1
|
Yu Q, Dehghani-Ghahnaviyeh S, Rasouli A, Sadurni A, Kowal J, Bang-Soerensen R, Wen PC, Tinzl-Zechner M, Irobalieva RN, Ni D, Stahlberg H, Altmann KH, Tajkhorshid E, Locher KP. Modulation of ABCG2 Transporter Activity by Ko143 Derivatives. ACS Chem Biol 2024; 19:2304-2313. [PMID: 39445888 PMCID: PMC11574751 DOI: 10.1021/acschembio.4c00353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
ABCG2 is a multidrug transporter that protects tissues from xenobiotics, affects drug pharmacokinetics, and contributes to multidrug resistance of cancer cells. Here, we present tetracyclic fumitremorgin C analog Ko143 derivatives, evaluate their in vitro modulation of purified ABCG2, and report four high-resolution cryo-EM structures and computational analyses to elucidate their interactions with ABCG2. We found that Ko143 derivatives that are based on a ring-opened scaffold no longer inhibit ABCG2-mediated transport activity. In contrast, closed-ring, tetracyclic analogs were highly potent inhibitors. Strikingly, the least potent of these compounds, MZ82, bound deeper into the central ABCG2 cavity than the other inhibitors and it led to partial closure of the transmembrane domains and increased flexibility of the nucleotide-binding domains. Minor structural modifications can thus convert a potent inhibitor into a compound that induces conformational changes in ABCG2 similar to those observed during binding of a substrate. Molecular dynamics simulations and free energy binding calculations further supported the correlation between reduced potency and distinct binding pose of the compounds. We introduce the highly potent inhibitor AZ99 that may exhibit improved in vivo stability.
Collapse
Affiliation(s)
- Qin Yu
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Sepehr Dehghani-Ghahnaviyeh
- Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Ali Rasouli
- Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Anna Sadurni
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich 8093, Switzerland
| | - Julia Kowal
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Rose Bang-Soerensen
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Po-Chao Wen
- Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Melanie Tinzl-Zechner
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich 8093, Switzerland
| | - Rossitza N Irobalieva
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Dongchun Ni
- Laboratory of Biological Electron Microscopy, Institute of Physics, SB, EPFL, Lausanne 1015, Switzerland
| | - Henning Stahlberg
- Laboratory of Biological Electron Microscopy, Institute of Physics, SB, EPFL, Lausanne 1015, Switzerland
| | - Karl-Heinz Altmann
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich 8093, Switzerland
| | - Emad Tajkhorshid
- Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Kaspar P Locher
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, Zurich 8093, Switzerland
| |
Collapse
|
2
|
Qi Q, Gu R, Zhu J, Anderson KE, Ma X. Roles of the ABCG2 Transporter in Protoporphyrin IX Distribution and Toxicity. Drug Metab Dispos 2024; 52:1201-1207. [PMID: 38351044 PMCID: PMC11495668 DOI: 10.1124/dmd.123.001582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/08/2024] [Indexed: 10/18/2024] Open
Abstract
ATP-binding cassette transporter subfamily G member 2 (ABCG2) is a membrane-bound transporter responsible for the efflux of various xenobiotics and endobiotics, including protoporphyrin IX (PPIX), an intermediate in the heme biosynthesis pathway. Certain genetic mutations and chemicals impair the conversion of PPIX to heme and/or increase PPIX production, leading to PPIX accumulation and toxicity. In mice, deficiency of ABCG2 protects against PPIX-mediated phototoxicity and hepatotoxicity by modulating PPIX distribution. In addition, in vitro studies revealed that ABCG2 inhibition increases the efficacy of PPIX-based photodynamic therapy by retaining PPIX inside target cells. In this review, we discuss the roles of ABCG2 in modulating the tissue distribution of PPIX, PPIX-mediated toxicity, and PPIX-based photodynamic therapy. SIGNIFICANCE STATEMENT: This review summarized the roles of ABCG2 in modulating PPIX distribution and highlighted the therapeutic potential of ABCG2 inhibitors for the management of PPIX-mediated toxicity.
Collapse
Affiliation(s)
- Qian Qi
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Ruizhi Gu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Karl E Anderson
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| |
Collapse
|
3
|
Ivanova E, Osipova M, Kadyrov Y, Karpov S, Markova S, Zazhivihina E, Umanova L, Vasilieva T, Mitrasov Y, Smolkina Y, Nasakin O. Reactions of Tetracyanoethylene with Aliphatic and Aromatic Amines and Hydrazines and Chemical Transformations of Tetracyanoethylene Derivatives. Molecules 2024; 29:4727. [PMID: 39407655 PMCID: PMC11477996 DOI: 10.3390/molecules29194727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
The significant synthetic potential and reactivity of tetracyanoethylene (TCNE) have captured the interest of numerous chemical communities. One of the most promising, readily achievable, yet least explored pathways for the reactivity of TCNE involves its interaction with arylamines. Typically, the reaction proceeds via tricyanovinylation (TCV); however, deviations from the standard chemical process have been observed in some instances. These include the formation of heterocyclic structures through tricyanovinyl intermediates, aliphatic dicarbonitriles through the cleavage of the C-C bond of a tetracyanoethyl substituent, complexation, and various pericyclic reactions. Therefore, the objective of this study is to review the diverse modes of interaction of TCNE with aromatic nitrogen-containing compounds and to focus the attention of the chemical community on the synthetic capabilities of this reagent, as well as the various biological and optical activities of the structures synthesized based on TCNE.
Collapse
Affiliation(s)
- Elizaveta Ivanova
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Margarita Osipova
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Yhtyyar Kadyrov
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Sergey Karpov
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Svetlana Markova
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Ekaterina Zazhivihina
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Lubov Umanova
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Tatyana Vasilieva
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Yurii Mitrasov
- Department of Scientific Chemistry Education, Yakovlev Chuvash State Pedagogical University, K. Marx Street, 38, 428000 Cheboksary, Russia
| | - Yulia Smolkina
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| | - Oleg Nasakin
- Organic and Pharmaceutical Chemistry Department, Ulyanov Chuvash State University, Moskovsky Prospect, 15, 428015 Cheboksary, Russia; (E.I.); (M.O.); (Y.K.); (S.K.); (S.M.); (E.Z.); (L.U.); (T.V.); (Y.S.)
| |
Collapse
|
4
|
Yasuda S, Svergja H, Olsen CE, Hoff BH. Promotion of Water as Solvent in Amination of 4-Chloropyrrolopyrimidines and Related Heterocycles under Acidic Conditions. ACS OMEGA 2024; 9:14142-14152. [PMID: 38559978 PMCID: PMC10976386 DOI: 10.1021/acsomega.3c09673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/22/2024] [Accepted: 02/28/2024] [Indexed: 04/04/2024]
Abstract
A switch of reaction medium from organic solvents to water can improve the safety and lower the cost of production processes. Hydrochloric acid-promoted amination of fused pyrimidines has been studied using 4-chloro-7H-pyrrolo[2,3-d]pyrimidine and aniline as model compounds. Higher rate was observed in water than in four alcoholic solvents and DMF. An important aspect is that the amount of acid should be kept low to minimize the competing solvolysis. The substrate scope for the amination in water was evaluated by reacting 4-chloro-7H-pyrrolo[2,3-d]pyrimidine with 20 aniline derivatives with variance in steric and electronic properties. Preparative useful reactions were seen for 14 of the 20 derivatives. Unsuited nucleophiles are ortho-substituted anilines with a pKa below 1. Amination of the corresponding quinazoline, thienopyrimidine, and purine also proceeded well in water. Highly lipophilic and crystalline compounds are more efficiently aminated in 2-propanol. Aliphatic and benzylic amines react poorly under acidic conditions, but these aminations can be done in water without acid.
