1
|
Hanssen KM, Haber M, Fletcher JI. Targeting multidrug resistance-associated protein 1 (MRP1)-expressing cancers: Beyond pharmacological inhibition. Drug Resist Updat 2021; 59:100795. [PMID: 34983733 DOI: 10.1016/j.drup.2021.100795] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/30/2021] [Accepted: 09/05/2021] [Indexed: 12/30/2022]
Abstract
Resistance to chemotherapy remains one of the most significant obstacles to successful cancer treatment. While inhibiting drug efflux mediated by ATP-binding cassette (ABC) transporters is a seemingly attractive and logical approach to combat multidrug resistance (MDR), small molecule inhibition of ABC transporters has so far failed to confer clinical benefit, despite considerable efforts by medicinal chemists, biologists, and clinicians. The long-sought treatment to eradicate cancers displaying ABC transporter overexpression may therefore lie within alternative targeting strategies. When aberrantly expressed, the ABC transporter multidrug resistance-associated protein 1 (MRP1, ABCC1) confers MDR, but can also shift cellular redox balance, leaving the cell vulnerable to select agents. Here, we explore the physiological roles of MRP1, the rational for targeting this transporter in cancer, the development of small molecule MRP1 inhibitors, and the most recent developments in alternative therapeutic approaches for targeting cancers with MRP1 overexpression. We discuss approaches that extend beyond simple MRP1 inhibition by exploiting the collateral sensitivity to glutathione depletion and ferroptosis, the rationale for targeting the shared transcriptional regulators of both MRP1 and glutathione biosynthesis, advances in gene silencing, and new molecules that modulate transporter activity to the detriment of the cancer cell. These strategies illustrate promising new approaches to address multidrug resistant disease that extend beyond the simple reversal of MDR and offer exciting routes for further research.
Collapse
Affiliation(s)
- Kimberley M Hanssen
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
2
|
Takubo H, Taniguchi T, Iwanaga K, Nomura Y. Evaluation of the changes in exposure to thiol compounds in chronic kidney disease patients using the PBPK model. Xenobiotica 2020; 51:31-39. [PMID: 32744915 DOI: 10.1080/00498254.2020.1805816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Targeted covalent inhibitors designed to bind covalently to a specific molecular target have recently been a focus of drug development. Among these inhibitors, thiol compounds bind covalently to endogenous thiols in the body through a process involving disulfide bonds. We investigated the predictability of changes in the exposure to captopril, tiopronin, the active form of dalcetrapib and the active metabolite of prasugrel, R-138727, all of which have a sulfhydryl group, in moderate and severe chronic kidney disease (CKD) patients using a constructed PBPK model. The changes in the exposure to captopril, tiopronin and the active form of dalcetrapib under CKD conditions were well predicted. However, the change in exposure to R-138727, which is a secondary metabolite of prasugrel, was overpredicted. Although these thiol compounds covalently bind to endogenous thiols, our study concluded that changes in exposure to these compounds under CKD conditions can probably be predicted, except for compounds with a complicated mechanism whereby the thiol metabolite is generated.
Collapse
Affiliation(s)
- Hiroaki Takubo
- Drug Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Toshio Taniguchi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| | - Kazunori Iwanaga
- Education and Research Center for Clinical Pharmacy, Osaka University of Pharmaceutical Sciences, Osaka, Japan
| | - Yukihiro Nomura
- Drug Metabolism and Pharmacokinetics Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan
| |
Collapse
|
3
|
Roth JS, Lee TD, Cheff DM, Gosztyla ML, Asawa RR, Danchik C, Michael S, Simeonov A, Klumpp-Thomas C, Wilson KM, Hall MD. Keeping It Clean: The Cell Culture Quality Control Experience at the National Center for Advancing Translational Sciences. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2020; 25:491-497. [PMID: 32233736 PMCID: PMC8506661 DOI: 10.1177/2472555220911451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Quality control monitoring of cell lines utilized in biomedical research is of utmost importance and is critical for the reproducibility of data. Two key pitfalls in tissue culture are 1) cell line authenticity and 2) Mycoplasma contamination. As a collaborative research institute, the National Center for Advancing Translational Sciences (NCATS) receives cell lines from a range of commercial and academic sources, which are adapted for high-throughput screening. Here, we describe the implementation of routine NCATS-wide Mycoplasma testing and short tandem repeat (STR) testing for cell lines. Initial testing identified a >10% Mycoplasma contamination rate. While the implementation of systematic testing has not fully suppressed Mycoplasma contamination rates, clearly defined protocols that include the immediate destruction of contaminated cell lines wherever possible has enabled rapid intervention and removal of compromised cell lines. Data for >2000 cell line samples tested over 3 years, and case studies are provided. STR testing of 186 cell lines with established STR profiles revealed only five misidentified cell lines, all of which were received from external labs. The data collected over the 3 years since implementation of this systematic testing demonstrate the importance of continual vigilance for rapid identification of "problem" cell lines, for ensuring reproducible data in translational science research.
Collapse
Affiliation(s)
- Jacob S. Roth
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Tobie D. Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Dorian M. Cheff
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Maya L. Gosztyla
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Rosita R. Asawa
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Carina Danchik
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Sam Michael
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Carleen Klumpp-Thomas
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Kelli M. Wilson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Matthew D. Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
4
|
Huff LM, Horibata S, Robey RW, Hall MD, Gottesman MM. Mycoplasma Infection Mediates Sensitivity of Multidrug-Resistant Cell Lines to Tiopronin: A Cautionary Tale. J Med Chem 2020; 63:1434-1439. [PMID: 31702923 DOI: 10.1021/acs.jmedchem.9b00484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We previously reported that some, but not all, multidrug-resistant cells that overexpressed various drug-resistance transporters were collaterally sensitive to tiopronin. In recent follow-up studies, we discovered that sensitivity to tiopronin in the original study was mediated by infection of the cells by a human-specific strain of mycoplasma. These results strongly support the need to constantly monitor cells for mycoplasma infection and keep stored samples of all cells that are used for in vitro studies.
