1
|
Marques AVL, Ruginsk BE, Prado LDO, de Lima DE, Daniel IW, Moure VR, Valdameri G. The association of ABC proteins with multidrug resistance in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119878. [PMID: 39571941 DOI: 10.1016/j.bbamcr.2024.119878] [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: 08/26/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024]
Abstract
Multidrug resistance (MDR) poses one of the primary challenges for cancer treatment, especially in cases of metastatic disease. Various mechanisms contribute to MDR, including the overexpression of ATP-binding cassette (ABC) proteins. In this context, we reviewed the literature to establish a correlation between the overexpression of ABC proteins and MDR in cancer, considering both in vitro and clinical studies. Initially, we presented an overview of the seven subfamilies of ABC proteins, along with the subcellular localization of each protein. Subsequently, we identified a panel of 20 ABC proteins (ABCA1-3, ABCA7, ABCB1-2, ABCB4-6, ABCC1-5, ABCC10-11, ABCE1, ABCF2, ABCG1, and ABCG2) associated with MDR. We also emphasize the significance of drug sequestration by certain ABC proteins into intracellular compartments. Among the anticancer drugs linked to MDR, 29 were definitively identified as substrates for at least one of the three most crucial ABC transporters: ABCB1, ABCC1, and ABCG2. We further discussed that the most commonly used drugs in standard regimens for mainly breast cancer, lung cancer, and acute lymphoblastic leukemia could be subject to MDR mediated by ABC transporters. Collectively, these insights will aid in conducting new studies aimed at a deeper understanding of the clinical MDR mediated by ABC proteins and in designing more effective pharmacological treatments to enhance the objective response rate in cancer patients.
Collapse
Affiliation(s)
- Andrezza Viviany Lourenço Marques
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Bruna Estelita Ruginsk
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Larissa de Oliveira Prado
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Diogo Eugênio de Lima
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Isabelle Watanabe Daniel
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil
| | - Vivian Rotuno Moure
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil.
| | - Glaucio Valdameri
- Graduate Program in Pharmaceutical Sciences, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, Paraná, Brazil.
| |
Collapse
|
2
|
Kyaw TS, Zhang C, Sandy M, Trepka K, Zhang S, Ramirez Hernandez LA, Ramirez L, Goh JJ, Yu K, Dimassa V, Bess EN, Brockert JG, Dumlao DS, Bisanz JE, Turnbaugh PJ. Human gut Actinobacteria boost drug absorption by secreting P-glycoprotein ATPase inhibitors. iScience 2024; 27:110122. [PMID: 38947502 PMCID: PMC11214321 DOI: 10.1016/j.isci.2024.110122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/17/2024] [Accepted: 05/24/2024] [Indexed: 07/02/2024] Open
Abstract
Drug efflux transporters are a major determinant of drug efficacy and toxicity. A canonical example is P-glycoprotein (P-gp), an efflux transporter that controls the intestinal absorption of diverse compounds. Despite a rich literature on the dietary and pharmaceutical compounds that impact P-gp activity, its sensitivity to gut microbial metabolites remains an open question. Surprisingly, we found that the cardiac drug-metabolizing gut Actinobacterium Eggerthella lenta increases drug absorption in mice. Experiments in cell culture revealed that E. lenta produces a soluble factor that post-translationally inhibits P-gp ATPase efflux activity. P-gp inhibition is conserved in the Eggerthellaceae family but absent in other Actinobacteria. Comparative genomics identified genes associated with P-gp inhibition. Finally, activity-guided biochemical fractionation coupled to metabolomics implicated a group of small polar metabolites with P-gp inhibitory activity. These results highlight the importance of considering the broader relevance of the gut microbiome for drug disposition beyond first-pass metabolism.
Collapse
Affiliation(s)
- Than S. Kyaw
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Chen Zhang
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Moriah Sandy
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
- Quantitative Metabolite Analysis Center, Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kai Trepka
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Shenwei Zhang
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Luis A. Ramirez Hernandez
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Lorenzo Ramirez
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Janice J.N. Goh
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kristie Yu
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Vincent Dimassa
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Elizabeth N. Bess
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jacob G. Brockert
- Quantitative Metabolite Analysis Center, Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Darren S. Dumlao
- Quantitative Metabolite Analysis Center, Benioff Center for Microbiome Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jordan E. Bisanz
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Peter J. Turnbaugh
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA
- Chan-Zuckerberg Biohub-San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
3
|
ALEMZADEH EFFAT, ALLAHQOLI LEILA, MAZIDIMORADI AFROOZ, ALEMZADEH ESMAT, GHASEMI FAHIMEH, SALEHINIYA HAMID, ALKATOUT IBRAHIM. Deciphering resistance mechanisms and novel strategies to overcome drug resistance in ovarian cancer: a comprehensive review. Oncol Res 2024; 32:831-847. [PMID: 38686048 PMCID: PMC11055988 DOI: 10.32604/or.2024.031006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/09/2023] [Indexed: 05/02/2024] Open
Abstract
Ovarian cancer is among the most lethal gynecological cancers, primarily due to the lack of specific symptoms leading to an advanced-stage diagnosis and resistance to chemotherapy. Drug resistance (DR) poses the most significant challenge in treating patients with existing drugs. The Food and Drug Administration (FDA) has recently approved three new therapeutic drugs, including two poly (ADP-ribose) polymerase (PARP) inhibitors (olaparib and niraparib) and one vascular endothelial growth factor (VEGF) inhibitor (bevacizumab) for maintenance therapy. However, resistance to these new drugs has emerged. Therefore, understanding the mechanisms of DR and exploring new approaches to overcome them is crucial for effective management. In this review, we summarize the major molecular mechanisms of DR and discuss novel strategies to combat DR.
Collapse
Affiliation(s)
- EFFAT ALEMZADEH
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - LEILA ALLAHQOLI
- Department of Midwifery, Ministry of Health and Medical Education, Tehran, Iran
| | - AFROOZ MAZIDIMORADI
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - ESMAT ALEMZADEH
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Department of Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - FAHIMEH GHASEMI
- Department of Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - HAMID SALEHINIYA
- Social Determinants of Health Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - IBRAHIM ALKATOUT
- Kiel School of Gynaecological Endoscopy, Campus Kiel, University Hospitals Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
4
|
Żesławska E, Kucwaj-Brysz K, Kincses A, Spengler G, Szymańska E, Czopek A, Marć MA, Kaczor A, Nitek W, Domínguez-Álvarez E, Latacz G, Kieć-Kononowicz K, Handzlik J. An insight into the structure of 5-spiro aromatic derivatives of imidazolidine-2,4-dione, a new group of very potent inhibitors of tumor multidrug resistance in T-lymphoma cells. Bioorg Chem 2021; 109:104735. [PMID: 33640632 DOI: 10.1016/j.bioorg.2021.104735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 01/07/2023]
Abstract
A series of 17 arylpiperazine derivatives of the 5-spiroimidazolidine-2,4-diones (6-22) has been explored, including variations in (i) the number of aromatic rings at position 5, (ii) the length of the linker, as well as (iii) the kind and position of the linked arylpiperazine terminal fragment. Synthesis (6-16) and X-ray crystallographic studies for representative compounds (8, 10, 14 and 18) have been performed. The ability to inhibit the tumor multidrug resistance (MDR) efflux pump P-glycoprotein (P-gp, ABCB1) overexpressed in mouse T-lymphoma cells was investigated. The cytotoxic and antiproliferative actions of the compounds on both the reference and the ABCB1-overproducing cells were also examined. The pharmacophore-based molecular modeling studies have been performed. ADMET properties in vitro of selected most active derivatives (6, 11 and 12) have been determined. All compounds, excluding 18, inhibited the cancer P-gp efflux pump with higher potency than that of reference verapamil. The spirofluorene derivatives with amine alkyl substituents at position 1, and the methyl group at position 3 (6-16), occurred the most potent P-gp inhibitors in the MDR T-lymphoma cell line. In particular, compounds 7 and 12 were 100-fold more potent than verapamil. Crystallography-supported pharmacophore-based SAR analysis has postulated specific structural properties that could explain this excellent cancer MDR-inhibitory action.
