1
|
Arshad JZ, Hanif M. Hydroxypyrone derivatives in drug discovery: from chelation therapy to rational design of metalloenzyme inhibitors. RSC Med Chem 2022; 13:1127-1149. [PMID: 36325396 PMCID: PMC9579940 DOI: 10.1039/d2md00175f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/28/2022] [Indexed: 07/31/2023] Open
Abstract
The versatile structural motif of hydroxypyrone is found in natural products and can be easily converted into hydroxypyridone and hydroxythiopyridone analogues. The favourable toxicity profile and ease of functionalization to access a vast library of compounds make them an ideal structural scaffold for drug design and discovery. This versatile scaffold possesses excellent metal chelating properties that can be exploited for chelation therapy in clinics. Deferiprone [1,2-dimethyl-3-hydroxy-4(1H)-one] was the first orally active chelator to treat iron overload in thalassemia major. Metal complexes of hydroxy-(thio)pyr(id)ones have been investigated as magnetic resonance imaging contrast agents, and anticancer and antidiabetic agents. In recent years, this compound class has demonstrated potential in discovering and developing metalloenzyme inhibitors. This review article summarizes recent literature on hydroxy-(thio)pyr(id)ones as inhibitors for metalloenzymes such as histone deacetylases, tyrosinase and metallo-β-lactamase. Different approaches to the design of hydroxy-(thio)pyr(id)ones and their biological properties against selected metalloenzymes are discussed.
Collapse
Affiliation(s)
- Jahan Zaib Arshad
- Department of Chemistry, Government College Women University Sialkot Sialkot Pakistan
| | - Muhammad Hanif
- School of Chemical Sciences, University of Auckland Private Bag 92019 Auckland 1142 New Zealand (+64) 9 373 7599 ext. 87422
- MacDiarmid Institute for Advanced Materials and Nanotechnology Wellington New Zealand
| |
Collapse
|
2
|
Design concepts of half-sandwich organoruthenium anticancer agents based on bidentate bioactive ligands. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213950] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
3
|
Shakil MS, Parveen S, Rana Z, Walsh F, Movassaghi S, Söhnel T, Azam M, Shaheen MA, Jamieson SMF, Hanif M, Rosengren RJ, Hartinger CG. High Antiproliferative Activity of Hydroxythiopyridones over Hydroxypyridones and Their Organoruthenium Complexes. Biomedicines 2021; 9:biomedicines9020123. [PMID: 33513800 PMCID: PMC7912191 DOI: 10.3390/biomedicines9020123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Hydroxypyr(id)ones are a pharmaceutically important class of compounds that have shown potential in diverse areas of drug discovery. We investigated the 3-hydroxy-4-pyridones 1a-1c and 3-hydroxy-4-thiopyridones 1d-1f as well as their Ru(η6-p-cymene)Cl complexes 2a-2f, and report here the molecular structures of 1b and 1d as determined by X-ray diffraction analysis. Detailed cell biological investigations revealed potent cytotoxic activity, in particular of the 3-hydroxy-4-thiopyridones 1d-1f, while the Ru complexes of both compound types were less potent, despite still showing antiproliferative activity in the low μM range. The compounds did not modulate the cell cycle distribution of cancer cells but were cytostatic in A549 and cytotoxic in NCI-H522 non-small lung cancer cells, among other effects on cancer cells.
Collapse
Affiliation(s)
- Md. Salman Shakil
- Department of Pharmacology and Toxicology, University of Otago, PO Box 56, Dunedin 9016, New Zealand; (M.S.S.); (Z.R.); (M.A.)
| | - Shahida Parveen
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (S.P.); (F.W.); (S.M.); (T.S.)
- Department of Chemistry, University of Sargodha, Sargodha 40100, Pakistan;
| | - Zohaib Rana
- Department of Pharmacology and Toxicology, University of Otago, PO Box 56, Dunedin 9016, New Zealand; (M.S.S.); (Z.R.); (M.A.)
| | - Fearghal Walsh
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (S.P.); (F.W.); (S.M.); (T.S.)
| | - Sanam Movassaghi
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (S.P.); (F.W.); (S.M.); (T.S.)
| | - Tilo Söhnel
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (S.P.); (F.W.); (S.M.); (T.S.)
| | - Mayur Azam
- Department of Pharmacology and Toxicology, University of Otago, PO Box 56, Dunedin 9016, New Zealand; (M.S.S.); (Z.R.); (M.A.)
| | | | - Stephen M. F. Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand;
| | - Muhammad Hanif
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (S.P.); (F.W.); (S.M.); (T.S.)
- Correspondence: (M.H.); (R.J.R.); (C.G.H.)
| | - Rhonda J. Rosengren
- Department of Pharmacology and Toxicology, University of Otago, PO Box 56, Dunedin 9016, New Zealand; (M.S.S.); (Z.R.); (M.A.)
- Correspondence: (M.H.); (R.J.R.); (C.G.H.)
| | - Christian G. Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; (S.P.); (F.W.); (S.M.); (T.S.)
- Correspondence: (M.H.); (R.J.R.); (C.G.H.)
| |
Collapse
|
4
|
Lin L, Turner LD, Šilhár P, Pellett S, Johnson EA, Janda KD. Identification of 3-hydroxy-1,2-dimethylpyridine-4(1 H)-thione as a metal-binding motif for the inhibition of botulinum neurotoxin A. RSC Med Chem 2021; 12:137-143. [PMID: 34046606 PMCID: PMC8130615 DOI: 10.1039/d0md00320d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/27/2020] [Indexed: 12/17/2022] Open
Abstract
Botulinum neurotoxin serotype A (BoNT/A) is an important therapeutic target owing to its extremely potent nature, but also has potential use as a biowarfare agent. Currently, no therapeutic exists to reverse the long-lasting paralysis caused by BoNT/A. Herein, we describe the identification of 3-hydroxy-1,2-dimethylpyridine-4(1H)-thione (3,4-HOPTO) as a metal binding warhead for the inhibition of BoNT/A1. An initial screen of 96 metal binding fragments identified three derivatives containing the 3,4-HOPTO scaffold to inhibit the BoNT/A1 light chain (LC) at >95% at 1 mM. Additional screening of a 3,4-HOPTO sub-library identified structure-activity relationships (SARs) between N-substituted 3,4-HOPTO derivatives and the BoNT/A1 LC. Subsequent synthesis was conducted to improve on inhibitory potency - achieving low μM biochemical IC50 values. Representative compounds were evaluated in a cellular-based assay and showed promising μM activity.
