1
|
Yin F, Zhao R, Gorja DR, Fu X, Lu N, Huang H, Xu B, Chen H, Shim JH, Liu K, Li Z, Laster KV, Dong Z, Lee MH. Novel dual inhibitor for targeting PIM1 and FGFR1 kinases inhibits colorectal cancer growth in vitro and patient-derived xenografts in vivo. Acta Pharm Sin B 2022; 12:4122-4137. [PMID: 36386480 PMCID: PMC9643289 DOI: 10.1016/j.apsb.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/15/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022] Open
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer-related death in the world. The pro-viral integration site for Moloney murine leukemia virus 1 (PIM1) is a proto-oncogene and belongs to the serine/threonine kinase family, which are involved in cell proliferation, migration, and apoptosis. Fibroblast growth factor receptor 1 (FGFR1) is a tyrosine kinase that has been implicated in cell proliferation, differentiation and migration. Small molecule HCI-48 is a derivative of chalcone, a class of compounds known to possess anti-tumor, anti-inflammatory and antibacterial effects. However, the underlying mechanism of chalcones against colorectal cancer remains unclear. This study reports that HCI-48 mainly targets PIM1 and FGFR1 kinases, thereby eliciting antitumor effects on colorectal cancer growth in vitro and in vivo. HCI-48 inhibited the activity of both PIM1 and FGFR1 kinases in an ATP-dependent manner, as revealed by computational docking models. Cell-based assays showed that HCI-48 inhibited cell proliferation in CRC cells (HCT-15, DLD1, HCT-116 and SW620), and induced cell cycle arrest in the G2/M phase through modulation of cyclin A2. HCI-48 also induced cellular apoptosis, as evidenced by an increase in the expression of apoptosis biomarkers such as cleaved PARP, cleaved caspase 3 and cleaved caspase 7. Moreover, HCI-48 attenuated the activation of downstream components of the PIM1 and FGFR1 signaling pathways. Using patient-derived xenograft (PDX) murine tumor models, we found that treatment with HCI-48 diminished the PDX tumor growth of implanted CRC tissue expressing high protein levels of PIM1 and FGFR1. This study suggests that the inhibitory effect of HCI-48 on colorectal tumor growth is mainly mediated through the dual-targeting of PIM1 and FGFR1 kinases. This work provides a theoretical basis for the future application of HCI-48 in the treatment of clinical CRC.
Collapse
|
2
|
Kapse P, Chikhale RV, Khan MR, Wabaidur SM, Islam MA. Synthesis of novel cycloheptylbenzothiazole-2-carboxamides and biological evaluation as human estrogen receptor modulators. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
3
|
Novel 1-methoxyindole- and 2-alkoxyindole-based chalcones: design, synthesis, characterization, antiproliferative activity and DNA, BSA binding interactions. Med Chem Res 2021. [DOI: 10.1007/s00044-020-02690-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
4
|
The importance of indole and azaindole scaffold in the development of antitumor agents. Eur J Med Chem 2020; 203:112506. [PMID: 32688198 DOI: 10.1016/j.ejmech.2020.112506] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/14/2020] [Accepted: 05/25/2020] [Indexed: 12/24/2022]
Abstract
With some indoles and azaindoles being successfully developed as anticancer drugs, the design and synthesis of indole and azaindole derivatives with remarkable antitumor activity has received increasing attention and significant progress has been made. This paper reviews the recent progress in the study of tumorigenesis, mechanism of actions and structure activity relationships about anticancer indole and azindole derivatives. Combining structure activity relationships and molecular targets-related knowledge, this review will help researchers design more effective, safe and cost-effective anticancer indoles and azindoles agents.
Collapse
|
5
|
Badria FA, Atef S, Al‐Majid AM, Ali M, Elshaier YAMM, Ghabbour HA, Islam MS, Barakat A. Synthesis and Inhibitory Effect of Some Indole-Pyrimidine Based Hybrid Heterocycles on α-Glucosidase and α-Amylase as Potential Hypoglycemic Agents. ChemistryOpen 2019; 8:1288-1297. [PMID: 31649838 PMCID: PMC6804419 DOI: 10.1002/open.201900240] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/18/2019] [Indexed: 11/09/2022] Open
Abstract
The Michael addition reaction of barbituric acid with chalcones incorporating the indole scaffold was achieved by using a highly efficient bimetallic Iron-palladium catalyst in the presence of acetylacetone (acac). This catalytic approach produced the desired products in a simple operation and low catalyst loading with acceptable yield of the new hybrids. All tested compounds were subjected for biological activity on α-glucosidase and α-amylase. The results revealed that all synthesized compounds exhibited very good activity against both enzymes when compared to positive control (acarbose). Moreover, compound 5o showed the best activity whereas its IC50 (μM) are 13.02+0.01 and 21.71+0.82 for α-glucosidase and α-amylase respectively. Both compounds 5o and 5l exhibited high similarity in binding mode and pose with amylase protein (4UAC). The obtained data may be used for developing potential hypoglycemic agents.