Collapse
|
5
|
Olson KR, Briggs A, Devireddy M, Xian M, Gao Y. Are the beneficial effects of 'antioxidant' lipoic acid mediated through metabolism of reactive sulfur species? Free Radic Biol Med 2020; 146:139-149. [PMID: 31676393 DOI: 10.1016/j.freeradbiomed.2019.10.410] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 12/28/2022]
Abstract
The health benefits of lipoic acid (LA) are generally attributed to mitigating the harmful effects of reactive oxygen species (ROS). ROS are chemically similar to reactive sulfur species (RSS) and signal through identical mechanisms. Here we examined the effects of LA on RSS in HEK293 cells using H2S and polysulfide (PS) specific fluorophores, AzMC and SSP4. We show that LA concentration-dependently increased both H2S and PS. Physioxia (5% O2) augmented the effects of LA on H2S production but decreased PS production. Thiosulfate, a known substrate for reduced LA, and an intermediate in the catabolism of H2S enhanced the effects of LA on H2S and PS production. Inhibiting peroxiredoxins with conoidin A and gluraredoxins with tiopronin augmented the effects of LA on PS and H2S, respectively while decreasing glutathione with buthionine-sulfoximine (BSO) or diethyl maleate (DEM) decreased the stimulatory effect of LA on H2S production but augmented LA's effect on PS. Aminooxyacetate (AOA) and propargylglycine (PPG), inhibitors of H2S production from cysteine partially inhibited LA augmentation of H2S production and further decreased the LA effect when applied concurrently with BSO and DEM. The selective and cell-permeable H2S scavenger, SS20, inhibited the effects of LA on cellular H2S. Estimates of single-cell H2S production suggest that 0.1-0.2% of O2 consumption is used to metabolize H2S and these requirements may increase to 1-2% with 1 mM LA. Collectively, these results suggest that LA rescues H2S from irreversible oxidation and that the effects of LA on RSS directly confer antioxidant, anti-inflammatory and cytoprotective responses. They also suggest that TS may be an effective supplement to increase the efficacy of LA in clinical settings.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine, South Bend Center, South Bend, IN, 46617, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | - Austin Briggs
- Indiana University School of Medicine, South Bend Center, South Bend, IN, 46617, USA
| | - Monesh Devireddy
- Indiana University School of Medicine, South Bend Center, South Bend, IN, 46617, USA
| | - Ming Xian
- Department of Chemistry, Washington State University, Pullman, WA, 99164, USA
| | - Yan Gao
- Indiana University School of Medicine, South Bend Center, South Bend, IN, 46617, USA
| |
Collapse
|
6
|
Efferth T, Saeed ME, Kadioglu O, Seo EJ, Shirooie S, Mbaveng AT, Nabavi SM, Kuete V. Collateral sensitivity of natural products in drug-resistant cancer cells. Biotechnol Adv 2020; 38:107342. [DOI: 10.1016/j.biotechadv.2019.01.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 01/17/2019] [Accepted: 01/26/2019] [Indexed: 01/25/2023]
|
7
|
Zuber G, Weiss E, Chiper M. Biocompatible gold nanoclusters: synthetic strategies and biomedical prospects. NANOTECHNOLOGY 2019; 30:352001. [PMID: 31071693 DOI: 10.1088/1361-6528/ab2088] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The latest advances concerning ultra-small gold nanoparticles (≤2 nm) commonly known as gold nanoclusters (AuNCs) are reviewed and discussed in the context of biological and biomedical applications (labeling, delivery, imaging and therapy). A great diversity of synthetic methods has been developed and optimized aiming to improve the chemical structures and physicochemical properties of the resulting AuNCs. The main synthetic approaches were surveyed with emphasis on methods leading to water-soluble AuNCs since aqueous solutions are the preferred media for biological applications. The most representative and recent experimental results are discussed in relationship to their potential for biomedical applications.
Collapse
Affiliation(s)
- Guy Zuber
- Molecular and Pharmaceutical Engineering of Biologics, CNRS-Université de Strasbourg UMR 7242, Boulevard Sebastien Brant, F-67412, Illkirch, France
| | | | | |
Collapse
|
8
|
Olson KR, Gao Y. Effects of inhibiting antioxidant pathways on cellular hydrogen sulfide and polysulfide metabolism. Free Radic Biol Med 2019; 135:1-14. [PMID: 30790656 DOI: 10.1016/j.freeradbiomed.2019.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 12/18/2022]
Abstract
Elaborate antioxidant pathways have evolved to minimize the threat of excessive reactive oxygen species (ROS) and to regulate ROS as signaling entities. ROS are chemically and functionally similar to reactive sulfur species (RSS) and both ROS and RSS have been shown to be metabolized by the antioxidant enzymes, superoxide dismutase and catalase. Here we use fluorophores to examine the effects of a variety of inhibitors of antioxidant pathways on metabolism of two important RSS, hydrogen sulfide (H2S with AzMC) and polysulfides (H2Sn, where n = 2-7, with SSP4) in HEK293 cells. Cells were exposed to inhibitors for up to 5 days in normoxia (21% O2) and hypoxia (5% O2), conditions also known to affect ROS production. Decreasing intracellular glutathione (GSH) with l-buthionine-sulfoximine (BSO) or diethyl maleate (DEM) decreased H2S production for 5 days but did not affect H2Sn. The glutathione reductase inhibitor, auranofin, initially decreased H2S and H2Sn but after two days H2Sn increased over controls. Inhibition of peroxiredoxins with conoidin A decreased H2S and increased H2Sn, whereas the glutathione peroxidase inhibitor, tiopronin, increased H2S. Aminoadipic acid, an inhibitor of cystine uptake did not affect either H2S or H2Sn. In buffer, the glutathione reductase and thioredoxin reductase inhibitor, 2-AAPA, the glutathione peroxidase mimetic, ebselen, and tiopronin variously reacted directly with AzMC and SSP4, reacted with H2S and H2S2, or optically interfered with AzMC or SSP4 fluorescence. Collectively these results show that antioxidant inhibitors, generally known for their ability to increase cellular ROS, have various effects on cellular RSS. These findings suggest that the inhibitors may affect cellular sulfur metabolism pathways that are not related to ROS production and in some instances they may directly affect RSS or the methods used to measure them. They also illustrate the importance of carefully evaluating RSS metabolism when biologically or pharmacologically attempting to manipulate ROS.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine - South Bend, South Bend, IN, 46617, USA; Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.