Collapse
Affiliation(s)
- Lucy Lin
- Department of Chemistry and the Skaggs Institute for Chemical Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - Lewis D Turner
- Department of Chemistry and the Skaggs Institute for Chemical Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - Peter Šilhár
- Department of Chemistry and the Skaggs Institute for Chemical Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin 1550 Linden Drive Madison Wisconsin 53706 USA
| | - Eric A Johnson
- Department of Bacteriology, University of Wisconsin 1550 Linden Drive Madison Wisconsin 53706 USA
| | - Kim D Janda
- Department of Chemistry and the Skaggs Institute for Chemical Biology, The Scripps Research Institute La Jolla CA 92037 USA
| |
Collapse
|
5
|
Zhou L, Ma YC, Tang X, Li WY, Ma Y, Wang RL. Identification of the potential dual inhibitor of protein tyrosine phosphatase sigma and leukocyte common antigen-related phosphatase by virtual screen, molecular dynamic simulations and post-analysis. J Biomol Struct Dyn 2019; 39:45-62. [PMID: 31842717 DOI: 10.1080/07391102.2019.1705913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Owing to their inhibitory role in regulating oligodendrocyte differentiation and apoptosis, protein tyrosine phosphatase sigma (PTPσ) and leukocyte common antigen-related phosphatase (LAR) play a crucial potential role in treating spinal cord injury (SCI) disease. In this research, the computer aided drug design (CADD) methods were applied to discover the potential dual-target drug involving virtual screen, molecular docking and molecular dynamic simulation. Initially, the top 20 compounds with higher docking score than the positive controls (ZINC13749892, ZINC14516161) were virtually screened out from NCI and ZINC databases, and then were submitted in ADMET to predict their drug properties. Among these potential compounds, ZINC72417086 showed a higher docking score and satisfied Lipinski's rule of five. In addition, the post-analysis demonstrated that when ZINC72417086 bound to PTPσ and LAR, it could stable proteins conformations and destroy the residues interactions between P-loop and other loop regions in active pocket. Meanwhile, residue ARG1595 and ARG1528 could play a crucial role in in the inhibition of PTPσ and LAR, respectively. This research offered a novel approach for rapid discovery of dual-target leads compounds to treat SCI.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Liang Zhou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yang-Chun Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xue Tang
- Tasly Research Institute, Tasly Holding Group Co., Ltd, Tianjin, China
| | - Wei-Ya Li
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Run-Ling Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
6
|
Petriček S. Syntheses and crystal structures of metal (Mn, Co, Ni) chloride complexes with 3-hydroxypyridin-2-one and contribution of O H⋯Cl hydrogen bonds to their structural diversity. Polyhedron 2019. [DOI: 10.1016/j.poly.2019.03.050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
7
|
Wei-Ya L, Yu-Qing D, Yang-Chun M, Xin-Hua L, Ying M, Wang RL. Exploring the cause of the inhibitor 4AX attaching to binding site disrupting protein tyrosine phosphatase 4A1 trimerization by molecular dynamic simulation. J Biomol Struct Dyn 2019; 37:4840-4851. [PMID: 30661451 DOI: 10.1080/07391102.2019.1567392] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Ectopic overexpression of protein tyrosine phosphatase of liver regeneration-1 (PTP4A1, also called PRL-1) markedly enhanced hepatocellular carcinoma (HCC) cells migration and invasion. The PTP4A1 trimerization played a vital role in mediating cell proliferation and motility. Biochemical and structural studies have proved that the compound 4AX, a well-known inhibitor for PRL1, directly binds to the PTP4A1 trimer interface and obstructs trimer formation of PTP4A1. However, the molecular basis of the ligand-4AX inhibition on PTP4A1 trimer conformations remains unclear. In this study, the docking analysis and the molecular dynamics simulation (MD simulation) study were performed to investigate how the molecule binding at each interface disrupted the trimer formation. The results suggested that the ligand-4AX attaching to the binding site changed the conformation of A:Q131, A:Q135 in the AC interface, C:R18, C:P96 in the CA interface and B:Q131 in the BA interface, leading to the weak interactions between subunits and thus resulting in the disruption of the PTP4A1 trimerization.
Collapse
Affiliation(s)
- Li Wei-Ya
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University , Tianjin , China
| | - Duan Yu-Qing
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College , Tianjin , China
| | - Ma Yang-Chun
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University , Tianjin , China
| | - Lu Xin-Hua
- National Microbial Medicine Engineering & Research Center, Hebei Industry Microbial Metabolic Engineering & Technology Research Center, New Drug Research & Development Center of North China Pharmaceutical Group Corporation , Shijiazhuang , China
| | - Ma Ying
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University , Tianjin , China
| | - Run-Ling Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University , Tianjin , China
| |
Collapse
|
8
|
Chen AY, Adamek RN, Dick BL, Credille CV, Morrison CN, Cohen SM. Targeting Metalloenzymes for Therapeutic Intervention. Chem Rev 2019; 119:1323-1455. [PMID: 30192523 PMCID: PMC6405328 DOI: 10.1021/acs.chemrev.8b00201] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Metalloenzymes are central to a wide range of essential biological activities, including nucleic acid modification, protein degradation, and many others. The role of metalloenzymes in these processes also makes them central for the progression of many diseases and, as such, makes metalloenzymes attractive targets for therapeutic intervention. Increasing awareness of the role metalloenzymes play in disease and their importance as a class of targets has amplified interest in the development of new strategies to develop inhibitors and ultimately useful drugs. In this Review, we provide a broad overview of several drug discovery efforts focused on metalloenzymes and attempt to map out the current landscape of high-value metalloenzyme targets.
