1
|
Sullivan H, Liang Y, Worthington K, Luo C, Gianneschi NC, Christman KL. Enzyme-Responsive Nanoparticles for the Targeted Delivery of an MMP Inhibitor to Acute Myocardial Infarction. Biomacromolecules 2023; 24:4695-4704. [PMID: 37695847 PMCID: PMC10646957 DOI: 10.1021/acs.biomac.3c00421] [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: 04/25/2023] [Revised: 08/21/2023] [Indexed: 09/13/2023]
Abstract
Herein, we have developed a drug-loaded matrix metalloproteinase (MMP)-responsive micellar nanoparticle (NP) intended for minimally invasive intravenous injection during the acute phase of myocardial infarction (MI) and prolonged retention in the heart for small-molecule drug delivery. Peptide-polymer amphiphiles (PPAs) bearing a small-molecule MMP inhibitor (MMPi), PD166793, were synthesized via ring-opening metathesis polymerization (ROMP) and formulated into spherical micelles by transitioning to aqueous solution. The resulting micellar NPs underwent MMP-induced aggregation, demonstrating enzyme responsiveness. Using a rat MI model, we observed that these NPs were capable of successfully extravasating into the infarcted region of the heart where they were retained due to the active, enzyme-mediated targeting, remaining detectable after 1 week post administration without increasing macrophage recruitment. Furthermore, in vitro studies show that these NPs demonstrated successful drug release following MMP treatment and maintained drug bioactivity as evidenced by comparable MMP inhibition to free MMPi. This work establishes a targeted NP platform for delivering small-molecule therapeutics to the heart after MI, opening possibilities for myocardial infarction treatment.
Collapse
Affiliation(s)
- Holly
L. Sullivan
- Shu
Chien-Gene Lay Department of Bioengineering and the Sanford Consortium
for Regenerative Medicine, University of
California San Diego, La Jolla, California 92093, United States
| | - Yifei Liang
- Department
of Chemistry, International Institute for Nanotechnology, Simpson-Querrey
Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Kendra Worthington
- Shu
Chien-Gene Lay Department of Bioengineering and the Sanford Consortium
for Regenerative Medicine, University of
California San Diego, La Jolla, California 92093, United States
| | - Colin Luo
- Shu
Chien-Gene Lay Department of Bioengineering and the Sanford Consortium
for Regenerative Medicine, University of
California San Diego, La Jolla, California 92093, United States
| | - Nathan C. Gianneschi
- Department
of Chemistry, International Institute for Nanotechnology, Simpson-Querrey
Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
- Departments
of Materials Science & Engineering, Biomedical Engineering and
Pharmacology, Northwestern University, Evanston, Illinois 60208, United States
- Department
of Chemistry & Biochemistry, University
of California San Diego, La Jolla, California 92093, United States
| | - Karen L. Christman
- Shu
Chien-Gene Lay Department of Bioengineering and the Sanford Consortium
for Regenerative Medicine, University of
California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
2
|
Paul Konken C, Beutel B, Schinor B, Song J, Gerwien H, Korpos E, Burmeister M, Riemann B, Schäfers M, Sorokin L, Haufe G. Influence of N-arylsulfonamido d-valine N-substituents on the selectivity and potency of matrix metalloproteinase inhibitors. Bioorg Med Chem 2023; 90:117350. [PMID: 37270903 DOI: 10.1016/j.bmc.2023.117350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/06/2023]
Abstract
To develop matrix metalloproteinase inhibitors (MMPIs) for both therapy and medicinal imaging by fluorescence-based techniques or positron-emission tomography (PET), a small library of eighteen N-substituted N-arylsulfonamido d-valines were synthesized and their potency to inhibit two gelatinases (MMP-2, and MMP-9), two collagenases (MMP-8, and MMP-13) and macrophage elastase (MMP-12) was determined in a Structure-Activity-Relation study with ({4-[3-(5-methylthiophen-2-yl)-1,2,4-oxadiazol-5-yl]phenyl}sulfonyl)-d-valine (1) as a lead. All compounds were shown to be more potent MMP-2/-9 inhibitors (nanomolar range) compared to other tested MMPs. This is a remarkable result considering that a carboxylic acid group is the zinc binding moiety. The compound with a terminal fluoropropyltriazole group at the furan ring (P1' substituent) was only four times less potent in inhibiting MMP-2 activity than the lead compound 1, making this compound a promising probe for PET application (after using a prosthetic group approach to introduce fluorine-18). Compounds with a TEG spacer and a terminal azide or even a fluorescein moiety at the sulfonylamide N atom (P2' substituent) were almost as active as the lead structure 1, making the latter derivative a suitable fluorescence imaging tool.
Collapse
Affiliation(s)
- Christian Paul Konken
- Organic Chemistry Institute, University of Münster, Corrensstraße 40, 48149 Münster, Germany; Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Bernd Beutel
- Organic Chemistry Institute, University of Münster, Corrensstraße 40, 48149 Münster, Germany
| | - Benjamin Schinor
- Organic Chemistry Institute, University of Münster, Corrensstraße 40, 48149 Münster, Germany
| | - Jian Song
- Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany; Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
| | - Hanna Gerwien
- Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany; Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
| | - Eva Korpos
- Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany; Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
| | - Miriam Burmeister
- Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany; Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
| | - Burkhard Riemann
- Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Michael Schäfers
- Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany; Department of Nuclear Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; European Institute for Molecular Imaging (EIMI), University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
| | - Lydia Sorokin
- Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany; Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149 Münster, Germany
| | - Günter Haufe
- Organic Chemistry Institute, University of Münster, Corrensstraße 40, 48149 Münster, Germany; Cells-in-Motion Interfaculty Centre (CiMIC), University of Münster, 48149 Münster, Germany.
| |
Collapse
|
3
|
Tabuse H, Abe-Sato K, Kanazawa H, Yashiro M, Tamura Y, Kamitani M, Hitaka K, Gunji E, Mitani A, Kojima N, Oka Y. Discovery of Highly Potent and Selective Matrix Metalloproteinase-7 Inhibitors by Hybridizing the S1' Subsite Binder with Short Peptides. J Med Chem 2022; 65:13253-13263. [PMID: 36137271 DOI: 10.1021/acs.jmedchem.2c01088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Matrix metalloproteinase-7 (MMP-7) has emerged as a protein playing important roles in both physiological and pathophysiological processes. Despite the growing interest in MMP-7 as a potential therapeutic target for diseases including cancer and fibrosis, potent and selective MMP-7 inhibitors have yet to be identified. Compound 1, previously reported by Edman and co-workers, binds to the S1' subsite of MMP-7, exhibiting moderate inhibitory activity and selectivity. To achieve both higher inhibitory activity and selectivity, we conceived hybridizing 1 with short peptides. The initially designed compound 6, which was a hybrid molecule between 1 and a tripeptide (Ala-Leu-Met) derived from an MMP-2-inhibitory peptide (APP-IP), showed enhanced MMP-7-inhibitory activity. Subsequent optimization of the peptide moiety led to the development of compound 18 with remarkable potency for MMP-7 and selectivity over other MMP subtypes.
Collapse
Affiliation(s)
- Hideaki Tabuse
- Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Kumi Abe-Sato
- Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Harumi Kanazawa
- Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Miyoko Yashiro
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Yunoshin Tamura
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Masafumi Kamitani
- Discovery Technologies Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Kosuke Hitaka
- Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Emi Gunji
- Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Akiko Mitani
- Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Naoki Kojima
- Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| | - Yusuke Oka
- Chemistry Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403, Yoshino-Cho, Kita-Ku, Saitama 331-9530, Japan
| |
Collapse
|
4
|
Kirchhain A, Zubrienė A, Kairys V, Vivaldi F, Bonini A, Biagini D, Santalucia D, Matulis D, Di Francesco F. Biphenyl substituted lysine derivatives as recognition elements for the matrix metalloproteinases MMP-2 and MMP-9. Bioorg Chem 2021; 115:105155. [PMID: 34303036 DOI: 10.1016/j.bioorg.2021.105155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/25/2022]
Abstract
Matrix metalloproteinases (MMPs) are an important factor in cancer progression and metastasis, especially gelatinases MMP-2 and MMP-9. A simple methodology for their detection and monitoring is highly desirable. Molecular probes have been very widely and successfully applied to study the activity of MMPs in cellular processes in vitro. We thus synthesized a small compound library of MMP-2 and MMP-9 binding probes based on drug molecules and endowed with free amine groups for the functionalization of transducer surfaces. In this study, we combined experimental results obtained by a kinetic fluorogenic peptide substrate cleavage assay with molecular modeling studies in order to assess the ability of the probe to bind to their target enzymes. The synthesized biphenyl substituted lysine derivatives showed IC50-values in the low nanomolar concentration range against MMP-2 (ligands 3a-d: 3 nM to 8 µM, ligands 4a-d: 45 nM to 350 µM) and low micromolar range against MMP-9 (ligands 3a-d: 350 nM to 60 µM, ligands 4a-d: 5 µM to 600 µM), with a selectivity up to more than 160-fold for MMP-2. The experimental results correlated well with molecular modelling with FleXAID and X-score functions. We showed that in our compound series, the side chain remained far away from the S1' cavity and the ligand for all the docked minima. Ligands 4a-d with their free amine group on the side chain may thus be bound to transducer surfaces for the fabrication of sensors, while retaining their activity against their target enzymes.
