K signaling plays an important role in cancer cell survival, angiogenesis and metastasis, making it a promising therapeutic target. There are several ongoing and completed clinical trials involving PI3K inhibitors (pan, isoform-specific and dual PI3K/mTOR) with the goal to find efficient PI3K inhibitors that could overcome resistance to current therapies. This review focuses on the current landscape of various PI3K inhibitors either as monotherapy or in combination therapies and the treatment outcomes involved in various phases of clinical trials in different cancer types. There is a discussion of the drug-related toxicities, challenges associated with these PI3K inhibitors and the adverse events leading to treatment failure. In addition, novel PI3K drugs that have potential to be translated in the clinic are highlighted.
Collapse
Affiliation(s)
| | | | | | | | - Joan T. Garrett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267-0514, USA; (R.M.); (H.P.); (S.A.); (M.K.K.)
| |
Collapse
|
30
|
Narayanan S, Gujarati NA, Wang JQ, Wu ZX, Koya J, Cui Q, Korlipara VL, Ashby CR, Chen ZS. The Novel Benzamide Derivative, VKNG-2, Restores the Efficacy of Chemotherapeutic Drugs in Colon Cancer Cell Lines by Inhibiting the ABCG2 Transporter. Int J Mol Sci 2021; 22:2463. [PMID: 33671108 PMCID: PMC7957563 DOI: 10.3390/ijms22052463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
The overexpression of ATP-binding cassette transporter, ABCG2, plays an important role in mediating multidrug resistance (MDR) in certain types of cancer cells. ABCG2-mediated MDR can significantly attenuate or abrogate the efficacy of anticancer drugs by increasing their efflux from cancer cells. In this study, we determined the efficacy of the novel benzamide derivative, VKNG-2, to overcome MDR due to the overexpression of the ABCG2 transporter in the colon cancer cell line, S1-M1-80. In vitro, 5 μM of VKNG-2 reversed the resistance of S1-M1-80 cell line to mitoxantrone (70-fold increase in efficacy) or SN-38 (112-fold increase in efficacy). In contrast, in vitro, 5 μM of VKNG-2 did not significantly alter either the expression of ABCG2, AKT, and PI3K p110β protein or the subcellular localization of the ABCG2 protein compared to colon cancer cells incubated with the vehicle. Molecular docking data indicated that VKNG-2 had a high docking score (-10.2 kcal/mol) for the ABCG2 transporter substrate-drug binding site whereas it had a low affinity on ABCB1 and ABCC1 transporters. Finally, VKNG-2 produced a significant concentration-dependent increase in ATPase activity (EC50 = 2.3 µM). In conclusion, our study suggests that in vitro, VKNG-2 reverses the resistance of S1-M1-80, a cancer cell line resistant to mitoxantrone and SN-38, by inhibiting the efflux function of the ABCG2 transporter.
Collapse
Affiliation(s)
- Silpa Narayanan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
| | - Nehaben A. Gujarati
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
| | - Jagadish Koya
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
| | - Qingbin Cui
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
- School of Pharmaceutical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Vijaya L. Korlipara
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (S.N.); (N.A.G.); (J.-Q.W.); (Z.-X.W.); (J.K.); (Q.C.); (V.L.K.); (C.R.A.J.)
| |
Collapse
|
31
|
Wang W, Liang Z, Ma P, Zhao Q, Dai M, Zhu J, Han X, Xu H, Chang Q, Zhen Y. Application of CRISPR/Cas9 System to Reverse ABC-Mediated Multidrug Resistance. Bioconjug Chem 2021; 32:73-81. [PMID: 33393280 DOI: 10.1021/acs.bioconjchem.0c00627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multidrug resistance (MDR) is the main obstacle in cancer chemotherapy. ATP-binding cassette (ABC) transporters can transport a wide range of antitumor drugs out of cells, which is the most common reason in the development of resistance to drugs. Currently, various therapeutic strategies are used to reverse MDR, among which CRISPR/Cas9 gene editing technique is expected to be an effective way. Here, we reviewed the research progress of reversing ABC-mediated drug resistance by CRISPR/Cas9 system.