| | - Yan Gao
- Indiana University School of Medicine - South Bend, South Bend, IN, 46617, USA
| |
Collapse
|
9
|
Cui Q, Wang JQ, Assaraf YG, Ren L, Gupta P, Wei L, Ashby CR, Yang DH, Chen ZS. Modulating ROS to overcome multidrug resistance in cancer. Drug Resist Updat 2018; 41:1-25. [DOI: 10.1016/j.drup.2018.11.001] [Citation(s) in RCA: 456] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023]
|
10
|
Ru nanoparticles coated with γ-Fe2O3 promoting and monitoring the differentiation of human mesenchymal stem cells via MRI tracking. Colloids Surf B Biointerfaces 2018; 170:701-711. [DOI: 10.1016/j.colsurfb.2018.05.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 01/13/2023]
|
11
|
Stefan SM, Wiese M. Small-molecule inhibitors of multidrug resistance-associated protein 1 and related processes: A historic approach and recent advances. Med Res Rev 2018; 39:176-264. [DOI: 10.1002/med.21510] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/05/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Sven Marcel Stefan
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| | - Michael Wiese
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| |
Collapse
|
12
|
Lemmerhirt H, Behnisch S, Bodtke A, Lillig CH, Pazderova L, Kasparkova J, Brabec V, Bednarski PJ. Effects of cytotoxic cis- and trans-diammine monochlorido platinum(II) complexes on selenium-dependent redox enzymes and DNA. J Inorg Biochem 2017; 178:94-105. [PMID: 29125948 DOI: 10.1016/j.jinorgbio.2017.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 10/16/2017] [Accepted: 10/25/2017] [Indexed: 11/16/2022]
Abstract
Here we present the preparation of 14 pairs of cis- and trans-diammine monochlorido platinum(II) complexes, coordinated to heterocycles (i.e., imidazole, 2-methylimidazole and pyrazole) and linked to various acylhydrazones, which were designed as potential inhibitors of the selenium-dependent enzymes glutathione peroxidase 1 (GPx-1) and thioredoxin reductase 1 (TrxR-1). However, no inhibition of bovine GPx-1 and only weak inhibition of murine TrxR-1 was observed in in vitro assays. Nonetheless, the cis configured diammine monochlorido Pt(II) complexes exhibited cytotoxic and apoptotic properties on various human cancer cell lines, whereas the trans configured complexes generally showed weaker potency with a few exceptions. On the other hand, the trans complexes were generally more likely to lack cross-resistance to cisplatin than the cis analogues. Platinum was found bound to the nuclear DNA of cancer cells treated with representative Pt complexes, suggesting that DNA might be a possible target. Thus, detailed in vitro binding experiments with DNA were conducted. Interactions of the compounds with calf thymus DNA were investigated, including Pt binding kinetics, circular dichroism (CD) spectral changes, changes in DNA melting temperatures, unwinding of supercoiled plasmids and ethidium bromide displacement in DNA. The CD results indicate that the most active cis configured pyrazole-derived complex causes unique structural changes in the DNA compared to the other complexes as well as to those caused by cisplatin, suggesting a denaturation of the DNA structure. This may be important for the antiproliferative activity of this compound in the cancer cells.
Collapse
Affiliation(s)
- Heidi Lemmerhirt
- Institute of Pharmacy, Ernst-Moritz-Arndt University of Greifswald, 17487 Greifswald, Germany
| | - Steven Behnisch
- Institute of Pharmacy, Ernst-Moritz-Arndt University of Greifswald, 17487 Greifswald, Germany
| | - Anja Bodtke
- Institute of Pharmacy, Ernst-Moritz-Arndt University of Greifswald, 17487 Greifswald, Germany
| | - Christopher H Lillig
- Institute of Medical Biochemistry and Molecular Biology, Ernst-Moritz-Arndt University of Greifswald, 17475 Greifswald, Germany
| | - Lucia Pazderova
- Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, 78371 Olomouc, Czech Republic
| | - Jana Kasparkova
- Department of Biophysics, Faculty of Science, Palacky University, Slechtitelu 27, 78371 Olomouc, Czech Republic
| | - Viktor Brabec
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Kralovopolska 135, 61265 Brno, Czech Republic
| | - Patrick J Bednarski
- Institute of Pharmacy, Ernst-Moritz-Arndt University of Greifswald, 17487 Greifswald, Germany.