Collapse
Affiliation(s)
- Arno Kirchhain
- Dipartimento di Chimica e Chimica Industriale, Via Giuseppe Moruzzi, 13, 56124 Pisa, Italy.
| | - Asta Zubrienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Visvaldas Kairys
- Department of Bioinformatics, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Federico Vivaldi
- Dipartimento di Chimica e Chimica Industriale, Via Giuseppe Moruzzi, 13, 56124 Pisa, Italy
| | - Andrea Bonini
- Dipartimento di Chimica e Chimica Industriale, Via Giuseppe Moruzzi, 13, 56124 Pisa, Italy
| | - Denise Biagini
- Dipartimento di Chimica e Chimica Industriale, Via Giuseppe Moruzzi, 13, 56124 Pisa, Italy
| | - Delio Santalucia
- Dipartimento di Chimica e Chimica Industriale, Via Giuseppe Moruzzi, 13, 56124 Pisa, Italy
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Fabio Di Francesco
- Dipartimento di Chimica e Chimica Industriale, Via Giuseppe Moruzzi, 13, 56124 Pisa, Italy
| |
Collapse
|
5
|
Kamdem N, Roske Y, Kovalskyy D, Platonov M, Balinskyi O, Kreuchwig A, Saupe J, Fang L, Diehl A, Schmieder P, Krause G, Rademann J, Heinemann U, Birchmeier W, Oschkinat H. Small-molecule inhibitors of the PDZ domain of Dishevelled proteins interrupt Wnt signalling. MAGNETIC RESONANCE (GOTTINGEN, GERMANY) 2021; 2:355-374. [PMID: 37904770 PMCID: PMC10539800 DOI: 10.5194/mr-2-355-2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/28/2021] [Indexed: 11/01/2023]
Abstract
Dishevelled (Dvl) proteins are important regulators of the Wnt signalling pathway, interacting through their PDZ domains with the Wnt receptor Frizzled. Blocking the Dvl PDZ-Frizzled interaction represents a potential approach for cancer treatment, which stimulated the identification of small-molecule inhibitors, among them the anti-inflammatory drug Sulindac and Ky-02327. Aiming to develop tighter binding compounds without side effects, we investigated structure-activity relationships of sulfonamides. X-ray crystallography showed high complementarity of anthranilic acid derivatives in the GLGF loop cavity and space for ligand growth towards the PDZ surface. Our best binding compound inhibits Wnt signalling in a dose-dependent manner as demonstrated by TOP-GFP assays (IC50 ∼ 50 µ M ) and Western blotting of β -catenin levels. Real-time PCR showed reduction in the expression of Wnt-specific genes. Our compound interacted with Dvl-1 PDZ (KD = 2.4 µ M ) stronger than Ky-02327 and may be developed into a lead compound interfering with the Wnt pathway.
Collapse
Affiliation(s)
- Nestor Kamdem
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| | - Yvette Roske
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Dmytro Kovalskyy
- Enamine Ltd., Chervonotkatska Street 78, Kyiv 02094, Ukraine
- ChemBio Ctr, Taras Shevchenko National University of Kyiv, 62 Volodymyrska, Kyiv 01033, Ukraine
| | - Maxim O. Platonov
- Enamine Ltd., Chervonotkatska Street 78, Kyiv 02094, Ukraine
- ChemBio Ctr, Taras Shevchenko National University of Kyiv, 62 Volodymyrska, Kyiv 01033, Ukraine
| | - Oleksii Balinskyi
- Enamine Ltd., Chervonotkatska Street 78, Kyiv 02094, Ukraine
- ChemBio Ctr, Taras Shevchenko National University of Kyiv, 62 Volodymyrska, Kyiv 01033, Ukraine
| | - Annika Kreuchwig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| | - Jörn Saupe
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| | - Liang Fang
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Anne Diehl
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Peter Schmieder
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Gerd Krause
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Jörg Rademann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Institut für Pharmazie, Freie Universität Berlin, Königin-Luise-Straße 2 + 4, 14195 Berlin, Germany
| | - Udo Heinemann
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Walter Birchmeier
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| | - Hartmut Oschkinat
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| |
Collapse
|
6
|
Polymerizable Matrix Metalloproteinases' Inhibitors with Potential Application for Dental Restorations. Biomedicines 2021; 9:biomedicines9040366. [PMID: 33807479 PMCID: PMC8065691 DOI: 10.3390/biomedicines9040366] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
Collagen cleavage by matrix metalloproteinase (MMP) is considered a major cause of dental resins long term failure. Most MMP inhibitors display significant toxicity and are unsuitable for dental resins’ applications. Here we report a study of a new class of inhibitors that display the unique property of being co-polymerizable with other vinyl compounds present in commercial dental resins, limiting their release and potential toxicity. Computational affinity towards the active site of different MMP-1; -2; -8; -9 and -13 of several compounds showed interesting properties and were synthesized. These free compounds were tested concerning their toxicity upon contact with two different cell types, with no substantial decrease in cell viability at high concentrations. Even so, compound’s safety can be further improved upon copolymerization with commercial dental resins, limiting their release.
Collapse
|
7
|
Arylsulfonamides and selectivity of matrix metalloproteinase-2: An overview. Eur J Med Chem 2017; 129:72-109. [DOI: 10.1016/j.ejmech.2017.02.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 02/02/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022]
|
8
|
Wang Z, Xu W, Song T, Guo Z, Liu L, Fan Y, Wang A, Zhang Z. Fragment-Based Design, Synthesis, and Biological Evaluation of 1-Substituted-indole-2-carboxylic Acids as Selective Mcl-1 Inhibitors. Arch Pharm (Weinheim) 2016; 350. [PMID: 27911011 DOI: 10.1002/ardp.201600251] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 11/10/2022]
Abstract
Based on a known selective Mcl-1 inhibitor, 6-chloro-3-(3-(4-chloro-3,5-dimethylphenoxy)propyl)-1H-indole-2-carboxylic acid, we applied a fragment-based approach to obtain new molecules that extended into the p1 pocket of the BH3 groove and then exhibited binding selectivity for the Mcl-1 over the Bcl-2 protein. After we deconstructed the 1H-indole-2-carboxylic acid from the parental molecule, a benzenesulfonyl was substituted at the 1-position to adopt a geometry preferred for accessing the p1 pocket according to the binding mode of the parental molecule identified by X-ray crystallography. A linear relationship between the free energy of ligand binding (ΔG) and the count of non-hydrogen heavy atoms (HAC) was maintained during the molecular growing to occupy the p1 pocket. Finally, we not only obtained compound 12 with a 7.5-fold selectivity to Mcl-1 (Ki = 0.48 µM by fluorescence polarization) over Bcl-2 (Ki = 3.6 µM), but also provided evidence that additional occupation of the p1 pocket is more favorable for Mcl-1 than for Bcl-2 binding, and contributes more to Mcl-1 inhibition than occupation of the p2 pocket. Compound 12 exhibited a selective killing ability on Mcl-1-dependent cancer cells.
Collapse
Affiliation(s)
- Ziqian Wang
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, People's Republic of China
| | - Wenjie Xu
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, People's Republic of China
| | - Ting Song
- School of Life Science and Technology, Dalian University of Technology, Dalian, People's Republic of China
| | - Zongwei Guo
- School of Life Science and Technology, Dalian University of Technology, Dalian, People's Republic of China
| | - Lu Liu
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, People's Republic of China
| | - Yudan Fan
- School of Life Science and Technology, Dalian University of Technology, Dalian, People's Republic of China
| | - Anhui Wang
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, People's Republic of China
| | - Zhichao Zhang
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, Dalian, People's Republic of China
| |
Collapse
|
9
|
Nara H, Sato K, Kaieda A, Oki H, Kuno H, Santou T, Kanzaki N, Terauchi J, Uchikawa O, Kori M. Design, synthesis, and biological activity of novel, potent, and highly selective fused pyrimidine-2-carboxamide-4-one-based matrix metalloproteinase (MMP)-13 zinc-binding inhibitors. Bioorg Med Chem 2016; 24:6149-6165. [PMID: 27825552 DOI: 10.1016/j.bmc.2016.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/03/2016] [Accepted: 09/06/2016] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinase-13 (MMP-13), a member of the collagenase family of enzymes, has been implicated to play a key role in the pathology of osteoarthritis. Recently, we have reported the discovery of a series of quinazoline-2-carboxamide based non-zinc-binding MMP-13 selective inhibitors, as exemplified by compound 1. We then continued our research of a novel class of zinc-binding inhibitors to obtain follow-up compounds with different physicochemical, pharmacokinetic, and biological activity profiles. In order to design selective MMP-13 inhibitors, we adopted a strategy of connecting a zinc-binding group with the quinazoline-2-carboxamide system, a unique S1' binder, by an appropriate linker. Among synthesized compounds, a triazolone inhibitor 35 exhibited excellent potency (IC50=0.071nM) and selectivity (greater than 170-fold) over other MMPs (MMP-1, 2, 3, 7, 8, 9, 10, 12, and 14) and tumor necrosis factor-α converting enzyme (TACE). In this article, the design, synthesis, and biological activity of novel zinc-binding MMP-13 inhibitors are described.
