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
|
Zhang M, She ML, Chen J, Zeng XQ, Xiong QQ, Cen YH, Ye JA, Qiu GB, Yang SY, Ren GH. Gilteritinib reverses ABCB1-mediated multidrug resistance: Preclinical in vitro and animal investigations. Biomed Pharmacother 2024; 180:117603. [PMID: 39471652 DOI: 10.1016/j.biopha.2024.117603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024] Open
Abstract
Multi-drug resistance (MDR) poses a significant challenge to cancer treatment. Targeting ATP-binding cassette subfamily B member 1 (ABCB1) is a viable strategy for overcoming MDR. This study examined the preclinical in vitro and animal studies that used gilteritinib, a FLT3 inhibitor that reverses ABCB1-mediated MDR. At nontoxic levels, gilteritinib significantly increased the susceptibility of cancer cells overexpressing ABCB1 to chemotherapeutic drugs. Furthermore, it impaired the development of drug-resistant cell colonies and 3D spheroids. Studies on the reversal mechanism have shown that gilteritinib can directly bind to the drug-binding site of ABCB1, inhibiting drug efflux activity. Consequently, the substrate's drug cytotoxicity increases in MDR cells. Furthermore, gilteritinib increased ATPase activity while leaving ABCB1 expression and subcellular distribution unchanged and inhibited AKT or ERK activation. Docking analysis indicated that Gilteritinib could interact with the drug-binding site of the ABCB1 transporter. In vivo studies have shown that gilteritinib improves the antitumor efficacy of paclitaxel in nude mice without obvious toxic effects. In conclusion, our preclinical investigations show that gilteritinib has the potential to successfully overcome ABCB1-mediated MDR in a clinical environment when combined with substrate medicines.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China
| | - Mei-Ling She
- Department of Gastroenterology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100020, China
| | - Jun Chen
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China
| | - Xiao-Qi Zeng
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China
| | - Qing-Quan Xiong
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China
| | - Ying-Huan Cen
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China
| | - Jia-An Ye
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China
| | - Guo-Bin Qiu
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China.
| | - Shu-Yi Yang
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China.
| | - Guang-Hui Ren
- Department of Thyroid And Breast Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong 510000, China.
| |
Collapse
|
3
|
Zhang M, Huang MN, Dong XD, Cui QB, Yan Y, She ML, Feng WG, Zhao XS, Wang DT. Overexpression of ABCB1 confers resistance to FLT3 inhibitor FN-1501 in cancer cells: in vitro and in vivo characterization. Am J Cancer Res 2023; 13:6026-6037. [PMID: 38187048 PMCID: PMC10767331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024] Open
Abstract
FN-1501 is a potent FLT3 inhibitor with antitumor activity. A phase 1 trial of FN-1501 monotherapy in patients with advanced solid tumors and R/R AML is in progress. Since one of the primary causes of multidrug resistance (MDR) is the overexpression of ATP-binding cassette superfamily B member 1 (ABCB1), the objective of this study was to investigate the potential relationship between FN-1501 and the ABCB1 transporter. We found ABCB1 overexpressing-cancer cells conferred FN-1501 resistance, which could be reversed by an ABCB1 inhibitor. Molecular docking study revealed that FN-1501 docked the ligand binding site with an affinity score of -9.77 kcal/mol, denoting a strong interaction between FN-1501 and ABCB1. Additionally, the ABCB1 ATPase assay indicated that FN-1501 could significantly stimulate ABCB1 ATPase activity. Furthermore, we observed a similar trend of ABCB1-facilated FN-1501 resistance in tumor-bearing mice model. In sum, we demonstrate that FN-1501 is a substrate of ABCB1 transporter from both in vivo and in vitro studies. Therefore, our findings provide new insight on the mechanism of chemoresistance due to ABCB1 overexpression.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Hospital of Southern Medical UniversityNo. 1333 Xinhu Road, Baoan, Shenzhen 510000, Guangdong, China
| | - Min-Na Huang
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine134 Research Park Dr, Columbia, MO 65211, USA
| | - Xing-Duo Dong
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s UniversityQueens, NY 11439, USA
| | - Qing-Bin Cui
- Department of Cancer Biology, University of Toledo College of Medicine and Life SciencesToledo, OH 43606, USA
| | - Yan Yan
- Department of Traditional Chinese Medicine, Shenzhen Hospital of Southern Medical UniversityNo. 1333 Xinhu Road, Baoan, Shenzhen 510000, Guangdong, China
| | - Mei-Ling She
- Department of Traditional Chinese Medicine, Shenzhen Hospital of Southern Medical UniversityNo. 1333 Xinhu Road, Baoan, Shenzhen 510000, Guangdong, China
| | - Wei-Guo Feng
- School of Life Science and Technology, Weifang Medical UniversityNo. 7166 Baotong Street, Weicheng, Weifang 261053, Shandong, China
| | - Xiao-Shan Zhao
- School of Traditional Chinese Medicine, Southern Medical UniversityNo. 1023 Satai South Road, Baiyun, Guangzhou 510000, Guangdong, China
| | - Dong-Tao Wang
- Department of Traditional Chinese Medicine, Shenzhen Hospital of Southern Medical UniversityNo. 1333 Xinhu Road, Baoan, Shenzhen 510000, Guangdong, China
- School of Traditional Chinese Medicine, Southern Medical UniversityNo. 1023 Satai South Road, Baiyun, Guangzhou 510000, Guangdong, China
| |
Collapse
|
4
|
Poustforoosh A, Moosavi F. Evaluation of the FDA-approved kinase inhibitors to uncover the potential repurposing candidates targeting ABC transporters in multidrug-resistant cancer cells: an in silico approach. J Biomol Struct Dyn 2023; 42:13650-13662. [PMID: 37942620 DOI: 10.1080/07391102.2023.2277848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
Multiple drug resistance (MDR) is characterized by the resistance of cancer cells to a broad spectrum of anticancer drugs. The main mechanism underlying the MDR phenotype is the overexpression of ATP-binding cassette (ABC) transporters by promoting active drug efflux from cancer cells. Some small-molecule protein kinase inhibitors have been found to overcome MDR by inhibiting ABC transporters as substrates or modulators. This study investigated the chemical activity of 58 FDA-approved anticancer kinase inhibitors against three multidrug resistance-related proteins. The studied proteins are ATP-Binding Cassette Sub-Family B Member 1 (ABCB1), ATP-Binding Cassette Subfamily C Member 1 (ABCC1), and ATP-binding cassette superfamily G member 2 (ABCG2). The drug-binding domain and ATP binding sites of the proteins were considered the kinase inhibitors' probable target. High-throughput virtual screening and molecular docking were employed to find the hit drugs, and the drugs with the highest binding affinity were further evaluated using the molecular dynamics (MD) simulation. The virtual screening revealed that several kinase inhibitors could be considered potential inhibitors of ABCB1, ABCC1, and ABCG2, among which larotrectinib, entrectinib, and infigratinib showed the highest binding affinity, respectively. Based on the obtained results from MD simulation, these drugs can form strong interactions with the essential residues of the target proteins. In silico investigation revealed that larotrectinib, entrectinib, and infigratinib can target the key residues of the studied proteins. Therefore, these approved kinase inhibitors could be considered potential therapies for MDR cancers by targeting these transporters.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
5
|
Wu CP, Hsiao SH, Wu YS. Perspectives on drug repurposing to overcome cancer multidrug resistance mediated by ABCB1 and ABCG2. Drug Resist Updat 2023; 71:101011. [PMID: 37865067 DOI: 10.1016/j.drup.2023.101011] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/23/2023]
Abstract
The overexpression of the human ATP-binding cassette (ABC) transporters in cancer cells is a common mechanism involved in developing multidrug resistance (MDR). Unfortunately, there are currently no approved drugs specifically designed to treat multidrug-resistant cancers, making MDR a significant obstacle to successful chemotherapy. Despite over two decades of research, developing transporter-specific inhibitors for clinical use has proven to be a challenging endeavor. As an alternative approach, drug repurposing has gained traction as a more practical method to discover clinically effective modulators of drug transporters. This involves exploring new indications for already-approved drugs, bypassing the lengthy process of developing novel synthetic inhibitors. In this context, we will discuss the mechanisms of ABC drug transporters ABCB1 and ABCG2, their roles in cancer MDR, and the inhibitors that have been evaluated for their potential to reverse MDR mediated by these drug transporters. Our focus will be on providing an up-to-date report on approved drugs tested for their inhibitory activities against these drug efflux pumps. Lastly, we will explore the challenges and prospects of repurposing already approved medications for clinical use to overcome chemoresistance in patients with high tumor expression of ABCB1 and/or ABCG2.