Collapse
Affiliation(s)
- Ewa Żesławska
- Institute of Biology, Pedagogical University, Podchorążych 2, 30-084 Kraków, Poland
| | - Katarzyna Kucwaj-Brysz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Annamária Kincses
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Gabriella Spengler
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Ewa Szymańska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Anna Czopek
- Department of Medicinal Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Małgorzata Anna Marć
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Aneta Kaczor
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Wojciech Nitek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Kraków, Poland
| | - Enrique Domínguez-Álvarez
- Instituto de Química Orgánica General, Consejo Superior de Investigaciones Científicas (IQOG-CSIC), Juan de la Cierva 3, 28006, Madrid, Spain
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Jadwiga Handzlik
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland.
| |
Collapse
|
5
|
Quan X, Du H, Xu J, Hou X, Gong X, Wu Y, Zhou Y, Jiang J, Lu L, Yuan S, Yang X, Shi L, Sun L. Novel Quinoline Compound Derivatives of NSC23925 as Potent Reversal Agents Against P-Glycoprotein-Mediated Multidrug Resistance. Front Chem 2020; 7:820. [PMID: 31921759 PMCID: PMC6931887 DOI: 10.3389/fchem.2019.00820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 11/12/2019] [Indexed: 12/02/2022] Open
Abstract
Multidrug resistance is a serious problem and a common cause of cancer treatment failure, leading to patient death. Although numerous reversal resistance inhibitors have been evaluated in preclinical or clinical trials, efficient and low-toxicity reversal agents have not been identified. In this study, a series of novel quinoline compound derivatives from NSC23925 were designed to inhibit P-glycoprotein (P-gp). Among them, YS-7a showed a stronger inhibitory effect against P-gp than verapamil, as a positive control, when co-incubated with chemotherapy drugs at minimally cytotoxic concentrations. YS-7a suppressed the P-gp transport function without affecting the expression of P-gp but stimulated the ATPase activity of P-gp in a dose-dependent manner. Next, molecular docking was used to predict the six most probable binding sites, namely, SER270, VAL273, VAL274, ILE354, VAL357, and PHE390. Moreover, YS-7a had no effect on cytochrome P450 3A4 activity and showed little toxicity to normal cells. In addition, combined treatment of YS-7a with vincristine showed a better inhibitory effect than the positive control verapamil in vivo without a negative effect on mouse weight. Overall, our results showed that YS-7a could reverse cancer multidrug resistance through the inhibition of P-gp transport function in vitro and in vivo, suggesting that YS-7a may be a novel therapeutic agent.
Collapse
Affiliation(s)
- Xingping Quan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jingjing Xu
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Xiaoying Hou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiaofeng Gong
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yao Wu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Yuqi Zhou
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Jingwei Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Ligong Lu
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiangyu Yang
- Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Lei Shi
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| | - Li Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
6
|
Rösner J, Merzendorfer H. Transcriptional plasticity of different ABC transporter genes from Tribolium castaneum contributes to diflubenzuron resistance. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 116:103282. [PMID: 31740345 DOI: 10.1016/j.ibmb.2019.103282] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 06/10/2023]
Abstract
The development of insecticide resistance challenges the sustainability of pest control and several studies have shown that ABC transporters contribute to this process. ABC transporters are known to transport a large range of chemically diverse molecules across cellular membranes, and therefore the identification of ABC transporters involved in insecticide resistance is difficult. Here, we describe a comprehensive strategy for the identification of whole sets of ABC transporters involved in insecticide resistance using the pest beetle, Tribolium castaneum (Tc) as a model. We analyzed the expression of ABCA to ABCC genes in different tissues and developmental stages using larvae that were sensitive or resistant to diflubenzuron (DFB). The mRNA levels of several ABC genes expressed in excretory or metabolic tissues such as midgut, Malpighian tubules or fat body were markedly upregulated in response to DFB. Next, we monitored mortality in the presence of the ABC inhibitor verapamil, and found that it causes sensitization to DFB. We furthermore established a competitive assay for the elimination of DFB, based on Texas Red (TR) fluorescence. We monitored TR elimination in larvae that were treated with DFB or different ABC inhibitors, and combinations of them. TR elimination was decreased significantly in the presence of DFB, verapamil and the ABCC inhibitor MK-571. The effect was synergized when DFB and verapamil were both present suggesting that the transport of TR and DFB involves overlapping sets of ABC transporters. Finally, we silenced the expression of DFB-responding ABC genes by RNA interference and then followed the survival rates after DFB exposure. Mortality increased particularly when specific ABCA and ABCC genes were silenced. Taken together, we were able to show that different ABC transporters expressed in metabolic and excretory tissues contribute to the elimination of DFB. Up- or down-regulation of gene expression occurs within a few days already at very low DFB concentrations. These results suggests that transcriptional plasticity of several ABC genes allows adaptation of the efflux capacity in different tissues to eliminate insecticides and/or their metabolites.
Collapse
Affiliation(s)
- Janin Rösner
- Department of Chemistry-Biology, University of Siegen, Adolf-Reichwein-Strasse 2, 57068, Siegen, Germany
| | - Hans Merzendorfer
- Department of Chemistry-Biology, University of Siegen, Adolf-Reichwein-Strasse 2, 57068, Siegen, Germany.
| |
Collapse
|
7
|
Chen R, Qiao Y, Hu W, Cheng Q, Xie H, Zhou L, Xu X, Zheng S, Jiang D. LY2228820 induces synergistic anti-cancer effects with anti-microtubule chemotherapeutic agents independent of P-glycoprotein in multidrug resistant cancer cells. Am J Cancer Res 2019; 9:2216-2232. [PMID: 31720084 PMCID: PMC6834474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 06/10/2023] Open
Abstract
Side-effects and resistance substantially limit the efficacy of chemotherapy. One possible solution to this persistent problem would be co-administration of targeted therapy and chemotherapy to achieve synergistic anti-cancer effects without extra toxicity. Here, we reported that LY2228820, a selective inhibitor of p38-MAPK signaling pathway, could induce synergistic anti-cancer effects with anti-microtubule (AMT) chemotherapy both in vitro and in vivo. In drug-resistant cancer cells, treatment with either LY2228820 or AMT drug alone was compatible with viability, while co-administration of both led to dramatic cytotoxicity, G2/M arrest and apoptosis. Moreover, co-treatment with LY2228820 notably improved the effectiveness of paclitaxel without exhibiting adverse effects in vivo. Mechanistic studies showed that LY2228820 sensitized cancer cells to AMT agents independent of P-gp. LY2228820 did not influence either the expression or the function of P-gp. Instead, it could inhibit p38-HSP27 signaling axis by down-regulating p-HSP27. Furthermore, LY2228820 blocked the p-HSP27 mediated protective response against AMT drugs in tumor cells, resulting in mitochondrial instability and the activation of mitochondrial death pathways. This P-gp-independent regime containing LY2228820 and AMT agents could produce synergistic anti-cancer effects without extra systematic toxicity. Our study offers a novel strategy for improving the therapeutic efficacy of AMT drugs by achieving a better balance between efficacy and toxicity. This new combination regime could be advantageous in patients who show little response to the maximal dosage of AMT chemotherapy, as well as those unable to tolerate the systematic toxicity of these agents in clinic.
Collapse
Affiliation(s)
- Ronggao Chen
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
| | - Yiting Qiao
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
| | - Wendi Hu
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
| | - Qiyang Cheng
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
| | - Haiyang Xie
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
- Key Laboratory of The Diagnosis and Treatment of Organ Transplantation, CAMSChina
- Key Laboratory of Organ TransplantationHangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseasesHangzhou 310000, Zhejiang Province, China
| | - Lin Zhou
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
- Key Laboratory of The Diagnosis and Treatment of Organ Transplantation, CAMSChina
- Key Laboratory of Organ TransplantationHangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseasesHangzhou 310000, Zhejiang Province, China
| | - Xiao Xu
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
- Key Laboratory of The Diagnosis and Treatment of Organ Transplantation, CAMSChina
- Key Laboratory of Organ TransplantationHangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseasesHangzhou 310000, Zhejiang Province, China
| | - Shusen Zheng
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
- Key Laboratory of The Diagnosis and Treatment of Organ Transplantation, CAMSChina
- Key Laboratory of Organ TransplantationHangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseasesHangzhou 310000, Zhejiang Province, China
| | - Donghai Jiang
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou 310000, Zhejiang Province, China
- NHFPC Key Laboratory of Combined Multi-Organ TransplantationHangzhou 310000, Zhejiang Province, China
- Key Laboratory of The Diagnosis and Treatment of Organ Transplantation, CAMSChina
- Key Laboratory of Organ TransplantationHangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for Diagnosis Treatment of Infectious DiseasesHangzhou 310000, Zhejiang Province, China
| |
Collapse
|
8
|
Mollazadeh S, Sahebkar A, Hadizadeh F, Behravan J, Arabzadeh S. Structural and functional aspects of P-glycoprotein and its inhibitors. Life Sci 2018; 214:118-123. [PMID: 30449449 DOI: 10.1016/j.lfs.2018.10.048] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/12/2018] [Accepted: 10/23/2018] [Indexed: 12/20/2022]
Abstract
P-glycoprotein (P-gp) is a member of ATP-binding cassette (ABC) superfamily which extrudes chemotherapeutic agents out of the cell. Suppression of this efflux activity has been the subject of numerous attempts to develop P-gp inhibitors. The aim of this review is to present up-to-date information on the structural and functional aspects of P-gp and its known inhibitors. The data presented also provide some information on drug discovery approaches for candidate P-gp inhibitors. Nucleotide-binding domains (NBDs) and drug-binding domains (DBDs) have been extensively studied to gain more information about P-gp inhibition and it looks that the ATPase activity of this pump has been the most attractive target for designing inhibitors. Hydrophobic and π-π (aromatic) interactions between P-gp binding domains and inhibitors are dominant intermolecular forces that have been reported in many studies using different methods. Many synthetic and natural products have been found to possess inhibitory or modulatory effects on drug transporter proteins. Log P value is an important factor in studying these inhibitors and has a crucial role on absorption, distribution, metabolism, and excretion (ADME) properties of candidate P-gp inhibitors.