Collapse
Affiliation(s)
- Hiroshi Nara
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kenjiro Sato
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Akira Kaieda
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hideyuki Oki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Haruhiko Kuno
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takashi Santou
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Naoyuki Kanzaki
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Jun Terauchi
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Osamu Uchikawa
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Masakuni Kori
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
10
|
Lorenzatti Hiles G, Bucheit A, Rubin JR, Hayward A, Cates AL, Day KC, El-Sawy L, Kunju LP, Daignault S, Lee CT, Liebert M, Hussain M, Day ML. ADAM15 Is Functionally Associated with the Metastatic Progression of Human Bladder Cancer. PLoS One 2016; 11:e0150138. [PMID: 26930657 PMCID: PMC4773041 DOI: 10.1371/journal.pone.0150138] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 02/09/2016] [Indexed: 12/21/2022] Open
Abstract
ADAM15 is a member of a family of catalytically active disintegrin membrane metalloproteinases that function as molecular signaling switches, shed membrane bound growth factors and/or cleave and inactivate cell adhesion molecules. Aberrant metalloproteinase function of ADAM15 may contribute to tumor progression through the release of growth factors or disruption of cell adhesion. In this study, we utilized human bladder cancer tissues and cell lines to evaluate the expression and function of ADAM15 in the progression of human bladder cancer. Examination of genome and transcriptome databases revealed that ADAM15 ranked in the top 5% of amplified genes and its mRNA was significantly overexpressed in invasive and metastatic bladder cancer compared to noninvasive disease. Immunostaining of a bladder tumor tissue array designed to evaluate disease progression revealed increased ADAM15 immunoreactivity associated with increasing cancer stage and exhibited significantly stronger staining in metastatic samples. About half of the invasive tumors and the majority of the metastatic cases exhibited high ADAM15 staining index, while all low grade and noninvasive cases exhibited negative or low staining. The knockdown of ADAM15 mRNA expression significantly inhibited bladder tumor cell migration and reduced the invasive capacity of bladder tumor cells through MatrigelTM and monolayers of vascular endothelium. The knockdown of ADAM15 in a human xenograft model of bladder cancer inhibited tumor growth by 45% compared to controls. Structural modeling of the catalytic domain led to the design of a novel ADAM15-specific sulfonamide inhibitor that demonstrated bioactivity and significantly reduced the viability of bladder cancer cells in vitro and in human bladder cancer xenografts. Taken together, the results revealed an undescribed role of ADAM15 in the invasion of human bladder cancer and suggested that the ADAM15 catalytic domain may represent a viable therapeutic target in patients with advanced disease.
Collapse
Affiliation(s)
- Guadalupe Lorenzatti Hiles
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Amanda Bucheit
- Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John R. Rubin
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Alexandra Hayward
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
- School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Angelica L. Cates
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kathleen C. Day
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Layla El-Sawy
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
- European Egyptian Pharmaceuticals, Alexandria, Egypt
| | - L. Priya Kunju
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Stephanie Daignault
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Cheryl T. Lee
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Monica Liebert
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Maha Hussain
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- School of Public Health, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mark L. Day
- Division of Urologic Oncology, Department of Urology, University of Michigan, Ann Arbor, Michigan, United States of America
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
11
|
Gupta SP. QSAR Studies on Hydroxamic Acids: A Fascinating Family of Chemicals with a Wide Spectrum of Activities. Chem Rev 2015; 115:6427-90. [DOI: 10.1021/cr500483r] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Satya P. Gupta
- Department of Applied Sciences, National Institute of Technical Teachers’ Training and Research, Shamla
Hills, Bhopal-462002, India
| |
Collapse
|
12
|
Kalva S, Vinod D, Saleena LM. Combined structure- and ligand-based pharmacophore modeling and molecular dynamics simulation studies to identify selective inhibitors of MMP-8. J Mol Model 2014; 20:2191. [DOI: 10.1007/s00894-014-2191-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 02/24/2014] [Indexed: 10/25/2022]
|
13
|
Burtea A, Salzameda NT. Discovery and SAR study of a sulfonamide hydroxamic acid inhibitor for the botulinum neurotoxin serotype A light chain. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00053f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Through the use of chemical synthesis and high throughput screening, we discovered a sulfonamide hydroxamic acid inhibitor for the botulinum neurotoxin serotype A light chain. A structure activity relationship study of the parent inhibitor resulted in the synthesis of a new inhibitor with an IC50of 0.95 ± 0.60 μM for the BoNT/A LC.
Collapse
Affiliation(s)
- Alexander Burtea
- Department of Chemistry & Biochemistry
- California State University
- Fullerton, USA
| | | |
Collapse
|
14
|
Atobe M, Maekawara N, Kawanishi M, Suzuki H, Tanaka E, Miyoshi S. Design, synthesis and SAR investigation of thienosultam derivatives as ADAMTS-5 (aggrecanase-2) inhibitors. Bioorg Med Chem Lett 2013; 23:2111-6. [DOI: 10.1016/j.bmcl.2013.01.120] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 01/24/2013] [Accepted: 01/28/2013] [Indexed: 11/28/2022]
|
15
|
SAVINOV ALEXEIY, STRONGIN ALEXY. Targeting the T-cell membrane type-1 matrix metalloproteinase-CD44 axis in a transferred type 1 diabetes model in NOD mice. Exp Ther Med 2013; 5:438-442. [PMID: 23403478 PMCID: PMC3570120 DOI: 10.3892/etm.2012.821] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 10/25/2012] [Indexed: 12/25/2022] Open
Abstract
This study tested the hypothesis that membrane-tethered type-1 matrix metalloproteinase (MT1-MMP)-induced proteolysis of T cell CD44 is important for defining the migration and function of autoreactive T cells, including diabetogenic, insulin-specific and K(d)-restricted IS-CD8(+) cells. To confirm the importance of MT1-MMP proteolysis of CD44 in type 1 diabetes (T1D), the anti-diabetic effects of three MMP inhibitors (3(S)-2,2-dimethyl-4[4-pyridin-4-yloxy-benzenesulfonyl]-thiomorpholine-3-carboxylic acid hydroxamate [AG3340], 2-(4-phenoxyphenylsulfonylmethyl) thiirane [SB-3CT] and epigallocatechin-3-gallate [EGCG]) were compared using an adoptive diabetes transfer model in non-obese diabetic (NOD) mice. Only AG3340 was capable of inhibiting both the activity of MT1-MMP and the shedding of CD44 in T cells; and the transendothelial migration and homing of IS-CD8(+) T cells into the pancreatic islets. SB-3CT and EGCG were incapable of inhibiting T cell MT1-MMP efficiently. As a result, AG3340 alone, but not SB-3CT or EGCG, delayed the onset of transferred diabetes in NOD mice. In summary, the results of the present study emphasize that the MT1-MMP-CD44 axis has a unique involvement in T1D development. Accordingly, we suggest that a potent small-molecule MT1-MMP antagonist is required for the design of novel therapies for T1D.
Collapse
Affiliation(s)
| | - ALEX Y. STRONGIN
- Sanford-Burnham Medical Research Institute, La Jolla, CA 92037,
USA
| |
Collapse
|
16
|
Development of triarylsulfonamides as novel anti-inflammatory agents. Bioorg Med Chem Lett 2013; 23:816-20. [DOI: 10.1016/j.bmcl.2012.11.067] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 11/16/2012] [Accepted: 11/18/2012] [Indexed: 11/24/2022]
|
17
|
Jain P, Saravanan C, Singh SK. Sulphonamides: Deserving class as MMP inhibitors? Eur J Med Chem 2012; 60:89-100. [PMID: 23287054 DOI: 10.1016/j.ejmech.2012.10.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 09/26/2012] [Accepted: 10/02/2012] [Indexed: 12/14/2022]
Abstract
The importance of sulphonamide moiety in medicinal chemistry cannot be ignored as it constitutes an important class of extensively used drugs. Recently, sulphonamides have also been reported for their matrix metalloproteinase (MMP) inhibitory activity. MMPs are calcium- and zinc-dependent endopeptidases, involved in both inter- and intra-cellular activity. This review documents the emergence of sulphonamides as matrix metalloproteinase inhibitors (MMPIs) from the first generation to the recent third generation MMPIs, their mode of action - how sulphonamides act on MMPs? as well as the structure activity relationship along with their therapeutic uses in chronic obstructive pulmonary disease (COPD), ulcer, asthma, arthritis and cancer. From this review, readers can get answer for the question- is sulphonamides a potential class of MMPIs?