Collapse
Affiliation(s)
- Wei Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Ze Liang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Pengfei Ma
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Qi Zhao
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Mengyuan Dai
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Jie Zhu
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Hong Xu
- College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Qingyan Chang
- Pharmacy Department, Dalian Sixth People Hospital of Dalian Medical University, Dalian 116031, China
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| |
Collapse
|
32
|
Gao HL, Gupta P, Cui Q, Ashar YV, Wu ZX, Zeng L, Lei ZN, Teng QX, Ashby CR, Guan Y, Chen ZS. Sapitinib Reverses Anticancer Drug Resistance in Colon Cancer Cells Overexpressing the ABCB1 Transporter. Front Oncol 2020; 10:574861. [PMID: 33163405 PMCID: PMC7581728 DOI: 10.3389/fonc.2020.574861] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Abstract
The efficacy of anti-cancer drugs in patients can be attenuated by the development of multi-drug resistance (MDR) due to ATP-binding cassette (ABC) transporters overexpression. In this in vitro study, we determined the reversal efficacy of the epidermal growth factor receptor (EFGR) inhibitor, saptinib, in SW620 and SW720/Ad300 colon cancer cells and HEK293/ABCB1 cells which overexpress the ABCB1 transporter. Sapitinib significantly increased the efficacy of paclitaxel and doxorubicin in ABCB1 overexpressing cells without altering the expression or the subcellular location of the ABCB1 transporter. Sapitinib significantly increased the accumulation of [3H]-paclitaxel in SW620/AD300 cells probably by stimulating ATPase activity which could competitively inhibit the uptake of [3H]-paclitaxel. Furthermore, sapitinib inhibited the growth of resistant multicellular tumor spheroids (MCTS). The docking study indicated that sapitinib interacted with the efflux site of ABCB1 transporter by π-π interaction and two hydrogen bonds. In conclusion, our study suggests that sapitinib surmounts MDR mediated by ABCB1 transporter in cancer cells.
Collapse
Affiliation(s)
- Hai-Ling Gao
- Department of Histology and Embryology, Weifang Medical University, Weifang, China.,Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Pranav Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Qingbin Cui
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Yunali V Ashar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Leli Zeng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States.,Tomas Lindahl Nobel Laureate Laboratory, Research Centre, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Yingjun Guan
- Department of Histology and Embryology, Weifang Medical University, Weifang, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| |
Collapse
|
33
|
Luo H, Cong S, Dong J, Jin L, Jiang D, Wang X, Chen Q, Li F. Paired‑related homeobox 1 overexpression promotes multidrug resistance via PTEN/PI3K/AKT signaling in MCF‑7 breast cancer cells. Mol Med Rep 2020; 22:3183-3190. [PMID: 32945446 PMCID: PMC7453582 DOI: 10.3892/mmr.2020.11414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 06/19/2020] [Indexed: 12/17/2022] Open
Abstract
Multidrug resistance (MDR) is a major cause of disease relapse and mortality in breast cancer. Paired‑related homeobox 1 (PRRX1) is associated with the epithelial‑mesenchymal transition (EMT), which is involved in tumor development, including cell invasion and MDR. However, the effect of PRRX1 on MDR had not clearly established. The present study investigated the influence of PRRX1 on MDR and the underlying molecular mechanisms in MCF‑7 breast cancer cells. MCF‑7 cells were divided into PRRX1+ group (cells transfected with a recombinant plasmid carrying the PRRX1 gene), negative control group (cells transfected with a blank vector) and blank group (untreated cells). It was found that the relative protein and mRNA expression levels of PRRX1, N‑cadherin, vimentin and P‑glycoprotein were significantly higher in PRRX1‑overexpressing MCF‑7 cells compared with those in control cells. The half‑maximal inhibitory concentration of three groups after treatment with docetaxel and cis‑platinum complexes were significantly higher in PRRX1‑overexpressing MCF‑7 cells compared with those in control cells. Furthermore, relative PTEN expression decreased significantly and levels of phosphorylated PI3K and AKT increased substantially in PRRX1‑overexpressing MCF‑7 cells. These results indicated that PRRX1 overexpression may induce MDR via PTEN/PI3K/AKT signaling in breast cancer. It is highly recommended that PRRX1 gene expression detection should be performed in patients with breast cancer to aid the selection of more appropriate treatments, which will lead to an improved prognosis in clinical practice.