Collapse
Affiliation(s)
- Shirin Mollazadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sepideh Arabzadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
van den Brand D, Mertens V, Massuger LF, Brock R. siRNA in ovarian cancer – Delivery strategies and targets for therapy. J Control Release 2018; 283:45-58. [DOI: 10.1016/j.jconrel.2018.05.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/21/2022]
|
10
|
Liu T, Li Z, Zhang Q, De Amorim Bernstein K, Lozano-Calderon S, Choy E, Hornicek FJ, Duan Z. Targeting ABCB1 (MDR1) in multi-drug resistant osteosarcoma cells using the CRISPR-Cas9 system to reverse drug resistance. Oncotarget 2018; 7:83502-83513. [PMID: 27835872 PMCID: PMC5347784 DOI: 10.18632/oncotarget.13148] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 10/16/2016] [Indexed: 12/14/2022] Open
Abstract
Background Multi-drug resistance (MDR) remains a significant obstacle to successful chemotherapy treatment for osteosarcoma patients. One of the central causes of MDR is the overexpression of the membrane bound drug transporter protein P-glycoprotein (P-gp), which is the protein product of the MDR gene ABCB1. Though several methods have been reported to reverse MDR in vitro and in vivo when combined with anticancer drugs, they have yet to be proven useful in the clinical setting. Results The meta-analysis demonstrated that a high level of P-gp may predict poor survival in patients with osteosarcoma. The expression of P-gp can be efficiently blocked by the clustered regularly interspaced short palindromic repeats (CRISPR)-associated Cas9 system (CRISPR-Cas9). Inhibition of ABCB1 was associated with reversing drug resistance in osteosarcoma MDR cell lines (KHOSR2 and U-2OSR2) to doxorubicin. Materials and Methods We performed a meta-analysis to investigate the relationship between P-gp expression and survival in patients with osteosarcoma. Then we adopted the CRISPR-Cas9, a robust and highly efficient novel genome editing tool, to determine its effect on reversing drug resistance by targeting endogenous ABCB1 gene at the DNA level in osteosarcoma MDR cell lines. Conclusion These results suggest that the CRISPR-Cas9 system is a useful tool for the modification of ABCB1 gene, and may be useful in extending the long-term efficacy of chemotherapy by overcoming P-gp-mediated MDR in the clinical setting.
Collapse
Affiliation(s)
- Tang Liu
- Department of Orthopaedics, The 2nd Xiangya Hospital of Central South University, Changsha, Hunan, 410011, P.R. China.,Sarcoma Biology Laboratory, Department of Orthopaedic surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Zhihong Li
- Department of Orthopaedics, The 2nd Xiangya Hospital of Central South University, Changsha, Hunan, 410011, P.R. China
| | - Qing Zhang
- Department of Orthopaedics, The 2nd Xiangya Hospital of Central South University, Changsha, Hunan, 410011, P.R. China
| | - Karen De Amorim Bernstein
- Sarcoma Biology Laboratory, Department of Orthopaedic surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Santiago Lozano-Calderon
- Sarcoma Biology Laboratory, Department of Orthopaedic surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Edwin Choy
- Sarcoma Biology Laboratory, Department of Orthopaedic surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedic surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
11
|
Khan IU, Khan RU, Asif H, Alamgeer, Khalid SH, Asghar S, Saleem M, Shah KU, Shah SU, Rizvi SAA, Shahzad Y. Co-delivery strategies to overcome multidrug resistance in ovarian cancer. Int J Pharm 2017; 533:111-124. [PMID: 28947245 DOI: 10.1016/j.ijpharm.2017.09.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
Abstract
Cancer is one of the leading causes of death and equally strikes both genders. Among women, ovarian cancer is responsible for many deaths as it remains symptomless in the earlier stages and generally diagnosed in third stage. At this point it becomes difficult to carry out de-bulking surgery and treatment with different chemotherapeutic drugs has shown resistance, a phenomenon known as multidrug resistance (MDR). Different treatment choices are available for ovarian cancer; however, this article only focuses on various co-delivery strategies, where two different agents are encapsulated in a single carrier and act via different pathways to overcome cancer cell resistance. Ovarian cancer develops MDR via different pathways but majorly involving pump and the non-pump mechanisms in most cases. To overcome MDR it is imperative to strike malignant cells from various directions. Nanocarriers are known to strike the pump mechanism by avoiding the drug efflux pump located on cellular membrane. The efflux pump can also be blocked by blocking activity of ATP binding cassette (ABC) membrane transporters. To stop the non-pump mechanism one can use chemosensitizers, genes, apoptotic factor and others. Treatment of cancer cells could even more effective if the drug is combined with co-agents in a single carrier with targeting moiety. These co-agents along with nanocarriers, allow the drug to accumulate in high enough concentrations in ovarian cancer cells to kill them without affecting normal cells.
Collapse
Affiliation(s)
- Ikram Ullah Khan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan.
| | - Rizwan Ullah Khan
- Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry, Lahore, Pakistan
| | - Hira Asif
- Department of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Alamgeer
- Department of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Syed Haroon Khalid
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Sajid Asghar
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Mohammad Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Kifayat Ullah Shah
- Saulat Institute of Pharmaceutical Sciences and Drug Research, Quaid-i-Azam University, Islamabad, Pakistan
| | - Shefat Ullah Shah
- Department of Pharmaceutics, Faculty of Pharmacy, Gomal University Dera Ismail Khan, Pakistan
| | - Syed A A Rizvi
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Yasser Shahzad
- Faculty of Pharmacy, University of Central Punjab, Lahore, Pakistan.
| |
Collapse
|
12
|
Wang J, Seebacher N, Shi H, Kan Q, Duan Z. Novel strategies to prevent the development of multidrug resistance (MDR) in cancer. Oncotarget 2017; 8:84559-84571. [PMID: 29137448 PMCID: PMC5663620 DOI: 10.18632/oncotarget.19187] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022] Open
Abstract
The development of multidrug resistance (MDR) is one of the major challenges to the success of traditional chemotherapy treatment in cancer patients. Most studies to date have focused on strategies to reverse MDR following its development. However, agents utilizing this approach have proven to be of limited clinical use, failing to demonstrate an improvement in therapeutic efficacy with almost no significant survival benefits observed in cancer clinical trials. An alternative approach that has been applied is to prevent or delay MDR prior or early in its development. Recent investigations have shown that preventing the emergence of MDR at the onset of chemotherapy treatment, rather than reversing MDR once it has developed, may assist in overcoming drug resistance. In this review, we focus on a number of novel strategies used by small-molecule inhibitors to prevent the development of MDR. These agents hold great promise for prolonging the efficacy of chemotherapy treatment and improving the clinical outcomes of patients with cancers that are susceptible to MDR development.
Collapse
Affiliation(s)
- Jinglu Wang
- Department of Gynecologic Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, People's Republic of China.,Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Nicole Seebacher
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Huirong Shi
- Department of Gynecologic Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, People's Republic of China
| | - Quancheng Kan
- Department of Gynecologic Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, People's Republic of China
| | - Zhenfeng Duan
- Department of Gynecologic Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, People's Republic of China.,Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
13
|
Huang Y, Jiang D, Sui M, Wang X, Fan W. Fulvestrant reverses doxorubicin resistance in multidrug-resistant breast cell lines independent of estrogen receptor expression. Oncol Rep 2016; 37:705-712. [PMID: 28000875 PMCID: PMC5355712 DOI: 10.3892/or.2016.5315] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022] Open
Abstract
Drug resistance, a major obstacle to successful cancer chemotherapy, frequently occurs in recurrent or metastatic breast cancer and results in poor clinical response. Fulvestrant is a new type of selective estrogen receptor (ER) downregulator and a promising endocrine therapy for breast cancer. In this study, we evaluated the combination treatment of fulvestrant and doxorubicin in ER-negative multidrug-resistant (MDR) breast cancer cell lines Bads-200 and Bats-72. Fulvestrant potentiated doxorubicin-induced cytotoxicity, apoptosis and G2/M arrest with upregulation of cyclin B1. It functioned as a substrate for P-glycoprotein (P-gp) without affecting its expression. Furthermore, fulvestrant not only restored the intracellular accumulation of doxorubicin but also relocalized it to the nuclei in Bats-72 and Bads-200 cells, which may be another potential mechanism of reversal of P-gp mediated doxorubicin resistance. These results indicated that the combination of fulvestrant and doxorubicin-based chemotherapy may be feasible and effective for patients with advanced breast cancer.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Donghai Jiang
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Meihua Sui
- Center for Cancer Biology and Innovative Therapeutics, Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaojia Wang
- Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Weimin Fan
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
14
|
Xu L, Lei J, Jiang D, Zhou L, Wang S, Fan W. Reversal effects of Raloxifene on paclitaxel resistance in 2 MDR breast cancer cells. Cancer Biol Ther 2016; 16:1794-801. [PMID: 26529585 DOI: 10.1080/15384047.2015.1095409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Raloxifene hydrochloride (RAL), one of second generation of selective estrogen receptor modulators (SERMs), is usually used in preventing osteoporosis and breast cancer. The present study evaluated whether Raloxifene might sensitize multidrug resistant (MDR) breast cancers to chemotherapies, especially in estrogen receptor negative (ER-) breast cancer. The results showed that RAL could significantly sensitize ER- MDR breast tumors to paclitaxel both in vitro and in vivo. Combination of Raloxifene could significantly enhance paclitaxel-induced cell apoptosis, G2-M arrest as well as inhibition of cell proliferation in MDR tumors. Further studies showed that the combined treatment did not alter P-glycoprotein expression but increased P-gp ATPase activity. These results suggested that raloxifene might be a valuable chemosensitizer agent for breast cancer therapy.