Collapse
Affiliation(s)
- Pranjali Jain
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutics, Indian Institute of Technology (BHU), Varanasi 221005, India
| | | | | |
Collapse
|
18
|
Yoshida T, Hitaoka S, Mashima A, Sugimoto T, Matoba H, Chuman H. Combined QM/MM (ONIOM) and QSAR approach to the study of complex formation of matrix metalloproteinase‑9 with a series of biphenylsulfonamides−LERE-QSAR analysis (V). J Phys Chem B 2012; 116:10283-9. [PMID: 22845734 DOI: 10.1021/jp305476x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We previously proposed a novel QSAR (quantitative structure-activity relationship) procedure called LERE (linear expression by representative energy terms)-QSAR involving molecular calculations such as an ab initio fragment molecular orbital ones. In the present work, we applied LERE-QSAR to complex formation of matrix metalloproteinase-9 (MMP-9) with a series of substituted biphenylsulfonamides. The results shows that the overall free-energy change accompanying complex formation is due to predominantly the contribution from the electrostatic interaction with the zinc atom in the active site of MMP-9. Carbonic anhydrase (CA) belongs to the zinc-containing protease family. In contrast to the current case of MMP-9, the overall free-energy change during complex formation of CA with a series of benzenesulfonamides is due to the contributions from the solvation and dissociation free-energy changes, as previously reported. Comparison of the two sets of results indicates quantitative differences in the relative contributions of free-energy components to the overall free-energy change between the two data sets, corresponding with those in the respective classical QSAR equations. The LERE-QSAR procedure was demonstrated to quantitatively reveal differences in the binding mechanisms between the two cases involving similar but different zinc-containing proteins at the electronic and atomic levels.
Collapse
Affiliation(s)
- Tatsusada Yoshida
- Institute of Health Biosciences, The University of Tokushima Graduate School , 1-78 Shomachi, Tokushima 770-8505, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Wang M, Zhang J, Telljohann R, Jiang L, Wu J, Monticone RE, Kapoor K, Talan M, Lakatta EG. Chronic matrix metalloproteinase inhibition retards age-associated arterial proinflammation and increase in blood pressure. Hypertension 2012; 60:459-66. [PMID: 22689745 PMCID: PMC3537179 DOI: 10.1161/hypertensionaha.112.191270] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 05/09/2012] [Indexed: 12/21/2022]
Abstract
Age-associated arterial remodeling involves arterial wall collagen deposition and elastin fragmentation, as well as an increase in arterial pressure. This arterial remodeling is linked to proinflammatory signaling, including transforming growth factor-β1, monocyte chemoattractant protein 1, and proendothelin 1, activated by extracellular matrix metalloproteinases (MMPs) and orchestrated, in part, by the transcriptional factor ets-1. We tested the hypothesis that inhibition of MMP activation can decelerate the age-associated arterial proinflammation and its attendant increase in arterial pressure. Indeed, chronic administration of a broad-spectrum MMP inhibitor, PD166739, via a daily gavage, to 16-month-old rats for 8 months markedly blunted the expected age-associated increases in arterial pressure. This was accompanied by the following: (1) inhibition of the age-associated increases in aortic gelatinase and interstitial collagenase activity in situ; (2) preservation of the elastic fiber network integrity; (3) a reduction of collagen deposition; (4) a reduction of monocyte chemoattractant protein 1 and transforming growth factor-β1 activation; (5) a diminution in the activity of the profibrogenic signaling molecule SMAD-2/3 phosphorylation; (6) inhibition of proendothelin 1 activation; and (7) downregulation of expression of ets-1. Acute exposure of cultured vascular smooth muscle cells in vitro to proendothelin 1 increased both the transcription and translation of ets-1, and these effects were markedly reduced by MMP inhibition. Furthermore, infection of vascular smooth muscle cells with an adenovirus harboring a full-length ets-1 cDNA increased activities of both transforming growth factor-β1 and monocyte chemoattractant protein 1. Collectively, our results indicate that MMP inhibition retards age-associated arterial proinflammatory signaling, and this is accompanied by preservation of intact elastin fibers, a reduction in collagen, and blunting of an age-associated increase in blood pressure.
Collapse
Affiliation(s)
- Mingyi Wang
- Laboratory of Cardiovascular Science, Intramural Research Program, 5600 Nathan Shock Dr, National Institute on Aging-National Institutes of Health, Baltimore, MD 21030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Patil VM, Gupta SP. Quantitative structure-activity relationship studies on sulfonamide-based MMP inhibitors. EXPERIENTIA SUPPLEMENTUM (2012) 2012; 103:177-208. [PMID: 22642193 DOI: 10.1007/978-3-0348-0364-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Matrix metalloproteinases (MMPs) regulate a wide range of biological functions, but their overactivation leads to a wide array of disease processes such as rheumatoid arthritis, ostereoarthritis, tumor metastatis, multiple sclerosis, congestive heart failure, and a host of others. Therefore, the study of MMP inhibitors has evoked a great interest among scientists. As a result, different groups of compounds have been synthesized and studied for MMP inhibitions. Among them, a large number of structurally novel sulfonamide derivatives have been reported to be potential MMP inhibitors, but only a few have reached to the final stage of clinical trial. Many authors have made quantitative structure-activity relationship (QSAR) studies on them to provide the guidelines to design more potent MMP inhibitors. This article presents a comprehensive review on all such QSARs reported with critical assessment in order to provide a deeper insight into the structure-activity relationship of sulfonamides which can be used to synthesize highly potential drugs of pharmaceutical importance.
Collapse
Affiliation(s)
- Vaishali M Patil
- School of Pharmacy, Bharat Institute of Technology, Meerut 250103, India.
| | | |
Collapse
|
21
|
LI DAILIN, ZHENG QINGCHUAN, FANG XUEXUN, JI HAITAO, YANG JINGANG, ZHANG HONGXING. THEORETICAL STUDY ON POTENCY AND SELECTIVITY OF NOVEL NONPEPTIDE INHIBITORS OF MATRIX METALLOPROTEINASES MMP-2 AND MMP-9. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2011. [DOI: 10.1142/s0219633609004824] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Two novel matrix metalloproteinase (MMP) inhibitors, myricetin (m) and kaempferol (k), were found and the inhibitory activity is both in decreased order towards MMP-2 and MMP-9. To understand the mechanism during the processes when inhibitors bind to MMP-2 and MMP-9, molecular modeling, docking, and density functional theory (DFT) calculations were performed. The calculated results indicated that the hydroxyls on benzene ring of the inhibitors control the binding modes between inhibitors and MMPs, thus play an important role on the potency and selectivity. Besides coordinating with the N atoms of three His residues, Zn also interacts with a hydroxyl group of inhibitors by O – Zn distances of 2.66–2.78 Å in all of the docked complexes, so that the hydroxyl acts as a weak zinc binding group (ZBG). The DFT calculated results support the above analysis. The binding affinity calculations between inhibitors and MMPs present the total interaction energies in the m-MMP < k-MMP order and the solvation energy of myricetin is less than that of kaempferol, which reflect the experimental inhibitory activity.
