1
|
Skvortsov DA, Zhirkina IV, Ipatova DA, Vasilyeva LA, Ivanenkov YA, Rubtsova MP, Kartsev VG, Sergiev PV, Dontsova OA. Coculture-Based Screening Revealed Selective Cytostatic Effects of Pyrazol-Azepinoindoles. ChemMedChem 2025:e2500052. [PMID: 40159440 DOI: 10.1002/cmdc.202500052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
This work focuses on the search for new small molecules for anticancer therapy using the fluorescent cells cocultivation test (FCCT). This method allows the control of the specificity of the action of compounds from the earliest stages of drug development. For the FCCT, labeled MCF7' breast cancer cells and noncancerous breast MCF10A cells are cocultured. Screening of 2025 compounds in the above system and previously developed coculture of A549 with VA13 yields 16 selectively cytotoxic molecules. The results are confirmed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay for seven of these molecules. Few are known as potential antitumor agents: angelicin, coumarin, and colchicine derivatives. However, the structures of macrocycle 1, pyrazole-azepinoindole derivative 2, and complex heterocyclic derivative 3 are not described as anticancer compounds according to the PubChem and SciFinder databases. Structure-activity relationships are investigated for 2 and its derivatives. The indole with a caprolactam ring (tetrahydro-azepinoindolone core) together with the pyrazolyl at the third position is the key element of the pharmacophore. The optimized pyrazole-azepinoindole derivative 23 shows SI = 18 for HCT116 versus VA-13 on the expanded array of cell lines. Its effect is mainly mediated by the G1 arrest of the cell cycle.
Collapse
Affiliation(s)
- Dmitry A Skvortsov
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | - Irina V Zhirkina
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | - Daria A Ipatova
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | - Lilya A Vasilyeva
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | - Yan A Ivanenkov
- P. Hertsen Moscow Oncology Research Institute, Moscow, 3125284, Russian Federation
- The Federal State Unitary Enterprise Dukhov Automatics Research Institute, Moscow, 127055, Russian Federation
| | - Maria P Rubtsova
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
| | | | - Petr V Sergiev
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
- Center of Molecular and Cellular Biology, Moscow, 121205, Russian Federation
| | - Olga A Dontsova
- Chemistry Department and AN Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, Building 1/3 GSP-1, Moscow, 119991, Russian Federation
- Center of Molecular and Cellular Biology, Moscow, 121205, Russian Federation
| |
Collapse
|
2
|
Lerma Clavero A, Boqvist PL, Ingelshed K, Bosdotter C, Sedimbi S, Jiang L, Wermeling F, Vojtesek B, Lane DP, Kannan P. MDM2 inhibitors, nutlin-3a and navtemadelin, retain efficacy in human and mouse cancer cells cultured in hypoxia. Sci Rep 2023; 13:4583. [PMID: 36941277 PMCID: PMC10027891 DOI: 10.1038/s41598-023-31484-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/13/2023] [Indexed: 03/23/2023] Open
Abstract
Activation of p53 by small molecule MDM2 inhibitors can induce cell cycle arrest or death in p53 wildtype cancer cells. However, cancer cells exposed to hypoxia can develop resistance to other small molecules, such as chemotherapies, that activate p53. Here, we evaluated whether hypoxia could render cancer cells insensitive to two MDM2 inhibitors with different potencies, nutlin-3a and navtemadlin. Inhibitor efficacy and potency were evaluated under short-term hypoxic conditions in human and mouse cancer cells expressing different p53 genotypes (wild-type, mutant, or null). Treatment of wild-type p53 cancer cells with MDM2 inhibitors reduced cell growth by > 75% in hypoxia through activation of the p53-p21 signaling pathway; no inhibitor-induced growth reduction was observed in hypoxic mutant or null p53 cells except at very high concentrations. The concentration of inhibitors needed to induce the maximal p53 response was not significantly different in hypoxia compared to normoxia. However, inhibitor efficacy varied by species and by cell line, with stronger effects at lower concentrations observed in human cell lines than in mouse cell lines grown as 2D and 3D cultures. Together, these results indicate that MDM2 inhibitors retain efficacy in hypoxia, suggesting they could be useful for targeting acutely hypoxic cancer cells.
Collapse
Affiliation(s)
- Ada Lerma Clavero
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Medical Cell Biology, Uppsala University, 751 23, Uppsala, Sweden
| | - Paula Lafqvist Boqvist
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Katrine Ingelshed
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Cecilia Bosdotter
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Saikiran Sedimbi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
- Moderna Therapeutics, 200 Technology Square, Cambridge, MA, 02139, USA
| | - Long Jiang
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Fredrik Wermeling
- Department of Medicine Solna, Center for Molecular Medicine, Karolinska University Hospital and Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Borivoj Vojtesek
- RECAMO, Masaryk Memorial Cancer Institute, 656 53, Brno, Czech Republic
| | - David P Lane
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Pavitra Kannan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
3
|
Özvegy-Laczka C, Ungvári O, Bakos É. Fluorescence-based methods for studying activity and drug-drug interactions of hepatic solute carrier and ATP binding cassette proteins involved in ADME-Tox. Biochem Pharmacol 2023; 209:115448. [PMID: 36758706 DOI: 10.1016/j.bcp.2023.115448] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023]
Abstract
In humans, approximately 70% of drugs are eliminated through the liver. This process is governed by the concerted action of membrane transporters and metabolic enzymes. Transporters mediating hepatocellular uptake of drugs belong to the SLC (Solute carrier) superfamily of transporters. Drug efflux either toward the portal vein or into the bile is mainly mediated by active transporters of the ABC (ATP Binding Cassette) family. Alteration in the function and/or expression of liver transporters due to mutations, disease conditions, or co-administration of drugs or food components can result in altered pharmacokinetics. On the other hand, drugs or food components interacting with liver transporters may also interfere with liver function (e.g., bile acid homeostasis) and may even cause liver toxicity. Accordingly, certain transporters of the liver should be investigated already at an early stage of drug development. Most frequently radioactive probes are applied in these drug-transporter interaction tests. However, fluorescent probes are cost-effective and sensitive alternatives to radioligands, and are gaining wider application in drug-transporter interaction tests. In our review, we summarize our current understanding about hepatocyte ABC and SLC transporters affected by drug interactions. We provide an update of the available fluorescent and fluorogenic/activable probes applicable in in vitro or in vivo testing of these ABC and SLC transporters, including near-infrared transporter probes especially suitable for in vivo imaging. Furthermore, our review gives a comprehensive overview of the available fluorescence-based methods, not directly relying on the transport of the probe, suitable for the investigation of hepatic ABC or SLC-type drug transporters.
Collapse
Affiliation(s)
- Csilla Özvegy-Laczka
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary.
| | - Orsolya Ungvári
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary; Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Éva Bakos
- Institute of Enzymology, RCNS, Eötvös Loránd Research Network, H-1117 Budapest, Magyar tudósok krt. 2., Hungary
| |
Collapse
|
4
|
Ahmad S, Wood KC, Scott JE. A High Throughput Proliferation and Cytotoxicity Assay for Co-Cultured Isogenic Cell Lines. MethodsX 2022; 9:101927. [DOI: 10.1016/j.mex.2022.101927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
|
5
|
Cárdenas SD, Reznik CJ, Ranaweera R, Song F, Chung CH, Fertig EJ, Gevertz JL. Model-informed experimental design recommendations for distinguishing intrinsic and acquired targeted therapeutic resistance in head and neck cancer. NPJ Syst Biol Appl 2022; 8:32. [PMID: 36075912 PMCID: PMC9458753 DOI: 10.1038/s41540-022-00244-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022] Open
Abstract
The promise of precision medicine has been limited by the pervasive resistance to many targeted therapies for cancer. Inferring the timing (i.e., pre-existing or acquired) and mechanism (i.e., drug-induced) of such resistance is crucial for designing effective new therapeutics. This paper studies cetuximab resistance in head and neck squamous cell carcinoma (HNSCC) using tumor volume data obtained from patient-derived tumor xenografts. We ask if resistance mechanisms can be determined from this data alone, and if not, what data would be needed to deduce the underlying mode(s) of resistance. To answer these questions, we propose a family of mathematical models, with each member of the family assuming a different timing and mechanism of resistance. We present a method for fitting these models to individual volumetric data, and utilize model selection and parameter sensitivity analyses to ask: which member(s) of the family of models best describes HNSCC response to cetuximab, and what does that tell us about the timing and mechanisms driving resistance? We find that along with time-course volumetric data to a single dose of cetuximab, the initial resistance fraction and, in some instances, dose escalation volumetric data are required to distinguish among the family of models and thereby infer the mechanisms of resistance. These findings can inform future experimental design so that we can best leverage the synergy of wet laboratory experimentation and mathematical modeling in the study of novel targeted cancer therapeutics.