Collapse
Affiliation(s)
- Farid A. Badria
- Department of Pharmacognosy, Faculty of pharmacyMansura UniversityMansura35516Egypt
| | - Saleh Atef
- Department of Chemistry, College of ScienceKing Saud UniversityP. O. Box 2455Riyadh11451Saudi Arabia
| | | | - M. Ali
- Department of Chemistry, College of ScienceKing Saud UniversityP. O. Box 2455Riyadh11451Saudi Arabia
| | - Yaseen A. M. M. Elshaier
- Department of Organic and Medicinal chemistry, Faculty of PharmacyUniversity of Sadat CityMenoufiyaEgypt
| | - Hazem A. Ghabbour
- Department of Medicinal Chemistry, Faculty of PharmacyUniversity of MansouraMansoura35516Egypt
| | - Mohammad Shahidul Islam
- Department of Chemistry, College of ScienceKing Saud UniversityP. O. Box 2455Riyadh11451Saudi Arabia
| | - Assem Barakat
- Department of Chemistry, College of ScienceKing Saud UniversityP. O. Box 2455Riyadh11451Saudi Arabia
- Department of Chemistry, Faculty of ScienceAlexandria UniversityP.O. Box 426Ibrahimia, Alexandria21321Egypt
| |
Collapse
|
6
|
Zinser CM, Warren KG, Nahra F, Al-Majid A, Barakat A, Islam MS, Nolan SP, Cazin CSJ. Palladate Precatalysts for the Formation of C–N and C–C Bonds. Organometallics 2019. [DOI: 10.1021/acs.organomet.9b00326] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - Katie G. Warren
- School of Chemistry, University of St Andrews, St Andrews KY16 9ST, U.K
| | - Fady Nahra
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University, Krijgslaan 281, S-3, 9000 Ghent, Belgium
- VITO (Flemish Institute for Technological Research), Separation and Conversion Technology, Boeretang 200, B-2400 Mol, Belgium
| | - Abdullah Al-Majid
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Assem Barakat
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Mohammad Shahidul Islam
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Steven P. Nolan
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University, Krijgslaan 281, S-3, 9000 Ghent, Belgium
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Catherine S. J. Cazin
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University, Krijgslaan 281, S-3, 9000 Ghent, Belgium
| |
Collapse
|
7
|
Du S, Sarver JG, Trabbic CJ, Erhardt PW, Schroering A, Maltese WA. 6-MOMIPP, a novel brain-penetrant anti-mitotic indolyl-chalcone, inhibits glioblastoma growth and viability. Cancer Chemother Pharmacol 2018; 83:237-254. [PMID: 30426158 DOI: 10.1007/s00280-018-3726-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 11/03/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE 3-(6-Methoxy-2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propene-1-one (6-MOMIPP) is a novel indole-based chalcone that disrupts microtubules. The present study aims to define the mechanism through which 6-MOMIPP induces cell death and to evaluate the efficacy of the compound in penetrating the blood-brain barrier and inhibiting growth of glioblastoma xenografts. METHODS The effects of 6-MOMIPP were evaluated in cultured U251 glioblastoma cells, using viability, flow cytometry, and tubulin polymerization assays. Scintillation proximity and tubulin crosslinking methods were used to identify the binding site of 6-MOMIPP on tubulin, and western blots were performed to define the signaling pathways that contribute to cell death. LC/MS assays were used to study the pharmacokinetic behavior of 6-MOMIPP in mice. Subcutaneous and intracerebral xenograft models were utilized to assess the effects of 6-MOMIPP on growth of U251 glioblastoma in vivo. RESULTS The findings indicate that 6-MOMIPP targets the colchicine site on β-tubulin. At concentrations ≥ 250 nm, 6-MOMIPP induces mitotic arrest, caspase activation and loss of cell viability. Cells are protected by caspase inhibitors, pointing to an apoptotic mechanism of cell death. Loss of cell viability is preceded by activation of Cdk1(Cdc2) and phosphorylation of Bcl-2 and Bcl-xL. Inhibition of both events with a Cdk1 inhibitor prevents cell death. 6-MOMIPP has broad activity against the viability of multiple glioblastoma, melanoma and lung carcinoma cell lines. Viability of normal cells, including differentiated neurons, is not significantly affected at a drug concentration (1 µM) that reduces viability in most cancer lines. Pharmacokinetic studies in mice show that concentrations of 6-MOMIPP in the brain mirror those in the plasma, indicating that 6-MOMIPP readily penetrates the blood-brain barrier. Studies with mice bearing human U251 glioblastoma xenografts demonstrate that 6-MOMIPP is effective in suppressing growth of subcutaneous and intracerebral tumors without causing general toxicity. CONCLUSIONS The results indicate that 6-MOMIPP is a novel microtubule disruptor that targets the colchicine binding site on β-tubulin to induce mitotic arrest and cell death. The ability of 6-MOMIPP to penetrate the blood-brain barrier and inhibit growth of glioblastoma xenografts suggests that it warrants further preclinical evaluation as potential small-molecule therapeutic that may have advantages in treating primary and metastatic brain tumors.