Collapse
Affiliation(s)
- DAI-LIN LI
- State Key Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University, Changchun 130023, P. R. China
| | - QING-CHUAN ZHENG
- State Key Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University, Changchun 130023, P. R. China
| | - XUE-XUN FANG
- Key Laboratory for Molecular Enzymology and Enzyme, Engineering of Ministry of Education, Jilin University, Changchun 130023, P. R. China
| | - HAI-TAO JI
- Key Laboratory for Molecular Enzymology and Enzyme, Engineering of Ministry of Education, Jilin University, Changchun 130023, P. R. China
| | - JIN-GANG YANG
- Key Laboratory for Molecular Enzymology and Enzyme, Engineering of Ministry of Education, Jilin University, Changchun 130023, P. R. China
| | - HONG-XING ZHANG
- State Key Laboratory of Theoretical and Computational Chemistry, Institute of Theoretical Chemistry, Jilin University, Changchun 130023, P. R. China
| |
Collapse
|
22
|
Hu Y, Xing L, Thomason JR, Xiang J, Ipek M, Guler S, Li H, Sabatini J, Chockalingam P, Reifenberg E, Sheldon R, Morris EA, Georgiadis KE, Tam S. Continued exploration of biphenylsulfonamide scaffold as a platform for aggrecanase-1 inhibition. Bioorg Med Chem Lett 2011; 21:6800-3. [DOI: 10.1016/j.bmcl.2011.09.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 09/08/2011] [Accepted: 09/12/2011] [Indexed: 10/17/2022]
|
23
|
Discovery of biphenylketones as dual modulators of inflammation and bone loss. Bioorg Med Chem Lett 2010; 20:5548-51. [DOI: 10.1016/j.bmcl.2010.07.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Revised: 07/13/2010] [Accepted: 07/14/2010] [Indexed: 11/23/2022]
|
24
|
Nuti E, Casalini F, Avramova SI, Santamaria S, Fabbi M, Ferrini S, Marinelli L, La Pietra V, Limongelli V, Novellino E, Cercignani G, Orlandini E, Nencetti S, Rossello A. Potent Arylsulfonamide Inhibitors of Tumor Necrosis Factor-α Converting Enzyme Able to Reduce Activated Leukocyte Cell Adhesion Molecule Shedding in Cancer Cell Models. J Med Chem 2010; 53:2622-35. [DOI: 10.1021/jm901868z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Elisa Nuti
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Francesca Casalini
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Stanislava I. Avramova
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Salvatore Santamaria
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Marina Fabbi
- Istituto Nazionale per la Ricerca sul Cancro, Largo R. Benzi 10, 16132 Genova, Italy
| | - Silvano Ferrini
- Istituto Nazionale per la Ricerca sul Cancro, Largo R. Benzi 10, 16132 Genova, Italy
| | - Luciana Marinelli
- Dipartimento di Chimica Farmaceutica e Tossicologica, Università di Napoli “Federico II”, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Valeria La Pietra
- Dipartimento di Chimica Farmaceutica e Tossicologica, Università di Napoli “Federico II”, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Vittorio Limongelli
- Dipartimento di Chimica Farmaceutica e Tossicologica, Università di Napoli “Federico II”, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Ettore Novellino
- Dipartimento di Chimica Farmaceutica e Tossicologica, Università di Napoli “Federico II”, Via Domenico Montesano 49, 80131 Napoli, Italy
| | - Giovanni Cercignani
- Dipartimento di Biologia, Unità di Biochimica, Università di Pisa, Via San Zeno, 51, 56127 Pisa, Italy
| | - Elisabetta Orlandini
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Susanna Nencetti
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Armando Rossello
- Dipartimento di Scienze Farmaceutiche, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
25
|
Xi L, Du J, Li S, Li J, Liu H, Yao X. A combined molecular modeling study on gelatinases and their potent inhibitors. J Comput Chem 2010; 31:24-42. [DOI: 10.1002/jcc.21279] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
26
|
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: 228] [Impact Index Per Article: 14.3] [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
|
27
|
3,4-Disubstituted benzofuran P1′ MMP-13 inhibitors: Optimization of selectivity and reduction of protein binding. Bioorg Med Chem Lett 2009; 19:4546-50. [DOI: 10.1016/j.bmcl.2009.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 06/22/2009] [Accepted: 07/02/2009] [Indexed: 11/23/2022]
|
28
|
šille J, Remko M. Computational Study of the Sulfonylated Amino Acid Hydroxamates Binding to the Zinc Ion within the Active Site of Carbonic Anhydrase. J Biomol Struct Dyn 2009; 26:431-44. [DOI: 10.1080/07391102.2009.10507258] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
29
|
Savinov AY, Strongin AY. Matrix metalloproteinases, T cell homing and beta-cell mass in type 1 diabetes. VITAMINS AND HORMONES 2009; 80:541-62. [PMID: 19251049 DOI: 10.1016/s0083-6729(08)00618-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The pathogenesis of type 1 diabetes begins with the activation of autoimmune T killer cells and is followed by their homing into the pancreatic islets. After penetrating the pancreatic islets, T cells directly contact and destroy insulin-producing beta cells. This review provides an overview of the dynamic interactions which link T cell membrane type-1 matrix metalloproteinase (MT1-MMP) and the signaling adhesion CD44 receptor with T cell transendothelial migration and the subsequent homing of the transmigrated cells to the pancreatic islets. MT1-MMP regulates the functionality of CD44 in diabetogenic T cells. By regulating the functionality of T cell CD44, MT1-MMP mediates the transition of T cell adhesion to endothelial cells to the transendothelial migration of T cells, thus, controlling the rate at which T cells home into the pancreatic islets. As a result, the T cell MT1-MMP-CD44 axis controls the severity of the disease. Inhibition of MT1-MMP proteolysis of CD44 using highly specific and potent synthetic inhibitors, which have been clinically tested in cancer patients, reduces the rate of transendothelial migration and the homing of T cells. Result is a decrease in the net diabetogenic efficiency of T cells and a restoration of beta cell mass and insulin production in NOD mice. The latter is a reliable and widely used model of type I diabetes in humans. Overall, existing experimental evidence suggests that there is a sound mechanistic rationale for clinical trials of the inhibitors of T cell MT1-MMP in human type 1 diabetes patients.
Collapse
|
30
|
Rouffet M, Denhez C, Bourguet E, Bohr F, Guillaume D. In silico study of MMP inhibition. Org Biomol Chem 2009; 7:3817-25. [DOI: 10.1039/b910543c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
31
|
LeDour G, Moroy G, Rouffet M, Bourguet E, Guillaume D, Decarme M, ElMourabit H, Augé F, Alix AJ, Laronze JY, Bellon G, Hornebeck W, Sapi J. Introduction of the 4-(4-bromophenyl)benzenesulfonyl group to hydrazide analogs of Ilomastat leads to potent gelatinase B (MMP-9) inhibitors with improved selectivity. Bioorg Med Chem 2008; 16:8745-59. [DOI: 10.1016/j.bmc.2008.07.041] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Revised: 07/08/2008] [Accepted: 07/16/2008] [Indexed: 11/17/2022]
|
32
|
Kaludercic N, Lindsey ML, Tavazzi B, Lazzarino G, Paolocci N. Inhibiting metalloproteases with PD 166793 in heart failure: impact on cardiac remodeling and beyond. Cardiovasc Ther 2008; 26:24-37. [PMID: 18466418 DOI: 10.1111/j.1527-3466.2007.00034.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Metalloproteinases (MMPs, also called matrixins) are extracellular proteolytic enzymes involved in the degradation of both matrix and nonmatrix proteins. Currently, 25 MMPs have been identified in humans, and the overexpression of one or more MMPs has been implicated in several pathologies, spanning from cancer to rheumathoid arthritis to cardiovascular disease. While research over the past 20 years has focused on understanding MMP biology and selectively inhibiting MMP activity, key issues that remain to be addressed include MMP roles in the context of normal versus pathological conditions and whether globally inhibiting MMPs improves or deteriorates overall organ function. In terms of cardiovascular disease, increased MMP expression has been demonstrated in the setting of myocardial ischemia, reperfusion injury, and during the progression to congestive heart failure. MMPs are also major contributors to the progression of atherosclerotic lesions. In this review, we focus on cardiovascular effects produced by PD 166793, a wide-broad spectrum MMP inhibitor, originally developed by Parke-Davis (now Pfizer). We will briefly review its structure, mechanism of action, and inhibitory capacity. Finally, we will illustrate the cardiac contexts, both in vivo and in vitro, in which PD166793 administration has proven beneficial.
Collapse
Affiliation(s)
- Nina Kaludercic
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
33
|
Agrawal A, Romero-Perez D, Jacobsen JA, Villarreal FJ, Cohen SM. Zinc-binding groups modulate selective inhibition of MMPs. ChemMedChem 2008; 3:812-20. [PMID: 18181119 PMCID: PMC2836234 DOI: 10.1002/cmdc.200700290] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Indexed: 12/29/2022]
Abstract
The need for selective matrix metalloproteinase (MMP) inhibition is of interest because of the range of pathologies mediated by different MMP isoforms. The development of more selective MMP inhibitors (MMPi) may help to overcome some of the undesired side effects that have hindered the clinical success of these compounds. In an effort to devise new approaches to selective inhibitors, herein we describe several novel MMPi and show that their selectivity is dependent on the nature of the zinc-binding group (ZBG). This is in contrast to most current MMPi, which obtain isoform selectivity solely from the peptidomimetic backbone portion of the compound. In the present study, six different hydroxypyrone and hydroxypyridinone ZBGs were appended to a common biphenyl backbone and the inhibition efficiency of each inhibitor was determined in vitro (IC(50) values) against MMP-1, -2, -3, -7, -8, -9, -12, and -13. The results show that the selectivity profile of each inhibitor is different as a result of the various ZBGs. Computational modeling studies were used to explain some trends in the observed selectivity profiles. To assess the importance of the ZBG in a biological model, two of the semiselective, potent MMPi (and one control) were evaluated using an isolated perfused rat heart system. Hearts were subjected to ischemia reperfusion injury, and recovery of contractile function was examined. In this model, only one of the two MMPi showed significant and sustained heart recovery, demonstrating that the choice of ZBG can have a significant effect in a relevant pathophysiological endpoint.