Collapse
Affiliation(s)
- Santiago D Cárdenas
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA
| | - Constance J Reznik
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA
- Datacor, Inc., Florham Park, NJ, USA
| | - Ruchira Ranaweera
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Feifei Song
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Elana J Fertig
- Convergence Institute, Department of Oncology, Department of Biomedical Engineering, Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA.
| | - Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA.
| |
Collapse
|
6
|
Skvortsov DA, Kalinina MA, Zhirkina IV, Vasilyeva LA, Ivanenkov YA, Sergiev PV, Dontsova OA. From Toxicity to Selectivity: Coculture of the Fluorescent Tumor and Non-Tumor Lung Cells and High-Throughput Screening of Anticancer Compounds. Front Pharmacol 2021; 12:713103. [PMID: 34707495 PMCID: PMC8542663 DOI: 10.3389/fphar.2021.713103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
For the search of anticancer compounds in modern large chemical libraries, new approaches are of great importance. Cocultivation of the cells of tumor and non-tumor etiology may reveal specific action of chemicals on cancer cells and also take into account some effects of the tumor cell's microenvironment. The fluorescent cell cocultivation test (FCCT) has been developed for screening of substances that are selectively cytotoxic on cancerous cells. It is based on the mixed culture of lung carcinoma cells A549'_EGFP and noncancerous fibroblasts of lung VA13_Kat, expressing different fluorescent proteins. Analysis of the cells was performed with the high-resolution scanner to increase the detection rate. The combination of cocultivation of cells with scanning of fluorescence reduces the experimental protocol to three steps: cells seeding, addition of the substance, and signal detection. The FCCT analysis does not disturb the cells and is compatible with other cell-targeted assays. The suggested method has been adapted for a high-throughput format and applied for screening of 2,491 compounds. Three compounds were revealed to be reproducibly selective in the FCCT although they were invisible in cytotoxicity tests in individual lines. Six structurally diverse indole, coumarin, sulfonylthiazol, and rifampicin derivatives were found and confirmed with an independent assay (MTT) to be selectively cytotoxic to cancer cells in the studied model.
Collapse
Affiliation(s)
- D A Skvortsov
- Chemistry Department, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow, Russia
| | - M A Kalinina
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - I V Zhirkina
- Chemistry Department, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - L A Vasilyeva
- Chemistry Department, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Y A Ivanenkov
- Chemistry Department, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Institute of Biochemistry and Genetics Russian Academy of Science (IBG RAS), Ufa Scientific Centre, Ufa, Russia
| | - P V Sergiev
- Chemistry Department, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Skolkovo Institute of Science and Technology, Moscow, Russia
| | - O A Dontsova
- Chemistry Department, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia.,Skolkovo Institute of Science and Technology, Moscow, Russia.,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
7
|
Evolutionary dynamics of cancer multidrug resistance in response to olaparib and photodynamic therapy. Transl Oncol 2021; 14:101198. [PMID: 34418731 PMCID: PMC8387718 DOI: 10.1016/j.tranon.2021.101198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/15/2021] [Accepted: 08/08/2021] [Indexed: 12/20/2022] Open
Abstract
P-glycoprotein (P-gp) is an adenosine triphosphate (ATP)-dependent drug efflux protein commonly associated with multidrug resistance in cancer chemotherapy. In this report, we used a dual-fluorescent co-culture model to study the population dynamics of the drug sensitive human ovarian cancer cell line (OVCAR-8-DsRed2) and its resistant subline that overexpresses P-gp (NCI/ADR-RES-EGFP) during the course of a photodynamic therapy (PDT)-olaparib combination regimen. Without treatment, OVCAR-8-DsRed2 cells grew more rapidly than the NCI/ADR-RES-EGFP cells. Olaparib treatment reduced the total number of cancer cells by 70±4% but selected for the resistant NCI/ADR-RES-EGFP population since olaparib is an efflux substrate for the P-gp pump. This study used the FDA-approved benzoporphyrin derivative (BPD) photosensitizer or its lipidated formulation ((16:0)LysoPC-BPD) to kill OVCAR-8 cells and reduce the likelihood that olaparib-resistant cells would have selective advantage. Three cycles of PDT effectively reduced the total cell number by 66±3%, while stabilizing the population ratio of sensitive and resistant cells at approximately 1:1. The combination of olaparib treatment and PDT enhanced PARP cleavage and deoxyribonucleic acid (DNA) damage, further decreasing the total cancer cell number down to 10±2%. We also showed that the combination of olaparib and (16:0)LysoPC-BPD-based PDT is up to 18-fold more effective in mitigating the selection of resistant NCI/ADR-RES-EGFP cells, compared to using olaparib and BPD-based PDT. These studies suggest that PDT may improve the effectiveness of olaparib, and the use of a lipidated photosensitizer formulation holds promise in overcoming cancer drug resistance.
Collapse
|
8
|
Paczkowski M, Kretzschmar WW, Markelc B, Liu SK, Kunz-Schughart LA, Harris AL, Partridge M, Byrne HM, Kannan P. Reciprocal interactions between tumour cell populations enhance growth and reduce radiation sensitivity in prostate cancer. Commun Biol 2021; 4:6. [PMID: 33398023 PMCID: PMC7782740 DOI: 10.1038/s42003-020-01529-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/24/2020] [Indexed: 01/29/2023] Open
Abstract
Intratumoural heterogeneity (ITH) contributes to local recurrence following radiotherapy in prostate cancer. Recent studies also show that ecological interactions between heterogeneous tumour cell populations can lead to resistance in chemotherapy. Here, we evaluated whether interactions between heterogenous populations could impact growth and response to radiotherapy in prostate cancer. Using mixed 3D cultures of parental and radioresistant populations from two prostate cancer cell lines and a predator-prey mathematical model to investigate various types of ecological interactions, we show that reciprocal interactions between heterogeneous populations enhance overall growth and reduce radiation sensitivity. The type of interaction influences the time of regrowth after radiation, and, at the population level, alters the survival and cell cycle of each population without eliminating either one. These interactions can arise from oxygen constraints and from cellular cross-talk that alter the tumour microenvironment. These findings suggest that ecological-type interactions are important in radiation response and could be targeted to reduce local recurrence.
Collapse
Affiliation(s)
| | - Warren W Kretzschmar
- School of Engineering Sciences in Chemistry Biotechnology and Health, Department of Gene Technology, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Center for Hematology and Regenerative Medicine (HERM), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Center for Hematology and Regenerative Medicine (HERM), Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Bostjan Markelc
- CRUK and MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Stanley K Liu
- Sunnybrook Research Institute and Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Leoni A Kunz-Schughart
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum, Dresden, Rossendorf, Germany
- National Center for Tumor Diseases (NCT), Partner Site, Dresden, Germany
| | - Adrian L Harris
- CRUK and MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Mike Partridge
- CRUK and MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Helen M Byrne
- Mathematical Institute, University of Oxford, Oxford, UK.
| | - Pavitra Kannan
- CRUK and MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK.
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
9
|
Greene JM, Sanchez-Tapia C, Sontag ED. Mathematical Details on a Cancer Resistance Model. Front Bioeng Biotechnol 2020; 8:501. [PMID: 32656186 PMCID: PMC7325889 DOI: 10.3389/fbioe.2020.00501] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 04/29/2020] [Indexed: 01/02/2023] Open
Abstract
One of the most important factors limiting the success of chemotherapy in cancer treatment is the phenomenon of drug resistance. We have recently introduced a framework for quantifying the effects of induced and non-induced resistance to cancer chemotherapy (Greene et al., 2018a, 2019). In this work, we expound on the details relating to an optimal control problem outlined in Greene et al. (2018a). The control structure is precisely characterized as a concatenation of bang-bang and path-constrained arcs via the Pontryagin Maximum Principle and differential Lie algebraic techniques. A structural identifiability analysis is also presented, demonstrating that patient-specific parameters may be measured and thus utilized in the design of optimal therapies prior to the commencement of therapy. For completeness, a detailed analysis of existence results is also included.
Collapse
Affiliation(s)
- James M. Greene
- Department of Mathematics, Clarkson University, Potsdam, NY, United States
| | - Cynthia Sanchez-Tapia
- Department of Mathematics and Center for Quantitative Biology, Rutgers University, Piscataway, NJ, United States
| | - Eduardo D. Sontag
- Department of Electrical and Computer Engineering, Department of Bioengineering, Northeastern University, Boston, MA, United States
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Greene JM, Gevertz JL, Sontag ED. Mathematical Approach to Differentiate Spontaneous and Induced Evolution to Drug Resistance During Cancer Treatment. JCO Clin Cancer Inform 2020; 3:1-20. [PMID: 30969799 PMCID: PMC6873992 DOI: 10.1200/cci.18.00087] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Purpose Drug resistance is a major impediment to the success of cancer treatment. Resistance is typically thought to arise from random genetic mutations, after which mutated cells expand via Darwinian selection. However, recent experimental evidence suggests that progression to drug resistance need not occur randomly, but instead may be induced by the treatment itself via either genetic changes or epigenetic alterations. This relatively novel notion of resistance complicates the already challenging task of designing effective treatment protocols. Materials and Methods To better understand resistance, we have developed a mathematical modeling framework that incorporates both spontaneous and drug-induced resistance. Results Our model demonstrates that the ability of a drug to induce resistance can result in qualitatively different responses to the same drug dose and delivery schedule. We have also proven that the induction parameter in our model is theoretically identifiable and propose an in vitro protocol that could be used to determine a treatment’s propensity to induce resistance.