| |
Collapse
|
13
|
Dankó B, Tóth S, Martins A, Vágvölgyi M, Kúsz N, Molnár J, Chang FR, Wu YC, Szakács G, Hunyadi A. Synthesis and SAR Study of Anticancer Protoflavone Derivatives: Investigation of Cytotoxicity and Interaction with ABCB1 and ABCG2 Multidrug Efflux Transporters. ChemMedChem 2017; 12:850-859. [DOI: 10.1002/cmdc.201700225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Balázs Dankó
- Institute of Pharmacognosy; University of Szeged; Szeged Hungary
| | - Szilárd Tóth
- Institute of Enzymology; Research Centre for Natural Sciences, Hungarian Academy of Sciences; Budapest Hungary
| | - Ana Martins
- Department of Medical Microbiology and Immunobiology; Faculty of Medicine; University of Szeged; Szeged Hungary
- Current address: Synthetic Systems Biology Unit; Institute of Biochemistry; Biological Research Centre; Temesvári krt. 62 6726 Szeged Hungary
| | - Máté Vágvölgyi
- Institute of Pharmacognosy; University of Szeged; Szeged Hungary
| | - Norbert Kúsz
- Institute of Pharmacognosy; University of Szeged; Szeged Hungary
| | - Joseph Molnár
- Department of Medical Microbiology and Immunobiology; Faculty of Medicine; University of Szeged; Szeged Hungary
| | - Fang-Rong Chang
- Graduate Institute of Natural Products; Kaohsiung Medical University; Kaohsiung Taiwan, R.O.C
- Cancer Center; Kaohsiung Medical University Hospital; Kaohsiung Taiwan, R.O.C
- R&D Center of Chinese Herbal Medicines & New Drugs, College of Pharmacy; Kaohsiung Medical University; Kaohsiung Taiwan, R.O.C
| | - Yang-Chang Wu
- Graduate Institute of Natural Products; Kaohsiung Medical University; Kaohsiung Taiwan, R.O.C
- Research Center for Natural Products and Drug Development; Kaohsiung Medical University; Kaohsiung Taiwan, R.O.C
- Department of Medical Research; Kaohsiung Medical University Hospital; Kaohsiung Taiwan, R.O.C
| | - Gergely Szakács
- Institute of Enzymology; Research Centre for Natural Sciences, Hungarian Academy of Sciences; Budapest Hungary
- Institute of Cancer Research; Medical University Vienna; Vienna Austria
| | - Attila Hunyadi
- Institute of Pharmacognosy; University of Szeged; Szeged Hungary
- Interdisciplinary Centre for Natural Products; University of Szeged; Szeged Hungary
| |
Collapse
|
14
|
DuHadaway J, Prendergast GC. Antimetabolite TTL-315 selectively kills glucose-deprived cancer cells and enhances responses to cytotoxic chemotherapy in preclinical models of cancer. Oncotarget 2016; 7:7372-80. [PMID: 26840263 PMCID: PMC4884924 DOI: 10.18632/oncotarget.7058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/23/2016] [Indexed: 11/26/2022] Open
Abstract
Maintaining thiol homeostasis is an imperative for cancer cell survival in the nutrient-deprived microenvironment of solid tumors. Despite this metabolic vulnerability, a selective approach has yet to be developed to disrupt thiol homeostasis in solid tumors for therapeutic purposes. In this study, we report the identification of 2-mercaptopropionyl glycine disulfide (TTL-315) as a novel antimetabolite that blocks cell survival in a manner conditional on glucose deprivation. In the presence of glucose, TTL-315 lacks cytotoxic effects in normal cells where it is detoxified by reduction to 2-mercaptopropionyl glycine, a compound with known clinical pharmacologic and safety profiles. In several rodent models of aggressive breast, lung and skin cancers, TTL-315 blocked tumor growth and cooperated with the DNA damaging drug cisplatin to trigger tumor regression. Our results offer preclinical proof of concept for TTL-315 as a novel antimetabolite to help selectively eradicate solid tumors by exploiting the glucose-deprived conditions of the tumor microenvironment.
Collapse
Affiliation(s)
| | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA, USA.,Sidney Kimmel Cancer Center and Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelpia, PA, USA
| |
Collapse
|
15
|
Pape VF, Tóth S, Füredi A, Szebényi K, Lovrics A, Szabó P, Wiese M, Szakács G. Design, synthesis and biological evaluation of thiosemicarbazones, hydrazinobenzothiazoles and arylhydrazones as anticancer agents with a potential to overcome multidrug resistance. Eur J Med Chem 2016; 117:335-54. [DOI: 10.1016/j.ejmech.2016.03.078] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/22/2016] [Accepted: 03/25/2016] [Indexed: 12/16/2022]
|
16
|
England CG, Gobin AM, Frieboes HB. Evaluation of uptake and distribution of gold nanoparticles in solid tumors. EUROPEAN PHYSICAL JOURNAL PLUS 2015; 130:231. [PMID: 27014559 PMCID: PMC4800753 DOI: 10.1140/epjp/i2015-15231-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Although nanotherapeutics offer a targeted and potentially less toxic alternative to systemic chemotherapy in cancer treatment, nanotherapeutic transport is typically hindered by abnormal characteristics of tumor tissue. Once nanoparticles targeted to tumor cells arrive in the circulation of tumor vasculature, they must extravasate from irregular vessels and diffuse through the tissue to ideally reach all malignant cells in cytotoxic concentrations. The enhanced permeability and retention effect can be leveraged to promote extravasation of appropriately sized particles from tumor vasculature; however, therapeutic success remains elusive partly due to inadequate intra-tumoral transport promoting heterogeneous nanoparticle uptake and distribution. Irregular tumor vasculature not only hinders particle transport but also sustains hypoxic tissue kregions with quiescent cells, which may be unaffected by cycle-dependent chemotherapeutics released from nanoparticles and thus regrow tumor tissue following nanotherapy. Furthermore, a large proportion of systemically injected nanoparticles may become sequestered by the reticuloendothelial system, resulting in overall diminished efficacy. We review recent work evaluating the uptake and distribution of gold nanoparticles in pre-clinical tumor models, with the goal to help improve nanotherapy outcomes. We also examine the potential role of novel layered gold nanoparticles designed to address some of these critical issues, assessing their uptake and transport in cancerous tissue.