Collapse
Affiliation(s)
- Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, 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; Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan.
| | - Sung-Han Hsiao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan.
| |
Collapse
|
6
|
Solana-Altabella A, Megías-Vericat JE, Ballesta-López O, Martínez-Cuadrón D, Montesinos P. Drug-drug interactions associated with FLT3 inhibitors for acute myeloblastic leukemia: current landscape. Expert Rev Clin Pharmacol 2023; 16:133-148. [PMID: 36708283 DOI: 10.1080/17512433.2023.2174523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION FLT3 inhibitors (FLT3i) are drugs in which there is limited experience and not yet enough information on the mechanisms of absorption, transport, and elimination; but especially on the potential drug-drug interactions (DDIs). There are therefore risks in the management of FLT3i DDIs (i.e. sorafenib, ponatinib, crenolanib, midostaurin, quizartinib, and gilteritinib) and ignoring them can compromise therapeutic success in acute myeloid leukemia (AML) treatment, in complex patients and secondary pathologies. AREAS COVERED This review summarizes the DDIs of FLT3i with P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), organic anion transporting (OAT), organic cationic transporting (OCT), cytochrome P450 (CYP) subunits, and other minor metabolic/transport pathways. EMBASE, PubMed, the Cochrane Central Register and the Web of Science were searched. The last literature search was performed on the 14 February 2022. EXPERT OPINION FLT3i will be combined with other therapeutic agents (supportive care, doublet, or triplet therapy) and in different clinical settings, which means a greater chance of controlling and even eradicating the disease effectively, but also an increased risk to patients due to potential DDIs. Healthcare professionals should be aware of the potential interactions that may occur and be vigilant in monitoring those patients who are receiving any potentially interacting drug.
Collapse
Affiliation(s)
- Antonio Solana-Altabella
- Servicio de Farmacia Área del Medicamento, Hospital Universitari i Politècnic La Fe Av. Valencia, Spain.,Grupo de Investigación en Hematología y Hemoterapia, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain
| | | | - Octavio Ballesta-López
- Servicio de Farmacia Área del Medicamento, Hospital Universitari i Politècnic La Fe Av. Valencia, Spain.,Grupo de Investigación en Hematología y Hemoterapia, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain
| | - David Martínez-Cuadrón
- Grupo de Investigación en Hematología y Hemoterapia, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain.,Servicio de Hematología y Hemoterapia Hospital Universitari i Politècnic La Fe. Valencia Spain
| | - Pau Montesinos
- Grupo de Investigación en Hematología y Hemoterapia, Instituto de Investigación Sanitaria La Fe (IISLAFE), Valencia, Spain.,Servicio de Hematología y Hemoterapia Hospital Universitari i Politècnic La Fe. Valencia Spain
| |
Collapse
|
7
|
Wu CP, Hsieh YJ, Tseng HY, Huang YH, Li YQ, Hung TH, Wang SP, Wu YS. The WD repeat-containing protein 5 (WDR5) antagonist WDR5-0103 restores the efficacy of cytotoxic drugs in multidrug-resistant cancer cells overexpressing ABCB1 or ABCG2. Biomed Pharmacother 2022; 154:113663. [PMID: 36081287 DOI: 10.1016/j.biopha.2022.113663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/02/2022] Open
Abstract
The development of multidrug resistance (MDR) is one of the major challenges in the treatment of cancer which is caused by the overexpression of the ATP-binding cassette (ABC) transporters ABCB1 (P-glycoprotein) and/or ABCG2 (BCRP/MXR/ABCP) in cancer cells. These transporters are capable of reducing the efficacy of cytotoxic drugs by actively effluxing them out of cancer cells. Since there is currently no approved treatment for patients with multidrug-resistant tumors, the drug repurposing approach provides an alternative route to identify agents to reverse MDR mediated by ABCB1 and/or ABCG2 in multidrug-resistant cancer cells. WDR5-0103 is a histone H3 lysine 4 (H3K4) methyltransferase inhibitor that disrupts the interaction between the WD repeat-containing protein 5 (WDR5) and mixed-lineage leukemia (MLL) protein. In this study, the effect of WDR5-0103 on MDR mediated by ABCB1 and ABCG2 was determined. We found that in a concentration-dependent manner, WDR5-0103 could sensitize ABCB1- and ABCG2-overexpressing multidrug-resistant cancer cells to conventional cytotoxic drugs. Our results showed that WDR5-0103 reverses MDR and improves drug-induced apoptosis in multidrug-resistant cancer cells by inhibiting the drug-efflux function of ABCB1 and ABCG2, without altering the protein expression of ABCB1 or ABCG2. The potential sites of interactions of WDR5-0103 with the drug-binding pockets of ABCB1 and ABCG2 were predicted by molecular docking. In conclusion, the MDR reversal activity of WDR5-0103 demonstrated here indicates that it could be used in combination therapy to provide benefits to a subset of patients with tumor expressing high levels of ABCB1 or ABCG2.