Collapse
Affiliation(s)
| | | | - Eduardo D Sontag
- Northeastern University, Boston, MA.,Harvard Medical School, Cambridge, MA
| |
Collapse
|
11
|
Lee TD, Lee OW, Brimacombe KR, Chen L, Guha R, Lusvarghi S, Tebase BG, Klumpp-Thomas C, Robey RW, Ambudkar SV, Shen M, Gottesman MM, Hall MD. A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein. Mol Pharmacol 2019; 96:629-640. [PMID: 31515284 PMCID: PMC6790066 DOI: 10.1124/mol.119.115964] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 09/06/2019] [Indexed: 12/21/2022] Open
Abstract
The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs. Although US Food and Drug Administration guidelines require that potential interactions of investigational drugs with P-gp be explored, often this information does not enter the literature. In response, we developed a high-throughput screen to identify substrates of P-gp from a series of chemical libraries, testing a total of 10,804 compounds, most of which have known mechanisms of action. We used the CellTiter-Glo viability assay to test library compounds against parental KB-3-1 human cervical adenocarcinoma cells and the colchicine-selected subline KB-8-5-11 that overexpresses P-gp. KB-8-5-11 cells were also tested in the presence of a P-gp inhibitor (tariquidar) to assess reversibility of transporter-mediated resistance. Of the tested compounds, a total of 90 P-gp substrates were identified, including 55 newly identified compounds. Substrates were confirmed using an orthogonal killing assay against human embryonic kidney-293 cells overexpressing P-gp. We confirmed that AT7159 (cyclin-dependent kinase inhibitor), AT9283, (Janus kinase 2/3 inhibitor), ispinesib (kinesin spindle protein inhibitor), gedatolisib (PKI-587, phosphoinositide 3-kinase/mammalian target of rampamycin inhibitor), GSK-690693 (AKT inhibitor), and KW-2478 (heat-shock protein 90 inhibitor) were substrates. In addition, we assessed direct ATPase stimulation. ABCG2 was also found to confer high levels of resistance to AT9283, GSK-690693, and gedatolisib, whereas ispinesib, AT7519, and KW-2478 were weaker substrates. Combinations of P-gp substrates and inhibitors were assessed to demonstrate on-target synergistic cell killing. These data identified compounds whose oral bioavailability or brain penetration may be affected by P-gp. SIGNIFICANCE STATEMENT: The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to be expressed at barrier sites, where it acts to limit oral bioavailability and brain penetration of substrates. In order to identify novel compounds that are transported by P-gp, we developed a high-throughput screen using the KB-3-1 cancer cell line and its colchicine-selected subline KB-8-5-11. We screened the Mechanism Interrogation Plate (MIPE) library, the National Center for Advancing Translational Science (NCATS) pharmaceutical collection (NPC), the NCATS Pharmacologically Active Chemical Toolbox (NPACT), and a kinase inhibitor library comprising 977 compounds, for a total of 10,804 compounds. Of the 10,804 compounds screened, a total of 90 substrates were identified of which 55 were novel. P-gp expression may adversely affect the oral bioavailability or brain penetration of these compounds.
Collapse
Affiliation(s)
- Tobie D Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Olivia W Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Kyle R Brimacombe
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Lu Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Rajarshi Guha
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Sabrina Lusvarghi
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Bethilehem G Tebase
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Carleen Klumpp-Thomas
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Robert W Robey
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Suresh V Ambudkar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Michael M Gottesman
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (T.D.L., O.W.L., K.R.B., L.C., R.G., C.K.-T., M.S., M.D.H.) and Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (S.L., B.G.T., R.W.R., S.V.A., M.M.G.)
| |
Collapse
|
12
|
Xin X, Yang ST. Development of a dual fluorescence system for simultaneous detection of two cell populations in a 3D coculture. Process Biochem 2019. [DOI: 10.1016/j.procbio.2019.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Xin X, Yang ST. A Dual Fluorescent 3-D Multicellular Coculture of Breast Cancer MCF-7 and Fibroblast NIH-3T3 Cells for High Throughput Cancer Drug Screening. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
14
|
Berguetti T, Quintaes LSP, Hancio T, Robaina MC, Cruz ALS, Maia RC, de Souza PS. TNF-α Modulates P-Glycoprotein Expression and Contributes to Cellular Proliferation via Extracellular Vesicles. Cells 2019; 8:cells8050500. [PMID: 31137684 PMCID: PMC6562596 DOI: 10.3390/cells8050500] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/15/2022] Open
Abstract
P-glycoprotein (Pgp/ABCB1) overexpression is associated with multidrug resistance (MDR) phenotype and, consequently, failure in cancer chemotherapy. However, molecules involved in cell death deregulation may also support MDR. Tumor necrosis factor-alpha (TNF-α) is an important cytokine that may trigger either death or tumor growth. Here, we examined the role of cancer cells in self-maintenance and promotion of cellular malignancy through the transport of Pgp and TNF-α molecules by extracellular vesicles (membrane microparticles (MP)). By using a classical MDR model in vitro, we identified a positive correlation between endogenous TNF-α and Pgp, which possibly favored a non-cytotoxic effect of recombinant TNF-α (rTNF-α). We also found a positive feedback involving rTNF-α incubation and TNF-α regulation. On the other hand, rTNF-α induced a reduction in Pgp expression levels and contributed to a reduced Pgp efflux function. Our results also showed that parental and MDR cells spontaneously released MP containing endogenous TNF-α and Pgp. However, these MP were unable to transfer their content to non-cancer recipient cells. Nevertheless, MP released from parental and MDR cells elevated the proliferation index of non-tumor cells. Collectively, our results suggest that Pgp and endogenous TNF-α positively regulate cancer cell malignancy and contribute to changes in normal cell behavior through MP.
Collapse
Affiliation(s)
- Tandressa Berguetti
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil.
- Programa de Pós-Graduação Strictu Sensu em Oncologia, INCA, Rio de Janeiro 20231-050, Brazil.
| | - Lucas S P Quintaes
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil.
| | - Thais Hancio
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil.
- Programa de Pós-Graduação Strictu Sensu em Oncologia, INCA, Rio de Janeiro 20231-050, Brazil.
| | - Marcela C Robaina
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil.
| | - André L S Cruz
- Laboratório de Fisiopatologia, Polo Novo Cavaleiros, Campus UFRJ-Macaé, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil.
| | - Raquel C Maia
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil.
| | - Paloma Silva de Souza
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro 20231-050, Brazil.
| |
Collapse
|
15
|
Wulftange WJ, Rose MA, Garmendia-Cedillos M, da Silva D, Poprawski JE, Srinivasachar D, Sullivan T, Lim L, Bliskovsky VV, Hall MD, Pohida TJ, Robey RW, Morgan NY, Gottesman MM. Spatial control of oxygen delivery to three-dimensional cultures alters cancer cell growth and gene expression. J Cell Physiol 2019; 234:20608-20622. [PMID: 31012116 DOI: 10.1002/jcp.28665] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/22/2019] [Indexed: 01/01/2023]
Abstract
Commonly used monolayer cancer cell cultures fail to provide a physiologically relevant environment in terms of oxygen delivery. Here, we describe a three-dimensional (3D) bioreactor system where cancer cells are grown in Matrigel in modified six-well plates. Oxygen is delivered to the cultures through a polydimethylsiloxane (PDMS) membrane at the bottom of the wells, with microfabricated PDMS pillars to control oxygen delivery. The plates receive 3% oxygen from below and 0% oxygen at the top surface of the media, providing a gradient of 3-0% oxygen. We compared growth and transcriptional profiles for cancer cells grown in Matrigel in the bioreactor, 3D cultures grown in 21% oxygen, and cells grown in a standard hypoxia chamber at 3% oxygen. Additionally, we compared gene expression of conventional two-dimensional monolayer culture and 3D Matrigel culture in 21% oxygen. We conclude that controlled oxygen delivery may provide a more physiologically relevant 3D system.