Collapse
Affiliation(s)
- Shuhei Yasuda
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), Ho̷gskoleringen 5, NO-7491 Trondheim, Norway
| | | | - Cecilie Elisabeth Olsen
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), Ho̷gskoleringen 5, NO-7491 Trondheim, Norway
| | - Bård Helge Hoff
- Department of Chemistry, Norwegian University of Science and Technology (NTNU), Ho̷gskoleringen 5, NO-7491 Trondheim, Norway
| |
Collapse
|
5
|
Zhu J, Lei S, Lu J, Hao Y, Qian Q, Devanathan AS, Feng Z, Xie XQ, Wipf P, Ma X. Metabolism-guided development of Ko143 analogs as ABCG2 inhibitors. Eur J Med Chem 2023; 259:115666. [PMID: 37482017 PMCID: PMC10529637 DOI: 10.1016/j.ejmech.2023.115666] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
ATP-binding cassette subfamily G member 2 (ABCG2), an efflux transporter, is involved in multiple pathological processes. Ko143 is a potent ABCG2 inhibitor; however, it is quickly metabolized through carboxylesterase 1-mediated hydrolysis of its t-butyl ester moiety. The current work aimed to develop more metabolically stable ABCG2 inhibitors. Novel Ko143 analogs were designed and synthesized by replacing the unstable t-butyl ester moiety in Ko143 with an amide group. The synthesized Ko143 analogs were evaluated for their ABCG2 inhibitory activity, binding mode with ABCG2, cytotoxicity, and metabolic stability. We found that the amide modification of Ko143 led to metabolically stable ABCG2 inhibitors. Among these Ko143 analogs, K2 and K34 are promising candidates with favorable oral pharmacokinetic profiles in mice. In summary, we synthesized novel Ko143 analogs with improved metabolic stability, which can potentially be used as lead compounds for the future development of ABCG2 inhibitors.
Collapse
Affiliation(s)
- Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Saifei Lei
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jie Lu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yixuan Hao
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qi Qian
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aaron S Devanathan
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter Wipf
- Department of Chemistry and Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Chandratre S, Olsen J, Howley R, Chen B. Targeting ABCG2 transporter to enhance 5-aminolevulinic acid for tumor visualization and photodynamic therapy. Biochem Pharmacol 2023; 217:115851. [PMID: 37858868 PMCID: PMC10842008 DOI: 10.1016/j.bcp.2023.115851] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
5-Aminolevulinic acid (ALA) has been approved by the U. S. FDA for fluorescence-guided resection of high-grade glioma and photodynamic therapy (PDT) of superficial skin precancerous and cancerous lesions. As a prodrug, ALA administered orally or topically is metabolized in the heme biosynthesis pathway to produce protoporphyrin IX (PpIX), the active drug with red fluorescence and photosensitizing property. Preferential accumulation of PpIX in tumors after ALA administration enables the use of ALA for PpIX-mediated tumor fluorescence diagnosis and PDT, functioning as a photo-theranostic agent. Extensive research is currently underway to further enhance ALA-mediated PpIX tumor disposition for better tumor visualization and treatment. Particularly, the discovery of PpIX as a specific substrate of ATP binding cassette subfamily G member 2 (ABCG2) opens the door to therapeutic enhancement with ABCG2 inhibitors. Studies with human tumor cell lines and human tumor samples have demonstrated ABCG2 as an important biological determinant of reduced ALA-PpIX tumor accumulation, inhibition of which greatly enhances ALA-PpIX fluorescence and PDT response. These studies strongly support targeting ABCG2 as an effective therapeutic enhancement approach. In this review, we would like to summarize current research of ABCG2 as a drug efflux transporter in multidrug resistance, highlight previous works on targeting ABCG2 for therapeutic enhancement of ALA, and provide future perspectives on how to translate this ABCG2-targeted therapeutic enhancement strategy from bench to bedside.
Collapse
Affiliation(s)
- Sharayu Chandratre
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, USA
| | - Jordyn Olsen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, USA
| | - Richard Howley
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, USA
| | - Bin Chen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, USA; Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Cai CY, Teng QX, Murakami M, Ambudkar SV, Chen ZS, Korlipara VL. Design, Synthesis and Biological Evaluation of Quinazolinamine Derivatives as Breast Cancer Resistance Protein and P-Glycoprotein Inhibitors with Improved Metabolic Stability. Biomolecules 2023; 13:biom13020253. [PMID: 36830622 PMCID: PMC9953095 DOI: 10.3390/biom13020253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/03/2023] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
A series of twenty-two quinazolinamine derivatives showing potent inhibitory activities on breast cancer resistance protein (BCRP) and p-glycoprotein (P-gp) were synthesized. A cyclopropyl-containing quinazolinamine 22 was identified as a dual BCRP and P-gp inhibitor, while azide-containing quinazolinamine 33 showed BCRP inhibitory activity. These lead compounds were further investigated in a battery of mechanistic experiments. Compound 22 changed the localization of BCRP and P-gp in cells, thus inhibiting the efflux of anticancer drugs by the two ATP-binding cassette (ABC) transporters. In addition, both 22 and 33 significantly stimulated the ATP hydrolysis of the BCRP transporter, indicating that they can be competitive substrates of the BCRP transporter, and thereby increase the accumulation of mitoxantrone in BCRP-overexpressing H460/MX20 cells. Azide derivative 33, exhibited a greater inhibitory effect on BCRP after UV activation and can serve as a valuable probe for investigating the interactions of quinazolinamine derivatives with BCRP. Notably, the dual BCRP and P-gp inhibitors 4-5, 22-24, 27, and BCRP inhibitor 33 showed improved metabolic stability compared to Ko143.