Collapse
Affiliation(s)
- Christopher G England
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA
| | - André M Gobin
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Hermann B Frieboes
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY 40292, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40292, USA; Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
17
|
Saab AM, Guerrini A, Zeino M, Wiench B, Rossi D, Gambari R, Sacchetti G, Greten HJ, Efferth T. Laurus nobilisL. Seed Extract Reveals Collateral Sensitivity in Multidrug-Resistant P-Glycoprotein-Expressing Tumor Cells. Nutr Cancer 2015; 67:664-75. [DOI: 10.1080/01635581.2015.1019632] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
18
|
Lorendeau D, Dury L, Genoux-Bastide E, Lecerf-Schmidt F, Simões-Pires C, Carrupt PA, Terreux R, Magnard S, Di Pietro A, Boumendjel A, Baubichon-Cortay H. Collateral sensitivity of resistant MRP1-overexpressing cells to flavonoids and derivatives through GSH efflux. Biochem Pharmacol 2014; 90:235-45. [DOI: 10.1016/j.bcp.2014.05.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/16/2014] [Accepted: 05/19/2014] [Indexed: 10/25/2022]
|
19
|
Huo S, Jin S, Ma X, Xue X, Yang K, Kumar A, Wang PC, Zhang J, Hu Z, Liang XJ. Ultrasmall gold nanoparticles as carriers for nucleus-based gene therapy due to size-dependent nuclear entry. ACS NANO 2014; 8:5852-62. [PMID: 24824865 PMCID: PMC4076024 DOI: 10.1021/nn5008572] [Citation(s) in RCA: 289] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/13/2014] [Indexed: 05/19/2023]
Abstract
The aim of this study was to determine the size-dependent penetration ability of gold nanoparticles and the potential application of ultrasmall gold nanoparticles for intranucleus delivery and therapy. We synthesized gold nanoparticles with diameters of 2, 6, 10, and 16 nm and compared their intracellular distribution in MCF-7 breast cancer cells. Nanoparticles smaller than 10 nm (2 and 6 nm) could enter the nucleus, whereas larger ones (10 and 16 nm) were found only in the cytoplasm. We then investigated the possibility of using ultrasmall 2 nm nanoparticles as carriers for nuclear delivery of a triplex-forming oligonucleotide (TFO) that binds to the c-myc promoter. Compared to free TFO, the nanoparticle-conjugated TFO was more effective at reducing c-myc RNA and c-myc protein, which resulted in reduced cell viability. Our result demonstrated that the entry of gold nanoparticles into the cell nucleus is critically dependent on the size of the nanoparticles. We developed a strategy for regulating gene expression, by directly delivering TFOs into the nucleus using ultrasmall gold nanoparticles. More importantly, guidelines were provided to choose appropriate nanocarriers for different biomedical purposes.
Collapse
Affiliation(s)
- Shuaidong Huo
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, China 100190
- University of Chinese Academy of Sciences, Beijing, China 100049
| | - Shubin Jin
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, China 100190
- University of Chinese Academy of Sciences, Beijing, China 100049
| | - Xiaowei Ma
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, China 100190
- University of Chinese Academy of Sciences, Beijing, China 100049
| | - Xiangdong Xue
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, China 100190
- University of Chinese Academy of Sciences, Beijing, China 100049
| | - Keni Yang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, China 100190
| | - Anil Kumar
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, China 100190
| | - Paul C. Wang
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington D.C. 20060, United States
| | - Jinchao Zhang
- College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, China
| | - Zhongbo Hu
- University of Chinese Academy of Sciences, Beijing, China 100049
- Address correspondence to ,
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No.11, First North Road, Zhongguancun, Beijing, China 100190
- Address correspondence to ,
| |
Collapse
|
20
|
Fung KL, Tepede AK, Pluchino KM, Pouliot LM, Pixley JN, Hall MD, Gottesman MM. Uptake of compounds that selectively kill multidrug-resistant cells: the copper transporter SLC31A1 (CTR1) increases cellular accumulation of the thiosemicarbazone NSC73306. Mol Pharm 2014; 11:2692-702. [PMID: 24800945 PMCID: PMC4137994 DOI: 10.1021/mp500114e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Acquired drug resistance in cancer continues to be a challenge in cancer therapy, in part due to overexpression of the drug efflux transporter P-glycoprotein (P-gp, MDR1, ABCB1). NSC73306 is a thiosemicarbazone compound that displays greater toxicity against cells expressing functional P-gp than against other cells. Here, we investigate the cellular uptake of NSC73306, and examine its interaction with P-gp and copper transporter 1 (CTR1, SLC31A1). Overexpression of P-gp sensitizes LLC-PK1 cells to NSC73306. Cisplatin (IC50 = 77 μM), cyclosporin A (IC50 = 500 μM), and verapamil (IC50 = 700 μM) inhibited cellular accumulation of [(3)H]NSC73306. Cellular hypertoxicity of NSC73306 to P-gp-expressing cells was inhibited by cisplatin in a dose-dependent manner. Cells transiently expressing the cisplatin uptake transporter CTR1 (SLC31A1) showed increased [(3)H]NSC73306 accumulation. In contrast, CTR1 knockdown decreased [(3)H]NSC73306 accumulation. The presence of NSC73306 reduced CTR1 levels, similar to the negative feedback of CTR1 levels by copper or cisplatin. Surprisingly, although cisplatin is a substrate of CTR1, we found that CTR1 protein was overexpressed in high-level cisplatin-resistant KB-CP20 and BEL7404-CP20 cell lines. We confirmed that the CTR1 protein was functional, as uptake of NSC73306 was increased in KB-CP20 cells compared to their drug-sensitive parental cells, and downregulation of CTR1 in KB-CP20 cells reduced [(3)H]NSC73306 accumulation. These results suggest that NSC73306 is a transport substrate of CTR1.