Collapse
Affiliation(s)
- Liang Xu
- a Program of Innovative Cancer Therapeutics; Department of Surgery.,b Clinical Research Center; First Affiliated Hospital of Zhejiang University School of Medicine ; Hangzhou , China
| | - Jingyu Lei
- a Program of Innovative Cancer Therapeutics; Department of Surgery
| | - Donghai Jiang
- a Program of Innovative Cancer Therapeutics; Department of Surgery
| | - Lin Zhou
- a Program of Innovative Cancer Therapeutics; Department of Surgery
| | - Shu Wang
- a Program of Innovative Cancer Therapeutics; Department of Surgery.,c Department of Biological Sciences ; National University of Singapore ; Singapore
| | - Weimin Fan
- a Program of Innovative Cancer Therapeutics; Department of Surgery.,d Department of Pathology and Laboratory Medicine ; Medical University of South Carolina ; SC , USA
| |
Collapse
|
15
|
Novel mechanisms and approaches to overcome multidrug resistance in the treatment of ovarian cancer. Biochim Biophys Acta Rev Cancer 2016; 1866:266-275. [PMID: 27717733 DOI: 10.1016/j.bbcan.2016.10.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/16/2016] [Accepted: 10/03/2016] [Indexed: 12/20/2022]
Abstract
Ovarian cancer remains the leading cause of gynecological cancer-related mortality despite the advances in surgical techniques and chemotherapy drugs over the past three decades. Multidrug resistance (MDR) to chemotherapy is the major cause of treatment failure. Previous research has focused mainly on strategies to reverse MDR by targeting the MDR1 gene encoded P-glycoprotein (Pgp) with small molecular compound inhibitors. However, prior Pgp inhibitors have shown very limited clinical success because these agents have relatively low potency and high toxicity. Therefore, identification of more specific and potent new inhibitors would be useful. In addition, emerging evidence suggests that cancer stem cells (CSCs), deregulated non-coding RNA (ncRNA), autophagy, and tumor heterogeneity also contribute significantly to drug sensitivity/resistance in ovarian cancer. This review summarizes these novel mechanisms of MDR and evaluates several new concepts to overcome MDR in the treatment of ovarian cancer. These new strategies include overcoming MDR with more potent and specific Pgp inhibitors, targeting CSCs and ncRNA, modulating autophagy signaling pathway, and targeting tumor heterogeneity.
Collapse
|
16
|
Pharmacokinetics and tolerability of NSC23925b, a novel P-glycoprotein inhibitor: preclinical study in mice and rats. Sci Rep 2016; 6:25659. [PMID: 27157103 PMCID: PMC4860631 DOI: 10.1038/srep25659] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/19/2016] [Indexed: 12/19/2022] Open
Abstract
Overexpression of P-glycoprotein (Pgp) increases multidrug resistance (MDR) in cancer, which greatly impedes satisfactory clinical treatment and outcomes of cancer patients. Due to unknown pharmacokinetics, the use of Pgp inhibitors to overcome MDR in the clinical setting remains elusive despite promising in vitro results. The purpose of our current preclinical study is to investigate the pharmacokinetics and tolerability of NSC23925b, a novel and potent P-glycoprotein inhibitor, in rodents. Plasma pharmacokinetic studies of single-dose NSC23925b alone or in combination with paclitaxel or doxorubicin were conducted in male BALB/c mice and Sprague-Dawley rats. Additionally, inhibition of human cytochrome P450 (CYP450) by NSC23925b was examined in vitro. Finally, the maximum tolerated dose (MTD) of NSC23925b was determined. NSC23925b displayed favorable pharmacokinetic profiles after intraperitoneal/intravenous (I.P./I.V.) injection alone or combined with chemotherapeutic drugs. The plasma pharmacokinetic characteristics of the chemotherapy drugs were not affected when co-administered with NSC23925b. All the animals tolerated the I.P./I.V. administration of NSC23925b. Moreover, the enzymatic activity of human CYP450 was not inhibited by NSC23925b. Our results demonstrated that Pgp inhibitor NSC23925b exhibits encouraging preclinical pharmacokinetic characteristics and limited toxicity in vivo. NSC23925b has the potential to treat cancer patients with MDR in the future.
Collapse
|
17
|
YOSHIKAWA NOBUHISA, KAJIYAMA HIROAKI, NAKAMURA KAE, UTSUMI FUMI, NIIMI KAORU, MITSUI HIROKO, SEKIYA RYUICHIRO, SUZUKI SHIRO, SHIBATA KIYOSUMI, CALLEN DAVID, KIKKAWA FUMITAKA. PRIMA-1MET induces apoptosis through accumulation of intracellular reactive oxygen species irrespective of p53 status and chemo-sensitivity in epithelial ovarian cancer cells. Oncol Rep 2016; 35:2543-52. [PMID: 26986846 PMCID: PMC4811399 DOI: 10.3892/or.2016.4653] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/07/2016] [Indexed: 12/13/2022] Open
Abstract
There is an intensive need for the development of novel drugs for the treatment of epithelial ovarian cancer (EOC), the most lethal gynecologic malignancy due to the high recurrence rate. TP53 mutation is a common event in EOC, particularly in high-grade serous ovarian cancer, where it occurs in more than 90% of cases. Recently, PRIMA-1 and PRIMA‑1MET (p53 reactivation and induction of massive apoptosis and its methylated form) were shown to have an antitumor effect on several types of cancer. Despite that PRIMA-1MET is the first compound evaluated in clinical trials, the antitumor effects of PRIMA-1MET on EOC remain unclear. In this study, we investigated the therapeutic potential of PRIMA-1MET for the treatment of EOC cells. PRIMA-1MET treatment of EOC cell lines (n=13) resulted in rapid apoptosis at various concentrations (24 h IC50 2.6-20.1 µM). The apoptotic response was independent of the p53 status and chemo-sensitivity. PRIMA‑1MET treatment increased intracellular reactive oxygen species (ROS), and PRIMA-1MET-induced apoptosis was rescued by an ROS scavenger. Furthermore, RNA expression analysis revealed that the mechanism of action of PRIMA‑1MET may be due to inhibition of antioxidant enzymes, such as Prx3 and GPx-1. In conclusion, our results suggest that PRIMA-1MET represents a novel therapeutic strategy for the treatment of ovarian cancer irrespective of p53 status and chemo-sensitivity.
Collapse
Affiliation(s)
- NOBUHISA YOSHIKAWA
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - HIROAKI KAJIYAMA
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - KAE NAKAMURA
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - FUMI UTSUMI
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - KAORU NIIMI
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - HIROKO MITSUI
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - RYUICHIRO SEKIYA
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - SHIRO SUZUKI
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - KIYOSUMI SHIBATA
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - DAVID CALLEN
- Centre for Personalized Cancer Medicine, Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - FUMITAKA KIKKAWA
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
18
|
Ye S, Zhang J, Shen J, Gao Y, Li Y, Choy E, Cote G, Harmon D, Mankin H, Gray NS, Hornicek FJ, Duan Z. NVP-TAE684 reverses multidrug resistance (MDR) in human osteosarcoma by inhibiting P-glycoprotein (PGP1) function. Br J Pharmacol 2016; 173:613-26. [PMID: 26603906 PMCID: PMC4728419 DOI: 10.1111/bph.13395] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/23/2015] [Accepted: 11/19/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Increased expression of P-glycoprotein (PGP1) is one of the major causes of multidrug resistance (MDR) in cancer, including in osteosarcoma, which eventually leads to the failure of cancer chemotherapy. Thus, there is an urgent need to develop effective therapeutic strategies to override the expression and function of PGP1 to counter MDR in cancer patients. EXPERIMENTAL APPROACH In an effort to search for new chemical entities targeting PGP1-associated MDR in osteosarcoma, we screened a 500+ compound library of known kinase inhibitors with established kinase selectivity profiles. We aimed to discover potential drug synergistic effects among kinase inhibitors and general chemotherapeutics by combining inhibitors with chemotherapy drugs such as doxorubicin and paclitaxel. The human osteosarcoma MDR cell lines U2OSR2 and KHOSR2 were used for the initial screen and secondary mechanistic studies. KEY RESULTS After screening 500+ kinase inhibitors, we identified NVP-TAE684 as the most effective MDR reversing agent. NVP-TAE684 significantly reversed chemoresistance when used in combination with doxorubicin, paclitaxel, docetaxel, vincristine, ET-743 or mitoxantrone. NVP-TAE684 itself is not a PGP1 substrate competitive inhibitor, but it can increase the intracellular accumulation of PGP1 substrates in PGP1-overexpressing cell lines. NVP-TAE684 was found to inhibit the function of PGP1 by stimulating PGP1 ATPase activity, a phenomenon reported for other PGP1 inhibitors. CONCLUSIONS AND IMPLICATIONS The application of NVP-TAE684 to restore sensitivity of osteosarcoma MDR cells to the cytotoxic effects of chemotherapeutics will be useful for further study of PGP1-mediated MDR in human cancer and may ultimately benefit cancer patients.