Collapse
Affiliation(s)
- Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Molecular and Medicine Research Center, Chang Gung University, Taoyuan, Taiwan; Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.
| | - Ya-Ju Hsieh
- Molecular and Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.
| | - Han-Yu Tseng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Yang-Hui Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Yan-Qing Li
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan; Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan.
| | - Shun-Ping Wang
- Department of Orthopedics, Taichung Veterans General Hospital, Taichung, Taiwan.
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung, Taiwan.
| |
Collapse
|
8
|
Morell A, Budagaga Y, Vagiannis D, Zhang Y, Laštovičková L, Novotná E, Haddad A, Haddad M, Portillo R, Hofman J, Wsól V. Isocitrate dehydrogenase 2 inhibitor enasidenib synergizes daunorubicin cytotoxicity by targeting aldo-keto reductase 1C3 and ATP-binding cassette transporters. Arch Toxicol 2022; 96:3265-3277. [PMID: 35972551 DOI: 10.1007/s00204-022-03359-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/03/2022] [Indexed: 12/01/2022]
Abstract
Targeting mutations that trigger acute myeloid leukaemia (AML) has emerged as a refined therapeutic approach in recent years. Enasidenib (Idhifa) is the first selective inhibitor of mutated forms of isocitrate dehydrogenase 2 (IDH2) approved against relapsed/refractory AML. In addition to its use as monotherapy, a combination trial of enasidenib with standard intensive induction therapy (daunorubicin + cytarabine) is being evaluated. This study aimed to decipher enasidenib off-target molecular mechanisms involved in anthracycline resistance, such as reduction by carbonyl reducing enzymes (CREs) and drug efflux by ATP-binding cassette (ABC) transporters. We analysed the effect of enasidenib on daunorubicin (Daun) reduction by several recombinant CREs and different human cell lines expressing aldo-keto reductase 1C3 (AKR1C3) exogenously (HCT116) or endogenously (A549 and KG1a). Additionally, A431 cell models overexpressing ABCB1, ABCG2, or ABCC1 were employed to evaluate enasidenib modulation of Daun efflux. Furthermore, the potential synergism of enasidenib over Daun cytotoxicity was quantified amongst all the cell models. Enasidenib selectively inhibited AKR1C3-mediated inactivation of Daun in vitro and in cell lines expressing AKR1C3, as well as its extrusion by ABCB1, ABCG2, and ABCC1 transporters, thus synergizing Daun cytotoxicity to overcome resistance. This work provides in vitro evidence on enasidenib-mediated targeting of the anthracycline resistance actors AKR1C3 and ABC transporters under clinically achievable concentrations. Our findings may encourage its combination with intensive chemotherapy and even suggest that the effectiveness of enasidenib as monotherapy against AML could lie beyond the targeting of mIDH2.
Collapse
Affiliation(s)
- Anselm Morell
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Youssif Budagaga
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Dimitrios Vagiannis
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Yu Zhang
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Lenka Laštovičková
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Eva Novotná
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Andrew Haddad
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Melodie Haddad
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Ramon Portillo
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Jakub Hofman
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Vladimír Wsól
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic.
| |
Collapse
|
9
|
Zheng Y, Yuan D, Zhang F, Tang R. A systematic pan-cancer analysis of the gasdermin (GSDM) family of genes and their correlation with prognosis, the tumor microenvironment, and drug sensitivity. Front Genet 2022; 13:926796. [PMID: 36003332 PMCID: PMC9393220 DOI: 10.3389/fgene.2022.926796] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Pyroptosis is a programmed cell death process mediated by the gasdermin (GSDM) protein. However, limited research has been conducted to comprehensively analyze the contribution of the GSDM family in a pan-cancer setting. Methods: We systematically evaluated the gene expression, genetic variations, and prognostic values of the GSDM family members. Furthermore, we investigated the association between the expression of GSDM genes and immune subtypes, the tumor microenvironment (TME), the stemness index, and cancer drug sensitivities by means of a pan-cancer analysis. Results: GSDM genes were highly upregulated in most of the tested cancers. Low-level mutation frequencies within GSDM genes were common across the examined types of cancer, and their expression levels were associated with prognosis, clinical characteristics, TME features, and stemness scores in several cancer types, particularly those of the urinary system. Importantly, we found that the expressions of GSDMB, GSDMC, and GSDMD were higher in kidney carcinomas, and specifically kidney renal clear cell carcinoma (KIRC); which adversely impacted the patient outcome. We showed that GSDMD was potentially the most useful biomarker for KIRC. The drug sensitivity analysis demonstrated that the expressions of GSDM genes were correlated with the sensitivity of tumor cells to treatment with chemotherapy drugs nelarabine, fluphenazine, dexrazoxane, bortezomib, midostaurin, and vincristine. Conclusion: GSDM genes were associated with tumor behaviors and may participate in carcinogenesis. The results of this study may therefore provide new directions for further investigating the role of GSDM genes as therapeutic targets in a pan-cancer setting.
Collapse
|
10
|
Sucha S, Sorf A, Svoren M, Vagiannis D, Ahmed F, Visek B, Ceckova M. ABCB1 as a potential beneficial target of midostaurin in acute myeloid leukemia. Biomed Pharmacother 2022; 150:112962. [PMID: 35462331 DOI: 10.1016/j.biopha.2022.112962] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 11/25/2022] Open
Abstract
Low curability of patients diagnosed with acute myeloid leukemia (AML) must be seen as a call for better understanding the disease's mechanisms and improving the treatment strategy. Therapeutic outcome of the crucial anthracycline-based induction therapy often can be compromised by a resistant phenotype associated with overexpression of ABCB1 transporters. Here, we evaluated clinical relevance of ABCB1 in a context of the FMS-like tyrosine kinase 3 (FLT3) inhibitor midostaurin in a set of 28 primary AML samples. ABCB1 gene expression was absolutely quantified, confirming its association with CD34 positivity, adverse cytogenetic risk, and unachieved complete remission (CR). Midostaurin, identified as an ABCB1 inhibitor, increased anthracycline accumulation in peripheral blood mononuclear cells (PBMC) of CD34+ AML patients and those not achieving CR. This effect was independent of FLT3 mutation, indicating even FLT3- AML patients might benefit from midostaurin therapy. In line with these data, midostaurin potentiated proapoptotic processes in ABCB1-overexpressing leukemic cells when combined with anthracyclines. Furthermore, we report a direct linkage of miR-9 to ABCB1 efflux activity in the PBMC and propose miR-9 as a useful prognostic marker in AML. Overall, we highlight the therapeutic value of midostaurin as more than just a FLT3 inhibitor, suggesting its maximal therapeutic outcomes might be very sensitive to proper timing and well-optimized dosage schemes based upon patient's characteristics, such as CD34 positivity and ABCB1 activity. Moreover, we suggest miR-9 as a predictive ABCB1-related biomarker that could be immensely helpful in identifying ABCB1-resistant AML phenotype to enable optimized therapeutic regimen and improved treatment outcome.