Collapse
Affiliation(s)
- King Leung Fung
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland 20892, United States
| | | | | | | | | | | | | |
Collapse
|
21
|
Hall MD, Marshall TS, Kwit ADT, Miller Jenkins LM, Dulcey AE, Madigan JP, Pluchino KM, Goldsborough AS, Brimacombe KR, Griffiths GL, Gottesman MM. Inhibition of glutathione peroxidase mediates the collateral sensitivity of multidrug-resistant cells to tiopronin. J Biol Chem 2014; 289:21473-89. [PMID: 24930045 DOI: 10.1074/jbc.m114.581702] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Multidrug resistance (MDR) is a major obstacle to the successful chemotherapy of cancer. MDR is often the result of overexpression of ATP-binding cassette transporters following chemotherapy. A common ATP-binding cassette transporter that is overexpressed in MDR cancer cells is P-glycoprotein, which actively effluxes drugs against a concentration gradient, producing an MDR phenotype. Collateral sensitivity (CS), a phenomenon of drug hypersensitivity, is defined as the ability of certain compounds to selectively target MDR cells, but not the drug-sensitive parent cells from which they were derived. The drug tiopronin has been previously shown to elicit CS. However, unlike other CS agents, the mechanism of action was not dependent on the expression of P-glycoprotein in MDR cells. We have determined that the CS activity of tiopronin is mediated by the generation of reactive oxygen species (ROS) and that CS can be reversed by a variety of ROS-scavenging compounds. Specifically, selective toxicity of tiopronin toward MDR cells is achieved by inhibition of glutathione peroxidase (GPx), and the mode of inhibition of GPx1 by tiopronin is shown in this report. Why MDR cells are particularly sensitive to ROS is discussed, as is the difficulty in exploiting this hypersensitivity to tiopronin in the clinic.
Collapse
Affiliation(s)
- Matthew D Hall
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Travis S Marshall
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Alexandra D T Kwit
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Lisa M Miller Jenkins
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Andrés E Dulcey
- the Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - James P Madigan
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Kristen M Pluchino
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Andrew S Goldsborough
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Kyle R Brimacombe
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| | - Gary L Griffiths
- the Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Michael M Gottesman
- From the Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892 and
| |
Collapse
|
22
|
Szakács G, Hall MD, Gottesman MM, Boumendjel A, Kachadourian R, Day BJ, Baubichon-Cortay H, Di Pietro A. Targeting the Achilles heel of multidrug-resistant cancer by exploiting the fitness cost of resistance. Chem Rev 2014; 114:5753-74. [PMID: 24758331 PMCID: PMC4059772 DOI: 10.1021/cr4006236] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences , Magyar tudósok körútja 2, Budapest 1117, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Callaghan R, Luk F, Bebawy M. Inhibition of the multidrug resistance P-glycoprotein: time for a change of strategy? Drug Metab Dispos 2014; 42:623-31. [PMID: 24492893 PMCID: PMC3965902 DOI: 10.1124/dmd.113.056176] [Citation(s) in RCA: 310] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 01/31/2014] [Indexed: 01/30/2023] Open
Abstract
P-glycoprotein (P-gp) is a key player in the multidrug-resistant phenotype in cancer. The protein confers resistance by mediating the ATP-dependent efflux of an astonishing array of anticancer drugs. Its broad specificity has been the subject of numerous attempts to inhibit the protein and restore the efficacy of anticancer drugs. The general strategy has been to develop compounds that either compete with anticancer drugs for transport or act as direct inhibitors of P-gp. Despite considerable in vitro success, there are no compounds currently available to "block" P-gp-mediated resistance in the clinic. The failure may be attributed to toxicity, adverse drug interaction, and numerous pharmacokinetic issues. This review provides a description of several alternative approaches to overcome the activity of P-gp in drug-resistant cells. These include 1) drugs that specifically target resistant cells, 2) novel nanotechnologies to provide high-dose, targeted delivery of anticancer drugs, 3) compounds that interfere with nongenomic transfer of resistance, and 4) approaches to reduce the expression of P-gp within tumors. Such approaches have been developed through the pursuit of greater understanding of resistance mediators such as P-gp, and they show considerable potential for further application.
Collapse
Affiliation(s)
- Richard Callaghan
- Division of Biomedical Science & Biochemistry, Research School of Biology, College of Medicine, Biology & Environment, The Australian National University, Canberra, New South Wales, Australia (R.C.); and School of Pharmacy, Graduate School of Health, The University of Technology, Sydney, New South Wales, Australia (F.L., M.B.)
| | | | | |
Collapse
|
24
|
Huo S, Jin S, Zheng K, He S, Wang D, Liang X. Preparation and characterization of doxorubicin functionalized tiopronin-capped gold nanorods for cancer therapy. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s11434-013-5918-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Huo S, Ma H, Huang K, Liu J, Wei T, Jin S, Zhang J, He S, Liang XJ. Superior penetration and retention behavior of 50 nm gold nanoparticles in tumors. Cancer Res 2013; 73:319-30. [PMID: 23074284 DOI: 10.1158/0008-5472.can-12-2071] [Citation(s) in RCA: 252] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nanoparticles offer potential as drug delivery systems for chemotherapeutics based on certain advantages of molecular drugs. In this study, we report that particle size exerts great influence on the penetration and retention behavior of nanoparticles entering tumors. On comparing gold-coated Au@tiopronin nanoparticles that were prepared with identical coating and surface properties, we found that 50 nanoparticles were more effective in all in vitro, ex vivo, and in vivo assays conducted using MCF-7 breast cells as a model system. Beyond superior penetration in cultured cell monolayers, 50 nm Au@tiopronin nanoparticles also penetrated more deeply into tumor spheroids ex vivo and accumulated more effectively in tumor xenografts in vivo after a single intravenous dose. In contrast, larger gold-coated nanoparticles were primarily localized in the periphery of the tumor spheroid and around blood vessels, hindering deep penetration into tumors. We found multicellular spheroids to offer a simple ex vivo tumor model to simulate tumor tissue for screening the nanoparticle penetration behavior. Taken together, our findings define an optimal smaller size for nanoparticles that maximizes their effective accumulation in tumor tissue.