Collapse
Affiliation(s)
- Shunan Ye
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Jianming Zhang
- Cutaneous Biology Research Center, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Jacson Shen
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Yan Gao
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Ying Li
- Cutaneous Biology Research Center, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Edwin Choy
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Gregory Cote
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - David Harmon
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Henry Mankin
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | | | - Francis J Hornicek
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| | - Zhenfeng Duan
- Center for Sarcoma and Connective Tissue Oncology, Massachusetts General HospitalHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
19
|
Nakamichi N, Ishimoto T, Yamauchi Y, Masuo Y, Kato Y. Screening to Identify Multidrug Resistance-Associated Protein Inhibitors with Neuroblastoma-Selective Cytotoxicity. Biol Pharm Bull 2016; 39:1638-1645. [DOI: 10.1248/bpb.b16-00319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Noritaka Nakamichi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Takahiro Ishimoto
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Yoshihide Yamauchi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Yusuke Masuo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University
| |
Collapse
|
20
|
Feng X, Zhu W, Schurig-Briccio LA, Lindert S, Shoen C, Hitchings R, Li J, Wang Y, Baig N, Zhou T, Kim BK, Crick DC, Cynamon M, McCammon JA, Gennis RB, Oldfield E. Antiinfectives targeting enzymes and the proton motive force. Proc Natl Acad Sci U S A 2015; 112:E7073-82. [PMID: 26644565 PMCID: PMC4697371 DOI: 10.1073/pnas.1521988112] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
There is a growing need for new antibiotics. Compounds that target the proton motive force (PMF), uncouplers, represent one possible class of compounds that might be developed because they are already used to treat parasitic infections, and there is interest in their use for the treatment of other diseases, such as diabetes. Here, we tested a series of compounds, most with known antiinfective activity, for uncoupler activity. Many cationic amphiphiles tested positive, and some targeted isoprenoid biosynthesis or affected lipid bilayer structure. As an example, we found that clomiphene, a recently discovered undecaprenyl diphosphate synthase inhibitor active against Staphylococcus aureus, is an uncoupler. Using in silico screening, we then found that the anti-glioblastoma multiforme drug lead vacquinol is an inhibitor of Mycobacterium tuberculosis tuberculosinyl adenosine synthase, as well as being an uncoupler. Because vacquinol is also an inhibitor of M. tuberculosis cell growth, we used similarity searches based on the vacquinol structure, finding analogs with potent (∼0.5-2 μg/mL) activity against M. tuberculosis and S. aureus. Our results give a logical explanation of the observation that most new tuberculosis drug leads discovered by phenotypic screens and genome sequencing are highly lipophilic (logP ∼5.7) bases with membrane targets because such species are expected to partition into hydrophobic membranes, inhibiting membrane proteins, in addition to collapsing the PMF. This multiple targeting is expected to be of importance in overcoming the development of drug resistance because targeting membrane physical properties is expected to be less susceptible to the development of resistance.
Collapse
Affiliation(s)
- Xinxin Feng
- Department of Chemistry, University of Illinois, Urbana, IL 61801
| | - Wei Zhu
- Department of Chemistry, University of Illinois, Urbana, IL 61801
| | | | - Steffen Lindert
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH 43210
| | - Carolyn Shoen
- Central New York Research Corporation, Veterans Affairs Medical Center, Syracuse, NY 13210
| | - Reese Hitchings
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523
| | - Jikun Li
- Department of Chemistry, University of Illinois, Urbana, IL 61801
| | - Yang Wang
- Department of Chemistry, University of Illinois, Urbana, IL 61801
| | - Noman Baig
- Department of Chemistry, University of Illinois, Urbana, IL 61801
| | - Tianhui Zhou
- Department of Chemistry, University of Illinois, Urbana, IL 61801
| | - Boo Kyung Kim
- Department of Chemistry, University of Illinois, Urbana, IL 61801
| | - Dean C Crick
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523
| | - Michael Cynamon
- Central New York Research Corporation, Veterans Affairs Medical Center, Syracuse, NY 13210
| | - J Andrew McCammon
- Department of Pharmacology and Department of Chemistry & Biochemistry, University of California San Diego, La Jolla, CA 92093; Howard Hughes Medical Institute, University of California San Diego, La Jolla, CA 92093; National Biomedical Computation Resource, University of California San Diego, La Jolla, CA 92093;
| | - Robert B Gennis
- Department of Chemistry, University of Illinois, Urbana, IL 61801; Department of Biochemistry, University of Illinois, Urbana, IL 61801; Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Eric Oldfield
- Department of Chemistry, University of Illinois, Urbana, IL 61801; Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
21
|
Cross-reacting material 197 reverses the resistance to paclitaxel in paclitaxel-resistant human ovarian cancer. Tumour Biol 2015; 37:5521-8. [PMID: 26572150 DOI: 10.1007/s13277-015-4412-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/09/2015] [Indexed: 01/22/2023] Open
Abstract
Heparin-binding epidermal growth factor-like growth factor (HB-EGF) has been proven to be a promising chemotherapeutic target for ovarian cancer. Our previous studies have demonstrated that inhibition of HB-EGF by the special inhibitor, cross-reacting material 197 (CRM197), potently inhibits the anti-tumor activity in paclitaxel-resistant ovarian cancer. Here, we found that inhibition of HB-EGF by CRM197 significantly reverses the resistance to paclitaxel in paclitaxel-resistant ovarian carcinoma cell line (A2780/Taxol). A2780/Taxol cells over-expressed HB-EGF and epidermal growth factor receptor (EGFR) and CRM197 notably suppressed the expression of HB-EGF and EGFR. Experiments performed in vitro and in vivo further suggested that CRM197 markedly down-regulated the ATP-binding cassette sub-family B member 1 (ABCB1/MDR1) messenger RNA (mRNA) expression (P = 0.01), plasma membrane glycoprotein (P-gp) protein (P = 0.009), and P-gp-mediated efflux (P = 0.007) through inhibition of nuclear factor-κB (NF-κB) expression, which were classical chemoresistance-related targets with respect to paclitaxel therapy. Meanwhile, inhibition of HB-EGF enhanced caspase-3 activity to induce apoptosis via MDR1 inhibition in A2780/Taxol cells (P = 0.038). Collectively, HB-EGF is a molecular target for the resistance of ovarian cancer to paclitaxel and CRM197 as a HB-EGF-targeted agent might be a chemosensitizing agent for paclitaxel-resistant ovarian carcinoma. Our findings provide novel possible mechanisms for HB-EGF to be a target to restore the chemosensitivity to paclitaxel.
Collapse
|
22
|
Ween MP, Armstrong MA, Oehler MK, Ricciardelli C. The role of ABC transporters in ovarian cancer progression and chemoresistance. Crit Rev Oncol Hematol 2015; 96:220-56. [PMID: 26100653 DOI: 10.1016/j.critrevonc.2015.05.012] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 04/08/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
Over 80% of ovarian cancer patients develop chemoresistance which results in a lethal course of the disease. A well-established cause of chemoresistance involves the family of ATP-binding cassette transporters, or ABC transporters that transport a wide range of substrates including metabolic products, nutrients, lipids, and drugs across extra- and intra-cellular membranes. Expressions of various ABC transporters, shown to reduce the intracellular accumulation of chemotherapy drugs, are increased following chemotherapy and impact on ovarian cancer survival. Although clinical trials to date using ABC transporter inhibitors have been disappointing, ABC transporter inhibition remains an attractive potential adjuvant to chemotherapy. A greater understanding of their physiological functions and role in ovarian cancer chemoresistance will be important for the development of more effective targeted therapies. This article will review the role of the ABC transporter family in ovarian cancer progression and chemoresistance as well as the clinical attempts used to date to reverse chemoresistance.
Collapse
Affiliation(s)
- M P Ween
- Lung Research, Hanson Institute and Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide
| | - M A Armstrong
- Data Management and Analysis Centre, University of Adelaide, Australia
| | - M K Oehler
- Gynaecological Oncology Department, Royal Adelaide Hospital, Australia; School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Australia
| | - C Ricciardelli
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Australia.
| |
Collapse
|
23
|
Yang X, Shen J, Gao Y, Feng Y, Guan Y, Zhang Z, Mankin H, Hornicek FJ, Duan Z. Nsc23925 prevents the development of paclitaxel resistance by inhibiting the introduction of P-glycoprotein and enhancing apoptosis. Int J Cancer 2015; 137:2029-39. [PMID: 25904021 DOI: 10.1002/ijc.29574] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 03/13/2015] [Accepted: 04/14/2015] [Indexed: 12/12/2022]
Abstract
Strategies to prevent the emergence of drug resistance will increase the effectiveness of chemotherapy treatment and prolong survival of women with ovarian cancer. The aim of our study is to determine the effects of NSC23925 on preventing the development of paclitaxel resistance in ovarian cancer both in cultured cells in vitro and in mouse xenograft models in vivo, and to further elucidate these underlying mechanisms. We first developed a paclitaxel-resistant ovarian cancer cell line, and demonstrated that NSC23925 could prevent the introduction of paclitaxel resistance by specifically inhibiting the overexpression of P-glycoprotein (Pgp) in vitro. The paclitaxel-resistant ovarian cancer cells were then established in a mouse model by continuous paclitaxel treatment in combination with or without NSC23925 administration in the mice. The majority of mice continuously treated with paclitaxel alone eventually developed paclitaxel resistance with overexpression of Pgp and antiapoptotic proteins, whereas mice remained sensitivity to paclitaxel and displayed lower expression levels of Pgp and antiapoptotic proteins after administered continuously with combination of paclitaxel-NSC23925. Paclitaxel-NSC23925-treated mice experienced significantly longer overall survival time than paclitaxel-treated mice. Furthermore, the combination of paclitaxel and NSC23925 therapy did not induce obvious toxicity as measured by mice body weight changes, blood cell counts and histology of internal organs. Collectively, our observations provide evidence that NSC23925 in combination with paclitaxel may prevent the onset of Pgp or antiapoptotic-mediated paclitaxel resistance, and improve the long-term clinical outcome in patients with ovarian cancer.