Collapse
Affiliation(s)
- Simona Sucha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| | - Ales Sorf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| | - Martin Svoren
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| | - Dimitrios Vagiannis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| | - Fahda Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| | - Benjamin Visek
- 4th Department of Internal Medicine - Hematology, University Hospital Hradec Kralove, Sokolska 581, 50005 Hradec Kralove, Czech Republic
| | - Martina Ceckova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic.
| |
Collapse
|
11
|
Pinto-Merino Á, Labrador J, Zubiaur P, Alcaraz R, Herrero MJ, Montesinos P, Abad-Santos F, Saiz-Rodríguez M. Role of Pharmacogenetics in the Treatment of Acute Myeloid Leukemia: Systematic Review and Future Perspectives. Pharmaceutics 2022; 14:pharmaceutics14030559. [PMID: 35335935 PMCID: PMC8954545 DOI: 10.3390/pharmaceutics14030559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease characterized by remarkable toxicity and great variability in response to treatment. Plenteous pharmacogenetic studies have already been published for classical therapies, such as cytarabine or anthracyclines, but such studies remain scarce for newer drugs. There is evidence of the relevance of polymorphisms in response to treatment, although most studies have limitations in terms of cohort size or standardization of results. The different responses associated with genetic variability include both increased drug efficacy and toxicity and decreased response or resistance to treatment. A broad pharmacogenetic understanding may be useful in the design of dosing strategies and treatment guidelines. The aim of this study is to perform a review of the available publications and evidence related to the pharmacogenetics of AML, compiling those studies that may be useful in optimizing drug administration.
Collapse
Affiliation(s)
| | - Jorge Labrador
- Research Unit, Fundación Burgos por la Investigación de la Salud (FBIS), Hospital Universitario de Burgos, 09006 Burgos, Spain; (J.L.); (R.A.)
- Haematology Department, Hospital Universitario de Burgos, 09006 Burgos, Spain
- Facultad de Ciencias de la Salud, Universidad Isabel I, 09003 Burgos, Spain
| | - Pablo Zubiaur
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain; (P.Z.); (F.A.-S.)
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IP), Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
| | - Raquel Alcaraz
- Research Unit, Fundación Burgos por la Investigación de la Salud (FBIS), Hospital Universitario de Burgos, 09006 Burgos, Spain; (J.L.); (R.A.)
| | - María José Herrero
- Pharmacogenetics Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain;
| | - Pau Montesinos
- Haematology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain;
| | - Francisco Abad-Santos
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain; (P.Z.); (F.A.-S.)
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IP), Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
| | - Miriam Saiz-Rodríguez
- Department of Health Sciences, University of Burgos, 09001 Burgos, Spain;
- Research Unit, Fundación Burgos por la Investigación de la Salud (FBIS), Hospital Universitario de Burgos, 09006 Burgos, Spain; (J.L.); (R.A.)
- Correspondence: ; Tel.: +34-947-281-800 (ext. 36078)
| |
Collapse
|
12
|
Faltracco M, Damian M, Ruijter E. Synthesis of Carbazoles and Dihydrocarbazoles by a Divergent Cascade Reaction of Donor-Acceptor Cyclopropanes. Org Lett 2021; 23:7592-7596. [PMID: 34543040 PMCID: PMC8491164 DOI: 10.1021/acs.orglett.1c02795] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
An alkylation/olefination
cascade of indolecarboxaldehydes and
phosphonate-functionalized donor–acceptor cyclopropanes affords
functionalized dihydrocarbazoles and cyclohepta[cd]indoles in formal (3 + 3) and (4 + 3) cycloadditions. A minor modification
to the reaction conditions also allows access to the fully aromatic
heterocyclic scaffolds by thermal loss of an electron-rich aryl moiety.
Collapse
Affiliation(s)
- Matteo Faltracco
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecular & Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Matteo Damian
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecular & Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Eelco Ruijter
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecular & Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
13
|
The Second-Generation PIM Kinase Inhibitor TP-3654 Resensitizes ABCG2-Overexpressing Multidrug-Resistant Cancer Cells to Cytotoxic Anticancer Drugs. Int J Mol Sci 2021; 22:ijms22179440. [PMID: 34502348 PMCID: PMC8431370 DOI: 10.3390/ijms22179440] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022] Open
Abstract
Human ATP-binding cassette (ABC) subfamily G member 2 (ABCG2) mediates the transport of a wide variety of conventional cytotoxic anticancer drugs and molecular targeted agents. Consequently, the overexpression of ABCG2 in cancer cells is linked to the development of the multidrug resistance (MDR) phenotype. TP-3654 is an experimental second-generation inhibitor of PIM kinase that is currently under investigation in clinical trials to treat advanced solid tumors and myelofibrosis. In this study, we discovered that by attenuating the drug transport function of ABCG2, TP-3654 resensitizes ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic ABCG2 substrate drugs topotecan, SN-38 and mitoxantrone. Moreover, our results indicate that ABCG2 does not mediate resistance to TP-3654 and may not play a major role in the induction of resistance to TP-3654 in cancer patients. Taken together, our findings reveal that TP-3654 is a selective, potent modulator of ABCG2 drug efflux function that may offer an additional combination therapy option for the treatment of multidrug-resistant cancers.
Collapse
|
14
|
Wu CP, Murakami M, Wu YS, Chi YC, Hsiao SH, Huang YH, Hung TH, Ambudkar SV. Branebrutinib (BMS-986195), a Bruton's Tyrosine Kinase Inhibitor, Resensitizes P-Glycoprotein-Overexpressing Multidrug-Resistant Cancer Cells to Chemotherapeutic Agents. Front Cell Dev Biol 2021; 9:699571. [PMID: 34350184 PMCID: PMC8326665 DOI: 10.3389/fcell.2021.699571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/28/2021] [Indexed: 12/02/2022] Open
Abstract
The overexpression of P-glycoprotein (P-gp/ABCB1), an ATP-binding cassette (ABC) drug transporter, often contributes to the development of multidrug resistance (MDR) in cancer cells. P-gp mediates the ATP hydrolysis-dependent efflux of a wide range of chemotherapeutic agents out of cancer cells, thereby reducing the intracellular drug accumulation and decreasing the chemosensitivity of these multidrug-resistant cancer cells. Studies with tyrosine kinase inhibitors (TKIs) in P-gp-overexpressing cells have shown that certain TKIs could reverse MDR mediated by P-gp, while some TKIs are transported by P-gp. In the present work, we explored the prospect of repositioning branebrutinib (BMS-986195), a highly selective inhibitor of Bruton’s tyrosine kinase (BTK), to resensitize P-gp-overexpressing multidrug-resistant cancer cells to chemotherapeutic agents. Our results demonstrated that branebrutinib is capable of reversing P-gp-mediated MDR at sub-toxic concentrations, most likely by directly inhibiting the drug transport function of P-gp. Our findings were supported by the result of branebrutinib stimulating the ATPase activity of P-gp in a concentration-dependent manner and the in silico study of branebrutinib binding to the substrate-binding pocket of P-gp. In addition, we found that branebrutinib is equally cytotoxic to drug-sensitive parental cell lines and the respective P-gp-overexpressing multidrug-resistant variants, suggesting that it is unlikely that the overexpression of P-gp in cancer cells plays a significant role in reduced susceptibility or resistance to branebrutinib. In summary, we discovered an additional pharmacological action of branebrutinib against the activity of P-gp, which should be investigated further in future drug combination studies.