Collapse
Affiliation(s)
- Allie Y Chen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Rebecca N Adamek
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Benjamin L Dick
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Cy V Credille
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Christine N Morrison
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| | - Seth M Cohen
- Department of Chemistry and Biochemistry , University of California, San Diego , La Jolla , California 92093 , United States
| |
Collapse
|
9
|
Chen AY, Thomas PW, Stewart AC, Bergstrom A, Cheng Z, Miller C, Bethel CR, Marshall SH, Credille CV, Riley CL, Page RC, Bonomo RA, Crowder MW, Tierney DL, Fast W, Cohen SM. Dipicolinic Acid Derivatives as Inhibitors of New Delhi Metallo-β-lactamase-1. J Med Chem 2017; 60:7267-7283. [PMID: 28809565 PMCID: PMC5599375 DOI: 10.1021/acs.jmedchem.7b00407] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The efficacy of β-lactam antibiotics is threatened by the emergence and global spread of metallo-β-lactamase (MBL) mediated resistance, specifically New Delhi metallo-β-lactamase-1 (NDM-1). By utilization of fragment-based drug discovery (FBDD), a new class of inhibitors for NDM-1 and two related β-lactamases, IMP-1 and VIM-2, was identified. On the basis of 2,6-dipicolinic acid (DPA), several libraries were synthesized for structure-activity relationship (SAR) analysis. Inhibitor 36 (IC50 = 80 nM) was identified to be highly selective for MBLs when compared to other Zn(II) metalloenzymes. While DPA displayed a propensity to chelate metal ions from NDM-1, 36 formed a stable NDM-1:Zn(II):inhibitor ternary complex, as demonstrated by 1H NMR, electron paramagnetic resonance (EPR) spectroscopy, equilibrium dialysis, intrinsic tryptophan fluorescence emission, and UV-vis spectroscopy. When coadministered with 36 (at concentrations nontoxic to mammalian cells), the minimum inhibitory concentrations (MICs) of imipenem against clinical isolates of Eschericia coli and Klebsiella pneumoniae harboring NDM-1 were reduced to susceptible levels.
Collapse
Affiliation(s)
- Allie Y Chen
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | - Pei W Thomas
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, University of Texas , Austin, Texas 78712, United States
| | - Alesha C Stewart
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, University of Texas , Austin, Texas 78712, United States
| | - Alexander Bergstrom
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio 45056, United States
| | - Zishuo Cheng
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio 45056, United States
| | - Callie Miller
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio 45056, United States
| | - Christopher R Bethel
- Research Services, Louis Stokes Cleveland Department of Veterans Affairs Medical Center , Cleveland, Ohio 44106, United States
| | - Steven H Marshall
- Research Services, Louis Stokes Cleveland Department of Veterans Affairs Medical Center , Cleveland, Ohio 44106, United States
| | - Cy V Credille
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | - Christopher L Riley
- Department of Molecular Biosciences, University of Texas , Austin, Texas 78712, United States
| | - Richard C Page
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio 45056, United States
| | - Robert A Bonomo
- Research Services, Louis Stokes Cleveland Department of Veterans Affairs Medical Center , Cleveland, Ohio 44106, United States
- Department of Medicine, Department of Molecular Biology and Microbiology, Department of Biochemistry, and Department of Pharmacology, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Michael W Crowder
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio 45056, United States
| | - David L Tierney
- Department of Chemistry and Biochemistry, Miami University , Oxford, Ohio 45056, United States
| | - Walter Fast
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, University of Texas , Austin, Texas 78712, United States
| | - Seth M Cohen
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| |
Collapse
|
10
|
Hanif M, Meier SM, Adhireksan Z, Henke H, Martic S, Movassaghi S, Labib M, Kandioller W, Jamieson SMF, Hejl M, Jakupec MA, Kraatz HB, Davey CA, Keppler BK, Hartinger CG. Functionalization of Ruthenium(II)(η 6 -p-cymene)(3-hydroxy-2-pyridone) Complexes with (Thio)Morpholine: Synthesis and Bioanalytical Studies. Chempluschem 2017; 82:841-847. [PMID: 31961568 DOI: 10.1002/cplu.201700050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/16/2017] [Indexed: 02/06/2023]
Abstract
Hydroxypyr(id)ones constitute an emerging platform for the design of drug molecules, owing to their favorable biocompatibility and toxicity profiles. Herein, [RuII (η6 -p-cymene)] complexes with 3-hydroxy-2-pyridinone functionalized with morpholine and thiomorpholine, as a means often used in medicinal chemistry to alter the physicochemical properties of drug compounds, are reported. The compounds underwent hydrolysis of the Ru-Cl bond and the aqua species were stable for up to 48 h in aqueous solution, as observed by 1 H NMR spectroscopy and ESI-MS. The compounds formed adducts with amino acids and proteins through cleavage of the pyridinone ligand. Binding experiments to the nucleosome core particle by means of X-ray crystallography revealed similar reactivity and exclusive binding to histidine moieties of the histone proteins. Preliminary cyclin-dependent kinase 2 (CDK2)/cyclin A kinase inhibitory studies revealed promising activity similar to that of structurally related organometallic compounds.