Collapse
Affiliation(s)
- Shengnan Du
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA
| | - Jeffrey G Sarver
- Center for Drug Design and Development, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2810 W. Bancroft Street, Toledo, OH, 43606, USA
| | - Christopher J Trabbic
- Center for Drug Design and Development, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2810 W. Bancroft Street, Toledo, OH, 43606, USA
| | - Paul W Erhardt
- Center for Drug Design and Development, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2810 W. Bancroft Street, Toledo, OH, 43606, USA
| | - Allen Schroering
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA
| | - William A Maltese
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA.
| |
Collapse
|
8
|
Bhale PS, Chavan HV, Dongare SB, Shringare SN, Mule YB, Nagane SS, Bandgar BP. Synthesis of extended conjugated indolyl chalcones as potent anti-breast cancer, anti-inflammatory and antioxidant agents. Bioorg Med Chem Lett 2017; 27:1502-1507. [DOI: 10.1016/j.bmcl.2017.02.052] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/19/2017] [Accepted: 02/21/2017] [Indexed: 01/29/2023]
|
9
|
Martel-Frachet V, Keramidas M, Nurisso A, DeBonis S, Rome C, Coll JL, Boumendjel A, Skoufias DA, Ronot X. IPP51, a chalcone acting as a microtubule inhibitor with in vivo antitumor activity against bladder carcinoma. Oncotarget 2016; 6:14669-86. [PMID: 26036640 PMCID: PMC4546496 DOI: 10.18632/oncotarget.4144] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 04/08/2015] [Indexed: 12/18/2022] Open
Abstract
We previously identified 1-(2,4-dimethoxyphenyl)-3-(1-methylindolyl) propenone (IPP51), a new chalcone derivative that is capable of inducing prometaphase arrest and subsequent apoptosis of bladder cancer cells. Here, we demonstrate that IPP51 selectively inhibits proliferation of tumor-derived cells versus normal non-tumor cells. IPP51 interfered with spindle formation and mitotic chromosome alignment. Accumulation of cyclin B1 and mitotic checkpoint proteins Bub1 and BubR1 on chromosomes in IPP51 treated cells indicated the activation of spindle-assembly checkpoint, which is consistent with the mitotic arrest. The antimitotic actions of other chalcones are often associated with microtubule disruption. Indeed, IPP51 inhibited tubulin polymerization in an in vitro assay with purified tubulin. In cells, IPP51 induced an increase in soluble tubulin. Furthermore, IPP51 inhibited in vitro capillary-like tube formation by endothelial cells, indicating that it has anti-angiogenic activity. Molecular docking showed that the indol group of IPP51 can be accommodated in the colchicine binding site of tubulin. This characteristic was confirmed by an in vitro competition assay demonstrating that IPP51 can compete for colchicine binding to soluble tubulin. Finally, in a human bladder xenograft mouse model, IPP51 inhibited tumor growth without signs of toxicity. Altogether, these findings suggest that IPP51 is an attractive new microtubule-targeting agent with potential chemotherapeutic value.