Collapse
Affiliation(s)
- Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Megumi Murakami
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung, Taiwan
| | - Ya-Chen Chi
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Sung-Han Hsiao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Yang-Hui Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
15
|
Faltracco M, Ortega-Rosales S, Janssen E, Cioc RC, Vande Velde CML, Ruijter E. Synthesis of Carbazoles by a Diverted Bischler-Napieralski Cascade Reaction. Org Lett 2021; 23:3100-3104. [PMID: 33787266 PMCID: PMC8056386 DOI: 10.1021/acs.orglett.1c00785] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
An unforeseen twist
in a seemingly trivial Bischler–Napieralski
reaction led to the selective formation of an unexpected carbazole
product. The reaction proved to be general, providing access to a
range of diversely substituted carbazoles from readily available substrates.
Judicious variation of substituents revealed a complex cascade mechanism
comprising no less than 10 elementary steps, that could be diverted
in multiple ways toward various other carbazole derivatives.
Collapse
Affiliation(s)
- Matteo Faltracco
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecular & Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Said Ortega-Rosales
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecular & Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Elwin Janssen
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecular & Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Răzvan C Cioc
- Organic Chemistry and Catalysis, Debye Institute for Nanomaterials Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Christophe M L Vande Velde
- Faculty of Applied Engineering, iPRACS, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerpen, Belgium
| | - Eelco Ruijter
- Department of Chemistry & Pharmaceutical Sciences, Amsterdam Institute of Molecular & Life Sciences (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
16
|
Zhu YX, Jia HR, Duan QY, Wu FG. Nanomedicines for combating multidrug resistance of cancer. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1715. [PMID: 33860622 DOI: 10.1002/wnan.1715] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Chemotherapy typically involves the use of specific chemodrugs to inhibit the proliferation of cancer cells, but the frequent emergence of a variety of multidrug-resistant cancer cells poses a tremendous threat to our combat against cancer. The fundamental causes of multidrug resistance (MDR) have been studied for decades, and can be generally classified into two types: one is associated with the activation of diverse drug efflux pumps, which are responsible for translocating intracellular drug molecules out of the cells; the other is linked with some non-efflux pump-related mechanisms, such as antiapoptotic defense, enhanced DNA repair ability, and powerful antioxidant systems. To overcome MDR, intense efforts have been made to develop synergistic therapeutic strategies by introducing MDR inhibitors or combining chemotherapy with other therapeutic modalities, such as phototherapy, gene therapy, and gas therapy, in the hope that the drug-resistant cells can be sensitized toward chemotherapeutics. In particular, nanotechnology-based drug delivery platforms have shown the potential to integrate multiple therapeutic agents into one system. In this review, the focus was on the recent development of nanostrategies aiming to enhance the efficiency of chemotherapy and overcome the MDR of cancer in a synergistic manner. Different combinatorial strategies are introduced in detail and the advantages as well as underlying mechanisms of why these strategies can counteract MDR are discussed. This review is expected to shed new light on the design of advanced nanomedicines from the angle of materials and to deepen our understanding of MDR for the development of more effective anticancer strategies. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Ya-Xuan Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Hao-Ran Jia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Qiu-Yi Duan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
17
|
Bruton's Tyrosine Kinase Inhibitors Ibrutinib and Acalabrutinib Counteract Anthracycline Resistance in Cancer Cells Expressing AKR1C3. Cancers (Basel) 2020; 12:cancers12123731. [PMID: 33322571 PMCID: PMC7764606 DOI: 10.3390/cancers12123731] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 02/08/2023] Open
Abstract
Simple Summary The enzyme aldo-keto reductase 1C3 (AKR1C3) is present in several cancers, in which it is capable of actively metabolising different chemotherapy drugs and decreasing their cytotoxic effects. Therefore, the combination with specific inhibitors of AKR1C3 might prevent drug metabolism and increase its efficacy. We investigated the ability of Bruton’s tyrosine kinase inhibitors ibrutinib and acalabrutinib to block the AKR1C3 mediated inactivation of the anthracycline daunorubicin. Experimentation with recombinant AKR1C3 and different cancer cells expressing this enzyme outlined BTK-inhibitors as potential partners to synergise daunorubicin cytotoxicity in vitro. This evidence could be useful to improve the clinical outcome of anthracycline-based chemotherapies. Abstract Over the last few years, aldo-keto reductase family 1 member C3 (AKR1C3) has been associated with the emergence of multidrug resistance (MDR), thereby hindering chemotherapy against cancer. In particular, impaired efficacy of the gold standards of induction therapy in acute myeloid leukaemia (AML) has been correlated with AKR1C3 expression, as this enzyme metabolises several drugs including anthracyclines. Therefore, the development of selective AKR1C3 inhibitors may help to overcome chemoresistance in clinical practice. In this regard, we demonstrated that Bruton’s tyrosine kinase (BTK) inhibitors ibrutinib and acalabrutinib efficiently prevented daunorubicin (Dau) inactivation mediated by AKR1C3 in both its recombinant form as well as during its overexpression in cancer cells. This revealed a synergistic effect of BTK inhibitors on Dau cytotoxicity in cancer cells expressing AKR1C3 both exogenously and endogenously, thus reverting anthracycline resistance in vitro. These findings suggest that BTK inhibitors have a novel off-target action, which can be exploited against leukaemia through combination regimens with standard chemotherapeutics like anthracyclines.