Collapse
Affiliation(s)
- Muhammad Hanif
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria
| | - Samuel M Meier
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria.,Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 38, 1090, Vienna, Austria
| | - Zenita Adhireksan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Helena Henke
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria
| | - Sanela Martic
- Department of Chemistry, Oakland University, 2200 North Squirrel Road, Rochester, MI, 48309, USA
| | - Sanam Movassaghi
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Mahmoud Labib
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, M5S 3H2, Canada
| | - Wolfgang Kandioller
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria.,Research Platform "Translational Cancer Therapy Research", University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Michaela Hejl
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria
| | - Michael A Jakupec
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria.,Research Platform "Translational Cancer Therapy Research", University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria
| | - Heinz-Bernhard Kraatz
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada.,Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, M1C 1A4, Canada
| | - Curt A Davey
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.,NTU Institute of Structural Biology, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria.,Research Platform "Translational Cancer Therapy Research", University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria
| | - Christian G Hartinger
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, 1090, Vienna, Austria
| |
Collapse
|
11
|
Zhang X, Amin EA. Highly predictive support vector machine (SVM) models for anthrax toxin lethal factor (LF) inhibitors. J Mol Graph Model 2016; 63:22-8. [PMID: 26615468 PMCID: PMC4713341 DOI: 10.1016/j.jmgm.2015.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 10/07/2015] [Accepted: 11/06/2015] [Indexed: 01/08/2023]
Abstract
Anthrax is a highly lethal, acute infectious disease caused by the rod-shaped, Gram-positive bacterium Bacillus anthracis. The anthrax toxin lethal factor (LF), a zinc metalloprotease secreted by the bacilli, plays a key role in anthrax pathogenesis and is chiefly responsible for anthrax-related toxemia and host death, partly via inactivation of mitogen-activated protein kinase kinase (MAPKK) enzymes and consequent disruption of key cellular signaling pathways. Antibiotics such as fluoroquinolones are capable of clearing the bacilli but have no effect on LF-mediated toxemia; LF itself therefore remains the preferred target for toxin inactivation. However, currently no LF inhibitor is available on the market as a therapeutic, partly due to the insufficiency of existing LF inhibitor scaffolds in terms of efficacy, selectivity, and toxicity. In the current work, we present novel support vector machine (SVM) models with high prediction accuracy that are designed to rapidly identify potential novel, structurally diverse LF inhibitor chemical matter from compound libraries. These SVM models were trained and validated using 508 compounds with published LF biological activity data and 847 inactive compounds deposited in the Pub Chem BioAssay database. One model, M1, demonstrated particularly favorable selectivity toward highly active compounds by correctly predicting 39 (95.12%) out of 41 nanomolar-level LF inhibitors, 46 (93.88%) out of 49 inactives, and 844 (99.65%) out of 847 Pub Chem inactives in external, unbiased test sets. These models are expected to facilitate the prediction of LF inhibitory activity for existing molecules, as well as identification of novel potential LF inhibitors from large datasets.
Collapse
Affiliation(s)
- Xia Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, MN 55414-2959, United States
| | - Elizabeth Ambrose Amin
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware St. SE, Minneapolis, MN 55414-2959, United States; Minnesota Supercomputing Institute for Advanced Computational Research, 117 Pleasant St SE, Minneapolis, MN, United States.
| |
Collapse
|
12
|
Martin DP, Blachly PG, McCammon JA, Cohen SM. Exploring the influence of the protein environment on metal-binding pharmacophores. J Med Chem 2014; 57:7126-35. [PMID: 25116076 PMCID: PMC4148168 DOI: 10.1021/jm500984b] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The binding of a series of metal-binding pharmacophores (MBPs) related to the ligand 1-hydroxypyridine-2-(1H)-thione (1,2-HOPTO) in the active site of human carbonic anhydrase II (hCAII) has been investigated. The presence and/or position of a single methyl substituent drastically alters inhibitor potency and can result in coordination modes not observed in small-molecule model complexes. It is shown that this unexpected binding mode is the result of a steric clash between the methyl group and a highly ordered water network in the active site that is further stabilized by the formation of a hydrogen bond and favorable hydrophobic contacts. The affinity of MBPs is dependent on a large number of factors including donor atom identity, orientation, electrostatics, and van der Waals interactions. These results suggest that metal coordination by metalloenzyme inhibitors is a malleable interaction and that it is thus more appropriate to consider the metal-binding motif of these inhibitors as a pharmacophore rather than a "chelator". The rational design of inhibitors targeting metalloenzymes will benefit greatly from a deeper understanding of the interplay between the variety of forces governing the binding of MBPs to active site metal ions.
Collapse
Affiliation(s)
- David P Martin
- Departments of Chemistry and Biochemistry, ‡Pharmacology, and §Howard Hughes Medical Institute, University of California, San Diego , 9500 Gilman Drive, MC 0358, La Jolla, California 92093, United States
| | | | | | | |
Collapse
|
13
|
Williams JD, Khan AR, Cardinale SC, Butler MM, Bowlin TL, Peet NP. Small molecule inhibitors of anthrax lethal factor toxin. Bioorg Med Chem 2013; 22:419-34. [PMID: 24290062 DOI: 10.1016/j.bmc.2013.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/29/2013] [Accepted: 11/05/2013] [Indexed: 10/26/2022]
Abstract
This manuscript describes the preparation of new small molecule inhibitors of Bacillus anthracis lethal factor. Our starting point was the symmetrical, bis-quinolinyl compound 1 (NSC 12155). Optimization of one half of this molecule led to new LF inhibitors that were desymmetrized to afford more drug-like compounds.
Collapse
Affiliation(s)
- John D Williams
- Microbiotix, Inc., Department of Medicinal Chemistry, One Innovation Drive, Worcester, MA 01605, United States; Microbiotix, Inc., Department of Molecular Biology, One Innovation Drive, Worcester, MA 01605, United States
| | - Atiyya R Khan
- Microbiotix, Inc., Department of Medicinal Chemistry, One Innovation Drive, Worcester, MA 01605, United States; Microbiotix, Inc., Department of Molecular Biology, One Innovation Drive, Worcester, MA 01605, United States
| | - Steven C Cardinale
- Microbiotix, Inc., Department of Medicinal Chemistry, One Innovation Drive, Worcester, MA 01605, United States; Microbiotix, Inc., Department of Molecular Biology, One Innovation Drive, Worcester, MA 01605, United States
| | - Michelle M Butler
- Microbiotix, Inc., Department of Medicinal Chemistry, One Innovation Drive, Worcester, MA 01605, United States; Microbiotix, Inc., Department of Molecular Biology, One Innovation Drive, Worcester, MA 01605, United States
| | - Terry L Bowlin
- Microbiotix, Inc., Department of Medicinal Chemistry, One Innovation Drive, Worcester, MA 01605, United States; Microbiotix, Inc., Department of Molecular Biology, One Innovation Drive, Worcester, MA 01605, United States
| | - Norton P Peet
- Microbiotix, Inc., Department of Medicinal Chemistry, One Innovation Drive, Worcester, MA 01605, United States; Microbiotix, Inc., Department of Molecular Biology, One Innovation Drive, Worcester, MA 01605, United States.