Collapse
Affiliation(s)
- Xiaoqian Yang
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jacson Shen
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Yan Gao
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Yong Feng
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Yichun Guan
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.,Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhan Zhang
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Henry Mankin
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
24
|
Yang X, Feng Y, Gao Y, Shen J, Choy E, Cote G, Harmon D, Zhang Z, Mankin H, Hornicek FJ, Duan Z. NSC23925 prevents the emergence of multidrug resistance in ovarian cancer in vitro and in vivo. Gynecol Oncol 2015; 137:134-42. [PMID: 25677062 DOI: 10.1016/j.ygyno.2015.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/03/2015] [Indexed: 01/04/2023]
Abstract
OBJECTIVE The development of multidrug resistance (MDR) remains the significant clinical challenge in ovarian cancer therapy; however, relatively little is known about how to prevent the emergence of MDR during chemotherapy treatment. NSC23925 previously has been shown to prevent the development of MDR in osteosarcoma cells in vitro. The purpose of this study was to evaluate the effects of NSC23925 on the prevention of MDR in ovarian cancer, especially in vivo. METHODS Human ovarian cancer cells were treated with paclitaxel alone or in combination with NSC23925 in vitro and in vivo. MDR ovarian cancer cells were established both in cultured cells and mouse models. The expression levels of Pgp and MDR1 were evaluated in various selected cell sublines by Western blot and real-time PCR. Pgp activity was also determined. RESULTS Paclitaxel treated cells eventually developed MDR with overexpression of Pgp and MDR1, and with high activity of Pgp, while paclitaxel-NSC23925 co-treated cells remained sensitive to chemotherapeutic agents in both in vitro and in vivo models. There was no observed increase in expression level and activity of Pgp in paclitaxel-NSC23925 co-treated cells. Additionally, there were no changes in the sensitivity to chemotherapeutic agents, nor expression of Pgp, in cells cultured with NSC23925. CONCLUSION Our findings suggest that NSC23925 can prevent the emergence of MDR in ovarian cancer both in vitro and in vivo. The clinical use of NSC2395 at the onset of chemotherapy may prevent the development of MDR and improve the clinical outcome of patients with ovarian cancer.
Collapse
Affiliation(s)
- Xiaoqian Yang
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yong Feng
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yan Gao
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jacson Shen
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Edwin Choy
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Gregory Cote
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - David Harmon
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Zhan Zhang
- Department of Gynaecology and Obstetrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Henry Mankin
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
25
|
Yang X, Iyer AK, Singh A, Milane L, Choy E, Hornicek FJ, Amiji MM, Duan Z. Cluster of Differentiation 44 Targeted Hyaluronic Acid Based Nanoparticles for MDR1 siRNA Delivery to Overcome Drug Resistance in Ovarian Cancer. Pharm Res 2014; 32:2097-109. [PMID: 25515492 DOI: 10.1007/s11095-014-1602-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/08/2014] [Indexed: 12/18/2022]
Abstract
PURPOSE Approaches for the synthesis of biomaterials to facilitate the delivery of "biologics" is a major area of research in cancer therapy. Here we designed and characterized a hyaluronic acid (HA) based self-assembling nanoparticles that can target CD44 receptors overexpressed on multidrug resistance (MDR) ovarian cancer. The nanoparticle system is composed of HA-poly(ethyleneimine)/HA-poly(ethylene glycol) (HA-PEI/HA-PEG) designed to deliver MDR1 siRNA for the treatment of MDR in an ovarian cancer model. METHODS HA-PEI/HA-PEG nanoparticles were synthesized and characterized, then the cellular uptake and knockdown efficiency of HA-PEI/HA-PEG/MDR1 siRNA nanoparticles was further determined. A human xenograft MDR ovarian cancer model was established to evaluate the effects of the combination of HA-PEI/HA-PEG/MDR1 siRNA nanoparticles and paclitaxel on MDR tumor growth. RESULTS Our results demonstrated that HA-PEI/HA-PEG nanoparticles successfully targeted CD44 and delivered MDR1 siRNA into OVCAR8TR (established paclitaxel resistant) tumors. Additionally, HA-PEI/HA-PEG nanoparticles loaded with MDR1 siRNA efficiently down-regulated the expression of MDR1 and P-glycoprotein (Pgp), inhibited the functional activity of Pgp, and subsequently increased cell sensitivity to paclitaxel. HA-PEI/HA-PEG/MDR1 siRNA nanoparticle therapy followed by paclitaxel treatment inhibited tumor growth in MDR ovarian cancer mouse models. CONCLUSIONS These findings suggest that this CD44 targeted HA-PEI/HA-PEG nanoparticle platform may be a clinicaly relevant gene delivery system for systemic siRNA-based anticancer therapeutics for the treatment of MDR cancers.
Collapse
Affiliation(s)
- Xiaoqian Yang
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, 100 Blossom St, Jackson 1115, Boston, Massachusetts, 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Chang SY, Liu FF, Dong XY, Sun Y. Molecular insight into conformational transmission of human P-glycoprotein. J Chem Phys 2014; 139:225102. [PMID: 24329094 DOI: 10.1063/1.4832740] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
P-glycoprotein (P-gp), a kind of ATP-binding cassette transporter, can export candidates through a channel at the two transmembrane domains (TMDs) across the cell membranes using the energy released from ATP hydrolysis at the two nucleotide-binding domains (NBDs). Considerable evidence has indicated that human P-gp undergoes large-scale conformational changes to export a wide variety of anti-cancer drugs out of the cancer cells. However, molecular mechanism of the conformational transmission of human P-gp from the NBDs to the TMDs is still unclear. Herein, targeted molecular dynamics simulations were performed to explore the atomic detail of the conformational transmission of human P-gp. It is confirmed that the conformational transition from the inward- to outward-facing is initiated by the movement of the NBDs. It is found that the two NBDs move both on the two directions (x and y). The movement on the x direction leads to the closure of the NBDs, while the movement on the y direction adjusts the conformations of the NBDs to form the correct ATP binding pockets. Six key segments (KSs) protruding from the TMDs to interact with the NBDs are identified. The relative movement of the KSs along the y axis driven by the NBDs can be transmitted through α-helices to the rest of the TMDs, rendering the TMDs to open towards periplasm in the outward-facing conformation. Twenty eight key residue pairs are identified to participate in the interaction network that contributes to the conformational transmission from the NBDs to the TMDs of human P-gp. In addition, 9 key residues in each NBD are also identified. The studies have thus provided clear insight into the conformational transmission from the NBDs to the TMDs in human P-gp.
Collapse
Affiliation(s)
- Shan-Yan Chang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Fu-Feng Liu
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Xiao-Yan Dong
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
27
|
Prevention of multidrug resistance (MDR) in osteosarcoma by NSC23925. Br J Cancer 2014; 110:2896-904. [PMID: 24853187 PMCID: PMC4056062 DOI: 10.1038/bjc.2014.254] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/10/2014] [Accepted: 04/14/2014] [Indexed: 12/11/2022] Open
Abstract
Background: The major limitation to the success of chemotherapy in osteosarcoma is the development of multidrug resistance (MDR). Preventing the emergence of MDR during chemotherapy treatment has been a high priority of clinical and investigational oncology, but it remains an elusive goal. The NSC23925 has recently been identified as a novel and potent MDR reversal agent. However, whether NSC23925 can prevent the development of MDR in cancer is unknown. Therefore, this study aims to evaluate the effects of NSC23925 on prevention of the development of MDR in osteosarcoma. Methods: Human osteosarcoma cell lines U-2OS and Saos were exposed to increasing concentrations of paclitaxel alone or in combination with NSC23925 for 6 months. Cell sublines selected at different time points were evaluated for their drug sensitivity, drug transporter P-glycoprotein (Pgp) expression and activity. Results: We observed that tumour cells selected with increasing concentrations of paclitaxel alone developed MDR with resistance to paclitaxel and other Pgp substrates, whereas cells cultured with paclitaxel–NSC23925 did not develop MDR and cells remained sensitive to chemotherapeutic agents. Paclitaxel-resistant cells showed high expression and activity of the Pgp, whereas paclitaxel–NSC23925-treated cells did not express Pgp. No changes in IC50 and Pgp expression and activity were observed in cells grown with the NSC23925 alone. Conclusions: Our findings suggest that NSC23925 may prevent the development of MDR by specifically preventing the overexpression of Pgp. Given the significant incidence of MDR in osteosarcoma and the lack of effective agents for prevention of MDR, NSC23925 and derivatives hold the potential to improve the outcome of cancer patients with poor prognosis due to drug resistance.