Collapse
Affiliation(s)
- Chao-Yun Cai
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York, NY 11439, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York, NY 11439, USA
| | - Megumi Murakami
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Suresh V. Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York, NY 11439, USA
- Correspondence: (Z.-S.C.); (V.L.K.)
| | - Vijaya L. Korlipara
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York, NY 11439, USA
- Correspondence: (Z.-S.C.); (V.L.K.)
| |
Collapse
|
8
|
Puri S, Stefan K, Khan SL, Pahnke J, Stefan SM, Juvale K. Indole Derivatives as New Structural Class of Potent and Antiproliferative Inhibitors of Monocarboxylate Transporter 1 (MCT1; SLC16A1). J Med Chem 2023; 66:657-676. [PMID: 36584238 PMCID: PMC9841531 DOI: 10.1021/acs.jmedchem.2c01612] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Indexed: 12/31/2022]
Abstract
The solute carrier (SLC) monocarboxylate transporter 1 (MCT1; SLC16A1) represents a promising target for the treatment of cancer; however, the MCT1 modulator landscape is underexplored with only roughly 100 reported compounds. To expand the knowledge about MCT1 modulation, we synthesized a library of 16 indole-based molecules and subjected these to a comprehensive biological assessment platform. All compounds showed functional inhibitory activities against MCT1 at low nanomolar concentrations and great antiproliferative activities against the MCT1-expressing cancer cell lines A-549 and MCF-7, while the compounds were selective over MCT4 (SLC16A4). Lead compound 24 demonstrated a greater potency than the reference compound, and molecular docking revealed strong binding affinities to MCT1. Compound 24 led to cancer cell cycle arrest as well as apoptosis, and it showed to sensitize these cancer cells toward an antineoplastic agent. Strikingly, compound 24 had also significant inhibitory power against the multidrug transporter ABCB1 and showed to reverse ABCB1-mediated multidrug resistance (MDR).
Collapse
Affiliation(s)
- Sachin Puri
- Shobhaben
Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s
NMIMS, V.L. Mehta Road,
Vile Parle (W), Mumbai400056, India
| | - Katja Stefan
- Department
of Pathology, Section of Neuropathology, Translational Neurodegeneration
Research and Neuropathology Lab (www.pahnkelab.eu), University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372Oslo, Norway
| | - Sharuk L. Khan
- Department
of Pharmaceutical Chemistry, N.B.S. Institute
of Pharmacy, Ausa413520, Maharashtra, India
| | - Jens Pahnke
- Department
of Pathology, Section of Neuropathology, Translational Neurodegeneration
Research and Neuropathology Lab (www.pahnkelab.eu), University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372Oslo, Norway
- Drug
Development and Chemical Biology Lab, Lübeck Institute of Experimental
Dermatology (LIED), University of Lübeck
and University Medical Center Schleswig-Holstein, Ratzeburger Allee 160, 23538Lübeck, Germany
- Department
of Pharmacology, Faculty of Medicine, University
of Latvia, Jelgavas iela
4, 1004Ri̅ga, Latvia
| | - Sven Marcel Stefan
- Department
of Pathology, Section of Neuropathology, Translational Neurodegeneration
Research and Neuropathology Lab (www.pahnkelab.eu), University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372Oslo, Norway
- Drug
Development and Chemical Biology Lab, Lübeck Institute of Experimental
Dermatology (LIED), University of Lübeck
and University Medical Center Schleswig-Holstein, Ratzeburger Allee 160, 23538Lübeck, Germany
| | - Kapil Juvale
- Shobhaben
Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s
NMIMS, V.L. Mehta Road,
Vile Parle (W), Mumbai400056, India
| |
Collapse
|
9
|
Zhang W, Guang J, Fan W, Huang D. Three-Component Cycloaddition of Nitriles: Construction of Bicyclic 4-Aminopyrimidines and Their Photophysical Studies. J Org Chem 2022; 87:13598-13604. [PMID: 36223211 DOI: 10.1021/acs.joc.2c01206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A base-induced synthesis of bicyclic 4-aminopyrimidines by the cycloaddition of three types of nitriles is reported. The scope of the method is demonstrated with 44 examples. Products are found to have luminescence properties and show potential applications as organic luminescent layer materials.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | - Jin Guang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | - Weibin Fan
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | - Deguang Huang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| |
Collapse
|
10
|
Updated chemical scaffolds of ABCG2 inhibitors and their structure-inhibition relationships for future development. Eur J Med Chem 2022; 241:114628. [DOI: 10.1016/j.ejmech.2022.114628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/07/2022] [Accepted: 07/21/2022] [Indexed: 11/19/2022]
|
11
|
Qi B, Wang F, He H, Fan M, Hu L, Xiong L, Gong G, Shi S, Song X. Identification of (S)-1-(2-(2,4-difluorophenyl)-4-oxothiazolidin-3-yl)-3-(4-((7-(3-(4-ethylpiperazin-1-yl)propoxy)-6-methoxyquinolin-4-yl)oxy)-3,5-difluorophenyl)urea as a potential anti-colorectal cancer agent. Eur J Med Chem 2022; 239:114561. [PMID: 35763868 DOI: 10.1016/j.ejmech.2022.114561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/12/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
In our previous study, 1-(2-(2,6-difluorophenyl)-4-oxothiazolidin-3-yl)-3-(4-((7-(3-(4-ethylpiperazin-1-yl)propoxy)-6-methoxyquinolin-4-yl)oxy)-3,5-difluorophenyl)urea (1) was obtained as a potent tyrosine kinase inhibitor. Further structural optimization was performed in this investigation, and a series of novel quinoline derivates were designed, synthesized and evaluated for their biological activity. Among them, compound 8m possessed nanomolar c-Met and Ron inhibitory activity, with IC50 values of 4.32 nM and 2.39 nM, respectively. Kinase profile study demonstrated that it could also inhibit ABL, PDGFRβ, AXL, RET, and FLT3 with submicromolar potency. It also exhibited moderate to excellent cytotoxic activity against different types of human cancer cell lines, especially against COLO 205 cells (IC50 = 0.035 μM) which was remarkably superior to that of Cabozantinib (IC50 = 6.6 μM) and Fruquintinib (IC50 > 10.0 μM). Compared to ( ± )-8m, isomer (S)-8m and (R)-8m showed similar kinase inhibitory activity against c-Met/RON and in vitro anticancer activity against COLO 205 cells. Differently, compound (S)-8m showed an over 238-fold selectivity toward COLO 205 (IC50 = 0.042 μM) cells to FHC cells (IC50 > 10.0 μM), which indicated its low cytotoxicity against human normal tissue cells. Flow cytometry study demonstrated that compound (S)-8m could significantly induce apoptosis in COLO 205 cells in a dose-dependent manner. Cell cycle arrest assays showed that compound (S)-8m could not arrest the cell-cycle progression due to the massive dead cells.