| |
Collapse
|
14
|
Martin DP, Hann ZS, Cohen SM. Metalloprotein-inhibitor binding: human carbonic anhydrase II as a model for probing metal-ligand interactions in a metalloprotein active site. Inorg Chem 2013; 52:12207-15. [PMID: 23706138 DOI: 10.1021/ic400295f] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An ever-increasing number of metalloproteins are being discovered that play essential roles in physiological processes. Inhibitors of these proteins have significant potential for the treatment of human disease, but clinical success of these compounds has been limited. Herein, zinc(II)-dependent metalloprotein inhibitors in clinical use are reviewed, and the potential for using novel metal-binding groups (MBGs) in the design of these inhibitors is discussed. By using human carbonic anhydrase II as a model system, the nuances of MBG-metal interactions in the context of a protein environment can be probed. Understanding how metal coordination influences inhibitor binding may help in the design of new therapeutics targeting metalloproteins.
Collapse
Affiliation(s)
- David P Martin
- Department of Chemistry and Biochemistry, University of California, San Diego , La Jolla, California 92093, United States
| | | | | |
Collapse
|
15
|
Henke H, Kandioller W, Hanif M, Keppler BK, Hartinger CG. Organometallic ruthenium and osmium compounds of pyridin-2- and -4-ones as potential anticancer agents. Chem Biodivers 2013; 9:1718-27. [PMID: 22976964 DOI: 10.1002/cbdv.201200005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Organometallic Ru(II) compounds are among the most widely studied anticancer agents. Functionalizing metal centers with biomolecule-derived ligands has been shown to be a promising strategy to improve the antiproliferative activity of metal-based chemotherapeutics. Herein, the synthesis of a series of novel 3-hydroxypyridin-2-one-derived ligands and their M(II)(η(6)-p-cymene) half-sandwich complexes (M = Ru, Os) is described. The compounds were characterized by 1D- and 2D-NMR spectroscopy, and elemental analysis.
Collapse
Affiliation(s)
- Helena Henke
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, AT-1090 Vienna
| | | | | | | | | |
Collapse
|
16
|
Calugi C, Trabocchi A, Lalli C, Guarna A. d-Proline-based peptidomimetic inhibitors of anthrax lethal factor. Eur J Med Chem 2012; 56:96-107. [DOI: 10.1016/j.ejmech.2012.08.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 08/17/2012] [Accepted: 08/20/2012] [Indexed: 11/27/2022]
|
17
|
Garner AL, Struss AK, Fullagar JL, Agrawal A, Moreno AY, Cohen SM, Janda KD. 3-Hydroxy-1-alkyl-2-methylpyridine-4(1H)-thiones: Inhibition of the Pseudomonas aeruginosa Virulence Factor LasB. ACS Med Chem Lett 2012. [PMID: 23181168 DOI: 10.1021/ml300128f] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Bacterial resistance coupled to our current arsenal of antibiotics presents us with a growing threat to public health, thus warranting the exploration of alternative antibacterial strategies. In particular, the targeting of virulence factors has been regarded as a "second generation" antibiotic approach. In Pseudomonas aeruginosa, a Zn(2+) metalloprotease virulence factor, LasB or P. aeruginosa elastase, has been implicated in the development of P. aeruginosa-related keratitis, pneumonia and burn infection. Moreover, the enzyme also plays a critical role in swarming and biofilm formation, both of which are processes that have been linked to antibiotic resistance. To further validate the importance of LasB in P. aeruginosa infection, we describe our efforts toward the discovery of non-peptidic small molecule inhibitors of LasB. Using identified compounds, we have confirmed the role that LasB plays in P. aeruginosa swarming and demonstrate the potential for LasB-targeted small molecules in studying antimicrobial resistant P. aeruginosa phenotypes.
Collapse
Affiliation(s)
- Amanda L. Garner
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Anjali K. Struss
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Jessica L. Fullagar
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Arpita Agrawal
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Amira Y. Moreno
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| | - Seth M. Cohen
- Department of Chemistry and
Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, United States
| | - Kim D. Janda
- Departments of Chemistry and
Immunology and Microbial Science, The Skaggs Institute for Chemical
Biology, The Worm Institute for Research and Medicine, The Scripps Research Institute, 10550 North Torrey
Pines Road, La Jolla, California, United States
| |
Collapse
|
18
|
Chiu TL, Amin EA. Development of a comprehensive, validated pharmacophore hypothesis for anthrax toxin lethal factor (LF) inhibitors using genetic algorithms, Pareto scoring, and structural biology. J Chem Inf Model 2012; 52:1886-97. [PMID: 22697455 PMCID: PMC3477282 DOI: 10.1021/ci300121p] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Anthrax is an acute infectious disease caused by the spore-forming bacterium Bacillus anthracis. The anthrax toxin lethal factor (LF), an 89-kDa zinc hydrolase secreted by the bacilli, is the toxin component chiefly responsible for pathogenesis and has been a popular target for rational and structure-based drug design. Although hundreds of small-molecule compounds have been designed to target the LF active site, relatively few reported inhibitors have exhibited activity in cell-based assays, and no LF inhibitor is currently available to treat or prevent anthrax. This study presents a new pharmacophore map assembly, validated by experiment, designed to rapidly identify and prioritize promising LF inhibitor scaffolds from virtual compound libraries. The new hypothesis incorporates structural information from all five available LF enzyme-inhibitor complexes deposited in the Protein Data Bank (PDB) and is the first LF pharmacophore map reported to date that includes features representing interactions involving all three key subsites of the LF catalytic binding region. In a wide-ranging validation study on all 546 compounds for which published LF biological activity data exist, this model displayed strong selectivity toward nanomolar-level LF inhibitors, successfully identifying 72.1% of existing nanomolar-level compounds in an unbiased test set, while rejecting 100% of weakly active (>100 μM) compounds. In addition to its capabilities as a database searching tool, this comprehensive model points to a number of key design principles and previously unidentified ligand-receptor interactions that are likely to influence compound potency.