Collapse
|
28
|
Kitambi SS, Toledo EM, Usoskin D, Wee S, Harisankar A, Svensson R, Sigmundsson K, Kalderén C, Niklasson M, Kundu S, Aranda S, Westermark B, Uhrbom L, Andäng M, Damberg P, Nelander S, Arenas E, Artursson P, Walfridsson J, Forsberg Nilsson K, Hammarström LGJ, Ernfors P. RETRACTED: Vulnerability of glioblastoma cells to catastrophic vacuolization and death induced by a small molecule. Cell 2014; 157:313-328. [PMID: 24656405 DOI: 10.1016/j.cell.2014.02.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/18/2013] [Accepted: 02/06/2014] [Indexed: 12/25/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies.
Collapse
Affiliation(s)
- Satish Srinivas Kitambi
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Enrique M Toledo
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Dmitry Usoskin
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Shimei Wee
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Aditya Harisankar
- Department of Medicine, HERM, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Richard Svensson
- Department of Pharmacy, UDOPP, Chemical Biology Consortium Sweden, Uppsala University, 751 05 Uppsala, Sweden
| | - Kristmundur Sigmundsson
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine & Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Christina Kalderén
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine & Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mia Niklasson
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Soumi Kundu
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Sergi Aranda
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Bengt Westermark
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Lene Uhrbom
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Michael Andäng
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Peter Damberg
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Sven Nelander
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Per Artursson
- Department of Pharmacy, UDOPP, Chemical Biology Consortium Sweden, Uppsala University, 751 05 Uppsala, Sweden
| | - Julian Walfridsson
- Department of Medicine, HERM, Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Karin Forsberg Nilsson
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Lars G J Hammarström
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine & Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
29
|
Jiang D, Huang Y, Han N, Xu M, Xu L, Zhou L, Wang S, Fan W. Fulvestrant, a selective estrogen receptor down-regulator, sensitizes estrogen receptor negative breast tumors to chemotherapy. Cancer Lett 2014; 346:292-9. [PMID: 24462822 DOI: 10.1016/j.canlet.2014.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 01/13/2014] [Accepted: 01/15/2014] [Indexed: 01/08/2023]
Abstract
Drug resistance frequently results in poor prognosis and high 5-year recurrence rate in estrogen receptor-negative (ER-) breast cancer patients. Herein, we examined the reversal effects of fulvestrant on multidrug resistance (MDR) in ER- breast cancer cells. Co-administration of fulvestrant significantly sensitized ER- MDR tumors to paclitaxel both in vitro and in vivo. Further analyses indicated that fulvestrant did not affect P-gp expression, but could inhibit P-gp function and subsequently reverse P-gp mediated drug resistance in ER- breast cancer cells. These results showed that combination of fulvestrant and chemotherapeutic agents might provide an effective treatment for ER- MDR breast cancers.
Collapse
Affiliation(s)
- Donghai Jiang
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Huang
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Department of Chemotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ning Han
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Mingjie Xu
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Xu
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Zhou
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shu Wang
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Weimin Fan
- Program of Innovative Cancer Therapeutics, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, USA.
| |
Collapse
|
30
|
Tang H, Shen DR, Han YH, Kong Y, Balimane P, Marino A, Gao M, Wu S, Xie D, Soars MG, O’Connell JC, Rodrigues AD, Zhang L, Cvijic ME. Development of Novel, 384-Well High-Throughput Assay Panels for Human Drug Transporters. ACTA ACUST UNITED AC 2013; 18:1072-83. [DOI: 10.1177/1087057113494807] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Transporter proteins are known to play a critical role in affecting the overall absorption, distribution, metabolism, and excretion characteristics of drug candidates. In addition to efflux transporters (P-gp, BCRP, MRP2, etc.) that limit absorption, there has been a renewed interest in influx transporters at the renal (OATs, OCTs) and hepatic (OATPs, BSEP, NTCP, etc.) organ level that can cause significant clinical drug-drug interactions (DDIs). Several of these transporters are also critical for hepatobiliary disposition of bilirubin and bile acid/salts, and their inhibition is directly implicated in hepatic toxicities. Regulatory agencies took action to address transporter-mediated DDI with the goal of ensuring drug safety in the clinic and on the market. To meet regulatory requirements, advanced bioassay technology and automation solutions were implemented for high-throughput transporter screening to provide structure-activity relationship within lead optimization. To enhance capacity, several functional assay formats were miniaturized to 384-well throughput including novel fluorescence-based uptake and efflux inhibition assays using high-content image analysis as well as cell-based radioactive uptake and vesicle-based efflux inhibition assays. This high-throughput capability enabled a paradigm shift from studying transporter-related issues in the development space to identifying and dialing out these concerns early on in discovery for enhanced mechanism-based efficacy while circumventing DDIs and transporter toxicities.
Collapse
Affiliation(s)
- Huaping Tang
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Ding Ren Shen
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Yong-Hae Han
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Yan Kong
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Praveen Balimane
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Anthony Marino
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Mian Gao
- Department of Protein Science, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Sophie Wu
- Department of Protein Science, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Dianlin Xie
- Department of Protein Science, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Matthew G. Soars
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Jonathan C. O’Connell
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - A. David Rodrigues
- Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Litao Zhang
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| | - Mary Ellen Cvijic
- Department of Leads Discovery and Optimization, Bristol-Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
31
|
Antczak C, Wee B, Radu C, Bhinder B, Holland EC, Djaballah H. A high-content assay strategy for the identification and profiling of ABCG2 modulators in live cells. Assay Drug Dev Technol 2013; 12:28-42. [PMID: 23992118 DOI: 10.1089/adt.2013.521] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
ABCG2 is a member of the ATP-binding cassette (ABC) family of transporters, the overexpression of which has been implicated in resistance to various chemotherapeutic agents. Though a number of cell-based assays to screen for inhibitors have been reported, they do not provide a content-rich platform to discriminate toxic and autofluorescent compounds. To fill this gap, we developed a live high-content cell-based assay to identify inhibitors of ABCG2-mediated transport and, at the same time, assess their cytotoxic effect and potential optical interference. We used a pair of isogenic U87MG human glioblastoma cell lines, with one stably overexpressing the ABCG2 transporter. JC-1 (J-aggregate-forming lipophilic cation 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazol carbocyanine iodide) was selected as the optimal reporter substrate for ABCG2 activity, and the resulting assay was characterized by a Z' value of 0.50 and a signal-to-noise (S/N) ratio of 14 in a pilot screen of ∼ 7,000 diverse chemicals. The screen led to the identification of 64 unique nontoxic positives, yielding an initial hit rate of 1%, with 58 of them being confirmed activity. In addition, treatment with two selected confirmed positives suppressed the side population of U87MG-ABCG2 cells that was able to efflux the Hoechst dye as measured by flow cytometry, confirming that they constitute potent new ABCG2 transporter inhibitors. Our results demonstrate that our live cell and content-rich platform enables the rapid identification and profiling of ABCG2 modulators, and this new strategy opens the door to the discovery of compounds targeting the expression and/or trafficking of ABC transporters as an alternative to functional inhibitors that failed in the clinic.
Collapse
Affiliation(s)
- Christophe Antczak
- 1 High-Throughput Screening Core Facility, Memorial Sloan-Kettering Cancer Center , New York, New York
| | | | | | | | | | | |
Collapse
|
32
|
Fox JT, Myung K. Cell-based high-throughput screens for the discovery of chemotherapeutic agents. Oncotarget 2012; 3:581-5. [PMID: 22653910 PMCID: PMC3388188 DOI: 10.18632/oncotarget.513] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
With modern advances in robotics and data processing, high-throughput screening (HTS) is playing an increasingly growing role in the drug discovery process. The ultimate success of HTS depends upon the development of assays that are robust and reproducible in miniaturized formats, have low false-positive rates, and can identify drugs that offer improvements over those currently on the market. One example of such an assay is the ATAD5-luciferase HTS assay, which identified three antioxidants that could kill cancer cells without inducing mutagenesis. Here we discuss the ATAD5-luciferase assay and expand upon the value of HTS in identifying other potential cancer drugs, focusing on cell-based assays that involve DNA damage or repair pathways.
Collapse
Affiliation(s)
- Jennifer T Fox
- Genome Instability Section, Genetics and Molecular Biology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
33
|
Wang X, Deng R, Lu Y, Xu Q, Yan M, Ye D, Chen W. Gambogic acid as a non-competitive inhibitor of ATP-binding cassette transporter B1 reverses the multidrug resistance of human epithelial cancers by promoting ATP-binding cassette transporter B1 protein degradation. Basic Clin Pharmacol Toxicol 2012; 112:25-33. [PMID: 22759348 DOI: 10.1111/j.1742-7843.2012.00921.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 06/20/2012] [Indexed: 12/13/2022]
Abstract
Gambogic acid (GA) is known for its anti-cancer activity in a phase II clinical trial. However, the detailed molecular mechanisms of its anti-multidrug resistance remain unclear. The present study was designed to study the relationship between GA and multidrug-resistant protein ATP-binding cassette transporter B1 (ABCB1). GA dose dependently inhibited ABCB1 activity levels in the in vitro Pgp-Glo assay system and increased the cellular accumulation of ABCB1 substrate adriamycin. Although GA had no significant influence on ABCB1 mRNA in the real-time PCR assay, Western blot detection indicated the compound reduced ABCB1 protein levels. Further study showed the proteasome inhibitor MG-132 reversed the GA-decreased ABCB1 level and prolonged half-life of ABCB1. It was also found that GA coordinated with other anti-cancer drugs (such as adriamycin, docetaxel, verapamil and protopanaxadiol) to enhance cellular cytotoxicity on human epithelial cancer cell lines with higher ABCB1 expression levels. These data suggest that GA functions as a non-competitive inhibitor of ABCB1 by directly inhibiting and reducing its expression levels by promoting protein degradation through post-translational proteasome pathway. The results of this study will aid in the understanding of the synergistic effects of combining GA with other drugs as a new anti-multidrug-resistant agent.