Collapse
Affiliation(s)
- Véronique Martel-Frachet
- Université Joseph Fourier, AGIM CNRS FRE, EPHE, GRENOBLE Cedex 9. Université Joseph Fourier, Grenoble, France
| | - Michelle Keramidas
- Unité INSERM/UJF U823, Centre de recherche Albert Bonniot, Grenoble, France
| | - Alessandra Nurisso
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Quai Ernest-Ansermet, Geneva, Switzerland
| | | | - Claire Rome
- Unité Inserm, Grenoble Institute of Neuroscience, Site Santé, Grenoble, France
| | - Jean-Luc Coll
- Unité INSERM/UJF U823, Centre de recherche Albert Bonniot, Grenoble, France
| | - Ahcène Boumendjel
- Université de Grenoble/CNRS, UMR, Département de Pharmacochimie Moléculaire, Grenoble Cedex, France
| | | | - Xavier Ronot
- Université Joseph Fourier, AGIM CNRS FRE, EPHE, GRENOBLE Cedex 9. Université Joseph Fourier, Grenoble, France
| |
Collapse
|
10
|
Trabbic CJ, Overmeyer JH, Alexander EM, Crissman EJ, Kvale HM, Smith MA, Erhardt PW, Maltese WA. Synthesis and biological evaluation of indolyl-pyridinyl-propenones having either methuosis or microtubule disruption activity. J Med Chem 2015; 58:2489-512. [PMID: 25654321 PMCID: PMC4360382 DOI: 10.1021/jm501997q] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Methuosis is a form of nonapoptotic cell death characterized by an accumulation of macropinosome-derived vacuoles with eventual loss of membrane integrity. Small molecules inducing methuosis could offer significant advantages compared to more traditional anticancer drug therapies that typically rely on apoptosis. Herein we further define the effects of chemical substitutions at the 2- and 5-indolyl positions on our lead compound 3-(5-methoxy-2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propene-1-one (MOMIPP). We have identified a number of compounds that induce methuosis at similar potencies, including an interesting analogue having a hydroxypropyl substituent at the 2-position. In addition, we have discovered that certain substitutions on the 2-indolyl position redirect the mode of cytotoxicity from methuosis to microtubule disruption. This switch in activity is associated with an increase in potency as large as 2 orders of magnitude. These compounds appear to represent a new class of potent microtubule-active anticancer agents.
Collapse
Affiliation(s)
- Christopher J Trabbic
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences , 3000 Arlington Avenue, Toledo, Ohio 43614, United States
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Gao W, Lan S, Li Y, Zhang H, Chang M. A Facile synthesis of Novel Indole-Based Chalcones ( E)-1-(2-Chloro-1-Methyl-1 H-Indol-3-Yl)-3-Arylprop-2-En-1-Ones. JOURNAL OF CHEMICAL RESEARCH 2014. [DOI: 10.3184/174751914x14108592918139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A facile and general synthesis of 23 novel indole-based chalcones, ( E)-1-(2-chloro-1-methyl-1 H-indol-3-yl)-3-arylprop-2-en-1-ones, has been achieved in good yields of 71–89% by the Claisen–Schmidt condensation reaction of 3-acetyl-2-chloro- N-methylindole with variously substituted araldehydes using 1,4-dioxane as solvent in the presence of 5% aq. KOH. A similar reaction using furan-2- or thiophene-2-carbaldehyde gave analogous products in good yield, but an unexpected aldol reaction occurred with 2-nitrobenzaldehyde and the stable aldol product was isolated as the major product in a good yield of 73%.
Collapse
Affiliation(s)
- Wentao Gao
- Institute of Superfine Chemicals, Bohai University, Jinzhou 121000, P.R. China
| | - Shuai Lan
- Institute of Superfine Chemicals, Bohai University, Jinzhou 121000, P.R. China
| | - Yang Li
- Institute of Superfine Chemicals, Bohai University, Jinzhou 121000, P.R. China
| | - Hong Zhang
- College of Chemistry & Chemical Engineering, Bohai University, Jinzhou 121000, P.R. China
| | - Mingqin Chang
- College of Chemistry & Chemical Engineering, Bohai University, Jinzhou 121000, P.R. China
| |
Collapse
|
12
|
Champelovier P, Chauchet X, Hazane-Puch F, Vergnaud S, Garrel C, Laporte F, Boutonnat J, Boumendjel A. Cellular and molecular mechanisms activating the cell death processes by chalcones: Critical structural effects. Toxicol In Vitro 2013; 27:2305-15. [PMID: 24134853 DOI: 10.1016/j.tiv.2013.09.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/06/2013] [Accepted: 09/24/2013] [Indexed: 02/06/2023]
Abstract
Chalcones are naturally occurring compounds with diverse pharmacological activities. Chalcones derive from the common structure: 1,3-diphenylpropenone. The present study aims to better understand the mechanistic pathways triggering chalcones anticancer effects and providing evidences that minor structural difference could lead to important difference in mechanistic effect. We selected two recently investigated chalcones (A and B) and investigated them on glioblastoma cell lines. It was found that chalcone A induced an apoptotic process (type I PCD), via the activation of caspase-3, -8 and -9. Chalcone A also increased CDK1/cyclin B ratios and decreased the mitochondrial transmembrane potential (ΔΨm). Chalcone B induced an autophagic cell death process (type II PCD), ROS-related but independent of both caspases and protein synthesis. Both chalcones increased Bax/Bcl2 ratios and decreased Ki67 and CD71 antigen expressions. The present investigation reveals that despite the close structure of chalcones A and B, significant differences in mechanism of effect were found.