Collapse
Affiliation(s)
- Baohui Qi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China.
| | - Fei Wang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Huan He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Mengmeng Fan
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Liping Hu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Li Xiong
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Guowei Gong
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Shengmin Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| | - Xiaomeng Song
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China; Key Laboratory of Biocatalysis & Chiral Drug Synthesis of Guizhou Province, Zunyi Medical University, Zunyi, 563000, China
| |
Collapse
|
12
|
Zuo B, Shao H, Zheng Y, Ma Y, Li W, Huang M, Deng Q. The Core‐Shell Magnetic Mesoporous Microspheres Immobilized NHC‐Palladacycles: An Efficient and Recyclable Catalyst for Suzuki–Miyaura Cross‐Coupling of Pharmaceutical Synthesis. ASIAN J ORG CHEM 2022. [DOI: 10.1002/ajoc.202200018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Bin Zuo
- University of Shanghai for Science and Technology School of Materials and Chemistry No. 334 Jungong Road, Shanghai, 200093, P.R. China 200093 Shanghai CHINA
| | - Han Shao
- University of Shanghai for Science and Technology School of Materials and Chemistry Shanghai 200093, China. Shanghai CHINA
| | - Yan Zheng
- University of Shanghai for Science and Technology School of Materials and Chemistry Shanghai 200093, China. Shanghai CHINA
| | - Yunhua Ma
- University of Shanghai for Science and Technology School of Materials and Chemistry Shanghai 200093, China. Shanghai CHINA
| | - Wanfang Li
- University of Shanghai for Science and Technology School of Materials and Chemistry Shanghai 200093, China. Shanghai CHINA
| | - Mingxian Huang
- University of Shanghai for Science and Technology School of Materials and Chemistry Shanghai 200093, China. Shanghai CHINA
| | - Qinyue Deng
- University of Shanghai for Science and Technology School of Materials and Chemistry No. 334 Jungong Road 200093 Shanghai CHINA
| |
Collapse
|
13
|
Szewc M, Radzikowska-Bűchner E, Wdowiak P, Kozak J, Kuszta P, Niezabitowska E, Matysiak J, Kubiński K, Masłyk M. MSCs as Tumor-Specific Vectors for the Delivery of Anticancer Agents-A Potential Therapeutic Strategy in Cancer Diseases: Perspectives for Quinazoline Derivatives. Int J Mol Sci 2022; 23:2745. [PMID: 35269887 PMCID: PMC8911180 DOI: 10.3390/ijms23052745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are considered to be a powerful tool in the treatment of various diseases. Scientists are particularly interested in the possibility of using MSCs in cancer therapy. The research carried out so far has shown that MSCs possess both potential pro-oncogenic and anti-oncogenic properties. It has been confirmed that MSCs can regulate tumor cell growth through a paracrine mechanism, and molecules secreted by MSCs can promote or block a variety of signaling pathways. These findings may be crucial in the development of new MSC-based cell therapeutic strategies. The abilities of MSCs such as tumor tropism, deep migration and immune evasion have evoked considerable interest in their use as tumor-specific vectors for small-molecule anticancer agents. Studies have shown that MSCs can be successfully loaded with chemotherapeutic drugs such as gemcitabine and paclitaxel, and can release them at the site of primary and metastatic neoplasms. The inhibitory effect of MSCs loaded with anti-cancer agents on the proliferation of cancer cells has also been observed. However, not all known chemotherapeutic agents can be used in this approach, mainly due to their cytotoxicity towards MSCs and insufficient loading and release capacity. Quinazoline derivatives appear to be an attractive choice for this therapeutic solution due to their biological and pharmacological properties. There are several quinazolines that have been approved for clinical use as anticancer drugs by the US Food and Drug Administration (FDA). It gives hope that the synthesis of new quinazoline derivatives and the development of methods of their application may contribute to the establishment of highly effective therapies for oncological patients. However, a deeper understanding of interactions between MSCs and tumor cells, and the exploration of the possibilities of using quinazoline derivatives in MSC-based therapy is necessary to achieve this goal. The aim of this review is to discuss the prospects for using MSC-based cell therapy in cancer treatment and the potential use of quinazolines in this procedure.
Collapse
Affiliation(s)
- Monika Szewc
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (P.W.); (J.K.); (P.K.)
| | - Elżbieta Radzikowska-Bűchner
- Department of Plastic, Reconstructive and Maxillary Surgery, Central Clinical Hospital MSWiA, 02-507 Warsaw, Poland;
| | - Paulina Wdowiak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (P.W.); (J.K.); (P.K.)
| | - Joanna Kozak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (P.W.); (J.K.); (P.K.)
| | - Piotr Kuszta
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (P.W.); (J.K.); (P.K.)
| | - Ewa Niezabitowska
- Department of Urology and Urological Oncology, Multidisciplinary Hospital in Lublin, 20-400 Lublin, Poland;
| | - Joanna Matysiak
- Department of Chemistry, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Konrad Kubiński
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, 20-708 Lublin, Poland;
| | - Maciej Masłyk
- Department of Molecular Biology, The John Paul II Catholic University of Lublin, 20-708 Lublin, Poland;
| |
Collapse
|
14
|
Nishimura RHV, Dos Santos T, Murie VE, Furtado LC, Costa-Lotufo LV, Clososki GC. Efficient N-arylation of 4-chloroquinazolines en route to novel 4-anilinoquinazolines as potential anticancer agents. Beilstein J Org Chem 2022; 17:2968-2975. [PMID: 35003373 PMCID: PMC8712970 DOI: 10.3762/bjoc.17.206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/08/2021] [Indexed: 12/02/2022] Open
Abstract
Microwave-mediated N-arylation of 4-chloroquinazolines in THF/H2O rapidly and efficiently afforded a library of novel 6-halo-2-phenyl-substituted 4-anilinoquinazolines. The methodology was compatible with numerous ortho-, meta-, and para-substituted N-methylanilines as well as substituted anilines and furnished the corresponding 4-anilinoquinazolines in good yields. Preliminary screening of the synthesized compounds against tumor cells (HCT-116 and T98G) showed promising antiproliferative properties.
Collapse
Affiliation(s)
- Rodolfo H V Nishimura
- Núcleo de Pesquisas em Produtos Naturais e Sintéticos, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café s/n, Ribeirão Preto-SP 14040-903, Brazil.,Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-SP, Universidade de São Paulo, Av. dos Bandeirantes 3900, Ribeirão Preto-SP 14090-901, Brazil
| | - Thiago Dos Santos
- Núcleo de Pesquisas em Produtos Naturais e Sintéticos, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café s/n, Ribeirão Preto-SP 14040-903, Brazil
| | - Valter E Murie
- Núcleo de Pesquisas em Produtos Naturais e Sintéticos, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café s/n, Ribeirão Preto-SP 14040-903, Brazil
| | - Luciana C Furtado
- Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Lineu Prestes 1524, 05508-900 São Paulo-SP, Brazil
| | - Leticia V Costa-Lotufo
- Instituto de Ciências Biomédicas, Universidade de São Paulo, Av. Lineu Prestes 1524, 05508-900 São Paulo-SP, Brazil
| | - Giuliano C Clososki
- Núcleo de Pesquisas em Produtos Naturais e Sintéticos, Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café s/n, Ribeirão Preto-SP 14040-903, Brazil.,Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto-SP, Universidade de São Paulo, Av. dos Bandeirantes 3900, Ribeirão Preto-SP 14090-901, Brazil
| |
Collapse
|
15
|
Sahoo S, Pal S. Copper-Catalyzed One-Pot Synthesis of Quinazolinones from 2-Nitrobenzaldehydes with Aldehydes: Application toward the Synthesis of Natural Products. J Org Chem 2021; 86:18067-18080. [PMID: 34813342 DOI: 10.1021/acs.joc.1c02343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A novel, efficient, and atom-economical approach for the construction of quinazolinones from 2-nitrobenzaldehydes has been unveiled via copper-catalyzed nitrile formation, hydrolysis, and reduction in one pot for the first time. In this reaction, urea is used as a source of nitrogen for nitrile formation, hydrazine hydrate is used for both the reduction of the nitro group and the hydrolysis of nitrile, and atmospheric oxygen is used as the sole oxidant. The method portrays a wide substrate scope with good functional group tolerances. Moreover, this method was applied for the synthesis of schizocommunin, tryptanthrin, phaitanthrin-A, phaitanthrin-B, and 8H-quinazolino[4,3-b]quinazolin-8-one.