Collapse
|
18
|
Selective inhibition of aldo-keto reductase 1C3: a novel mechanism involved in midostaurin and daunorubicin synergism. Arch Toxicol 2020; 95:67-78. [PMID: 33025066 DOI: 10.1007/s00204-020-02884-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022]
Abstract
Midostaurin is an FMS-like tyrosine kinase 3 receptor (FLT3) inhibitor that provides renewed hope for treating acute myeloid leukaemia (AML). The limited efficacy of this compound as a monotherapy contrasts with that of its synergistic combination with standard cytarabine and daunorubicin (Dau), suggesting a therapeutic benefit that is not driven only by FLT3 inhibition. In an AML context, the activity of the enzyme aldo-keto reductase 1C3 (AKR1C3) is a crucial factor in chemotherapy resistance, as it mediates the intracellular transformation of anthracyclines to less active hydroxy metabolites. Here, we report that midostaurin is a potent inhibitor of Dau inactivation mediated by AKR1C3 in both its recombinant form as well as during its overexpression in a transfected cell model. Likewise, in the FLT3- AML cell line KG1a, midostaurin was able to increase the cellular accumulation of Dau and significantly decrease its metabolism by AKR1C3 simultaneously. The combination of those mechanisms increased the nuclear localization of Dau, thus synergizing its cytotoxic effects on KG1a cells. Our results provide new in vitro evidence of how the therapeutic activity of midostaurin could operate beyond targeting the FLT3 receptor.
Collapse
|
19
|
Megías-Vericat JE, Solana-Altabella A, Ballesta-López O, Martínez-Cuadrón D, Montesinos P. Drug-drug interactions of newly approved small molecule inhibitors for acute myeloid leukemia. Ann Hematol 2020; 99:1989-2007. [PMID: 32683457 DOI: 10.1007/s00277-020-04186-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/13/2020] [Indexed: 01/13/2023]
Abstract
Several small molecule inhibitors (SMIs) have been recently approved for AML patients. These targeted therapies could be more tolerable than classical antineoplastics, but potential drug-drug interactions (DDI) are relatively frequent. Underestimation or lack of appropriate awareness and management of DDIs with SMIs can jeopardize therapeutic success in AML patients, which often require multiple concomitant medications in the context of prior comorbidities or for the prevention and treatment of infectious and other complications. In this systematic review, we analyze DDIs of glasdegib, venetoclax, midostaurin, quizartinib, gilteritinib, enasidenib, and ivosidenib. CYP3A4 is the main enzyme responsible for SMIs metabolism, and strong CYP3A4 inhibitors, such azoles, could increase drug exposure and toxicity; therefore dose adjustments (venetoclax, quizartinib, and ivosidenib) or alternative therapies or close monitoring (glasdegib, midostaurin, and gilteritinib) are recommended. Besides, coadministration of strong CYP3A4 inducers with SMIs should be avoided due to potential decrease of efficacy. Regarding tolerability, QTc prolongation is frequently observed for most of approved SMIs, and drugs with a potential to prolong the QTc interval and CYP3A4 inhibitors should be avoided and replaced by alternative treatments. In this study, we critically assess the DDIs of SMIs, and we summarize best management options for these new drugs and concomitant medications.
Collapse
Affiliation(s)
- Juan Eduardo Megías-Vericat
- Servicio de Farmacia, Área del Medicamento, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Antonio Solana-Altabella
- Servicio de Farmacia, Área del Medicamento, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - Octavio Ballesta-López
- Servicio de Farmacia, Área del Medicamento, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, 106, 46026, Valencia, Spain
| | - David Martínez-Cuadrón
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, 106, 46026, Valencia, Spain.,CIBERONC, Instituto Carlos III, Madrid, Spain
| | - Pau Montesinos
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, 106, 46026, Valencia, Spain. .,CIBERONC, Instituto Carlos III, Madrid, Spain.
| |
Collapse
|
20
|
Wu ZX, Yang Y, Wang G, Wang JQ, Teng QX, Sun L, Lei ZN, Lin L, Chen ZS, Zou C. Dual TTK/CLK2 inhibitor, CC-671, selectively antagonizes ABCG2-mediated multidrug resistance in lung cancer cells. Cancer Sci 2020; 111:2872-2882. [PMID: 32478948 PMCID: PMC7419038 DOI: 10.1111/cas.14505] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
One pivotal factor that leads to multidrug resistance (MDR) is the overexpression of ABCG2. Therefore, tremendous effort has been devoted to the search of effective reversal agents to overcome ABCG2‐mediated MDR. CC‐671 is a potent and selective inhibitor of both TTK (human protein kinase monopolar spindle 1 [hMps1]) and CDC like kinase 2 (CLK2). It represents a new class of cancer therapeutic drugs. In this study, we show that CC‐671 is an effective ABCG2 reversal agent that enhances the efficacy of chemotherapeutic drugs in ABCG2‐overexpressing lung cancer cells. Mechanistic studies show that the reversal effect of CC‐671 is primarily attributed to the inhibition of the drug efflux activity of ABCG2, which leads to an increased intracellular level of chemotherapeutic drugs. In addition, CC‐671 does not alter the protein expression or subcellular localization of ABCG2. The computational molecule docking analysis suggests CC‐671 has high binding affinity to the drug‐binding site of ABCG2. In conclusion, we reveal the interaction between CC‐671 and ABCG2, providing a rationale for the potential combined use of CC‐671 with ABCG2 substrate to overcome MDR.
Collapse
Affiliation(s)
- Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Guangsuo Wang
- Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Lingling Sun
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Lizhu Lin
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Chang Zou
- Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen, China.,Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| |
Collapse
|
21
|
Erdafitinib Resensitizes ABCB1-Overexpressing Multidrug-Resistant Cancer Cells to Cytotoxic Anticancer Drugs. Cancers (Basel) 2020; 12:cancers12061366. [PMID: 32466597 PMCID: PMC7352346 DOI: 10.3390/cancers12061366] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 01/17/2023] Open
Abstract
The development of multidrug resistance (MDR) in cancer patients, which is often associated with the overexpression of ABCB1 (MDR1, P-glycoprotein) in cancer cells, remains a significant problem in cancer chemotherapy. ABCB1 is one of the major adenosine triphosphate (ATP)-binding cassette (ABC) transporters that can actively efflux a range of anticancer drugs out of cancer cells, causing MDR. Given the lack of Food and Drug Administration (FDA)-approved treatment for multidrug-resistant cancers, we explored the prospect of repurposing erdafitinib, the first fibroblast growth factor receptor (FGFR) kinase inhibitor approved by the FDA, to reverse MDR mediated by ABCB1. We discovered that by reducing the function of ABCB1, erdafitinib significantly resensitized ABCB1-overexpressing multidrug-resistant cancer cells to therapeutic drugs at sub-toxic concentrations. Results of erdafitinib-stimulated ABCB1 ATPase activity and in silico docking analysis of erdafitinib binding to the substrate-binding pocket of ABCB1 further support the interaction between erdafitinib and ABCB1. Moreover, our data suggest that ABCB1 is not a major mechanism of resistance to erdafitinib in cancer cells. In conclusion, we revealed an additional action of erdafitinib as a potential treatment option for multidrug-resistant cancers, which should be evaluated in future drug combination trials.