Collapse
Affiliation(s)
- William J Wulftange
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland.,Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michelle A Rose
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Marcial Garmendia-Cedillos
- Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, Maryland
| | - Davi da Silva
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Joanna E Poprawski
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Dhruv Srinivasachar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Taylor Sullivan
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Langston Lim
- Confocal Microscopy Core Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Valery V Bliskovsky
- CCR Genomics Core, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland
| | - Thomas J Pohida
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nicole Y Morgan
- Trans-NIH Shared Resources on Biomedical Engineering and Physical Sciences (BEPS), National Institutes of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, Maryland
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
16
|
Windt T, Tóth S, Patik I, Sessler J, Kucsma N, Szepesi Á, Zdrazil B, Özvegy-Laczka C, Szakács G. Identification of anticancer OATP2B1 substrates by an in vitro triple-fluorescence-based cytotoxicity screen. Arch Toxicol 2019; 93:953-964. [PMID: 30863990 PMCID: PMC6510822 DOI: 10.1007/s00204-019-02417-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/13/2018] [Indexed: 12/21/2022]
Abstract
Membrane transporters play an important role in the absorption, distribution, metabolism and excretion of drugs. The cellular accumulation of many drugs is the result of the net function of efflux and influx transporters. Efflux transporters such as P-glycoprotein/ABCB1 have been shown to confer multidrug resistance in cancer. Although expression of uptake transporters has been confirmed in cancer cells, their role in chemotherapy response has not been systematically investigated. In the present study we have adapted a fluorescence-based cytotoxic assay to characterize the influence of key drug-transporters on the toxicity of approved anticancer drugs. Co-cultures of fluorescently labeled parental and transporter-expressing cells (expressing ABCB1, ABCG2 or OATP2B1) were screened against 101 FDA-approved anticancer drugs, using a novel, automated, triple fluorescence-based cytotoxicity assay. By measuring the survival of parental and transporter-expressing cells in co-cultures, we identify those FDA-approved anticancer drugs, whose toxicity is influenced by ABCB1, ABCG2 or OATP2B1. In addition to confirming known substrates of ABCB1 and ABCG2, the fluorescence-based cytotoxicity assays identified anticancer agents whose toxicity was increased in OATP2B1 expressing cells. Interaction of these compounds with OATP2B1 was verified in dedicated transport assays using cell-impermeant fluorescent substrates. Understanding drug-transporter interactions is needed to increase the efficacy of chemotherapeutic agents. Our results highlight the potential of the fluorescence-based HT screening system for identifying transporter substrates, opening the way for the design of therapeutic approaches based on the inhibition or even the exploitation of transporters in cancer cells.
Collapse
Affiliation(s)
- Tímea Windt
- Institute of Enzymology, Research Centre for National Sciences, HAS, Budapest, 1117, Hungary
| | - Szilárd Tóth
- Institute of Enzymology, Research Centre for National Sciences, HAS, Budapest, 1117, Hungary.
| | - Izabel Patik
- Institute of Enzymology, Research Centre for National Sciences, HAS, Budapest, 1117, Hungary
| | - Judit Sessler
- Institute of Enzymology, Research Centre for National Sciences, HAS, Budapest, 1117, Hungary
| | - Nóra Kucsma
- Institute of Enzymology, Research Centre for National Sciences, HAS, Budapest, 1117, Hungary
| | - Áron Szepesi
- Institute of Enzymology, Research Centre for National Sciences, HAS, Budapest, 1117, Hungary
| | - Barbara Zdrazil
- Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria
| | - Csilla Özvegy-Laczka
- Institute of Enzymology, Research Centre for National Sciences, HAS, Budapest, 1117, Hungary
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for National Sciences, HAS, Budapest, 1117, Hungary.
- Institute of Cancer Research, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
17
|
|
18
|
Cytotoxicity Test Based on Human Cells Labeled with Fluorescent Proteins: Fluorimetry, Photography, and Scanning for High-Throughput Assay. Mol Imaging Biol 2017; 20:368-377. [DOI: 10.1007/s11307-017-1152-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
19
|
Konovalova EV, Shulga AA, Chumakov SP, Khodarovich YM, Woo EJ, Deev SM. Stably Fluorescent Cell Line of Human Ovarian Epithelial Cancer Cells SK-OV-3ip-red. Bull Exp Biol Med 2017; 164:99-101. [PMID: 29124539 DOI: 10.1007/s10517-017-3933-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Indexed: 01/16/2023]
Abstract
Stable red fluorescing line of human ovarian epithelial cancer cells SK-OV-3ip-red was generated expressing gene coding for protein TurboFP635 (Katushka) fluorescing in the far-red spectrum region with excitation and emission peaks at 588 and 635 nm, respectively. Fluorescence of SK-OV-3ip-red line remained high during long-term cell culturing and after cryogenic freezing. The obtained cell line SK-OV-3ip-red can serve a basis for a model of a scattered tumor with numerous/extended metastases and used both for testing anticancer drugs inhibiting metastasis growth and for non-invasive monitoring of the growth dynamics with high precision.
Collapse
Affiliation(s)
- E V Konovalova
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - A A Shulga
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - S P Chumakov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yu M Khodarovich
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Eui-Jeon Woo
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
| | - S M Deev
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
20
|
Becker M, Levy D. Modeling the Transfer of Drug Resistance in Solid Tumors. Bull Math Biol 2017; 79:2394-2412. [PMID: 28852953 DOI: 10.1007/s11538-017-0334-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/08/2017] [Indexed: 11/30/2022]
Abstract
ABC efflux transporters are a key factor leading to multidrug resistance in cancer. Overexpression of these transporters significantly decreases the efficacy of anti-cancer drugs. Along with selection and induction, drug resistance may be transferred between cells, which is the focus of this paper. Specifically, we consider the intercellular transfer of P-glycoprotein (P-gp), a well-known ABC transporter that was shown to confer resistance to many common chemotherapeutic drugs. In a recent paper, Durán et al. (Bull Math Biol 78(6):1218-1237, 2016) studied the dynamics of mixed cultures of resistant and sensitive NCI-H460 (human non-small lung cancer) cell lines. As expected, the experimental data showed a gradual increase in the percentage of resistance cells and a decrease in the percentage of sensitive cells. The experimental work was accompanied with a mathematical model that assumed P-gp transfer from resistant cells to sensitive cells, rendering them temporarily resistant. The mathematical model provided a reasonable fit to the experimental data. In this paper, we develop a new mathematical model for the transfer of drug resistance between cancer cells. Our model is based on incorporating a resistance phenotype into a model of cancer growth (Greene et al. in J Theor Biol 367:262-277, 2015). The resulting model for P-gp transfer, written as a system of integro-differential equations, follows the dynamics of proliferating, quiescent, and apoptotic cells, with a varying resistance phenotype. We show that this model provides a good match to the dynamics of the experimental data of Durán et al. (2016). The mathematical model shows a better fit when resistant cancer cells have a slower division rate than the sensitive cells.
Collapse
Affiliation(s)
- Matthew Becker
- Department of Mathematics, University of Maryland, College Park, MD, 20742, USA
| | - Doron Levy
- Department of Mathematics and Center for Scientific Computation and Mathematical Modeling (CSCAMM), University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
21
|
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]
|
22
|
Greene JM, Levy D, Herrada SP, Gottesman MM, Lavi O. Mathematical Modeling Reveals That Changes to Local Cell Density Dynamically Modulate Baseline Variations in Cell Growth and Drug Response. Cancer Res 2016; 76:2882-90. [PMID: 26933088 DOI: 10.1158/0008-5472.can-15-3232] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 02/17/2016] [Indexed: 11/16/2022]
Abstract
Cell-to-cell variations contribute to drug resistance with consequent therapy failure in cancer. Experimental techniques have been developed to monitor tumor heterogeneity, but estimates of cell-to-cell variation typically fail to account for the expected spatiotemporal variations during the cell growth process. To fully capture the extent of such dynamic variations, we developed a mechanistic mathematical model supported by in vitro experiments with an ovarian cancer cell line. We introduce the notion of dynamic baseline cell-to-cell variation, showing how the emerging spatiotemporal heterogeneity of one cell population can be attributed to differences in local cell density and cell cycle. Manipulation of the geometric arrangement and spatial density of cancer cells revealed that given a fixed global cell density, significant differences in growth, proliferation, and paclitaxel-induced apoptosis rates were observed based solely on cell movement and local conditions. We conclude that any statistical estimate of changes in the level of heterogeneity should be integrated with the dynamics and spatial effects of the baseline system. This approach incorporates experimental and theoretical methods to systematically analyze biologic phenomena and merits consideration as an underlying reference model for cell biology studies that investigate dynamic processes affecting cancer cell behavior. Cancer Res; 76(10); 2882-90. ©2016 AACR.