Collapse
Affiliation(s)
- Subrata Sahoo
- Organic Chemistry Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Argul, Odisha 752050, India
| | - Shantanu Pal
- Organic Chemistry Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Argul, Odisha 752050, India
| |
Collapse
|
16
|
Rhodium(III)-catalyzed [4 + 2] annulation of N-arylbenzamidines with 1,4,2-dioxazol-5-ones: Easy access to 4-aminoquinazolines via highly selective C H bond activation. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.02.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
Namasivayam V, Silbermann K, Pahnke J, Wiese M, Stefan SM. Scaffold fragmentation and substructure hopping reveal potential, robustness, and limits of computer-aided pattern analysis (C@PA). Comput Struct Biotechnol J 2021; 19:3269-3283. [PMID: 34141145 PMCID: PMC8193046 DOI: 10.1016/j.csbj.2021.05.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023] Open
Abstract
Computer-aided pattern analysis (C@PA) was recently presented as a powerful tool to predict multitarget ABC transporter inhibitors. The backbone of this computational methodology was the statistical analysis of frequently occurring molecular features amongst a fixed set of reported small-molecules that had been evaluated toward ABCB1, ABCC1, and ABCG2. As a result, negative and positive patterns were elucidated, and secondary positive substructures could be suggested that complemented the multitarget fingerprints. Elevating C@PA to a non-statistical and exploratory level, the concluded secondary positive patterns were extended with potential positive substructures to improve C@PA's prediction capabilities and to explore its robustness. A small-set compound library of known ABCC1 inhibitors with a known hit rate for triple ABCB1, ABCC1, and ABCG2 inhibition was taken to virtually screen for the extended positive patterns. In total, 846 potential broad-spectrum ABCB1, ABCC1, and ABCG2 inhibitors resulted, from which 10 have been purchased and biologically evaluated. Our approach revealed 4 novel multitarget ABCB1, ABCC1, and ABCG2 inhibitors with a biological hit rate of 40%, but with a slightly lower inhibitory power than derived from the original C@PA. This is the very first report about discovering novel broad-spectrum inhibitors against the most prominent ABC transporters by improving C@PA.
Collapse
Key Words
- ABC transporter, ATP-binding cassette transporter
- ABCB1 (P-gp)
- ABCC1 (MRP1)
- ABCG2 (BCRP)
- ATP, adenosine-triphosphate
- Alzheimer's disease (AD)
- BCRP, breast cancer resistance protein (ABCG2)
- C@PA, computer-aided pattern analysis
- F1–5, pharmacophore features 1–5
- IC50, half-maximal inhibition concentration
- MDR, multidrug resistance
- MOE, molecular operating environment
- MRP1, multidrug resistance-associated protein 1 (ABCC1)
- Multidrug resistance (MDR)
- Multitarget fingerprints
- P-gp, P-glycoprotein (ABCB1)
- Pan-ABC inhibition / antagonism / blockage (PANABC)
- Pattern analysis (C@PA)
- SEM, standard error of the mean
- SMILES, simplified molecular input line entry specification
- Tc, Tanimotto coefficient
- Triple / multitarget / broad-spectrum / promiscuous inhibitor / antagonist
- Under-studied ABC transporters (e.g., ABCA7)
- Well-studied ABC transporters
- calcein AM, calcein acetoxymethyl
Collapse
Affiliation(s)
- Vigneshwaran Namasivayam
- Department of Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Katja Silbermann
- Department of Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Jens Pahnke
- Department of Neuro-/Pathology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- LIED, University of Lübeck, Ratzenburger Allee 160, 23538 Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Jelgavas iela 1, 1004 Rīga, Latvia
- Department of Bioorganic Chemistry, Leibniz-Institute of Plant Biochemistry, Weinberg 3, 06120 Halle, Germany
| | - Michael Wiese
- Department of Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Sven Marcel Stefan
- Department of Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
- Department of Neuro-/Pathology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Kolling Builging, 10 Westbourne Street, Sydney, New South Wales 2065, Australia
| |
Collapse
|
18
|
Novosjolova I, Turks M, Jeminejs A, Goliškina SM, Stepanovs D, Bizdēna Ē. Application of Azide-Tetrazole Tautomerism and Arylsulfanyl Group Dance in the Synthesis of Thiosubstituted Tetrazoloquinazolines. SYNTHESIS-STUTTGART 2021. [DOI: 10.1055/s-0040-1706568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
AbstractNucleophilic aromatic substitution reaction between 4-arylthio-2-chloroquinazolines and NaN3 takes place with an unusual sulfanyl group dance and leads to the formation of 5-(arylthio)tetrazolo[1,5-c]-quinazolines, which do not form the azide tautomer and do not undergo CuAAC reactions with alkynes. On the other hand, 5-azidotetrazolo[1,5-a]quinazoline (formally described as 2,4-diazidoquinazoline) undergoes regioselective nucleophilic aromatic substitution with thiols at C5 and forms 5-(alkyl/arylthio)tetrazolo[1,5-a]quinazolines, the structure of which has been proved by X-ray crystallography. The latter exist in tautomeric equilibrium with their 2-azidoquinazoline form, which provides possibility for copper-catalyzed azide–alkyne 1,3-dipolar cycloaddition reaction, leading to the 4-alkyl/arylthio-2-(1H-1,2,3-triazol-1-yl)quinazolines.
Collapse
Affiliation(s)
- Irina Novosjolova
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University
| | - Māris Turks
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University
| | - Andris Jeminejs
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University
| | - Svetlana M. Goliškina
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University
| | | | - Ērika Bizdēna
- Institute of Technology of Organic Chemistry, Faculty of Materials Science and Applied Chemistry, Riga Technical University
| |
Collapse
|
19
|
Namasivayam V, Silbermann K, Wiese M, Pahnke J, Stefan SM. C@PA: Computer-Aided Pattern Analysis to Predict Multitarget ABC Transporter Inhibitors. J Med Chem 2021; 64:3350-3366. [PMID: 33724808 PMCID: PMC8041314 DOI: 10.1021/acs.jmedchem.0c02199] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Based on literature reports of the last two decades, a computer-aided pattern analysis (C@PA) was implemented for the discovery of novel multitarget ABCB1 (P-gp), ABCC1 (MRP1), and ABCG2 (BCRP) inhibitors. C@PA included basic scaffold identification, substructure search and statistical distribution, as well as novel scaffold extraction to screen a large virtual compound library. Over 45,000 putative and novel broad-spectrum ABC transporter inhibitors were identified, from which 23 were purchased for biological evaluation. Our investigations revealed five novel lead molecules as triple ABCB1, ABCC1, and ABCG2 inhibitors. C@PA is the very first successful computational approach for the discovery of promiscuous ABC transporter inhibitors.