Collapse
Affiliation(s)
- Haoyue Luo
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Shaobo Cong
- Department of Biochemistry and Molecular Biology, Medical College of Qingdao University, Qingdao, Shandong 266021, P.R. China
| | - Jiaojiao Dong
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Litao Jin
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Dandan Jiang
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Xingang Wang
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Qingfeng Chen
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Funian Li
- Department of Breast Disease Diagnosis, Treatment Centre, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
34
|
Feng W, Zhang M, Wu ZX, Wang JQ, Dong XD, Yang Y, Teng QX, Chen XY, Cui Q, Yang DH. Erdafitinib Antagonizes ABCB1-Mediated Multidrug Resistance in Cancer Cells. Front Oncol 2020; 10:955. [PMID: 32670878 PMCID: PMC7330633 DOI: 10.3389/fonc.2020.00955] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/15/2020] [Indexed: 12/21/2022] Open
Abstract
ABCB1 overexpression is known to contribute to multidrug resistance (MDR) in cancers. Therefore, it is critical to find effective drugs to target ABCB1 and overcome MDR. Erdafitinib is a tyrosine kinase inhibitor (TKI) of fibroblast growth factor receptor (FGFR) that is approved by the FDA to treat urothelial carcinoma. Previous studies have demonstrated that some TKIs exhibit MDR reversal effect. In this work, we examined whether erdafitinib could reverse MDR mediated by ABCB1. The results of reversal experiments showed that erdafitinib remarkably reversed ABCB1-mediated MDR without affecting ABCG2-mediated MDR. The results of immunofluorescence and Western blot analysis demonstrated that erdafitinib did not affect the expression of ABCB1 or its cellular localization. Further study revealed that erdafitinib inhibited ABCB1 efflux function leading to increasing intracellular drug accumulation, thereby reversing MDR. Furthermore, ATPase assay indicated that erdafitinib activated the ABCB1 ATPase activity. Docking study suggested that erdafitinib interacted with ABCB1 on the drug-binding sites. In summary, this study demonstrated that erdafitinib can reverse MDR mediated by ABCB1, suggesting that combination of erdafitinib and ABCB1-substrate conventional chemotherapeutic drugs could potentially be used to overcome MDR mediated by ABCB1.
Collapse
Affiliation(s)
- Weiguo Feng
- College of Bioscience and Technology, Weifang Medical University, Weifang, China.,Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Meng Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States.,First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Xing-Duo Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Xuan-Yu Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States.,College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| | - Qingbin Cui
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Dong-Hua Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| |
Collapse
|
35
|
Inhibition of Phosphatidylinositol 3-Kinase γ by IPI-549 Attenuates Abdominal Aortic Aneurysm Formation in Mice. Eur J Vasc Endovasc Surg 2020; 60:254-263. [PMID: 32423743 DOI: 10.1016/j.ejvs.2020.03.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/28/2020] [Accepted: 03/15/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signalling pathway plays a pivotal role in abdominal aortic aneurysm (AAA). However, systemic inhibition of this pathway causes serious side effects, thus limiting the clinical use of pan-PI3K inhibitors. In this study, it was hypothesised that the γ subunit of PI3K plays an important role in the PI3K/AKT signalling pathway during AAA, and that specifically targeting PI3Kγ may prevent this process. METHODS Aortic specimens were collected from AAA patients and organ donors. Furthermore, a classical AAA model in male C57BL/6 mice was created via an intra-aortic porcine pancreatic elastase (PPE) infusion and aortas were collected. A specific PI3Kγ inhibitor, IPI-549, was administered to mice orally. The protein expression level of PI3Kγ was examined by immunohistochemistry and western blotting. The aortic leukocytes were examined by immunohistochemistry and flow cytometry. RESULTS PI3Kγ protein levels were elevated in the aortas of AAA patients and PPE infused mice. Three color immunofluorescence staining revealed the predominant area of PI3Kγ by T cells and macrophages in aneurysmal aortas. IPI-549 treatment significantly prevented AAA formation in mice. Aortic macrophages, T cells and neo-angiogenesis were significantly reduced in mice treated with IPI-549 compared with vehicle treated PPE infused mice. Flow cytometry analysis also revealed that CD45+ leukocytes and CD45+ F4/80+ macrophages in IPI-549 treated mouse aortas decreased dramatically. Additionally, IPI-549 treatment inhibited the phosphorylation of AKT in experimental aneurysmal lesions. CONCLUSION Specific inhibition of PI3Kγ limits AAA formation. Targeting PI3Kγ prevents inflammatory cell infiltration through inhibition of AKT phosphorylation in AAA.