Collapse
Affiliation(s)
- Arpita Agrawal
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093 (USA)
| | - Diego Romero-Perez
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093 (USA)
| | - Jennifer A. Jacobsen
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093 (USA)
| | - Francisco J. Villarreal
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093 (USA)
| | - Seth M. Cohen
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093 (USA)
| |
Collapse
|
34
|
Wang J, Hu X, Jiang J, Gou S, Huang X, Liu X, Feng X. Asymmetric Activation oftropos 2,2′-Biphenol with Cinchonine Generates an Effective Catalyst for the Asymmetric Strecker Reaction ofN-Tosyl-Protected Aldimines and Ketoimines. Angew Chem Int Ed Engl 2007. [DOI: 10.1002/ange.200703188] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
35
|
Wang J, Hu X, Jiang J, Gou S, Huang X, Liu X, Feng X. Asymmetric Activation oftropos 2,2′-Biphenol with Cinchonine Generates an Effective Catalyst for the Asymmetric Strecker Reaction ofN-Tosyl-Protected Aldimines and Ketoimines. Angew Chem Int Ed Engl 2007; 46:8468-70. [PMID: 17768752 DOI: 10.1002/anie.200703188] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jun Wang
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu 610064, P.R. China
| | | | | | | | | | | | | |
Collapse
|
36
|
Johnson AR, Pavlovsky AG, Ortwine DF, Prior F, Man CF, Bornemeier DA, Banotai CA, Mueller WT, McConnell P, Yan C, Baragi V, Lesch C, Roark WH, Wilson M, Datta K, Guzman R, Han HK, Dyer RD. Discovery and Characterization of a Novel Inhibitor of Matrix Metalloprotease-13 That Reduces Cartilage Damage in Vivo without Joint Fibroplasia Side Effects. J Biol Chem 2007; 282:27781-91. [PMID: 17623656 DOI: 10.1074/jbc.m703286200] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Matrix metalloproteinase-13 (MMP13) is a Zn(2+)-dependent protease that catalyzes the cleavage of type II collagen, the main structural protein in articular cartilage. Excess MMP13 activity causes cartilage degradation in osteoarthritis, making this protease an attractive therapeutic target. However, clinically tested MMP inhibitors have been associated with a painful, joint-stiffening musculoskeletal side effect that may be due to their lack of selectivity. In our efforts to develop a disease-modifying osteoarthritis drug, we have discovered MMP13 inhibitors that differ greatly from previous MMP inhibitors; they do not bind to the catalytic zinc ion, they are noncompetitive with respect to substrate binding, and they show extreme selectivity for inhibiting MMP13. By structure-based drug design, we generated an orally active MMP13 inhibitor that effectively reduces cartilage damage in vivo and does not induce joint fibroplasias in a rat model of musculoskeletal syndrome side effects. Thus, highly selective inhibition of MMP13 in patients may overcome the major safety and efficacy challenges that have limited previously tested non-selective MMP inhibitors. MMP13 inhibitors such as the ones described here will help further define the role of this protease in arthritis and other diseases and may soon lead to drugs that safely halt cartilage damage in patients.
Collapse
Affiliation(s)
- Adam R Johnson
- Department of Inflammation Molecular Sciences, Pfizer Global Research and Development, Ann Arbor, Michigan 48105, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Greig IR, Idris AI, Ralston SH, van't Hof RJ. Development and characterization of biphenylsulfonamides as novel inhibitors of bone resorption. J Med Chem 2007; 49:7487-92. [PMID: 17149877 DOI: 10.1021/jm051236m] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Increased osteoclastic bone resorption plays a central role in the pathogenesis of many bone diseases, and osteoclast inhibitors are the most widely used treatments for these diseases. We have identified and characterized a series of novel biphenylsulfonamide derivatives that have potent inhibitory effects on osteoclastic bone resorption in vitro and that prevent ovariectomy-induced bone loss in vivo. A number of aromatic substituted derivatives were prepared and a QSAR model was generated, which allowed accurate prediction of compound potency. Using this model, we have prepared compounds able to inhibit osteoclast formation and bone resorption in vitro at concentrations in the nanomolar range. One such compound, 55 (ABD295) (Greig, I. R.; Mohamed, A. I.; Ralston, S. H.; van't Hof, R. J. Alkyl Aryl Sulfonamides as Therapeutic Agents for the Treatment of Bone Conditions. GB Patent WO2005118528, 2005), fully reversed ovariectomy-induced bone loss in mice at a dose of 5 (mg/kg)/day. In conclusion, biphenylsulfonamides like 55 form a new class of potent antiresorptive agents with possible therapeutic use in diseases characterized by increased bone resorption.
Collapse
Affiliation(s)
- Iain R Greig
- School of Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| | | | | | | |
Collapse
|
38
|
Verma RP, Hansch C. Matrix metalloproteinases (MMPs): chemical-biological functions and (Q)SARs. Bioorg Med Chem 2007; 15:2223-68. [PMID: 17275314 DOI: 10.1016/j.bmc.2007.01.011] [Citation(s) in RCA: 542] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 01/09/2007] [Accepted: 01/11/2007] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases (MMPs) are a large family of calcium-dependent zinc-containing endopeptidases, which are responsible for the tissue remodeling and degradation of the extracellular matrix (ECM), including collagens, elastins, gelatin, matrix glycoproteins, and proteoglycan. They are regulated by hormones, growth factors, and cytokines, and are involved in ovarian functions. MMPs are excreted by a variety of connective tissue and pro-inflammatory cells including fibroblasts, osteoblasts, endothelial cells, macrophages, neutrophils, and lymphocytes. These enzymes are expressed as zymogens, which are subsequently processed by other proteolytic enzymes (such as serine proteases, furin, plasmin, and others) to generate the active forms. Matrix metalloproteinases are considered as promising targets for the treatment of cancer due to their strong involvement in malignant pathologies. Clinical/preclinical studies on MMP inhibition in tumor models brought positive results raising the idea that the development of strategies to inhibit MMPs may be proved to be a powerful tool to fight against cancer. However, the presence of an inherent flexibility in the MMP active-site limits dramatically the accurate modeling of MMP-inhibitor complexes. The interest in the application of quantitative structure-activity relationships (QSARs) has steadily increased in recent decades and we hope it may be useful in elucidating the mechanisms of chemical-biological interactions for this enzyme. In the present review, an attempt has been made to explore the in-depth knowledge from the classification of this enzyme to the clinical trials of their inhibitors. A total number of 92 QSAR models (44 published and 48 new formulated QSAR models) have also been presented to understand the chemical-biological interactions. QSAR results on the inhibition of various compound series against MMP-1, -2, -3, -7, -8, -9, -12, -13, and -14 reveal a number of interesting points. The most important of these are hydrophobicity and molar refractivity, which are the most important determinants of the activity.
Collapse
Affiliation(s)
- Rajeshwar P Verma
- Department of Chemistry, Pomona College, 645 North College Avenue, Claremont, CA 91711, USA.
| | | |
Collapse
|
39
|
Biasone A, Tortorella P, Campestre C, Agamennone M, Preziuso S, Chiappini M, Nuti E, Carelli P, Rossello A, Mazza F, Gallina C. α-Biphenylsulfonylamino 2-methylpropyl phosphonates: Enantioselective synthesis and selective inhibition of MMPs. Bioorg Med Chem 2007; 15:791-9. [PMID: 17088065 DOI: 10.1016/j.bmc.2006.10.047] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Revised: 10/17/2006] [Accepted: 10/23/2006] [Indexed: 11/26/2022]
Abstract
(R)-alpha-Biphenylsulfonylamino 2-methylpropyl phosphonates attain nM potency against several MMPs and are the most effective inhibitors based on phosphonate as zinc binding group. Since their preparation by direct N-acylation of expensive, enantiopure, alpha-aminophosphonic acids proceeds in low yields, we devised and evaluated a stereoselective and straightforward method of synthesis that avoids the unfavourable step of N-acylation. The key intermediate (R)-4-bromophenylsulfonylamino 2-methylpropyl phosphonate 9 was obtained by highly stereoselective addition of dibenzylphosphite to the enantiopure (S)-N-isobutylidene-p-bromobenzenesulfinamide 3, followed by oxidation with m-CPBA. Suzuki coupling of 9 with the desired arylboronic acids, gave the expected biphenylsulfonylamino derivatives in satisfactory yields. Liberation of the phosphonic group by hydrogenolysis led to the desired (R)-alpha-biphenylsulfonylamino 2-methylpropyl phosphonates 14a-i. Screening of the new compounds on MMP-1, -2, -3, -7, -8, -9, -13 and -14 showed IC(50) in the range of nM in most cases.
Collapse
Affiliation(s)
- Alessandro Biasone
- Dipartimento di Scienze del Farmaco, Università "G. d'Annunzio", Via dei Vestini 31, 66013 Chieti, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Aschi M, Besker N, Re N, Pochetti G, Coletti C, Gallina C, Mazza F. Stereoselectivity by Enantiomeric Inhibitors of Matrix Metalloproteinase-8: New Insights from Molecular Dynamics Simulations. J Med Chem 2006; 50:211-8. [PMID: 17228863 DOI: 10.1021/jm0608457] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular Dynamics simulations in aqueous solution were performed for the matrix metalloproteinase-8 (MMP-8) free catalytic domain and for its complexes with the (R)- and (S)-[1-(4'-methoxybiphenyl-4-sulfonylamino)-2-methylpropyl] phosphonate. The 144-155 loop of the enzyme undergoes a drastic decrease of mobility once complexed with both enantiomers. The two enantiomers induce a different decrease of conformational entropy upon complexation. The higher affinity of the R-enantiomer can be related to the lower loss of conformational entropy accompanying its binding. The differences in the dynamical behavior of the protein induced by the two enantiomers are discussed at molecular level and the mode of binding of the simulated complexes is compared with that previously determined by X-ray crystallography.