Collapse
Affiliation(s)
- James M Greene
- Department of Mathematics, Rutgers University, New Brunswick, New Jersey
| | - Doron Levy
- Department of Mathematics and Center for Scientific Computation and Mathematical Modeling, University of Maryland, College Park, Maryland
| | - Sylvia P Herrada
- Laboratory of Cell Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Orit Lavi
- Laboratory of Cell Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|
23
|
Bioluminescent imaging of ABCG2 efflux activity at the blood-placenta barrier. Sci Rep 2016; 6:20418. [PMID: 26853103 PMCID: PMC4745077 DOI: 10.1038/srep20418] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/04/2016] [Indexed: 11/09/2022] Open
Abstract
Physiologic barriers such as the blood placenta barrier (BPB) and the blood brain barrier protect the underlying parenchyma from pathogens and toxins. ATP-binding cassette (ABC) transporters are transmembrane proteins found at these barriers, and function to efflux xenobiotics and maintain chemical homeostasis. Despite the plethora of ex vivo and in vitro data showing the function and expression of ABC transporters, no imaging modality exists to study ABC transporter activity in vivo at the BPB. In the present study, we show that in vitro models of the placenta possess ABCG2 activity and can specifically transport D-luciferin, the endogenous substrate of firefly luciferase. To test ABCG2 transport activity at the BPB, we devised a breeding strategy to generate a bioluminescent pregnant mouse model to demonstrate transporter function in vivo. We found that coadministering the ABCG2 inhibitors Ko143 and gefitinib with D-luciferin increased bioluminescent signal from fetuses and placentae, whereas the control P-gp inhibitor DCPQ had no effect. We believe that our bioluminescent pregnant mouse model will facilitate greater understanding of the BPB and ABCG2 activity in health and disease.
Collapse
|
24
|
Weidner LD, Fung KL, Kannan P, Moen JK, Kumar JS, Mulder J, Innis RB, Gottesman MM, Hall MD. Tariquidar Is an Inhibitor and Not a Substrate of Human and Mouse P-glycoprotein. Drug Metab Dispos 2016; 44:275-82. [PMID: 26658428 PMCID: PMC4746486 DOI: 10.1124/dmd.115.067785] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/08/2015] [Indexed: 01/15/2023] Open
Abstract
Since its development, tariquidar (TQR; XR9576; N-[2-[[4-[2-(6,7-Dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)ethyl]phenyl]carbamoyl]-4,5-dimethoxyphenyl]quinoline-3-carboxamide) has been widely regarded as one of the more potent inhibitors of P-glycoprotein (P-gp), an efflux transporter of the ATP-binding cassette (ABC) transporter family. A third-generation inhibitor, TQR exhibits high affinity for P-gp, although it is also a substrate of another ABC transporter, breast cancer resistance protein (BCRP). Recently, several studies have questioned the mechanism by which TQR interfaces with P-gp, suggesting that TQR is a substrate for P-gp instead of a noncompetitive inhibitor. We investigated TQR and its interaction with human and mouse P-gp to determine if TQR is a substrate of P-gp in vitro. To address these questions, we used multiple in vitro transporter assays, including cytotoxicity, flow cytometry, accumulation, ATPase, and transwell assays. A newly generated BCRP cell line was used as a positive control that demonstrates TQR-mediated transport. Based on our results, we conclude that TQR is a potent inhibitor of both human and mouse P-gp and shows no signs of being a substrate at the concentrations tested. These in vitro data further support our position that the in vivo uptake of [(11)C]TQR into the brain can be explained by its high-affinity binding to P-gp and by it being a substrate of BCRP, followed by amplification of the brain signal by ionic trapping in acidic lysosomes.
Collapse
Affiliation(s)
- Lora D Weidner
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| | - King Leung Fung
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| | - Pavitra Kannan
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| | - Janna K Moen
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| | - Jeyan S Kumar
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| | - Jan Mulder
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| | - Michael M Gottesman
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| | - Matthew D Hall
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., P.K., R.B.I.); Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.L.F., J.K.M., J.S.K., M.M.G., M.D.H.); and Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.)
| |
Collapse
|
25
|
Weidner LD, Zoghbi SS, Lu S, Shukla S, Ambudkar SV, Pike VW, Mulder J, Gottesman MM, Innis RB, Hall MD. The Inhibitor Ko143 Is Not Specific for ABCG2. J Pharmacol Exp Ther 2015; 354:384-93. [PMID: 26148857 PMCID: PMC4538874 DOI: 10.1124/jpet.115.225482] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/26/2015] [Indexed: 11/22/2022] Open
Abstract
Imaging ATP-binding cassette (ABC) transporter activity in vivo with positron emission tomography requires both a substrate and a transporter inhibitor. However, for ABCG2, there is no inhibitor proven to be specific to that transporter alone at the blood-brain barrier. Ko143 [[(3S,6S,12aS)-1,2,3,4,6,7,12,12a-octahydro-9-methoxy-6-(2-methylpropyl)-1,4-dioxopyrazino[1',2':1,6]pyrido[3,4- b]indole-3-propanoic acid 1,1-dimethylethyl ester], a nontoxic analog of fungal toxin fumitremorgin C, is a potent inhibitor of ABCG2, although its specificity in mouse and human systems is unclear. This study examined the selectivity of Ko143 using human embryonic kidney cell lines transfected with ABCG2, ABCB1, or ABCC1 in several in vitro assays. The stability of Ko143 in rat plasma was measured using high performance liquid chromatography. Our results show that, in addition to being a potent inhibitor of ABCG2, at higher concentrations (≥1 μM) Ko143 also has an effect on the transport activity of both ABCB1 and ABCC1. Furthermore, Ko143 was found to be unstable in rat plasma. These findings indicate that Ko143 lacks specificity for ABCG2 and this should be taken into consideration when using Ko143 for both in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Lora D Weidner
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Shuiyu Lu
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Suneet Shukla
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Suresh V Ambudkar
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Jan Mulder
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Michael M Gottesman
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| | - Matthew D Hall
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland (L.D.W., S.S.Z., S.L., V.W.P., R.B.I.); Karolinska Institutet, Department of Neuroscience, Stockholm, Sweden (L.D.W., J.M.); and Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (S.S., S.V.A., M.M.G., M.D.H.)
| |
Collapse
|
26
|
Stukova M, Hall MD, Tsotsoros SD, Madigan JP, Farrell NP, Gottesman MM. Reduced accumulation of platinum drugs is not observed in drug-resistant ovarian cancer cell lines derived from cisplatin-treated patients. J Inorg Biochem 2015; 149:45-8. [PMID: 26021697 PMCID: PMC4467998 DOI: 10.1016/j.jinorgbio.2015.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 12/11/2022]
Abstract
The resistance of ovarian cancer towards front-line chemotherapy, usually cisplatin or carboplatin in combination with paclitaxel or docetaxel, remains a major clinical challenge. Resistance to these agents has been largely studied using cell lines selected for resistance to agents in vitro. We examined a series of paired cell lines derived from patients with ovarian cancer prior to chemotherapy (PEO1, PEO4, PEO14 and PEA1), and following the acquisition of resistance to a platinum-based chemotherapy regimen (PEO6, PEO23 and PEA2, respectively). All resistant patient lines showed resistance to cisplatin (2-5-fold), but this did not correspond with lowered accumulation. No general cross-resistance was observed for oxaliplatin, paclitaxel or docetaxel, and paclitaxel accumulation was not affected. PEO1 cells carrying BRCA2 mutations were hypersensitive to the PARP inhibitors olaparib and velaparib, but all other cell lines expressing functional forms of BRCA2 were less sensitive. While reduced drug accumulation was not observed, we believe these pairs of lines are of use to researchers studying Pt drug resistance and experimental therapeutics against drug-resistant ovarian cancer.
Collapse
Affiliation(s)
- Marina Stukova
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Matthew D Hall
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Samantha D Tsotsoros
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, United States
| | - James P Madigan
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nicholas P Farrell
- Department of Chemistry, Virginia Commonwealth University, Richmond, VA, United States
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
27
|
Souza PS, Madigan JP, Gillet JP, Kapoor K, Ambudkar SV, Maia RC, Gottesman MM, Fung KL. Expression of the multidrug transporter P-glycoprotein is inversely related to that of apoptosis-associated endogenous TRAIL. Exp Cell Res 2015; 336:318-28. [PMID: 26101157 DOI: 10.1016/j.yexcr.2015.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 12/27/2022]
Abstract
Multidrug resistance (MDR) has been associated with expression of ABC transporter genes including P-glycoprotein (Pgp, MDR1, ABCB1). However, deregulation of apoptotic pathways also renders cells resistant to chemotherapy. To discover apoptosis-related genes affected by Pgp expression, we used the HeLa MDR-off system. We found that using doxycycline to control Pgp expression has a significant advantage over tetracycline, in that doxycycline caused less endogenous gene expression modification/perturbation, and was more potent than tetracycline in suppressing Pgp expression. Cells overexpressing Pgp have lower TNFSF10 (TRAIL) expression than their parental cells. Controlled downregulation of Pgp increased endogenous TRAIL protein expression. Also, ectopic overexpression of TRAIL in Pgp-positive cells was associated with a reduction in Pgp levels. However, cells expressing a functionally defective mutant Pgp showed an increase in TRAIL expression, suggesting that Pgp function is required for TRAIL suppression. Cells in which Pgp is knocked down by upregulation of TRAIL expression are less susceptible to TRAIL ligand (sTRAIL)-induced apoptosis. Our findings reveal an inverse correlation between functional Pgp and endogenous TRAIL expression. Pgp function plays an important role in the TRAIL-mediated apoptosis pathway by regulating endogenous TRAIL expression and the TRAIL-mediated apoptosis pathway in MDR cancer cells.