Collapse
|
22
|
Yuan P, Zhou Q, Hu X. WS 2 Nanosheets at Noncytotoxic Concentrations Enhance the Cytotoxicity of Organic Pollutants by Disturbing the Plasma Membrane and Efflux Pumps. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:1698-1709. [PMID: 31916439 DOI: 10.1021/acs.est.9b05537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Emerging transition-metal dichalcogenide (TMDC) nanosheets, such as WS2 nanosheets, have shown tremendous potential for use in many fields such as intelligent manufacturing and environmental protection. However, considerable knowledge gaps still exist regarding the impact of TMDCs on environmental risks, especially risks involving organic pollutants. Here, a synergistic toxicity between WS2 nanosheets and organic pollutants (triclosan or tris(1,3-dichloro-2-propyl) phosphate) was found using the median-effect and combination index equations. In particular, the effect of synergy had a higher magnitude at low cytotoxicity levels and a noncytotoxic concentration of WS2 nanosheets clearly enhanced the cytotoxicity and intracellular accumulation of organic pollutants. On the one hand, WS2 nanosheets damaged the plasma membrane and cytoskeleton, resulting in increased membrane permeability and organic pollutant uptake. On the other hand, as shown by fluorescence substrate accumulation experiments and molecular dynamics simulations, WS2 nanosheets affected the secondary structure of the efflux pumps and competitively bound with efflux pumps, blocking xenobiotic removal. This work emphasized that TMDCs, especially at the noncytotoxic level, in combination with organic pollutants caused damage that cannot be ignored, providing insight into comprehensive safety assessment and the specific toxicological mechanisms of TMDCs that accompany organic pollutant exposure.
Collapse
Affiliation(s)
- Peng Yuan
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| | - Qixing Zhou
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| | - Xiangang Hu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education)/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering , Nankai University , Tianjin 300350 , China
| |
Collapse
|
23
|
Weisberg E, Meng C, Case AE, Tiv HL, Gokhale PC, Buhrlage SJ, Yang J, Liu X, Wang J, Gray N, Adamia S, Sattler M, Stone R, Griffin JD. Effects of the multi-kinase inhibitor midostaurin in combination with chemotherapy in models of acute myeloid leukaemia. J Cell Mol Med 2020; 24:2968-2980. [PMID: 31967735 PMCID: PMC7077552 DOI: 10.1111/jcmm.14927] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/04/2019] [Accepted: 12/09/2019] [Indexed: 01/08/2023] Open
Abstract
Recently, several targeted agents have been developed for specific subsets of patients with acute myeloid leukaemia (AML), including midostaurin, the first FDA‐approved FLT3 inhibitor for newly diagnosed patients with FLT3 mutations. However, in the initial Phase I/II clinical trials, some patients without FLT3 mutations had transient responses to midostaurin, suggesting that this multi‐targeted kinase inhibitor might benefit AML patients more broadly. Here, we demonstrate submicromolar efficacy of midostaurin in vitro and efficacy in vivo against wild‐type (wt) FLT3‐expressing AML cell lines and primary cells, and we compare its effectiveness with that of other FLT3 inhibitors currently in clinical trials. Midostaurin was found to synergize with standard chemotherapeutic drugs and some targeted agents against AML cells without mutations in FLT3. The mechanism may involve, in part, the unique kinase profile of midostaurin that includes proteins implicated in AML transformation, such as SYK or KIT, or inhibition of ERK pathway or proviability signalling. Our findings support further investigation of midostaurin as a chemosensitizing agent in AML patients without FLT3 mutations.
Collapse
Affiliation(s)
- Ellen Weisberg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Chengcheng Meng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Abigail E Case
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hong L Tiv
- Experimental Therapeutic Core, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Prafulla C Gokhale
- Experimental Therapeutic Core, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sara J Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jing Yang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Xiaoxi Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jinhua Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Nathanael Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Sophia Adamia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Richard Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - James D Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Wu CP, Lusvarghi S, Tseng PJ, Hsiao SH, Huang YH, Hung TH, Ambudkar SV. MY-5445, a phosphodiesterase type 5 inhibitor, resensitizes ABCG2-overexpressing multidrug-resistant cancer cells to cytotoxic anticancer drugs. Am J Cancer Res 2020; 10:164-178. [PMID: 32064159 PMCID: PMC7017726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/18/2019] [Indexed: 06/10/2023] Open
Abstract
The overexpression of one or multiple ATP-binding cassette (ABC) transporters such as ABCB1, ABCC1 or ABCG2 in cancer cells often leads to the development of multidrug resistance phenotype and consequent treatment failure. Therefore, these transporters constitute an important target to improve the therapeutic outcome in cancer patients. In this study, we employed a drug repurposing approach to identify MY-5445, a known phosphodiesterase type 5 inhibitor, as a selective modulator of ABCG2. We discovered that by inhibiting the drug transport function of ABCG2, MY-5445 potentiates drug-induced apoptosis in ABCG2-overexpressing multidrug-resistant cancer cells and resensitizes these cells to chemotherapeutic drugs. Our data of MY-5445 stimulating the ATPase activity of ABCG2 and molecular docking analysis of its binding to the substrate-binding pocket of ABCG2 provide additional insight into the manner in which MY-5445 interacts with ABCG2. Furthermore, we found that ABCG2 does not confer resistance to MY-5445 in human cancer cells. Overall, our study revealed an additional action of MY-5445 to resensitize ABCG2-overexpressing multidrug-resistant cancer cells to conventional anticancer drugs, and this should be evaluated in future drug combination trials.
Collapse
Affiliation(s)
- Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial HospitalTaipei, Taiwan
| | | | - Pin-Jung Tseng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan
| | - Sung-Han Hsiao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan
| | - Yang-Hui Huang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan
| | - Tai-Ho Hung
- Department of Chinese Medicine, College of Medicine, Chang Gung UniversityTao-Yuan, Taiwan
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial HospitalTaipei, Taiwan
| | | |
Collapse
|
25
|
The Selective Class IIa Histone Deacetylase Inhibitor TMP195 Resensitizes ABCB1- and ABCG2-Overexpressing Multidrug-Resistant Cancer Cells to Cytotoxic Anticancer Drugs. Int J Mol Sci 2019; 21:ijms21010238. [PMID: 31905792 PMCID: PMC6981391 DOI: 10.3390/ijms21010238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 12/16/2022] Open
Abstract
Multidrug resistance caused by the overexpression of the ATP-binding cassette (ABC) proteins in cancer cells remains one of the most difficult challenges faced by drug developers and clinical scientists. The emergence of multidrug-resistant cancers has driven efforts from researchers to develop innovative strategies to improve therapeutic outcomes. Based on the drug repurposing approach, we discovered an additional action of TMP195, a potent and selective inhibitor of class IIa histone deacetylase. We reveal that in vitro TMP195 treatment significantly enhances drug-induced apoptosis and sensitizes multidrug-resistant cancer cells overexpressing ABCB1 or ABCG2 to anticancer drugs. We demonstrate that TMP195 inhibits the drug transport function, but not the protein expression of ABCB1 and ABCG2. The interaction between TMP195 with these transporters was supported by the TMP195-stimulated ATPase activity of ABCB1 and ABCG2, and by in silico docking analysis of TMP195 binding to the substrate-binding pocket of these transporters. Furthermore, we did not find clear evidence of TMP195 resistance conferred by ABCB1 or ABCG2, suggesting that these transporters are unlikely to play a significant role in the development of resistance to TMP195 in cancer patients.