Collapse
|
36
|
Liu W, Zhou F, Xia D, Shiloach J. Expression of multidrug transporter P-glycoprotein in Pichia pastoris affects the host's methanol metabolism. Microb Biotechnol 2019; 12:1226-1236. [PMID: 31131547 PMCID: PMC6801151 DOI: 10.1111/1751-7915.13420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 12/05/2022] Open
Abstract
Pichia pastoris KM71H (MutS ) is an efficient producer of hard-to-express proteins such as the membrane protein P-glycoprotein (Pgp), an ATP-powered efflux pump which is expressed properly, but at very low concentration, using the conventional induction strategy. Evaluation of different induction strategies indicated that it was possible to increase Pgp expression by inducing the culture with 20% media containing 2.5% methanol. By quantifying methanol, formaldehyde, hydrogen peroxide and formate, and by measuring alcohol oxidase, catalase, formaldehyde dehydrogenase, formate dehydrogenase, malate dehydrogenase, isocitrate dehydrogenase and α-ketoglutarate dehydrogenases, it was possible to correlate Pgp expression to the induction strategy. Inducing the culture by adding methanol with fresh media was associated with decreases in formaldehyde and hydrogen peroxide, and increases in formaldehyde dehydrogenase, formate dehydrogenase, isocitrate dehydrogenase and α-ketoglutarate dehydrogenases. At these conditions, Pgp expression was 1400-fold higher, an indication that Pgp expression is affected by increases in formaldehyde and hydrogen peroxide. It is possible that Pgp is responsible for this behaviour, since the increased metabolite concentrations and decreased enzymatic activities were not observed when parental Pichia was subjected to the same growth conditions. This report adds information on methanol metabolism during expression of Pgp from P. pastoris MutS strain and suggests an expression procedure for hard-to-express proteins from P. pastoris.
Collapse
Affiliation(s)
- Wan‐cang Liu
- Biotechnology Core LaboratoryNational Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)National Institutes of Health (NIH)BethesdaMD20892USA
| | - Fei Zhou
- Laboratory of Cell BiologyCenter for Cancer Research (CCR)National Cancer Institute (NCI)National Institutes of Health (NIH)BethesdaMD20892USA
| | - Di Xia
- Laboratory of Cell BiologyCenter for Cancer Research (CCR)National Cancer Institute (NCI)National Institutes of Health (NIH)BethesdaMD20892USA
| | - Joseph Shiloach
- Biotechnology Core LaboratoryNational Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)National Institutes of Health (NIH)BethesdaMD20892USA
| |
Collapse
|
37
|
Wang J, Mao N, Liu Y, Xie X, Tian J, Li F, Chen J. Inhibition of miR-16 enhances the sensitivity of fibroblast-like synovial cells to methotrexate by restraining MDR1/P-gp expression via NF-κB pathway. RSC Adv 2019; 9:26619-26627. [PMID: 35528582 PMCID: PMC9070447 DOI: 10.1039/c9ra04991f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/16/2019] [Indexed: 01/19/2023] Open
Abstract
MicroRNAs (miRNAs) are demonstrated to contribute to the regulation of drug resistance in a number of diseases. Nevertheless, little is known about the role and the underlying mechanism of miR-16 in rheumatoid arthritis (RA) methotrexate resistance. In this study, we firstly examined the miR-16 expression in the serum and synovial fluid from RA patients who were unresponsive to methotrexate monotherapy (UR-MTX patients) and responsive RA patients (R-MTX patients). Secondly, the miR-16 expression was measured in both fibroblast-like synovial cells (FLS) and methotrexate resistance RA-FLS cells (FLS-MTX). FLS cells used in this study were isolated from synovial tissue specimens obtained from patients with RA who underwent total joint replacement. FLS-MTX cells were conducted by gradually increasing the concentration of methotrexate in the medium. The construction of FLS-MTX cells was confirmed by 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assay. Thirdly, in order to further investigate the role of miR-16 in FLS-MTX cells, we introduced miR-16 inhibitor into FLS-MTX cells to knockdown the expression of miR-16, used fluorescence quantitative PCR to detect the inhibition efficiency. The effects of miR-16 inhibition on cell viability, cell cycle arrest and apoptosis in FLS-MTX cells were monitored with MTT and flow cytometry analysis, respectively. And the regulation of miR-16 on P-glycoprotein (P-gp) was performed using qRT-PCR, western blotting, and immunofluorescence staining. Fourthly, ammonium pyrrolidinedithiocarbamate (PDTC), a NF-κB pathway inhibitor, was applied to verify the mechanism by which miR-16 involved