Collapse
Affiliation(s)
- Ting-Lan Chiu
- Department of Medicinal Chemistry and Minnesota Supercomputing Institute for Advanced Computational Research, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| | - Elizabeth A. Amin
- Department of Medicinal Chemistry and Minnesota Supercomputing Institute for Advanced Computational Research, University of Minnesota, 717 Delaware St. SE, Minneapolis, Minnesota 55414-2959
| |
Collapse
|
19
|
Dubois C, Haudecoeur R, Orio M, Belle C, Bochot C, Boumendjel A, Hardré R, Jamet H, Réglier M. Versatile effects of aurone structure on mushroom tyrosinase activity. Chembiochem 2012; 13:559-65. [PMID: 22307818 DOI: 10.1002/cbic.201100716] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Indexed: 11/09/2022]
Abstract
Elucidation of the binding modes of Ty inhibitors is an important step for in-depth studies on how to regulate tyrosinase activity. In this paper we highlight the extraordinarily versatile effects of the aurone structure on mushroom Ty activity. Depending on the position of the OH group on the B-ring, aurones can behave either as substrates or as hyperbolic activators. The synthesis of a hybrid aurone through combination of an aurone moiety with HOPNO (2-hydroxypyridine N-oxide), a good metal chelate, led us to a new, efficient, mixed inhibitor for mushroom tyrosinase. Another important feature pointed out by our study is the presence of more than one site for aurone compounds on mushroom tyrosinase. Because study of the binding of the hybrid aurone was difficult to perform with the enzyme, we undertook binding studies with tyrosinase functional models in order to elucidate the binding mode (chelating vs. bridging) on a dicopper(II) center. Use of EPR combined with theoretical DFT calculations allowed us to propose a preferred chelating mode for the interaction of the hybrid aurone with a dicopper(II) center.
Collapse
Affiliation(s)
- Carole Dubois
- Institut des Sciences Moléculaires de Marseille, Équipe BiosCiences, UMR-CNRS 7313, Aix-Marseille Université Avenue Escadrille Normandie-Niemen, 13397 Marseille Cedex 20, France
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
A series of HIV integrase (HIV-1 IN) inhibitors were synthesized to evaluate the role of the metal-binding group (MBG) in this class of metalloenzyme inhibitors. A total of 21 different raltegravir-chelator derivative (RCD) compounds were prepared that differed only in the nature of the MBG. These IN strand-transfer inhibitors (INSTIs) were evaluated in vitro in cell-free enzyme activity assays, and the in vitro results were further validated in cell culture experiments. All of the active compounds showed selective inhibition of the strand-transfer reaction over 3'-processing, suggesting a common mode of action with raltegravir. The results of the in vitro activity suggest that the nature of the MBG donor atoms, the overall MBG structure, and the specific arrangement of the MBG donor atom triad are essential for obtaining maximal HIV-1 IN inhibition. At least two compounds (RCD-4, RCD-5) containing a hydroxypyrone MBG were found to display superior strand-transfer inhibition when compared to an abbreviated analogue of raltegravir (RCD-1). By isolating and examining the role of the MBG in a series of INSTIs, we have identified a scaffold (hydroxypyrones) that may provide access to a unique class of HIV-1 IN inhibitors, and may help overcome rising raltegravir resistance.
Collapse
|
21
|
Schlesinger SR, Bruner B, Farmer PJ, Kim SK. Kinetic characterization of a slow-binding inhibitor of Bla2: thiomaltol. J Enzyme Inhib Med Chem 2012; 28:137-42. [DOI: 10.3109/14756366.2011.640632] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Britain Bruner
- Department of Chemistry and Biochemistry, Baylor University,
Waco, TX, USA
| | - Patrick J. Farmer
- Department of Chemistry and Biochemistry, Baylor University,
Waco, TX, USA
| | - Sung-Kun Kim
- Department of Chemistry and Biochemistry, Baylor University,
Waco, TX, USA
| |
Collapse
|
22
|
Jacobsen JA, Fullagar JL, Miller MT, Cohen SM. Identifying chelators for metalloprotein inhibitors using a fragment-based approach. J Med Chem 2010; 54:591-602. [PMID: 21189019 DOI: 10.1021/jm101266s] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fragment-based lead design (FBLD) has been used to identify new metal-binding groups for metalloenzyme inhibitors. When screened at 1 mM, a chelator fragment library (CFL-1.1) of 96 compounds produced hit rates ranging from 29% to 43% for five matrix metalloproteases (MMPs), 24% for anthrax lethal factor (LF), 49% for 5-lipoxygenase (5-LO), and 60% for tyrosinase (TY). The ligand efficiencies (LE) of the fragment hits are excellent, in the range of 0.4-0.8 kcal/mol. The MMP enzymes all generally elicit the same chelators as hits from CFL-1.1; however, the chelator fragments that inhibit structurally unrelated metalloenzymes (LF, 5-LO, TY) vary considerably. To develop more advanced hits, one hit from CFL-1.1, 8-hydroxyquinoline, was elaborated at four different positions around the ring system to generate new fragments. 8-Hydroxyquinoline fragments substituted at either the 5- or 7-positions gave potent hits against MMP-2, with IC(50) values in the low micromolar range. The 8-hydroxyquinoline represents a promising new chelator scaffold for the development of MMP inhibitors that was discovered by use of a metalloprotein-focused chelator fragment library.
Collapse
Affiliation(s)
- Jennifer A Jacobsen
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | | | | | | |
Collapse
|
23
|
Hanif M, Henke H, Meier SM, Martic S, Labib M, Kandioller W, Jakupec MA, Arion VB, Kraatz HB, Keppler BK, Hartinger CG. Is the reactivity of M(II)-arene complexes of 3-hydroxy-2(1H)-pyridones to biomolecules the anticancer activity determining parameter? Inorg Chem 2010; 49:7953-63. [PMID: 20704358 DOI: 10.1021/ic1009785] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Hydroxypyr(id)ones are versatile ligands for the synthesis of organometallic anticancer agents, equipping them with fine-tunable pharmacological properties. Herein, we report on the preparation, mode of action, and in vitro anticancer activity of Ru(II)- and Os(II)-arene complexes with alkoxycarbonylmethyl-3-hydroxy-2-pyridone ligands. The hydrolysis and binding to amino acids proceed quickly, as characterized by NMR spectroscopy and ESI mass spectrometry. However, the reaction with amino acids causes cleavage of the pyridone ligands from the metal center because the amino acids act as multidentate ligands. A similar behavior was also observed during the reactions with the model proteins ubiquitin and cytochrome c, yielding mainly [protein + M(eta(6)-p-cymene)] adducts (M = Ru, Os). Notably the ligand cleavage of the Os derivative was significantly slower than of its Ru analogue, which could explain its higher activity in in vitro anticancer assays. Furthermore, the reaction of the compounds to 5'-GMP was characterized and coordination to the N7 of the guanine moiety was demonstrated by (1)H NMR spectroscopy and X-ray diffraction analysis. CDK2/Cyclin A protein kinase inhibition studies revealed potent activity of the Ru and Os complexes.