Collapse
|
26
|
Chang L, Hou Y, Zhu L, Wang Z, Chen G, Shu C, Liu Y. Veliparib overcomes multidrug resistance in liver cancer cells. Biochem Biophys Res Commun 2019; 521:596-602. [PMID: 31679697 DOI: 10.1016/j.bbrc.2019.10.141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 10/19/2019] [Indexed: 01/30/2023]
Abstract
Overexpression of ATP-binding cassette (ABC) transporter is one of the most important factors taking responsibility for the progress of multidrug resistance (MDR) in multiple cancers. In this study, we investigated that veliparib, a PARP inhibitor which is in clinical development, could overcome ABCB1-mediated MDR in liver cancer cells. Veliparib could significantly enhance the cytotoxic effects of a series of conventional chemotherapeutic drugs in ABCB1-overexpression liver cancer cells. Mechanism study showed that veliparib could significantly enhance the accumulation of doxorubicin in ABCB1-overexpression liver cancer cells, without down-regulating the expression level of ABCB1. Finally, veliparib could significantly inhibit the ATPase activity of ABCB1 transporter. This study could provide information that combine veliparib with other chemotherapeutic drugs may benefit liver cancer patients.
Collapse
Affiliation(s)
- Lin Chang
- Department of Laboratory Medicine, Bishan Hospital, Chongqing, China.
| | - Yulan Hou
- Department of Laboratory Medicine, Bishan Hospital, Chongqing, China
| | - Lili Zhu
- Department of Laboratory Medicine, Bishan Hospital, Chongqing, China
| | - Zhixue Wang
- Department of Laboratory Medicine, Bishan Hospital, Chongqing, China
| | - Guotao Chen
- Department of Nephrology, Bishan Hospital, Chongqing, China
| | - Chengfeng Shu
- Department of Laboratory Medicine, Bishan Hospital, Chongqing, China
| | - Yali Liu
- Department of Laboratory Medicine, Bishan Hospital, Chongqing, China
| |
Collapse
|
27
|
Ji N, Yang Y, Cai CY, Wang JQ, Lei ZN, Wu ZX, Cui Q, Yang DH, Chen ZS, Kong D. Midostaurin Reverses ABCB1-Mediated Multidrug Resistance, an in vitro Study. Front Oncol 2019; 9:514. [PMID: 31275850 PMCID: PMC6591272 DOI: 10.3389/fonc.2019.00514] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Overexpression of ABC transporters in cancer cells is an underlying mechanism of multidrug resistance (MDR), leading to insensitive response to chemotherapeutic strategies. Thus, MDR is often results in treatment failure in the clinic. In this study, we found midostaurin, a Food and Drug Administration (FDA)-approved anti-leukemia drug, can antagonize ATP-binding cassette subfamily B member 1 (ABCB1)-mediated MDR. Our results indicated that midostaurin has the capacity to antagonize ABCB1-mediated MDR, while no significant reversal effect was found on ATP-binding cassette subfamily G member 2 (ABCG2)-mediated MDR. Our subsequent resistance mechanism studies showed that midostaurin directly inhibited the efflux function of the ABCB1 transporter without alteration of the expression level or the subcellular localization of ABCB1 transporter. In addition, midostaurin inhibited the ATPase activity of ABCB1 transporter in a dose-dependent manner. Moreover, our in silico docking study predicted that midostaurin could interact with the substrate-binding sites of ABCB1 transporter. This novel finding could provide a promising treatment strategy that co-administrating midostaurin with anticancer drugs in the clinic could overcome MDR and improve the efficiency of cancer treatment.
Collapse
Affiliation(s)
- Ning Ji
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.,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
| | - Chao-Yun Cai
- 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
| | - Zi-Ning Lei
- 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
| | - 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
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China.,Research Center, School of Medicine, Tianjin Tianshi College, Tianyuan University, Tianjin, China
| |
Collapse
|
28
|
Wu CP, Lusvarghi S, Wang JC, Hsiao SH, Huang YH, Hung TH, Ambudkar SV. Avapritinib: A Selective Inhibitor of KIT and PDGFRα that Reverses ABCB1 and ABCG2-Mediated Multidrug Resistance in Cancer Cell Lines. Mol Pharm 2019; 16:3040-3052. [PMID: 31117741 DOI: 10.1021/acs.molpharmaceut.9b00274] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The frequent occurrence of multidrug resistance (MDR) conferred by the overexpression of ATP-binding cassette (ABC) transporters ABCB1 and ABCG2 in cancer cells remains a therapeutic obstacle for scientists and clinicians. Consequently, developing or identifying modulators of ABCB1 and ABCG2 that are suitable for clinical practice is of great importance. Therefore, we have explored the drug repositioning approach to identify candidate modulators of ABCB1 and ABCG2 from tyrosine kinase inhibitors with known pharmacological properties and anticancer activities. In this study, we discovered that avapritinib (BLU-285), a potent, selective, and orally bioavailable tyrosine kinase inhibitor against mutant forms of KIT and platelet-derived growth factor receptor alpha (PDGFRA), attenuates the transport function of both ABCB1 and ABCG2. Moreover, avapritinib restores the chemosensitivity of ABCB1- and ABCG2-overexpressing MDR cancer cells at nontoxic concentrations. These findings were further supported by results of apoptosis induction assays, ATP hydrolysis assays, and docking of avapritinib in the drug-binding pockets of ABCB1 and ABCG2. Altogether, our study highlights an additional action of avapritinib on ABC drug transporters, and a combination of avapritinib with conventional chemotherapy should be further investigated in patients with MDR tumors.
Collapse
Affiliation(s)
- Chung-Pu Wu
- Department of Obstetrics and Gynecology , Taipei Chang Gung Memorial Hospital , Taipei 105 , Taiwan
| | - Sabrina Lusvarghi
- Laboratory of Cell Biology, Center for Cancer Research , National Cancer Institute , Bethesda , Maryland 20892 , United States
| | | | | | | | - Tai-Ho Hung
- Department of Obstetrics and Gynecology , Taipei Chang Gung Memorial Hospital , Taipei 105 , Taiwan
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research , National Cancer Institute , Bethesda , Maryland 20892 , United States
| |
Collapse
|