Collapse
Affiliation(s)
- Massimiliano Aschi
- Dipartimento di Chimica, Ingegneria Chimica e Materiali, Università di L'Aquila, Italia.
| | | | | | | | | | | | | |
Collapse
|
41
|
Oltenfreiter R, Staelens L, Kersemans V, Cornelissen B, Frankenne F, Foidart JM, Van de Wiele C, Slegers G. Valine-based biphenylsulphonamide matrix metalloproteinase inhibitors as tumor imaging agents. Appl Radiat Isot 2006; 64:677-85. [PMID: 16546398 DOI: 10.1016/j.apradiso.2006.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Revised: 01/24/2006] [Accepted: 01/24/2006] [Indexed: 10/24/2022]
Abstract
Among matrix metalloproteinases (MMPs), the subfamily of gelatinases (MMP-2, MMP-9) is of particular interest due to their ability to degrade type IV collagen and other non-fibrillar collagen domains and proteins such as fibronectin and laminin. Whilst malignant cells often over-express various MMPs, the gelatinases have been most consistently detected in malignant tissues and associated with tumor growth, metastatic potential and angiogenesis. Radiosynthesis of carboxylic (1') and hydroxamic (2') MMPIs resulted in radiochemical yields of 70 +/- 5% (n = 6) and 60 +/- 5% (n = 4), respectively. Evaluation in A549-inoculated athymic mice showed a tumor uptake of 2. 0+/- 0.7%ID/g (3 h p.i.), a tumor/blood ratio of 0.5 and a tumor/muscle ratio of 4.6 at 48 h p.i. for 1'. For compound 2' a tumor uptake of 0.7 +/- 0.2%ID/g (3 h p.i.), a tumor/blood ratio of 1.2 and a tumor/muscle ratio of 1.8 at 24 h p.i. were observed. HPLC analysis of the blood (plasma) showed no dehalogenation or other metabolites of 1' 2 h p.i. For compound 2', 65.4% of intact compound was found in the blood (plasma) and one polar metabolite (31%) was detected whereas in the tumor 91.8% of the accumulated activity was caused by intact compound and only 8.1% by the metabolite. Planar imaging, using a Toshiba GCA-9300A/hg SPECT camera, showed that tumor tissue could be visualized and that image quality improved by decreasing specific activity resulting in lower liver uptake, indicating some degree of saturable binding in the liver. In vivo evaluation of these radioiodinated carboxylic and hydroxamic MMP inhibitor tracers revealed that MMP inhibitors could have potential as tumor imaging agents, but that further research is necessary.
Collapse
Affiliation(s)
- Ruth Oltenfreiter
- Faculty of Pharmaceutical Sciences, Department of Radiopharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The search for an MMP inhibitor with anticancer efficacy is a nearly three-decade endeavor. This inhibitor is yet to be found. The reasons for this failure include shortcomings in the chemistry of these compounds (including broad MMP sub-type selectivity, metabolic lability, and toxicity) as well as the emerging, and arguably extraordinary, complexity of MMP cell (and cancer) biology. Together these suggest that the successful anticancer inhibitor must possess MMP selectivity against the MMP subtype whose involvement is critical, yet highly temporally (with respect to metastatic progression) and mechanistically (with respect to matrix degradation) regulated. This review summarizes the progression of chemical structure and mechanistic thinking toward these objectives, with emphasis on the disappointment, the perseverance, and the resilient optimism that such an inhibitor is there to be discovered.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556-5670, USA
| | | |
Collapse
|
43
|
Pochetti G, Gavuzzo E, Campestre C, Agamennone M, Tortorella P, Consalvi V, Gallina C, Hiller O, Tschesche H, Tucker PA, Mazza F. Structural Insight into the Stereoselective Inhibition of MMP-8 by Enantiomeric Sulfonamide Phosphonates. J Med Chem 2006; 49:923-31. [PMID: 16451058 DOI: 10.1021/jm050787+] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Potent and selective inhibitors of matrix metalloproteinases (MMPs), a family of zinc proteases that can degrade all the components of the extracellular matrix, could be useful for treatment of diseases such as cancer and arthritis. The most potent MMP inhibitors are based on hydroxamate as zinc-binding group (ZBG). alpha-Arylsulfonylamino phosphonates incorporate a particularly favorable combination of phosphonate as ZBG and arylsulfonylamino backbone so that their affinity exceptionally attains the nanomolar strength frequently observed for hydroxamate analogues. The detailed mode of binding of [1-(4'-methoxybiphenyl-4-sulfonylamino)-2-methylpropyl]phosphonate has been clarified by the crystal structures of the complexes that the R- and S-enantiomers respectively form with MMP-8. The reasons for the preferential MMP-8 inhibition by the R-phosphonate are underlined and the differences in the mode of binding of analogous alpha-arylsulfonylamino hydroxamates and carboxylates are discussed.
Collapse
Affiliation(s)
- Giorgio Pochetti
- Istituto di Cristallografia, C.N.R., Monterotondo Stazione, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mendes O, Kim HT, Stoica G. Expression of MMP2, MMP9 and MMP3 in breast cancer brain metastasis in a rat model. Clin Exp Metastasis 2005; 22:237-46. [PMID: 16158251 DOI: 10.1007/s10585-005-8115-6] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Accepted: 05/30/2005] [Indexed: 02/07/2023]
Abstract
In order to study the expression of MMP2, MMP3 and MMP9 in breast cancer brain metastasis, we used a syngeneic rat model of distant metastasis of ENU1564, a carcinogen-induced mammary adenocarcinoma cell line. At six weeks post inoculation we observed development of micro-metastasis in the brain. Immunohistochemistry and Western Blotting analyses showed that MMP-2, -3 and -9 proteins expressions are consistently significantly higher in neoplastic brain tissue compared to normal brain tissue. These results were confirmed by RT-PCR. In situ zymography revealed gelatinase activity within the brain metastasis. Gel zymography showed increase in MMP2 and MMP3 activity in brain metastasis. Furthermore, we were able to significantly decrease the development of breast cancer brain metastasis in animals by treatment with PD 166793, a selective synthetic MMP inhibitor. In addition, PD 166793 decreased the in vitro invasive cell behavior of ENU1546. Together our results suggest that MMP-2, -3 and -9 may be involved in the process of metastasis of breast cancer to the brain.
Collapse
Affiliation(s)
- Odete Mendes
- Department of Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas 77843, USA.
| | | | | |
Collapse
|
45
|
Oltenfreiter R, Staelens L, Labied S, Kersemans V, Frankenne F, Noël A, Van de Wiele C, Slegers G. Tryptophane-Based Biphenylsulfonamide Matrix Metalloproteinase Inhibitors as Tumor Imaging Agents. Cancer Biother Radiopharm 2005; 20:639-47. [PMID: 16398616 DOI: 10.1089/cbr.2005.20.639] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIM As a part of our efforts to use small organic matrix metalloproteinase (MMP) inhibitors with improved characteristics for the diagnosis and treatment of different kinds of tumor tissues, biphenylsulfonamide analogues were synthesized. This study reports on the in vivo biodistribution of iodine-123-labeled biphenylsulfonide and analogues in A549 lung carcinoma inoculated into athymic mice and the evaluation of their suitability as imaging agents using a single photon emission computed tomography (SPECT) camera. METHODS The radioiodinated carboxylic and hydroxamic MMP inhibitors 2-(4'- [(123)I]iodobiphenyl-4-sulfonylamino)-3-(1H-indol-3-yl)-propionic acid (1') and 2-(4'-[(123)I]iodobiphenyl-4- sulfonylamino)-3-(1H-indol-3-yl)-propionamide (2') were synthesized by electrophilic aromatic substitution of the tributylstannyl derivatives. Planar gamma camera imaging was performed in nu/nu athymic mice bearing an A549 tumor using a Toshiba GCA-9300A/hg SPECT camera in planar mode equipped with a high-resolution, parallel-hole collimator. RESULTS Radiosynthesis of (1') and (2') resulted in radiochemical yields of 60 +/- 5% (n +/- 3) and 70 +/- 5% (n = 6), respectively. Evaluation of tumors induced in athymic mice by the inoculation of non-small cell lung A549 carcinoma cells, showed a tumor uptake of 0.27-0.01 percent injected dose per gram (%ID/g) (3 hours-48 hours p.i.), a tumor-blood ratio of 0.7, a tumor-muscle ratio of 1.6, and a tumor-fat ratio of 0.5 at 24 hours (p.i.) for compound 1'. For compound 2' a tumor uptake of 0.7-0.04 %ID/g (3 hours-48 hours p.i.), a postinjection tumor-blood ratio of 1.2, a tumor-muscle ratio of 3.2, and a tumor-fat ratio of 2.4 at 48 hours p.i. was observed. SPECT evaluation confirmed the results obtained from biodistribution. CONCLUSION In vivo evaluation of these radioiodinated carboxylic and hydroxamic MMP inhibitor tracers revealed that they do not appear suitable as tumor-imaging agents.