Collapse
Affiliation(s)
- Muhammad Hanif
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Yuan H, Johnson SL, Chen LH, Wei J, Pellecchia M. A novel pharmacophore model for the design of anthrax lethal factor inhibitors. Chem Biol Drug Des 2010; 76:263-8. [PMID: 20572812 DOI: 10.1111/j.1747-0285.2010.01000.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study aims at the identification of novel structural features on the surface of the Zn-dependent metalloprotease lethal factor (LF) from anthrax onto which to design novel and selective inhibitors. We report that by targeting an unexplored region of LF that exhibits ligand-induced conformational changes, we could obtain inhibitors with at least 30-fold LF selectivity compared to two other most related human metalloproteases, MMP-2 and MMP-9. Based on these results, we propose a novel pharmacophore model that, together with the preliminarily identified compounds, should help the design of more potent and selective inhibitors against anthrax.
Collapse
Affiliation(s)
- Hongbin Yuan
- Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
25
|
Agrawal A, Johnson SL, Jacobsen JA, Miller MT, Chen LH, Pellecchia M, Cohen SM. Chelator fragment libraries for targeting metalloproteinases. ChemMedChem 2010; 5:195-9. [PMID: 20058293 DOI: 10.1002/cmdc.200900516] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Arpita Agrawal
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Hu M, Li J, Yao SQ. In situ "click" assembly of small molecule matrix metalloprotease inhibitors containing zinc-chelating groups. Org Lett 2009; 10:5529-31. [PMID: 19053720 DOI: 10.1021/ol802286g] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A panel of small molecule-based MMP inhibitors containing rhodanine warheads was assembled using "one-pot" click chemistry. Upon biological screening, moderate inhibitors were identified which specifically targets MMP-7 and MMP-13 over other MMPs.
Collapse
Affiliation(s)
- Mingyu Hu
- Department of Chemistry, NUS MedChem Program of the Office of Life Sciences, National University of Singapore, Singapore 117543
| | | | | |
Collapse
|
27
|
To bind zinc or not to bind zinc: an examination of innovative approaches to improved metalloproteinase inhibition. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1803:72-94. [PMID: 19712708 DOI: 10.1016/j.bbamcr.2009.08.006] [Citation(s) in RCA: 231] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 08/12/2009] [Accepted: 08/12/2009] [Indexed: 12/14/2022]
Abstract
This short review highlights some recent advances in matrix metalloproteinase inhibitor (MMPi) design and development. Three distinct approaches to improved MMP inhibition are discussed: (1) the identification and investigation of novel zinc-binding groups (ZBGs), (2) the study of non-zinc-binding MMPi, and (3) mechanism-based MMPi that form covalent adducts with the protein. Each of these strategies is discussed and their respective advantages and remaining challenges are highlighted. The studies discussed here bode well for the development of ever more selective, potent, and well-tolerated MMPi for treating several important disease pathologies.
Collapse
|
28
|
Kandioller W, Hartinger CG, Nazarov AA, Kuznetsov ML, John RO, Bartel C, Jakupec MA, Arion VB, Keppler BK. From Pyrone to Thiopyrone Ligands−Rendering Maltol-Derived Ruthenium(II)−Arene Complexes That Are Anticancer Active in Vitro. Organometallics 2009. [DOI: 10.1021/om900483t] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Wolfgang Kandioller
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
| | - Christian G. Hartinger
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
| | - Alexey A. Nazarov
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Maxim L. Kuznetsov
- Centro de Química Estrutural, Complexo I, Instituto Superior Técnico, Technical University of Lisbon, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Roland O. John
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
| | - Caroline Bartel
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
| | - Michael A. Jakupec
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
| | - Vladimir B. Arion
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- University of Vienna, Institute of Inorganic Chemistry, Waehringer Str. 42, A-1090 Vienna, Austria
| |
Collapse
|
29
|
Yan YL, Miller MT, Cao Y, Cohen SM. Synthesis of hydroxypyrone- and hydroxythiopyrone-based matrix metalloproteinase inhibitors: developing a structure-activity relationship. Bioorg Med Chem Lett 2009; 19:1970-6. [PMID: 19261472 PMCID: PMC2833267 DOI: 10.1016/j.bmcl.2009.02.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2009] [Accepted: 02/10/2009] [Indexed: 11/22/2022]
Abstract
The zinc(II)-dependent matrix metalloproteinases (MMPs) are associated with a variety of diseases. Development of inhibitors to modulate MMP activity has been an active area of investigation for therapeutic development. Hydroxypyrones and hydroxythiopyrones are alternative zinc-binding groups (ZBGs) that, when combined with peptidomimetic backbones, comprise a novel class of MMP inhibitors (MMPi). In this report, a series of hydroxypyrone- and hydroxythiopyrone-based MMPi with aryl backbones at the 2-, 5-, and 6-positions of the hydroxypyrone ring have been synthesized. Synthetic routes for developing inhibitors with substituents at two of these positions (so-called double-handed inhibitors) are also explored. The MMP inhibition profiles and structure-activity relationship of synthesized hydroxypyrones and hydroxythiopyrones have been analyzed. The results here show that the ZBG, the position of the backbone on the ZBG, and the nature of the linker between the ZBG and backbone are critical for MMPi activities.