Collapse
Affiliation(s)
- Paloma S Souza
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, USA; Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Brazil
| | - James P Madigan
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, USA
| | - Jean-Pierre Gillet
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, USA
| | - Khyati Kapoor
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, USA
| | - Raquel C Maia
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Pesquisa em Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Brazil
| | - Michael M Gottesman
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, USA.
| | - King Leung Fung
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, USA
| |
Collapse
|
28
|
de Souza PS, Cruz ALS, Viola JPB, Maia RC. Microparticles induce multifactorial resistance through oncogenic pathways independently of cancer cell type. Cancer Sci 2014; 106:60-8. [PMID: 25457412 PMCID: PMC4317771 DOI: 10.1111/cas.12566] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/24/2014] [Accepted: 10/27/2014] [Indexed: 12/15/2022] Open
Abstract
Multidrug resistance (MDR) is considered a multifactorial event that favors cancer cells becoming resistant to several chemotherapeutic agents. Numerous mechanisms contribute to MDR, such as P-glycoprotein (Pgp/ABCB1) activity that promotes drug efflux, overexpression of inhibitors of apoptosis proteins (IAP) that contribute to evasion of apoptosis, and oncogenic pathway activation that favors cancer cell survival. MDR molecules have been identified in membrane microparticles (MP) and can be transferred to sensitive cancer cells. By co-culturing MP derived from MDR-positive cells with recipient cells, we showed that sensitive cells accumulated Pgp, IAP proteins and mRNA. In addition, MP promoted microRNA transfer and NFκB and Yb-1 activation. Therefore, our results indicate that MP can induce a multifactorial phenotype in sensitive cancer cells.
Collapse
Affiliation(s)
- Paloma Silva de Souza
- Program of Hemato-Oncology Molecular, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
29
|
Modeling intrinsic heterogeneity and growth of cancer cells. J Theor Biol 2014; 367:262-277. [PMID: 25457229 DOI: 10.1016/j.jtbi.2014.11.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/20/2014] [Accepted: 11/14/2014] [Indexed: 02/02/2023]
Abstract
Intratumoral heterogeneity has been found to be a major cause of drug resistance. Cell-to-cell variation increases as a result of cancer-related alterations, which are acquired by stochastic events and further induced by environmental signals. However, most cellular mechanisms include natural fluctuations that are closely regulated, and thus lead to asynchronization of the cells, which causes intrinsic heterogeneity in a given population. Here, we derive two novel mathematical models, a stochastic agent-based model and an integro-differential equation model, each of which describes the growth of cancer cells as a dynamic transition between proliferative and quiescent states. These models are designed to predict variations in growth as a function of the intrinsic heterogeneity emerging from the durations of the cell-cycle and apoptosis, and also include cellular density dependencies. By examining the role all parameters play in the evolution of intrinsic tumor heterogeneity, and the sensitivity of the population growth to parameter values, we show that the cell-cycle length has the most significant effect on the growth dynamics. In addition, we demonstrate that the agent-based model can be approximated well by the more computationally efficient integro-differential equations when the number of cells is large. This essential step in cancer growth modeling will allow us to revisit the mechanisms of multidrug resistance by examining spatiotemporal differences of cell growth while administering a drug among the different sub-populations in a single tumor, as well as the evolution of those mechanisms as a function of the resistance level.
Collapse
|
30
|
Behbehani GK, Thom C, Zunder ER, Finck R, Gaudilliere B, Fragiadakis GK, Fantl WJ, Nolan GP. Transient partial permeabilization with saponin enables cellular barcoding prior to surface marker staining. Cytometry A 2014; 85:1011-9. [PMID: 25274027 DOI: 10.1002/cyto.a.22573] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/19/2014] [Accepted: 09/15/2014] [Indexed: 12/29/2022]
Abstract
Fluorescent cellular barcoding and mass-tag cellular barcoding are cytometric methods that enable high sample throughput, minimize inter-sample variation, and reduce reagent consumption. Previously employed barcoding protocols require that barcoding be performed after surface marker staining, complicating combining the technique with measurement of alcohol-sensitive surface epitopes. This report describes a method of barcoding fixed cells after a transient partial permeabilization with 0.02% saponin that results in efficient and consistent barcode staining with fluorescent or mass-tagged reagents while preserving surface marker staining. This approach simplifies barcoding protocols and allows direct comparison of surface marker staining of multiple samples without concern for variations in the antibody cocktail volume, antigen-antibody ratio, or machine sensitivity. Using this protocol, cellular barcoding can be used to reliably detect subtle differences in surface marker expression.
Collapse
Affiliation(s)
- Gregory K Behbehani
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California; Divisions of Hematology and Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Gacias M, Gerona-Navarro G, Plotnikov AN, Zhang G, Zeng L, Kaur J, Moy G, Rusinova E, Rodriguez Y, Matikainen B, Vincek A, Joshua J, Casaccia P, Zhou MM. Selective chemical modulation of gene transcription favors oligodendrocyte lineage progression. CHEMISTRY & BIOLOGY 2014; 21:841-854. [PMID: 24954007 PMCID: PMC4104156 DOI: 10.1016/j.chembiol.2014.05.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 04/28/2014] [Accepted: 05/01/2014] [Indexed: 01/07/2023]
Abstract
Lysine acetylation regulates gene expression through modulating protein-protein interactions in chromatin. Chemical inhibition of acetyl-lysine binding bromodomains of the major chromatin regulators BET (bromodomain and extraterminal domain) proteins has been shown to effectively block cell proliferation in cancer and inflammation. However, whether selective inhibition of individual BET bromodomains has distinctive functional consequences remains only partially understood. In this study, we show that selective chemical inhibition of the first bromodomain of BET proteins using our small-molecule inhibitor, Olinone, accelerated the progression of mouse primary oligodendrocyte progenitors toward differentiation, whereas inhibition of both bromodomains of BET proteins hindered differentiation. This effect was target specific, as it was not detected in cells treated with inactive analogs and independent of any effect on proliferation. Therefore, selective chemical modulation of individual bromodomains, rather than use of broad-based inhibitors, may enhance regenerative strategies in disorders characterized by myelin loss such as aging and neurodegeneration.
Collapse
Affiliation(s)
- Mar Gacias
- Departments of Neuroscience, and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Guillermo Gerona-Navarro
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
- Department of Chemistry, Brooklyn College, 2900 Bedford Avenue, Room 351 NE, Brooklyn, NY 11210, USA
| | - Alexander N. Plotnikov
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Guangtao Zhang
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Lei Zeng
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Jasbir Kaur
- Departments of Neuroscience, and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Gregory Moy
- Departments of Neuroscience, and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Elena Rusinova
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Yoel Rodriguez
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
- Department of Natural Sciences, Hostos Community College of CUNY, Bronx, NY 10451, USA
| | - Bridget Matikainen
- Departments of Neuroscience, and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Adam Vincek
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Jennifer Joshua
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Patrizia Casaccia
- Departments of Neuroscience, and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Ming-Ming Zhou
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| |
Collapse
|
32
|
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
|
33
|
The impact of cell density and mutations in a model of multidrug resistance in solid tumors. Bull Math Biol 2014; 76:627-53. [PMID: 24553772 DOI: 10.1007/s11538-014-9936-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 01/29/2014] [Indexed: 10/25/2022]
Abstract
In this paper we develop a mathematical framework for describing multidrug resistance in cancer. To reflect the complexity of the underlying interplay between cancer cells and the therapeutic agent, we assume that the resistance level is a continuous parameter. Our model is written as a system of integro-differential equations that are parameterized by the resistance level. This model incorporates the cell density and mutation dependence. Analysis and simulations of the model demonstrate how the dynamics evolves to a selection of one or more traits corresponding to different levels of resistance. The emerging limit distribution with nonzero variance is the desirable modeling outcome as it represents tumor heterogeneity.