Collapse
|
46
|
Bouissane L, El Kazzouli S, Léonce S, Pfeiffer B, Rakib EM, Khouili M, Guillaumet G. Synthesis and biological evaluation of N-(7-indazolyl)benzenesulfonamide derivatives as potent cell cycle inhibitors. Bioorg Med Chem 2005; 14:1078-88. [PMID: 16274996 DOI: 10.1016/j.bmc.2005.09.037] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Revised: 08/30/2005] [Accepted: 09/13/2005] [Indexed: 10/25/2022]
Abstract
We herein describe a new synthesis of N-(7-indazolyl)benzenesulfonamide derivatives. These compounds were evaluated for their antiproliferative activities toward L1210 murine leukemia cells. One of them, 4-methoxy-N-(3-chloro-7-indazolyl)benzenesulfonamide, was identified as the most potent with an IC(50) of 0.44 microM.
Collapse
Affiliation(s)
- L Bouissane
- Institut de Chimie Organique et Analytique, UMR CNRS 6005, Université d'Orléans, BP 6759, France
| | | | | | | | | | | | | |
Collapse
|
47
|
Oltenfreiter R, Staelens L, Hillaert U, Heremans A, Noël A, Frankenne F, Slegers G. Synthesis, radiosynthesis, in vitro and preliminary in vivo evaluation of biphenyl carboxylic and hydroxamic matrix metalloproteinase (MMP) inhibitors as potential tumor imaging agents. Appl Radiat Isot 2005; 62:903-13. [PMID: 15799868 DOI: 10.1016/j.apradiso.2004.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2004] [Revised: 12/17/2004] [Accepted: 12/18/2004] [Indexed: 11/23/2022]
Abstract
Excess matrix degradation is one of the hallmarks of cancer and is an important factor in the process of tumor progression. It is implicated in invasion, metastasis, growth, angiogenesis and migration. Many characteristics of matrix metalloproteinases (MMPs) make them attractive therapeutic and diagnostic targets. MMP expression is upregulated at the tumor site, with localization of activity in the tumor or the surrounding stroma, providing a target for medical imaging techniques. Radioiodinated carboxylic and hydroxamic MMP inhibitors 2-(4'-[123I] iodo-biphenyl-4-sulfonylamino)-3-methyl-butyric acid (9) and 2-(4'-[123I] iodo-biphenyl-4-sulfonylamino)-3-methyl-butyramide (11), their unlabelled standards and precursors were synthesized. Radioiodination was conducted by electrophilic aromatic substitution of the tributylstannyl precursors and resulted in radiochemical yields of 70+/-5% (n=6) and 60+/-5% (n=4), respectively. In vitro zymography and enzyme assays showed for both hydroxamic acid and carboxylic acid compounds a good inhibition activity and a high selectivity for MMP-2. In vivo biodistribution in NMRI mice showed no long-term accumulation in organs and the possibility to accumulate in the tumor in a later phase of this study.
Collapse
Affiliation(s)
- Ruth Oltenfreiter
- Laboratory of Radiopharmacy, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium.
| | | | | | | | | | | | | |
Collapse
|
48
|
Khandelwal A, Lukacova V, Comez D, Kroll DM, Raha S, Balaz S. A combination of docking, QM/MM methods, and MD simulation for binding affinity estimation of metalloprotein ligands. J Med Chem 2005; 48:5437-47. [PMID: 16107143 PMCID: PMC2896055 DOI: 10.1021/jm049050v] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To alleviate the problems in the receptor-based design of metalloprotein ligands due to inadequacies in the force-field description of coordination bonds, a four-tier approach was devised. Representative ligand-metalloprotein interaction energies are obtained by subsequent application of (1) docking with metal-binding-guided selection of modes, (2) optimization of the ligand-metalloprotein complex geometry by combined quantum mechanics and molecular mechanics (QM/MM) methods, (3) conformational sampling of the complex with constrained metal bonds by force-field-based molecular dynamics (MD), and (4) a single point QM/MM energy calculation for the time-averaged structures. The QM/MM interaction energies are, in a linear combination with the desolvation-characterizing changes in the solvent-accessible surface areas, correlated with experimental data. The approach was applied to structural correlation of published binding free energies of a diverse set of 28 hydroxamate inhibitors to zinc-dependent matrix metalloproteinase 9 (MMP-9). Inclusion of steps 3 and 4 significantly improved both correlation and prediction. The two descriptors explained 90% of variance in inhibition constants of all 28 inhibitors, ranging from 0.08 to 349 nM, with the average unassigned error of 0.318 log units. The structural and energetic information obtained from the time-averaged MD simulation results helped understand the differences in binding modes of related compounds.
Collapse
Affiliation(s)
- Akash Khandelwal
- Department of Pharmaceutical Sciences and Center for Protease Research, North Dakota State University, Fargo, ND 58105, USA
| | - Viera Lukacova
- Department of Pharmaceutical Sciences and Center for Protease Research, North Dakota State University, Fargo, ND 58105, USA
| | - Dogan Comez
- Department of Mathematics, North Dakota State University, Fargo, ND 58105, USA
| | - Daniel M. Kroll
- Department of Physics, North Dakota State University, Fargo, ND 58105, USA
| | - Soumyendu Raha
- Department of Computer Science, North Dakota State University, Fargo, ND 58105, USA
| | - Stefan Balaz
- Department of Pharmaceutical Sciences and Center for Protease Research, North Dakota State University, Fargo, ND 58105, USA
- CORRESPONDING AUTHOR FOOTNOTE: Stefan Balaz, North Dakota State University, College of Pharmacy, Sudro Hall Rm 8, Fargo, ND-58105; phone 701-231-7749; fax 701-231-8333; e-mail
| |
Collapse
|
49
|
Hu Y, Xiang JS, DiGrandi MJ, Du X, Ipek M, Laakso LM, Li J, Li W, Rush TS, Schmid J, Skotnicki JS, Tam S, Thomason JR, Wang Q, Levin JI. Potent, selective, and orally bioavailable matrix metalloproteinase-13 inhibitors for the treatment of osteoarthritis. Bioorg Med Chem 2005; 13:6629-44. [PMID: 16216515 DOI: 10.1016/j.bmc.2005.07.076] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2005] [Revised: 07/14/2005] [Accepted: 07/15/2005] [Indexed: 11/30/2022]
Abstract
Modification of alpha-biphenylsulfonamidocarboxylic acids led to potent and selective MMP-13 inhibitors. Compound 16 showed 100% oral bioavailability in rats and demonstrated >50% inhibition of bovine cartilage degradation at 10 ng/mL.
Collapse
Affiliation(s)
- Yonghan Hu
- Department of Chemical and Screening Sciences, Wyeth Research, Cambridge, MA 02140, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhou YP, Madjidi A, Wilson ME, Nothhelfer DA, Johnson JH, Palma JF, Schweitzer A, Burant C, Blume JE, Johnson JD. Matrix metalloproteinases contribute to insulin insufficiency in Zucker diabetic fatty rats. Diabetes 2005; 54:2612-9. [PMID: 16123349 DOI: 10.2337/diabetes.54.9.2612] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
To assess the molecular changes associated with pancreatic beta-cell dysfunction occurring during the onset of type 2 diabetes, we profiled pancreatic islet mRNAs from diabetic male and high-fat-fed female Zucker diabetic fatty (ZDF) rats and their nondiabetic lean counterparts on custom islet-specific oligonucleotide arrays. The most prominent changes in both the male and female models of type 2 diabetes were increases in the mRNAs encoding proteases and extracellular matrix components that are associated with tissue remodeling and fibrosis. The mRNAs for metalloproteinase (MMP)-2, -12, and -14 were sharply increased with the onset of islet dysfunction and diabetes. Zymography of islet extracts revealed a concurrent, >10-fold increase in MMP-2 protease activity in islets from 9-week-old male ZDF rats. Treatment of female ZDF rats receiving a diabetogenic diet with PD166793, a broad-spectrum MMP inhibitor, substantially prevented diabetes. The effect of this compound was due in part to marked beta-cell expansion. These studies indicate that MMPs contribute to islet fibrosis and insulin insufficiency in ZDF rats. Class-targeted protease inhibitors should be explored for their potential therapeutic utility in preservation of beta-cell mass in type 2 diabetes.
Collapse
Affiliation(s)
- Yun-Ping Zhou
- Metabolex, 3876 Bay Center Place, Hayward, CA 94583, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|