Collapse
Affiliation(s)
- Shuaidong Huo
- School of Materials Science and Engineering, Tianjin Polytechnic University, Tianjin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Multidrug Resistance in Cancer: A Tale of ABC Drug Transporters. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2013. [DOI: 10.1007/978-1-4614-7070-0_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
27
|
Saeed M, Greten HJ, Efferth T. Collateral Sensitivity in Drug-Resistant Tumor Cells. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2013. [DOI: 10.1007/978-1-4614-7070-0_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
28
|
Xia CQ, Smith PG. Drug efflux transporters and multidrug resistance in acute leukemia: therapeutic impact and novel approaches to mediation. Mol Pharmacol 2012; 82:1008-21. [PMID: 22826468 DOI: 10.1124/mol.112.079129] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Multidrug resistance (MDR), which is mediated by multiple drug efflux ATP-binding cassette (ABC) transporters, is a critical issue in the treatment of acute leukemia, with permeability glycoprotein (P-gp), multidrug resistance-associated protein 1, and breast cancer resistance protein (i.e., ABCG2) consistently being shown to be key effectors of MDR in cell line studies. Studies have demonstrated that intrinsic MDR can arise as a result of specific gene expression profiles and that drug-induced overexpression of P-gp and other MDR proteins can result in acquired resistance, with multiple ABC transporters having been shown to be overexpressed in cell lines selected for resistance to multiple drugs used to treat acute leukemia. Furthermore, numerous anticancer drugs, including agents commonly used for the treatment of acute leukemia (e.g., doxorubicin, vincristine, mitoxantrone, and methotrexate), have been shown to be P-gp substrates or to be susceptible to efflux mediated by other MDR proteins, and multiple clinical studies have demonstrated associations between P-gp or other MDR protein expression and responses to therapy or survival rates in acute leukemia. Here we review the importance of MDR in cancer, with a focus on acute leukemia, and we highlight the need for rapid accurate assessment of MDR status for optimal treatment selection. We also address the latest research on overcoming MDR, from inhibition of P-gp and other MDR proteins through various approaches (including direct antagonism and gene silencing) to the design of novel agents or novel delivery systems for existing therapeutic agents, to evade cellular efflux.
Collapse
Affiliation(s)
- Cindy Q Xia
- Millennium Pharmaceuticals, Inc., Cambridge, MA 02139, USA.
| | | |
Collapse
|
29
|
Huang K, Ma H, Liu J, Huo S, Kumar A, Wei T, Zhang X, Jin S, Gan Y, Wang PC, He S, Zhang X, Liang XJ. Size-dependent localization and penetration of ultrasmall gold nanoparticles in cancer cells, multicellular spheroids, and tumors in vivo. ACS NANO 2012; 6:4483-4493. [PMID: 22540892 DOI: 10.1021/nn301282m/suppl_file/nn301282m_si_001.pdf] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
This work demonstrated that ultrasmall gold nanoparticles (AuNPs) smaller than 10 nm display unique advantages over nanoparticles larger than 10 nm in terms of localization to, and penetration of, breast cancer cells, multicellular tumor spheroids, and tumors in mice. Au@tiopronin nanoparticles that have tunable sizes from 2 to 15 nm with identical surface coatings of tiopronin and charge were successfully prepared. For monolayer cells, the smaller the Au@tiopronin NPs, the more AuNPs found in each cell. In addition, the accumulation of Au NPs in the ex vivo tumor model was size-dependent: smaller AuNPs were able to penetrate deeply into tumor spheroids, whereas 15 nm nanoparticles were not. Owing to their ultrasmall nanostructure, 2 and 6 nm nanoparticles showed high levels of accumulation in tumor tissue in mice after a single intravenous injection. Surprisingly, both 2 and 6 nm Au@tiopronin nanoparticles were distributed throughout the cytoplasm and nucleus of cancer cells in vitro and in vivo, whereas 15 nm Au@tiopronin nanoparticles were found only in the cytoplasm, where they formed aggregates. The ex vivo multicellular spheroid proved to be a good model to simulate in vivo tumor tissue and evaluate nanoparticle penetration behavior. This work gives important insights into the design and functionalization of nanoparticles to achieve high levels of accumulation in tumors.
Collapse
Affiliation(s)
- Keyang Huang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Huang K, Ma H, Liu J, Huo S, Kumar A, Wei T, Zhang X, Jin S, Gan Y, Wang PC, He S, Zhang X, Liang XJ. Size-dependent localization and penetration of ultrasmall gold nanoparticles in cancer cells, multicellular spheroids, and tumors in vivo. ACS NANO 2012; 6:4483-93. [PMID: 22540892 PMCID: PMC3370420 DOI: 10.1021/nn301282m] [Citation(s) in RCA: 639] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
This work demonstrated that ultrasmall gold nanoparticles (AuNPs) smaller than 10 nm display unique advantages over nanoparticles larger than 10 nm in terms of localization to, and penetration of, breast cancer cells, multicellular tumor spheroids, and tumors in mice. Au@tiopronin nanoparticles that have tunable sizes from 2 to 15 nm with identical surface coatings of tiopronin and charge were successfully prepared. For monolayer cells, the smaller the Au@tiopronin NPs, the more AuNPs found in each cell. In addition, the accumulation of Au NPs in the ex vivo tumor model was size-dependent: smaller AuNPs were able to penetrate deeply into tumor spheroids, whereas 15 nm nanoparticles were not. Owing to their ultrasmall nanostructure, 2 and 6 nm nanoparticles showed high levels of accumulation in tumor tissue in mice after a single intravenous injection. Surprisingly, both 2 and 6 nm Au@tiopronin nanoparticles were distributed throughout the cytoplasm and nucleus of cancer cells in vitro and in vivo, whereas 15 nm Au@tiopronin nanoparticles were found only in the cytoplasm, where they formed aggregates. The ex vivo multicellular spheroid proved to be a good model to simulate in vivo tumor tissue and evaluate nanoparticle penetration behavior. This work gives important insights into the design and functionalization of nanoparticles to achieve high levels of accumulation in tumors.