Collapse
Affiliation(s)
- Vigneshwaran Namasivayam
- Department of Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Katja Silbermann
- Department of Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Michael Wiese
- Department of Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Jens Pahnke
- Department of Neuro-/Pathology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway.,LIED, University of Lübeck, Ratzenburger Allee 160, 23538 Lübeck, Germany.,Department of Pharmacology, Faculty of Medicine, University of Latvia, Jelgavas iela 1, 1004 Riga, Latvia.,Department of Bioorganic Chemistry, Leibniz-Institute of Plant Biochemistry, Weinberg 3, 06120 Halle, Germany
| | - Sven Marcel Stefan
- Department of Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany.,Department of Neuro-/Pathology, University of Oslo and Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway.,Cancer Drug Resistance and Stem Cell Program, University of Sydney, Kolling Building, 10 Westbourne Street, Sydney, New South Wales 2065, Australia
| |
Collapse
|
20
|
Amin S, Alam MM, Akhter M, Najmi AK, Siddiqui N, Husain A, Shaquiquzzaman M. A review on synthetic procedures and applications of phosphorus oxychloride (POCl 3) in the last biennial period (2018–19). PHOSPHORUS SULFUR 2021. [DOI: 10.1080/10426507.2020.1831499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Shaista Amin
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - M. Mumtaz Alam
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mymoona Akhter
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - A. K. Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Nadeem Siddiqui
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Asif Husain
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - M. Shaquiquzzaman
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
21
|
Kita DH, Guragossian N, Zattoni IF, Moure VR, Rego FGDM, Lusvarghi S, Moulenat T, Belhani B, Picheth G, Bouacida S, Bouaziz Z, Marminon C, Berredjem M, Jose J, Gonçalves MB, Ambudkar SV, Valdameri G, Le Borgne M. Mechanistic basis of breast cancer resistance protein inhibition by new indeno[1,2-b]indoles. Sci Rep 2021; 11:1788. [PMID: 33469044 PMCID: PMC7815716 DOI: 10.1038/s41598-020-79892-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
The ATP-binding cassette transporter ABCG2 mediates the efflux of several chemotherapeutic drugs, contributing to the development of multidrug resistance (MDR) in many cancers. The most promising strategy to overcome ABCG2-mediated MDR is the use of specific inhibitors. Despite many efforts, the identification of new potent and specific ABCG2 inhibitors remains urgent. In this study, a structural optimization of indeno[1,2-b]indole was performed and a new generation of 18 compounds was synthesized and tested as ABCG2 inhibitors. Most compounds showed ABCG2 inhibition with IC50 values below 0.5 µM. The ratio between cytotoxicity (IG50) and ABCG2 inhibition potency (IC50) was used to identify the best inhibitors. In addition, it was observed that some indeno[1,2-b]indole derivatives produced complete inhibition, while others only partially inhibited the transport function of ABCG2. All indeno[1,2-b]indole derivatives are not transported by ABCG2, and even the partial inhibitors are able to fully chemosensitize cancer cells overexpressing ABCG2. The high affinity of these indeno[1,2-b]indole derivatives was confirmed by the strong stimulatory effect on ABCG2 ATPase activity. These compounds did not affect the binding of conformation-sensitive antibody 5D3 binding, but stabilized the protein structure, as revealed by the thermostabilization assay. Finally, a docking study showed the indeno[1,2-b]indole derivatives share the same binding site as the substrate estrone-3-sulfate.
Collapse
Affiliation(s)
- Diogo Henrique Kita
- Pharmaceutical Sciences Graduate Program, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, 80210-170, Brazil.,Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nathalie Guragossian
- EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie - ISPB, SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, Université Claude Bernard Lyon 1, Univ Lyon, 69373, Lyon, France
| | - Ingrid Fatima Zattoni
- Pharmaceutical Sciences Graduate Program, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, 80210-170, Brazil
| | - Vivian Rotuno Moure
- Pharmaceutical Sciences Graduate Program, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, 80210-170, Brazil.,Department of Clinical Analysis, Federal University of Parana, Curitiba, PR, 80210-170, Brazil
| | | | - Sabrina Lusvarghi
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas Moulenat
- EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie - ISPB, SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, Université Claude Bernard Lyon 1, Univ Lyon, 69373, Lyon, France
| | - Billel Belhani
- Laboratory of Applied Organic Chemistry, Synthesis of Biomolecules and Molecular Modelling Group, Badji-Mokhtar-Annaba University, Box 12, 23000, Annaba, Algeria
| | - Geraldo Picheth
- Department of Clinical Analysis, Federal University of Parana, Curitiba, PR, 80210-170, Brazil
| | - Sofiane Bouacida
- Département Sciences de la Matière, Faculté des Sciences exactes et Sciences de la nature et de la vie, Université Larbi Ben M'hidi, Oum El Bouaghi, Algeria.,Research Unit for Chemistry of the Environment and Molecular Structural, University of Constantine 1, Constantine, Algeria
| | - Zouhair Bouaziz
- EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie - ISPB, SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, Université Claude Bernard Lyon 1, Univ Lyon, 69373, Lyon, France
| | - Christelle Marminon
- EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie - ISPB, SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, Université Claude Bernard Lyon 1, Univ Lyon, 69373, Lyon, France.,Small Molecules for Biological Targets Team, Centre de recherche en cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, Univ Lyon, 69373, Lyon, France
| | - Malika Berredjem
- Laboratory of Applied Organic Chemistry, Synthesis of Biomolecules and Molecular Modelling Group, Badji-Mokhtar-Annaba University, Box 12, 23000, Annaba, Algeria
| | - Joachim Jose
- Institut für Pharmazeutische und Medizinische Chemie, PharmaCampus, Westfälische Wilhelms-Universität Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Marcos Brown Gonçalves
- Department of Physics, Federal Technological University of Paraná, Curitiba, PR, 80230-901, Brazil
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Glaucio Valdameri
- Pharmaceutical Sciences Graduate Program, Laboratory of Cancer Drug Resistance, Federal University of Parana, Curitiba, PR, 80210-170, Brazil. .,Department of Clinical Analysis, Federal University of Parana, Curitiba, PR, 80210-170, Brazil.
| | - Marc Le Borgne
- EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie - ISPB, SFR Santé Lyon-Est CNRS UMS3453 - INSERM US7, Université Claude Bernard Lyon 1, Univ Lyon, 69373, Lyon, France. .,Small Molecules for Biological Targets Team, Centre de recherche en cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, Univ Lyon, 69373, Lyon, France.