Collapse
|
34
|
Fung KL, Pan J, Ohnuma S, Lund PE, Pixley JN, Kimchi-Sarfaty C, Ambudkar SV, Gottesman MM. MDR1 synonymous polymorphisms alter transporter specificity and protein stability in a stable epithelial monolayer. Cancer Res 2013; 74:598-608. [PMID: 24305879 DOI: 10.1158/0008-5472.can-13-2064] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The drug efflux function of P-glycoprotein (P-gp) encoded by MDR1 can be influenced by genetic polymorphisms, including two synonymous changes in the coding region of MDR1. Here we report that the conformation of P-gp and its drug efflux activity can be altered by synonymous polymorphisms in stable epithelial monolayers expressing P-gp. Several cell lines with similar MDR1 DNA copy number were developed and termed LLC-MDR1-WT (expresses wild-type P-gp), LLC-MDR1-3H (expresses common haplotype P-gp), and LLC-MDR1-3HA (a mutant that carries a different valine codon in position 3435). These cell lines express similar levels of recombinant mRNA and protein. P-gp in each case is localized on the apical surface of polarized cells. However, the haplotype and its mutant P-gps fold differently from the wild-type, as determined by UIC2 antibody shift assays and limited proteolysis assays. Surface biotinylation experiments suggest that the non-wild-type P-gps have longer recycling times. Drug transport assays show that wild-type and haplotype P-gp respond differently to P-gp inhibitors that block efflux of rhodamine 123 or mitoxantrone. In addition, cytotoxicity assays show that the LLC-MDR1-3H cells are more resistant to mitoxantrone than the LLC-MDR1-WT cells after being treated with a P-gp inhibitor. Expression of polymorphic P-gp, however, does not affect the host cell's morphology, growth rate, or monolayer formation. Also, ATPase activity assays indicate that neither basal nor drug-stimulated ATPase activities are affected in the variant P-gps. Taken together, our findings indicate that "silent" polymorphisms significantly change P-gp function, which would be expected to affect interindividual drug disposition and response.
Collapse
Affiliation(s)
- King Leung Fung
- Authors' Affiliations: Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH; and Center for Biologics Evaluation and Research, Division of Hematology, Food and Drug Administration, Bethesda, Maryland
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Jaeger AA, Das CK, Morgan NY, Pursley RH, McQueen PG, Hall MD, Pohida TJ, Gottesman MM. Microfabricated polymeric vessel mimetics for 3-D cancer cell culture. Biomaterials 2013; 34:8301-13. [PMID: 23911071 PMCID: PMC3759366 DOI: 10.1016/j.biomaterials.2013.07.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 07/03/2013] [Indexed: 12/13/2022]
Abstract
Modeling tumor growth in vitro is essential for cost-effective testing of hypotheses in preclinical cancer research. 3-D cell culture offers an improvement over monolayer culture for studying cellular processes in cancer biology because of the preservation of cell-cell and cell-ECM interactions. Oxygen transport poses a major barrier to mimicking in vivo environments and is not replicated in conventional cell culture systems. We hypothesized that we can better mimic the tumor microenvironment using a bioreactor system for controlling gas exchange in cancer cell cultures with silicone hydrogel synthetic vessels. Soft-lithography techniques were used to fabricate oxygen-permeable silicone hydrogel membranes containing arrays of micropillars. These membranes were inserted into a bioreactor and surrounded by basement membrane extract (BME) within which fluorescent ovarian cancer (OVCAR8) cells were cultured. Cell clusters oxygenated by synthetic vessels showed a ∼100μm drop-off to anoxia, consistent with in vivo studies of tumor nodules fed by the microvasculature. Oxygen transport in the bioreactor system was characterized by experimental testing with a dissolved oxygen probe and finite element modeling of convective flow. Our study demonstrates differing growth patterns associated with controlling gas distributions to better mimic in vivo conditions.
Collapse
Affiliation(s)
- Ashley A. Jaeger
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Chandan K. Das
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nicole Y. Morgan
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Randall H. Pursley
- Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Philip G. McQueen
- Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Matthew D. Hall
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas J. Pohida
- Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, MD, USA
| | - Michael M. Gottesman
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
36
|
Bakhsheshian J, Hall MD, Robey RW, Herrmann MA, Chen JQ, Bates SE, Gottesman MM. Overlapping substrate and inhibitor specificity of human and murine ABCG2. Drug Metab Dispos 2013; 41:1805-12. [PMID: 23868912 PMCID: PMC3781367 DOI: 10.1124/dmd.113.053140] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 07/18/2013] [Indexed: 01/16/2023] Open
Abstract
ABCG2 (also known as breast cancer resistance protein) is an ATP-binding cassette (ABC) transporter localized to the plasma membrane where it mediates the efflux of xenobiotics, including potential therapeutics. Studies investigating Abcg2 function at the blood-brain barrier in mouse models are often compared with human ABCG2 function. It is critical to understand the nature of species differences between mouse and human ABCG2, since extrapolations are made from murine data to humans. Two independent drug-selected cell line pairs expressing human or mouse ABCG2 were compared for efflux of fluorescent substrates using flow cytometry. To this end, we developed and characterized a new mouse Abcg2-expressing subline that demonstrated efflux of known fluorescent ABCG2 substrates and increased resistance to mitoxantrone, which is reduced in the presence of the ABCG2 inhibitor Ko143. Our results indicate that the substrate specificity of human and mouse ABCG2 is very similar. We identified a new human and mouse ABCG2 substrate, a porphyrin analog, purpurin-18 (Pp-18), which is not a substrate for P-glycoprotein or multidrug resistance protein 1. The ability of inhibitors to block efflux activity of ABCG2 was assessed using Pp-18. Inhibitors also demonstrated similar effects on human and mouse ABCG2. Chrysin, benzoflavone, and cyclosporin A inhibited Pp-18 efflux in both human and mouse ABCG2. The similarity of the substrate and inhibitor specificity of human and mouse ABCG2 supports interpretation of mouse models in understanding the clinical, pharmacological, and physiologic roles of ABCG2.
Collapse
Affiliation(s)
- Joshua Bakhsheshian
- Laboratory of Cell Biology (J.B., M.D.H., M.M.G.), Cancer Therapeutics Branch (R.W.R., S.E.B.), Collaborative Protein Technology Resource (M.A.H., J.-Q.C.), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | | | | | | | | |
Collapse
|
37
|
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
|
38
|
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
|
39
|
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] [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
|
40
|
Goldsborough AS, Handley MD, Dulcey AE, Pluchino KM, Kannan P, Brimacombe KR, Hall MD, Griffiths G, Gottesman MM. Collateral sensitivity of multidrug-resistant cells to the orphan drug tiopronin. J Med Chem 2011; 54:4987-97. [PMID: 21657271 PMCID: PMC3208667 DOI: 10.1021/jm2001663] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A major challenge in the treatment of cancer is multidrug resistance (MDR) that develops during chemotherapy. Here we demonstrate that tiopronin (1), a thiol-substituted N-propanoylglycine derivative, was selectively toxic to a series of cell lines expressing the drug efflux pump P-glycoprotein (P-gp, ABCB1) and MRP1 (ABCC1). Treatment of MDR cells with 1 led to instability of the ABCB1 mRNA and consequently a reduction in P-gp protein, despite functional assays demonstrating that tiopronin does not interact with P-gp. Long-term exposure of P-gp-expressing cells to 1 sensitized them to doxorubicin and paclitaxel, both P-gp substrates. Treatment of MRP1-overexpressing cells with tiopronin led to a significant reduction in MRP1 protein. Synthesis and screening of analogues of tiopronin demonstrated that the thiol functional group was essential for collateral sensitivity while substitution of the amino acid backbone altered but did not destroy specificity, pointing to future development of targeted analogues.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drug Screening Assays, Antitumor
- HEK293 Cells
- Humans
- Multidrug Resistance-Associated Proteins/metabolism
- Orphan Drug Production
- Paclitaxel/pharmacology
- RNA Stability
- RNA, Messenger/metabolism
- Structure-Activity Relationship
- Tiopronin/chemical synthesis
- Tiopronin/chemistry
- Tiopronin/pharmacology
Collapse
Affiliation(s)
- Andrew S Goldsborough
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Brózik A, Hegedüs C, Erdei Z, Hegedus T, Özvegy-Laczka C, Szakács G, Sarkadi B. Tyrosine kinase inhibitors as modulators of ATP binding cassette multidrug transporters: substrates, chemosensitizers or inducers of acquired multidrug resistance? Expert Opin Drug Metab Toxicol 2011; 7:623-42. [PMID: 21410427 DOI: 10.1517/17425255.2011.562892] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Anticancer tyrosine kinase inhibitors (TKIs) are small molecule hydrophobic compounds designed to arrest aberrant signaling pathways in malignant cells. Multidrug resistance (MDR) ATP binding cassette (ABC) transporters have recently been recognized as important determinants of the general ADME-Tox (absorption, distribution, metabolism, excretion, toxicity) properties of small molecule TKIs, as well as key factors of resistance against targeted anticancer therapeutics. AREAS COVERED The article summarizes MDR-related ABC transporter interactions with imatinib, nilotinib, dasatinib, gefitinib, erlotinib, lapatinib, sunitinib and sorafenib, including in vitro and in vivo observations. An array of methods developed to study such interactions is presented. Transporter-TKI interactions relevant to the ADME-Tox properties of TKI drugs, primary or acquired cancer TKI resistance, and drug-drug interactions are also reviewed. EXPERT OPINION Based on the concept presented in this review, TKI anticancer drugs are considered as compounds recognized by the cellular mechanisms handling xenobiotics. Accordingly, novel anticancer therapies should equally focus on the effectiveness of target inhibition and exploration of potential interactions of the designed molecules by membrane transporters. Thus, targeted hydrophobic small molecule compounds should also be screened to evade xenobiotic-sensing cellular mechanisms.