in to regulate the P-gp expression, and thus contributing to the methotrexate resistance in FLS-MTX cells. MiR-16 was upregulated in the in serum and synovial fluid from UR-MTX patients as well as in FLS-MTX cells. Inhibition of miR-16 re-sensitized the FLS-MTX cells to methotrexate by suppressing the cell viability, cell promoting cycle arrest at G0/G1 phase and enhancing apoptosis. Knockdown of miR-16 significantly reduced MDR1 mRNA expression and P-gp protein expression in FLS-MTX cells. Furthermore, inhibition of NF-κB pathway by PDTC reinforced the effect of miR-16 knockdown on P-gp expression, cell viability, cell cycle arrest and apoptosis. In conclusion, our study illustrated that inhibition of miR-16 in FLS-MTX cells alleviated methotrexate resistance by inhibiting MDR1/P-gp expression through inactivation of the NF-κB pathway. MicroRNAs (miRNAs) are demonstrated to contribute to the regulation of drug resistance in a number of diseases.![]()
Collapse
Affiliation(s)
- Jing Wang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province Kunming 650034 Yunnan Province China.,Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University No. 139 Middle Renmin Road Changsha 410011 Hunan Province China +86-731-85533525 +86-731-85295888
| | - Ni Mao
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University No. 139 Middle Renmin Road Changsha 410011 Hunan Province China +86-731-85533525 +86-731-85295888
| | - Yiming Liu
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University No. 139 Middle Renmin Road Changsha 410011 Hunan Province China +86-731-85533525 +86-731-85295888
| | - Xi Xie
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University No. 139 Middle Renmin Road Changsha 410011 Hunan Province China +86-731-85533525 +86-731-85295888
| | - Jing Tian
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University No. 139 Middle Renmin Road Changsha 410011 Hunan Province China +86-731-85533525 +86-731-85295888
| | - Fen Li
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University No. 139 Middle Renmin Road Changsha 410011 Hunan Province China +86-731-85533525 +86-731-85295888
| | - Jinwei Chen
- Department of Rheumatology and Immunology, The Second Xiangya Hospital of Central South University No. 139 Middle Renmin Road Changsha 410011 Hunan Province China +86-731-85533525 +86-731-85295888
| |
Collapse
|
38
|
Zhang W, Li M, Du W, Yang W, Li G, Zhang C, Liang X, Chen H. Tissue Distribution and Anti-Lung Cancer Effect of 10-Hydroxycamptothecin Combined with Platycodonis Radix and Glycyrrhizae Radix ET Rhizoma. Molecules 2019; 24:E2068. [PMID: 31151274 PMCID: PMC6600312 DOI: 10.3390/molecules24112068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022] Open
Abstract
10-Hydroxycamptothecin (HCPT) is a broad-spectrum chemotherapeutic drug, although its side effects and multidrug resistance (MDR) limit its clinical application. A range of drug delivery systems have been utilized to overcome its shortcomings and maintain its therapeutic efficacy, however the use of the transport effect of traditional Chinese medicines (TCMs) to improve the distribution of chemotherapeutic drugs has not been widely reported. Platycodonis Radix (JG) and Glycyrrhizae Radix ET Rhizoma (GC) are common TCMs in clinics and are often combined as drug pairs to act as "transport agents". In the present study, the effect of JG and GC (JGGC) on the distribution of HCPT in tissues and its antitumor efficacy after being combined as a therapy were investigated, for which ultrahigh-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) was used. Furthermore, the effect on the protein expression of multidrug resistance proteins (P-gp and LRP), and the immunomodulatory and synergistic antiapoptotic effect on Lewis lung cancer-bearing C57BL/6J mice were also evaluated. The results demonstrate that JGGC significantly increased the area under the concentration time curve (AUC) and mean residence time (MRT) and reduced the clearance rate (CL) of HCPT. In addition, the combined use of JGGC decreased the levels of LRP, P-gp and Bcl-2/Bax when treated with HCPT. JGGC also significantly elevated the levels of RBCs, PLTs, HGB, IL-2, and IFN-γ, and decreased IL-10 levels. In summary, an increased concentration of HCPT in tissues was observed when it was combined with JGGC through inhibition of efflux protein, with a synergistic enhancement of the anticancer effect observed through promotion of apoptosis and immunity due to a reversion of the Th1/Th2 shift. Our findings provide a reference for the feasibility of combining JGGC with chemotherapy drugs in clinical applications.