Collapse
Affiliation(s)
- Keyang Huang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | - Huili Ma
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | - Juan Liu
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | - Shuaidong Huo
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
- School of Materials Science and Engineering, Tianjin Polytechnic University, Tianjin, China 300387
| | - Anil Kumar
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | - Tuo Wei
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | - Xu Zhang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | - Shubin Jin
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | - Yaling Gan
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
| | - Paul C. Wang
- Laboratory of Molecular Imaging, Department of Radiology, Howard University, Washington, D.C. 20060, United States
| | - Shengtai He
- School of Materials Science and Engineering, Tianjin Polytechnic University, Tianjin, China 300387
- Address correspondence to ; ;
| | - Xiaoning Zhang
- Laboratory of Pharmaceutics, School of Medicine, Tsinghua University, Beijing, China 100084
- Address correspondence to ; ;
| | - Xing-Jie Liang
- CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11, First North Road, Zhongguancun, Beijing, China 100190
- Address correspondence to ; ;
| |
Collapse
|
31
|
Pluchino KM, Hall MD, Goldsborough AS, Callaghan R, Gottesman MM. Collateral sensitivity as a strategy against cancer multidrug resistance. Drug Resist Updat 2012; 15:98-105. [PMID: 22483810 DOI: 10.1016/j.drup.2012.03.002] [Citation(s) in RCA: 239] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
While chemotherapy remains the most effective treatment for disseminated tumors, acquired or intrinsic drug resistance accounts for approximately 90% of treatment failure. Multidrug resistance (MDR), the simultaneous resistance to drugs that differ both structurally and mechanistically, often results from drug efflux pumps in the cell membrane that reduce intracellular drug levels to less than therapeutic concentrations. Expression of the MDR transporter P-glycoprotein (P-gp, MDR1, ABCB1) has been shown to correlate with overall poor chemotherapy response and prognosis. This review will focus on collateral sensitivity (CS), the ability of compounds to kill MDR cells selectively over the parental cells from which they were derived. Insights into CS may offer an alternative strategy for the clinical resolution of MDR, as highly selective and potent CS agents may lead to drugs that are effective at MDR cell killing and tumor resensitization. Four main mechanistic hypotheses for CS will be reviewed, followed by a discussion on quantitative and experimental evaluation of CS.
Collapse
Affiliation(s)
- Kristen M Pluchino
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
32
|
Hall MD, Brimacombe KR, Varonka MS, Pluchino KM, Monda JK, Li J, Walsh MJ, Boxer MB, Warren TH, Fales HM, Gottesman MM. Synthesis and structure-activity evaluation of isatin-β-thiosemicarbazones with improved selective activity toward multidrug-resistant cells expressing P-glycoprotein. J Med Chem 2011; 54:5878-89. [PMID: 21721528 PMCID: PMC3201829 DOI: 10.1021/jm2006047] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer multidrug resistance (MDR) mediated by ATP-binding cassette (ABC) transporters presents a significant unresolved clinical challenge. One strategy to resolve MDR is to develop compounds that selectively kill cells overexpressing the efflux transporter P-glycoprotein (MDR1, P-gp, ABCB1). We have previously reported structure-activity studies based around the lead compound NSC73306 (1, 1-isatin-4-(4'-methoxyphenyl)-3-thiosemicarbazone, 4.3-fold selective). Here we sought to extend this work on MDR1-selective analogues by establishing whether 1 showed "robust" activity against a range of cell lines expressing P-gp. We further aimed to synthesize and test analogues with varied substitution at the N4-position, and substitution around the N4-phenyl ring of isatin-β-thiosemicarbazones (IBTs), to identify compounds with increased MDR1-selectivity. Compound 1 demonstrated MDR1-selectivity against all P-gp-expressing cell lines examined. This selectivity was reversed by inhibitors of P-gp ATPase activity. Structural variation at the 4'-phenyl position of 1 yielded compounds of greater MDR1-selectivity. Two of these analogues, 1-isatin-4-(4'-nitrophenyl)-3-thiosemicarbazone (22, 8.3-fold selective) and 1-isatin-4-(4'-tert-butyl phenyl)-3-thiosemicarbazone (32, 14.8-fold selective), were selected for further testing and were found to retain the activity profile of 1. These compounds are the most active IBTs identified to date.
Collapse
Affiliation(s)
- Matthew D. Hall
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kyle R. Brimacombe
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Matthew S. Varonka
- Department of Chemistry, Georgetown University, Box 571227, Washington, D.C. 20057
| | - Kristen M. Pluchino
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Julie K. Monda
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jiayang Li
- Laboratory of Applied Mass Spectrometry, National Heart, Lung & Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Martin J. Walsh
- NIH Chemical Genomics Center, NIH Center for Translational Therapeutics, National Institutes of Health, 9800 Medical Center Drive, MSC 3370, Bethesda, MD 20892
| | - Matthew B. Boxer
- NIH Chemical Genomics Center, NIH Center for Translational Therapeutics, National Institutes of Health, 9800 Medical Center Drive, MSC 3370, Bethesda, MD 20892
| | - Timothy H. Warren
- Department of Chemistry, Georgetown University, Box 571227, Washington, D.C. 20057
| | - Henry M. Fales
- Laboratory of Applied Mass Spectrometry, National Heart, Lung & Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michael M. Gottesman
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|