Collapse
Affiliation(s)
- Anna Brózik
- Hungarian Academy of Sciences and Semmelweis University, Membrane Biology, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
42
|
Kannan P, Telu S, Shukla S, Ambudkar SV, Pike VW, Halldin C, Gottesman MM, Innis RB, Hall MD. The "specific" P-glycoprotein inhibitor Tariquidar is also a substrate and an inhibitor for breast cancer resistance protein (BCRP/ABCG2). ACS Chem Neurosci 2011; 2:82-9. [PMID: 22778859 DOI: 10.1021/cn100078a] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 10/14/2010] [Indexed: 12/16/2022] Open
Abstract
Tariquidar was developed as a specific inhibitor of the efflux transporter ABCB1. Recent positron emission tomographic brain imaging studies using [(11)C]tariquidar to measure ABCB1 (P-gp, P-glycoprotein) density in mice indicate that the inhibitor may not be as specific as previously thought. We examined its selectivity as an inhibitor and a substrate for the human transporters P-gp, breast cancer resistance protein (BCRP, ABCG2), and multidrug resistance protein 1 (MRP1, ABCC1). Our results show that at low concentrations, tariquidar acts selectively as an inhibitor of P-gp and also as a substrate of BCRP. At much higher concentrations (≥100 nM), tariquidar acts as an inhibitor of both P-gp and BCRP. Thus, the in vivo specificity of tariquidar depends on concentration and the relative density and capacity of P-gp vs BCRP.
Collapse
Affiliation(s)
- Pavitra Kannan
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States
| | - Suneet Shukla
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, Maryland, United States
| | - Suresh V. Ambudkar
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, Maryland, United States
| | - Victor W. Pike
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States
| | - Christer Halldin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Michael M. Gottesman
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, Maryland, United States
| | - Robert B. Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, United States
| | - Matthew D. Hall
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, Maryland, United States
| |
Collapse
|
43
|
Shukla SJ, Huang R, Austin CP, Xia M. The future of toxicity testing: a focus on in vitro methods using a quantitative high-throughput screening platform. Drug Discov Today 2010; 15:997-1007. [PMID: 20708096 DOI: 10.1016/j.drudis.2010.07.007] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 06/08/2010] [Accepted: 07/30/2010] [Indexed: 01/16/2023]
Abstract
The US Tox21 collaborative program represents a paradigm shift in toxicity testing of chemical compounds from traditional in vivo tests to less expensive and higher throughput in vitro methods to prioritize compounds for further study, identify mechanisms of action and ultimately develop predictive models for adverse health effects in humans. The NIH Chemical Genomics Center (NCGC) is an integral component of the Tox21 collaboration owing to its quantitative high-throughput screening (qHTS) paradigm, in which titration-based screening is used to profile hundreds of thousands of compounds per week. Here, we describe the Tox21 collaboration, qHTS-based compound testing and the various Tox21 screening assays that have been validated and tested at the NCGC to date.
Collapse
Affiliation(s)
- Sunita J Shukla
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-3370, USA
| | | | | | | |
Collapse
|
44
|
Liu YY, Gupta V, Patwardhan GA, Bhinge K, Zhao Y, Bao J, Mehendale H, Cabot MC, Li YT, Jazwinski SM. Glucosylceramide synthase upregulates MDR1 expression in the regulation of cancer drug resistance through cSrc and beta-catenin signaling. Mol Cancer 2010; 9:145. [PMID: 20540746 PMCID: PMC2903501 DOI: 10.1186/1476-4598-9-145] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 06/11/2010] [Indexed: 12/04/2022] Open
Abstract
Background Drug resistance is the outcome of multiple-gene interactions in cancer cells under stress of anticancer agents. MDR1 overexpression is most commonly detected in drug-resistant cancers and accompanied with other gene alterations including enhanced glucosylceramide synthase (GCS). MDR1 encodes for P-glycoprotein that extrudes anticancer drugs. Polymorphisms of MDR1 disrupt the effects of P-glycoprotein antagonists and limit the success of drug resistance reversal in clinical trials. GCS converts ceramide to glucosylceramide, reducing the impact of ceramide-induced apoptosis and increasing glycosphingolipid (GSL) synthesis. Understanding the molecular mechanisms underlying MDR1 overexpression and how it interacts with GCS may find effective approaches to reverse drug resistance. Results MDR1 and GCS were coincidently overexpressed in drug-resistant breast, ovary, cervical and colon cancer cells; silencing GCS using a novel mixed-backbone oligonucleotide (MBO-asGCS) sensitized these four drug-resistant cell lines to doxorubicin. This sensitization was correlated with the decreased MDR1 expression and the increased doxorubicin accumulation. Doxorubicin treatment induced GCS and MDR1 expression in tumors, but MBO-asGCS treatment eliminated "in-vivo" growth of drug-resistant tumor (NCI/ADR-RES). MBO-asGCS suppressed the expression of MDR1 with GCS and sensitized NCI/ADR-RES tumor to doxorubicin. The expression of P-glycoprotein and the function of its drug efflux of tumors were decreased by 4 and 8 times after MBO-asGCS treatment, even though this treatment did not have a significant effect on P-glycoprotein in normal small intestine. GCS transient transfection induced MDR1 overexpression and increased P-glycoprotein efflux in dose-dependent fashion in OVCAR-8 cancer cells. GSL profiling, silencing of globotriaosylceramide synthase and assessment of signaling pathway indicated that GCS transfection significantly increased globo series GSLs (globotriaosylceramide Gb3, globotetraosylceramide Gb4) on GSL-enriched microdomain (GEM), activated cSrc kinase, decreased β-catenin phosphorylation, and increased nuclear β-catenin. These consequently increased MDR1 promoter activation and its expression. Conversely, MBO-asGCS treatments decreased globo series GSLs (Gb3, Gb4), cSrc kinase and nuclear β-catenin, and suppressed MDR-1 expression in dose-dependent pattern. Conclusion This study demonstrates, for the first time, that GCS upregulates MDR1 expression modulating drug resistance of cancer. GSLs, in particular globo series GSLs mediate gene expression of MDR1 through cSrc and β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yong-Yu Liu
- Department of Basic Pharmaceutical Sciences, University of Louisiana at Monroe, Monroe, Louisiana 71209, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kannan P, Brimacombe KR, Zoghbi SS, Liow JS, Morse C, Taku AK, Pike VW, Halldin C, Innis RB, Gottesman MM, Hall MD. N-desmethyl-loperamide is selective for P-glycoprotein among three ATP-binding cassette transporters at the blood-brain barrier. Drug Metab Dispos 2010; 38:917-22. [PMID: 20212014 PMCID: PMC2879961 DOI: 10.1124/dmd.109.031161] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 03/08/2010] [Indexed: 11/22/2022] Open
Abstract
[(11)C]N-desmethyl-Loperamide ([(11)C]dLop) is used in positron emission tomography (PET) to measure the in vivo activity of efflux transporters that block the passage of drugs across the blood-brain barrier. The three most prevalent ATP-binding cassette efflux transporters at the blood-brain barrier are P-glycoprotein (P-gp), multidrug resistance protein 1 (Mrp1), and breast cancer resistance protein (BCRP). We sought to measure the selectivity of dLop among these three transporters. The selectivity of dLop at low concentrations (< or =1 nM) was measured both as the accumulation of [(3)H]dLop in human cells that overexpress each transporter and as the uptake of [(11)C]dLop in brains of mice that lack genes encoding P-gp, Mrp1, or BCRP. The selectivity of dLop at high concentrations (> or =20 microM) was measured as the inhibition of uptake of a fluorescent substrate and the change in cytotoxicity of drugs effluxed at each transporter. Accumulation of [(3)H]dLop was lowest in cells overexpressing P-gp, and the uptake of [(11)C]dLop was highest in brains of mice lacking P-gp. At high concentrations, dLop selectively inhibited P-gp function and also decreased the resistance of only the P-gp-expressing cells to cytotoxic agents. dLop is selective for P-gp among these three transporters, but its activity is dependent on concentration. At low concentrations (< or =1 nM), dLop acts only as a substrate; at high concentrations (> or =20 microM), it acts as both a substrate and an inhibitor (i.e., a competitive substrate). Because low concentrations of radiotracer are used for PET imaging, [(11)C]dLop acts selectively and only as a substrate for P-gp.
Collapse
Affiliation(s)
- Pavitra Kannan
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|