Collapse
Affiliation(s)
- Yi-Long Yan
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, United States
| | - Melissa T. Miller
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, United States
| | - Yuchen Cao
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, United States
| | - Seth M. Cohen
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, United States
| |
Collapse
|
30
|
Agrawal A, de Oliveira CAF, Cheng Y, Jacobsen JA, McCammon JA, Cohen SM. Thioamide hydroxypyrothiones supersede amide hydroxypyrothiones in potency against anthrax lethal factor. J Med Chem 2009; 52:1063-74. [PMID: 19170530 PMCID: PMC2698031 DOI: 10.1021/jm8013212] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Anthrax lethal factor (LF) is a critical virulence factor in the pathogenesis of anthrax. A structure-activity relationship (SAR) of potential lethal factor inhibitors (LFi) is presented in which the zinc-binding group (ZBG), linker, and backbone moieties for a series of hydroxypyrone-based compounds were systematically varied. It was found that hydroxypyrothione ZBGs generate more potent inhibitors than hydroxypyrone ZBGs. Furthermore, coupling the hydroxypyrothione to a backbone group via a thioamide bond improves potency when compared to an amide linker. QM/MM studies show that the thioamide bond in these inhibitors allows for the formation of two additional hydrogen bonds with the protein active site. In both types of hydroxypyrothione compounds, ligand efficiencies of 0.29-0.54 kcal mol(-1) per heavy atom were achieved. The results highlight the need for a better understanding to optimize the interplay between the ZBG, linker, and backbone to get improved LFi.
Collapse
Affiliation(s)
- Arpita Agrawal
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093
| | - César Augusto F. de Oliveira
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093
- Howard Hughes Medical Institute, Center for Theoretical Biological Physics, Department of Chemistry and Biochemistry, Department of Pharmacology, University of California at San Diego, La Jolla, California 92093
| | - Yuhui Cheng
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093
- Howard Hughes Medical Institute, Center for Theoretical Biological Physics, Department of Chemistry and Biochemistry, Department of Pharmacology, University of California at San Diego, La Jolla, California 92093
| | - Jennifer A. Jacobsen
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093
- Howard Hughes Medical Institute, Center for Theoretical Biological Physics, Department of Chemistry and Biochemistry, Department of Pharmacology, University of California at San Diego, La Jolla, California 92093
| | - Seth M. Cohen
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093
| |
Collapse
|
31
|
Jacobsen FE, Lewis JA, Cohen SM. The design of inhibitors for medicinally relevant metalloproteins. ChemMedChem 2008; 2:152-71. [PMID: 17163561 DOI: 10.1002/cmdc.200600204] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A number of metalloproteins are important medicinal targets for conditions ranging from pathogenic infections to cancer. Many but not all of these metalloproteins contain a zinc(II) ion in the protein active site. Small-molecule inhibitors of these metalloproteins are designed to bind directly to the active site metal ions. In this review several metalloproteins of interest are discussed, including matrix metalloproteinases (MMPs), histone deacetylases (HDACs), anthrax lethal factor (LF), and others. Different strategies that have been employed to design effective inhibitors against these proteins are described, with an effort to highlight the strengths and drawbacks of each approach. An emphasis is placed on examining the bioinorganic chemistry of these metal active sites and how a better understanding of the coordination chemistry in these systems may lead to improved inhibitors. It is hoped that this review will help inspire medicinal, biological, and inorganic chemists to tackle this important problem by considering all aspects of metalloprotein inhibitor design.
Collapse
Affiliation(s)
- Faith E Jacobsen
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, USA
| | | | | |
Collapse
|
32
|
Yan YL, Cohen SM. Efficient synthesis of 5-amido-3-hydroxy-4-pyrones as inhibitors of matrix metalloproteinases. Org Lett 2007; 9:2517-20. [PMID: 17521196 PMCID: PMC2531216 DOI: 10.1021/ol0707665] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
3-hydroxy-4-pyrones are a class of important metal chelators with versatile medicinal applications. An efficient pathway for the preparation of new 5-amido-3-hydroxy-4-pyrone derivatives has been developed. The synthesized 5-amido-3-hydroxy-4-pyrones have been evaluated as inhibitors of matrix metalloproteinases.
Collapse
Affiliation(s)
- Yi-Long Yan
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0358
| | - Seth M. Cohen
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093-0358
| |
Collapse
|
33
|
Pelat T, Hust M, Laffly E, Condemine F, Bottex C, Vidal D, Lefranc MP, Dübel S, Thullier P. High-affinity, human antibody-like antibody fragment (single-chain variable fragment) neutralizing the lethal factor (LF) of Bacillus anthracis by inhibiting protective antigen-LF complex formation. Antimicrob Agents Chemother 2007; 51:2758-64. [PMID: 17517846 PMCID: PMC1932538 DOI: 10.1128/aac.01528-06] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The anthrax lethal toxin (LT) consists of two subunits, the protective antigen (PA) and the lethal factor (LF), and is essential for anthrax pathogenesis. Several recombinant antibodies directed against PA and intended for medical use have been obtained, but none against LF, despite the recommendations of anthrax experts. Here we describe an anti-LF single-chain variable fragment (scFv) that originated from an immunized macaque (Macaca fascicularis) and was obtained by phage display. Panning of the library of 1.8 x 10(8) clones allowed the isolation of 2LF, a high-affinity (equilibrium dissociation constant, 1.02 nM) scFv, which is highly neutralizing in the standardized in vitro assay (50% inhibitory concentration, 1.20 +/- 0.06 nM) and in an in vivo assay. The scFv neutralizes anthrax LT by inhibiting the formation of the LF-PA complex. The genes encoding 2LF are very similar to those of human immunoglobulin germ line genes, sharing substantial (84.2%) identity with their most similar, germinally encoded counterparts; this feature favors medical applications. These results, and others formerly published, demonstrate that our approach can generate antibody fragments suitable for prophylaxis and therapeutics.
Collapse
Affiliation(s)
- Thibaut Pelat
- Groupe de Biotechnologie des Anticorps, Département de Biologie des Agents Transmissibles, La Tronche, France
| | | | | | | | | | | | | | | | | |
Collapse
|