| |
Collapse
|
22
|
Silbermann K, Li J, Namasivayam V, Stefan SM, Wiese M. Rational drug design of 6-substituted 4-anilino-2-phenylpyrimidines for exploration of novel ABCG2 binding site. Eur J Med Chem 2020; 212:113045. [PMID: 33454462 DOI: 10.1016/j.ejmech.2020.113045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 01/24/2023]
Abstract
In the search for novel, highly potent, and nontoxic adjuvant chemotherapeutics to resolve the major issue of ABC transporter-mediated multidrug resistance (MDR), pyrimidines were discovered as a promising compound class of modern ABCG2 inhibitors. As ABCG2-mediated MDR is a major obstacle in leukemia, pancreatic carcinoma, and breast cancer chemotherapy, adjuvant chemotherapeutics are highly desired for future clinical oncology. Very recently, docking studies of one of the most potent reversers of ABCG2-mediated MDR were reported and revealed a putative second binding pocket of ABCG2. Based on this (sub)pocket, a series of 16 differently 6-substituted 4-anilino-2-phenylpyrimidines was designed and synthesized to explore the potential increase in inhibitory activity of these ABCG2 inhibitors. The compounds were assessed for their influence on the ABCG2-mediated pheophorbide A transport, as well as the ABCB1- and ABCC1-mediated transport of calcein AM. They were additionally evaluated in MDR reversal assays to determine their half-maximal reversal concentration (EC50). The 6-substitution did not only show increased toxicity against ABCG2-overexpressing cells in combination with SN-38 but also a negative influence on cell viability in general. Nevertheless, several candidates had EC50 values in the low double-digit nanomolar concentration range, qualifying them as some of the most potent reversers of ABCG2-mediated MDR. In addition, five novel multitarget ABCB1, ABCC1, and ABCG2 inhibitors were discovered, four of them exerting their inhibitory power against the three stated transporters at least in the single-digit micromolar concentration range.
Collapse
Affiliation(s)
- Katja Silbermann
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Jiyang Li
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Vigneshwaran Namasivayam
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Sven Marcel Stefan
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University of Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| | - Michael Wiese
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University of Bonn, An der Immenburg 4, 53121, Bonn, Germany.
| |
Collapse
|
23
|
Malasala S, Ahmad MN, Akunuri R, Shukla M, Kaul G, Dasgupta A, Madhavi YV, Chopra S, Nanduri S. Synthesis and evaluation of new quinazoline-benzimidazole hybrids as potent anti-microbial agents against multidrug resistant Staphylococcus aureus and Mycobacterium tuberculosis. Eur J Med Chem 2020; 212:112996. [PMID: 33190958 DOI: 10.1016/j.ejmech.2020.112996] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/12/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022]
Abstract
Owing to the rapid rise in antibiotic resistance, infectious diseases have become serious threat to public health. There is an urgent need to develop new antimicrobial agents with diverse chemical structures and novel mechanisms of action to overcome the resistance. In recent years, Quinazoline-benzimidazole hybrids have emerged as a new class of antimicrobial agents active against S. aureus and M. tuberculosis. In the current study, we designed and synthesized fifteen new Quinazoline-benzimidazole hybrids and evaluated them for their antimicrobial activity against S. aureus ATCC 29213 and M. tuberculosis H37Rv. These studies led to the identification of nine potent antibacterial agents 8a, 8b, 8c, 8d, 8f, 8g, 8h, 8i and 10c with MICs in the range of 4-64 μg/mL. Further, these selected compounds were found to possess potent antibacterial potential against a panel of drug-resistant clinical isolates which include methicillin and vancomycin-resistant S. aureus. The selected compounds were found to be less toxic to Vero cells (CC50 = 40-≥200 μg/mL) and demonstrated a favourable selectivity index. Based on the encouraging results obtained these new benzimidazol-2-yl quinazoline derivatives have emerged as promising antimicrobial agents for the treatment of MDR- S. aureus and Mycobacterial infections.
Collapse
Affiliation(s)
- Satyaveni Malasala
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, Telangana, India
| | - Md Naiyaz Ahmad
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, 226031, Uttar Pradesh, India; AcSIR, Ghaziabad, Sector 19, Kamla Nehru Nagar, Ghaziabad, 201002, Uttar Pradesh, India
| | - Ravikumar Akunuri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, Telangana, India
| | - Manjulika Shukla
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, 226031, Uttar Pradesh, India
| | - Grace Kaul
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, 226031, Uttar Pradesh, India; AcSIR, Ghaziabad, Sector 19, Kamla Nehru Nagar, Ghaziabad, 201002, Uttar Pradesh, India
| | - Arunava Dasgupta
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, 226031, Uttar Pradesh, India; AcSIR, Ghaziabad, Sector 19, Kamla Nehru Nagar, Ghaziabad, 201002, Uttar Pradesh, India
| | - Y V Madhavi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, Telangana, India
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Janakipuram Extension, Lucknow, 226031, Uttar Pradesh, India; AcSIR, Ghaziabad, Sector 19, Kamla Nehru Nagar, Ghaziabad, 201002, Uttar Pradesh, India.
| | - Srinivas Nanduri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500 037, Telangana, India.
| |
Collapse
|
24
|
Malasala S, Ahmad MN, Gour J, Shukla M, Kaul G, Akhir A, Gatadi S, Madhavi Y, Chopra S, Nanduri S. Synthesis, biological evaluation and molecular modelling insights of 2-arylquinazoline benzamide derivatives as anti-tubercular agents. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
25
|
Bao Z, Zhou ZY, Mao YT, Shao LX. N-heterocyclic carbene-Pd(II)-1-methylimidazole complex-catalyzed Suzuki-Miyaura coupling of 2-chloro-4-aminoquinazolines with arylboronic acids. Tetrahedron 2020. [DOI: 10.1016/j.tet.2020.131548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Silbermann K, Li J, Namasivayam V, Baltes F, Bendas G, Stefan SM, Wiese M. Superior Pyrimidine Derivatives as Selective ABCG2 Inhibitors and Broad-Spectrum ABCB1, ABCC1, and ABCG2 Antagonists. J Med Chem 2020; 63:10412-10432. [PMID: 32787102 DOI: 10.1021/acs.jmedchem.0c00961] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the search for highly effective modulators addressing ABCG2-mediated MDR, 23 pyrimidines were synthesized and biologically assessed. Seven derivatives with (a) nitrogen- and/or halogen-containing residue(s) had extraordinary potencies against ABCG2 (IC50 < 150 nM). The compounds competitively inhibited ABCG2-mediated Hoechst 33342 transport but were not substrates of ABCG2. The most potent MDR reverser, compound 19, concentration-dependently increased SN-38-mediated cancer cell death at 11 nM (EC50), time-dependently doubled SN-38 toxicity in a period of 7 days at 10 nM, and half-maximally accelerated cell death combined with SN-38 at 17 nM. No induction of ABCG2 was observed. Furthermore, 11 pyrimidines were revealed as triple ABCB1/ABCC1/ABCG2 inhibitors. Five possessed IC50 values below 10 μM against each transporter, classifying them as some of the 50 most potent multitarget ABC transporter inhibitors. The most promising representative, compound 37, reversed ABCB1-, ABCC1-, and ABCG2-mediated MDR, making it one of the three most potent ABC transporter inhibitors and reversers of ABC transporters-mediated MDR.