Collapse
Affiliation(s)
- Wugang Zhang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herb Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Mulan Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herb Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Wendi Du
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herb Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Wuliang Yang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Guofeng Li
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herb Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Chen Zhang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herb Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Xinli Liang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| | - Haifang Chen
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China.
| |
Collapse
|
39
|
Wu ZX, Teng QX, Cai CY, Wang JQ, Lei ZN, Yang Y, Fan YF, Zhang JY, Li J, Chen ZS. Tepotinib reverses ABCB1-mediated multidrug resistance in cancer cells. Biochem Pharmacol 2019; 166:120-127. [PMID: 31078601 DOI: 10.1016/j.bcp.2019.05.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
Abstract
Overexpression of ABCB1 transporters plays a crucial role in mediating multidrug resistance (MDR). Therefore, it is important to inhibit ABCB1 activity in order to maintain an effective intracellular level of chemotherapeutic drugs. Tepotinib is a MET tyrosine kinase inhibitor with potential anticancer effect and it is currently in clinical trials. In this study, we investigated whether tepotinib could antagonize ABC transporters-mediated MDR. Our results suggest that tepotinib significantly reversed ABCB1-mediated MDR but not ABCG2- or ABCC1-mediated MDR. Mechanistic studies show that tepotinib significantly reversed ABCB1-mediated MDR by attenuating the efflux activity of ABCB1 transporter. The ATPase assay showed that tepotinib inhibited the ATPase activity of ABCB1 in a concentration-dependent manner. Furthermore, treatment with tepotinib did not change protein expression or subcellular localization of ABCB1. Docking analysis indicated that tepotinib interacted with the drug-binding site of the ABCB1 transporter. Our study provides a potential chemotherapeutic strategy of co-administrating tepotinib with other conventional chemotherapeutic agents to overcome MDR and improve therapeutic effect.
Collapse
Affiliation(s)
- Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Chao-Yun Cai
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Ying-Fang Fan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jian-Ye Zhang
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Jun Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; Department of Otolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
40
|
Ciprofloxacin Enhances the Chemosensitivity of Cancer Cells to ABCB1 Substrates. Int J Mol Sci 2019; 20:ijms20020268. [PMID: 30641875 PMCID: PMC6358874 DOI: 10.3390/ijms20020268] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 12/21/2022] Open
Abstract
ABCB1 is one of the major drug efflux transporters that is known to cause multidrug resistance (MDR) in cancer patients receiving chemotherapy for the treatment of solid tumors and hematological malignancies. Inhibition of ABCB1 efflux function is important for maintaining the intracellular concentration of chemotherapeutic drugs. Here, we evaluated ciprofloxacin for its ability to reverse MDR caused by the overexpression of ABCB1. Cytotoxicity of ciprofloxacin was determined by the MTT assay. The chemosensitizing effects of ciprofloxacin were determined in combination with ABCB1 substrates. The intracellular accumulation and efflux of ABCB1 substrates was measured by a scintillation counter, and protein expression was determined by the Western blotting. Vanadate-sensitive ATPase assay was performed to determine the effect of ciprofloxacin on the ATPase activity of ABCB1, and docking analysis was done to determine the interaction of ciprofloxacin with ABCB1. Ciprofloxacin significantly potentiated the cytotoxic effects of ABCB1 substrates in ABCB1-overexpressing cells. Furthermore, ciprofloxacin increased the intracellular accumulation and decreased the efflux of [³H]-paclitaxel without altering the expression of ABCB1. Ciprofloxacin stimulated the ATPase activity of ABCB1 in a concentration-dependent manner. Our findings showed that ciprofloxacin potently inhibits the ABCB1 efflux function and it has potential to be developed as a combination anticancer therapy.
Collapse
|