Collapse
Affiliation(s)
- Xu Wang
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
INTRODUCTION Multidrug resistance (MDR) is the main cause of failure in cancer therapy. One mechanism responsible for MDR is the active efflux of drugs by ATP-binding cassette (ABC) transporters. Several agents have been developed to block transporter-mediated drug efflux and some of these compounds have entered Phase II/III clinical testing. Evidence is also emerging of the role played by ABC transporters in cancer cell signalling that is likely to be important in disease progression and which is distinct from MDR. AREAS COVERED This article reviews current literature to analyse the rationale for targeting ABC transporters in cancer. Preclinical and clinical results of ABC transporter inhibitors in early clinical trials, as single agents or in combination with other drugs, are described. The development of new strategies to target MDR and the emerging roles of ABC transporters in cancer signalling are discussed. EXPERT OPINION The intense active search for safe and effective inhibitors of ABC transporters has led to some success in MDR reversal in preclinical studies. However, there has been little impact on clinical outcome. The discovery of novel, potent and nontoxic inhibitors as well as new treatment strategies is therefore needed.
Collapse
Affiliation(s)
- Marco Falasca
- Queen Mary University of London, Blizard Institute, Barts and The London School of Medicine and Dentistry, Centre for Diabetes, Inositide Signalling Group, 4 Newark Street, London, UK.
| | | |
Collapse
|
35
|
Duan Z, Li X, Huang H, Yuan W, Zheng SL, Liu X, Zhang Z, Choy E, Harmon D, Mankin H, Hornicek F. Synthesis and Evaluation of (2-(4-Methoxyphenyl)-4-quinolinyl)(2-piperidinyl)methanol (NSC23925) Isomers To Reverse Multidrug Resistance in Cancer. J Med Chem 2012; 55:3113-21. [DOI: 10.1021/jm300117u] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Zhenfeng Duan
- Sarcoma Biology Laboratory, Center
for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, United
States
| | - Xin Li
- Chengdu ChemPartner Co., Ltd., Floor 3, Building 3, Tianfu Life Science
Park, No. 88, Keyuan South Road, Hi-Tech Zone, Chengdu, 610041, People's
Republic of China
| | - Haoxi Huang
- Chengdu ChemPartner Co., Ltd., Floor 3, Building 3, Tianfu Life Science
Park, No. 88, Keyuan South Road, Hi-Tech Zone, Chengdu, 610041, People's
Republic of China
| | - Wei Yuan
- Chengdu ChemPartner Co., Ltd., Floor 3, Building 3, Tianfu Life Science
Park, No. 88, Keyuan South Road, Hi-Tech Zone, Chengdu, 610041, People's
Republic of China
| | - Shao-Liang Zheng
- Department
of Chemistry and
Chemical Biology, Harvard University, Cambridge,
Massachusetts 02138, United States
| | - Xianzhe Liu
- Sarcoma Biology Laboratory, Center
for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, United
States
| | - Zhan Zhang
- Sarcoma Biology Laboratory, Center
for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, United
States
- The Third Affiliated Hospital of Zhengzhou University, People's Republic
of China
| | - Edwin Choy
- Sarcoma Biology Laboratory, Center
for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, United
States
| | - David Harmon
- Sarcoma Biology Laboratory, Center
for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, United
States
| | - Henry Mankin
- Sarcoma Biology Laboratory, Center
for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, United
States
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Center
for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts 02114, United
States
| |
Collapse
|
36
|
Hanušová V, Boušová I, Skálová L. Possibilities to increase the effectiveness of doxorubicin in cancer cells killing. Drug Metab Rev 2011; 43:540-57. [DOI: 10.3109/03602532.2011.609174] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Azzariti A, Quatrale AE, Porcelli L, Colabufo NA, Cantore M, Cassano G, Gasparre G, Iannelli G, Tommasi S, Panaro MA, Paradiso A. MC70 potentiates doxorubicin efficacy in colon and breast cancer in vitro treatment. Eur J Pharmacol 2011; 670:74-84. [PMID: 21925160 DOI: 10.1016/j.ejphar.2011.08.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 06/30/2011] [Accepted: 08/15/2011] [Indexed: 11/26/2022]
Abstract
A major limitation of cancer treatment is the ability of cancer cells to develop resistance to chemotherapeutic drugs, by the establishment of multidrug resistance. Here, we characterize MC70 as ABC transporters inhibitor and anticancer agent, alone or with chemotherapy. MC70 was analyzed for its interaction with ABCB1, ABCG2 and ABCC1 by specific transport assays. In breast and colon cancer cell lines, cell growth and apoptosis were measured by MTT assay and DNA laddering Elisa kit, respectively. Cell cycle perturbation and cellular targets modulation were analyzed by Flow-cytometry and Western blotting, respectively. MC70 interacted with ABC transporters. In breast cancer cells, MC70 slightly inhibited cell proliferation strongly enhancing doxorubicin effectiveness. By contrast, MC70 was found to inhibit cell growth in colon cancer cells without affecting doxorubicin efficacy and in combination with topoisomerase I inhibitors it could be a promising therapeutic approach. What is more, it was also observed that MC70 induced apoptosis, canceled in favor of necrosis when given in combination with high doses of doxorubicin. MC70 inhibited cell migration probably through its interaction with sigma-1 receptor. Modulations of i) cell cycle, ii) pAkt and the phosphorylation of the three MAPKs were highlighted, while any activity was excluded at transcription level, thus accounting for the phenotypic effects observed. MC70 might be considered as a new potential anticancer agent capable to i) enhance chemotherapy effectiveness and ii) to play a contributory role in the treatment of chemotherapy resistant tumors.
Collapse
Affiliation(s)
- Amalia Azzariti
- Clinical Experimental Oncology Laboratory, National Cancer Institute Giovanni Paolo II, Bari, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Role of cellular uptake in the reversal of multidrug resistance by PEG-b-PLA polymeric micelles. Biomaterials 2011; 32:5148-57. [DOI: 10.1016/j.biomaterials.2011.03.071] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 03/29/2011] [Indexed: 02/02/2023]
|
39
|
Kepp O, Galluzzi L, Lipinski M, Yuan J, Kroemer G. Cell death assays for drug discovery. Nat Rev Drug Discov 2011; 10:221-37. [PMID: 21358741 DOI: 10.1038/nrd3373] [Citation(s) in RCA: 405] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cell death has an important role in many human diseases, and strategies aimed at modulating the associated pathways have been successfully applied to treat various disorders. Indeed, several clinically promising cytotoxic and cytoprotective agents with potential applications in cancer, ischaemic and neurodegenerative diseases have recently been identified by high-throughput screening (HTS), based on appropriate cell death assays. Given that different cell death modalities may be dysregulated in different diseases, it is becoming increasingly clear that such assays need to not only quantify the extent of cell death, but they must also be able to distinguish between the various pathways. Here, we systematically describe approaches to accurately quantify distinct cell death pathways, discuss their advantages and pitfalls, and focus on those techniques that are amenable to HTS.
Collapse
|
40
|
Susa M, Choy E, Yang C, Schwab J, Mankin H, Hornicek F, Duan Z. Multidrug Resistance Reversal Agent, NSC77037, Identified with a Cell-Based Screening Assay. ACTA ACUST UNITED AC 2010; 15:287-96. [DOI: 10.1177/1087057109359422] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The development of multidrug resistance (MDR) remains a significant obstacle in treating cancer patients with chemotherapy. To identify small-molecule compounds that can reverse MDR, the authors used a cell-based screening assay with an MDR ovarian cancer cell line. Incubating MDR cells with a sublethal concentration of paclitaxel in combination with each of 2000 small-molecule compounds from the National Cancer Institute Diversity Set Library, they identified NSC77037. The cytotoxic activity of NSC77037 and the duration of its effect were evaluated in vitro using a panel of cancer cell lines expressing permeability glycoprotein (Pgp), multiple drug resistance protein 1 (MRP 1), and breast cancer resistance protein (BCRP). The mechanism of its effects was further analyzed by assessing the retention of calcein and Pgp-ATPase activity. The relative potency of MDR reversal by NSC77037 was significantly higher than that of frequently used MDR reversal agents such as verapamil and cyclosporine A. NSC77037 reversed Pgp without reversing MRP or BCRP-mediated MDR. NSC77037, at a concentration of >10 µM, moderately inhibited the proliferation of both sensitive and resistant cell lines, but the inhibitory effect of NSC77037 was not altered by coincubation with the Pgp inhibitor verapamil, suggesting that NSC77037 itself is not a substrate of Pgp. NSC77037 directly inhibited the function of Pgp in a dose-dependent manner, but it did not alter the protein expression level of Pgp. The use of NSC77037 to restore sensitivity to chemotherapy or to prevent resistance could be a potential treatment strategy for cancer patients.
Collapse
Affiliation(s)
- Michiro Susa
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Edwin Choy
- Division of Hematology Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Cao Yang
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Joseph Schwab
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Henry Mankin
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|