Collapse
Affiliation(s)
- Katja Silbermann
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Jiyang Li
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Vigneshwaran Namasivayam
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Fabian Baltes
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Gerd Bendas
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Sven Marcel Stefan
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Michael Wiese
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
27
|
He ZX, Zhao TQ, Gong YP, Zhang X, Ma LY, Liu HM. Pyrimidine: A promising scaffold for optimization to develop the inhibitors of ABC transporters. Eur J Med Chem 2020; 200:112458. [PMID: 32497962 DOI: 10.1016/j.ejmech.2020.112458] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/30/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022]
Abstract
The multidrug resistance (MDR) phenomenon in cancer cells is the major obstacle leading to failure of chemotherapy accompanied by the feature of intractable and recurrence of cancers. As significant contributors that cause MDR, ABC superfamily proteins can transport the chemotherapeutic drugs out of the tumor cells by the energy of adenosine triphosphate (ATP) hydrolysis, thereby reducing their intracellular accumulation. The ABC transports like ABCB1, ABCC1 and ABCG2 have been extensively studied to develop modulators for overcoming MDR. To date, no reversal agents have been successfully marketed for clinical application, and little information about the ABC proteins bound to specific inhibitors is known, which make the design of MDR inhibitors with potency, selectivity and low toxicity a major challenge. In recent years, it has been increasingly recognized that pyrimidine-based derivatives have the potential for reversing ABC-mediated MDR. In this review, we summarized the pyrimidine-based inhibitors of ABC transporters, and mainly focused on their structure optimizations, development strategies and structure-activity relationship studies in hope of providing a reference for medicinal chemists to develop new modulators of MDR with highly potency and fewer side effects.
Collapse
Affiliation(s)
- Zhang-Xu He
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Tao-Qian Zhao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Yun-Peng Gong
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Xin Zhang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Li-Ying Ma
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, PR China.
| | - Hong-Min Liu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Key Laboratory of Technology of Drug Preparation (Zhengzhou University), Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, PR China.
| |
Collapse
|
28
|
Horsey AJ, Briggs DA, Holliday ND, Briddon SJ, Kerr ID. Application of fluorescence correlation spectroscopy to study substrate binding in styrene maleic acid lipid copolymer encapsulated ABCG2. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183218. [PMID: 32057756 PMCID: PMC7156912 DOI: 10.1016/j.bbamem.2020.183218] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/16/2022]
Abstract
ABCG2 is one of a trio of human ATP binding cassette transporters that have the ability to bind and transport a diverse array of chemical substrates out of cells. This so-called "multidrug" transport has numerous physiological consequences including effects on how drugs are absorbed into and eliminated from the body. Understanding how ABCG2 is able to interact with multiple drug substrates remains an important goal in transporter biology. Most drugs are believed to interact with ABCG2 through the hydrophobic lipid bilayer and experimental systems for ABCG2 study need to incorporate this. We have exploited styrene maleic acid to solubilise ABCG2 from HEK293T cells overexpressing the transporter, and confirmed by dynamic light scattering and fluorescence correlation spectroscopy (FCS) that this results in the extraction of SMA lipid copolymer (SMALP) particles that are uniform in size and contain a dimer of ABCG2, which is the predominant physiological state. FCS was further employed to measure the diffusion of a fluorescent ABCG2 substrate (BODIPY-prazosin) in the presence and absence of SMALP particles of purified ABCG2. Autocorrelation analysis of FCS traces enabled the mathematical separation of free BODIPY-prazosin from drug bound to ABCG2 and allowed us to show that combining SMALP extraction with FCS can be used to study specific drug: transporter interactions.
Collapse
Affiliation(s)
- Aaron J Horsey
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Deborah A Briggs
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Nicholas D Holliday
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Stephen J Briddon
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK; Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK.
| | - Ian D Kerr
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| |
Collapse
|
29
|
Li WJ, Chen XH, Zeng JC, Duan LL, Liu ZH, Sheng XH. Theoretical insight into the multiple interactions of quinazoline inhibitors with breast cancer resistance protein (BCRP/ABCG2). J Biomol Struct Dyn 2019; 38:4336-4343. [DOI: 10.1080/07391102.2019.1677503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Wen-Jing Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Xin-Hui Chen
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Jia-Cheng Zeng
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| | - Li-Li Duan
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Zhao-Hua Liu
- Center for New Drug Safety Evaluation, School of Pharmaceutical Sciences of Shandong University, Jinan, China
| | - Xie-Huang Sheng
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, China
| |
Collapse
|
30
|
Silbermann K, Shah CP, Sahu NU, Juvale K, Stefan SM, Kharkar PS, Wiese M. Novel chalcone and flavone derivatives as selective and dual inhibitors of the transport proteins ABCB1 and ABCG2. Eur J Med Chem 2019; 164:193-213. [PMID: 30594677 DOI: 10.1016/j.ejmech.2018.12.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/25/2018] [Accepted: 12/09/2018] [Indexed: 02/05/2023]
Abstract
During cancer chemotherapy, certain cancers may become cross-resistant to structurally diverse antineoplastic agents. This so-called multidrug resistance (MDR) is highly associated with the overexpression of ATP-binding cassette (ABC) transport proteins. These membrane-bound efflux pumps export a broad range of structurally diverse endo- and xenobiotics, including chemically unrelated anticancer agents. This translocation of drugs from the inside to the outside of cancer cells is mediated at the expense of ATP. In the last 40 years, three ABC transporters - ABCB1 (P-gp), ABCC1 (MRP1), and ABCG2 (BCRP) - have mainly been attributed to the occurrence of MDR in cancer cells. One of the strategies to overcome MDR is to inhibit the efflux transporter function by small-molecule inhibitors. In this work, we investigated new chalcone- and flavone-based compounds for selective as well as broad-spectrum inhibition of the stated transport proteins. These include substituted chalcones with variations at rings A and B, and flavones with acetamido linker at position 3. The synthesized molecules were evaluated for their inhibitory potential against ABCB1, ABCC1, and ABCG2 in calcein AM and pheophorbide A assays. In further investigations with the most promising candidates from each class, we proved that ABCB1- and ABCG2-mediated MDR could be reversed by the compounds. Moreover, their intrinsic toxicity was found to be negligible in most cases. Altogether, our findings contribute to the understanding of ABC transport proteins and reveal new compounds for ongoing evaluation in the field of ABC transporter-mediated MDR.
Collapse
Affiliation(s)
- Katja Silbermann
- Pharmaceutical Chemistry II, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Chetan P Shah
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Niteshkumar U Sahu
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Kapil Juvale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Sven Marcel Stefan
- Pharmaceutical Chemistry II, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Prashant S Kharkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| | - Michael Wiese
- Pharmaceutical Chemistry II, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University of Bonn, An der Immenburg 4, 53121 Bonn, Germany.
| |
Collapse
|