Collapse
Affiliation(s)
- Pierre Champelovier
- Laboratoire de Cytologie, Département d'Anatomie et de Cytologie Pathologiques, Institut de Biologie et de Pathologie, Centre Hospitalier Universitaire de Grenoble, Hôpital A. Michallon, CS10217, 38043 Grenoble cedex 09, France.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Gao W, Liu R, Li Y, Cui P. Two efficient methods for the synthesis of novel indole-based chalcone derivatives. RESEARCH ON CHEMICAL INTERMEDIATES 2013. [DOI: 10.1007/s11164-013-1148-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
14
|
Valdameri G, Gauthier C, Terreux R, Kachadourian R, Day BJ, Winnischofer SMB, Rocha MEM, Frachet V, Ronot X, Di Pietro A, Boumendjel A. Investigation of chalcones as selective inhibitors of the breast cancer resistance protein: critical role of methoxylation in both inhibition potency and cytotoxicity. J Med Chem 2012; 55:3193-200. [PMID: 22449016 PMCID: PMC3983950 DOI: 10.1021/jm2016528] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
ABCG2 plays a major role in anticancer-drug efflux and related tumor multidrug resistance. Potent and selective ABCG2 inhibitors with low cytotoxicity were investigated among a series of 44 chalcones and analogues (1,3-diarylpropenones), by evaluating their inhibitory effect on the transport of mitoxantrone, a known ABCG2 substrate. Six compounds producing complete inhibition with IC(50) values below 0.5 μM and high selectivity for ABCG2 were identified. The number and position of methoxy substituents appeared to be critical for both inhibition and cytotoxicity. The best compounds, with potent inhibition and low toxicity, contained an N-methyl-1-indolyl (compound 38) or a 6'-hydroxyl-2',4'-dimethoxy-1-phenyl (compound 27) moiety (A-ring) and two methoxy groups at positions 2 and 6 of the 3-phenyl moiety (B-ring). Methoxy substitution contributed to inhibition at positions 3 and 5, but had a negative effect at position 4. Finally, methoxy groups at positions 3, 4, and 5 of the B-ring markedly increased cytotoxicity and, therefore, should be avoided.
Collapse
Affiliation(s)
- Glaucio Valdameri
- Equipe Labellisée Ligue 2012, BMSSI UMR 5086 CNRS/Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Charlotte Gauthier
- Equipe Labellisée Ligue 2012, BMSSI UMR 5086 CNRS/Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Raphaël Terreux
- Equipe Bioinformatique: structures et interactions, BMSSI UMR 5086 CNRS/Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Rémy Kachadourian
- Department of Medicine, National Jewish Health, Denver Colorado 80206, United States
| | - Brian J. Day
- Department of Medicine, National Jewish Health, Denver Colorado 80206, United States
| | - Sheila M. B. Winnischofer
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Maria E. M. Rocha
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Véronique Frachet
- AGing Imaging Modeling, FRE 3405, Université Joseph Fourier, CNRS, EPHE, Faculté de Médecine, La Tronche, France
| | - Xavier Ronot
- AGing Imaging Modeling, FRE 3405, Université Joseph Fourier, CNRS, EPHE, Faculté de Médecine, La Tronche, France
| | - Attilio Di Pietro
- Equipe Labellisée Ligue 2012, BMSSI UMR 5086 CNRS/Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Ahcène Boumendjel
- Université Joseph Fourier—Grenoble/CNRS, UMR 5063, Département de Pharmacochimie Moléculaire, Grenoble, France
| |
Collapse
|