1
|
Mehta NV, Abhyankar A, Degani MS. Elemental exchange: Bioisosteric replacement of phosphorus by boron in drug design. Eur J Med Chem 2023; 260:115761. [PMID: 37651875 DOI: 10.1016/j.ejmech.2023.115761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/12/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Continuous efforts are being directed toward the employment of boron in drug design due to its advantages and unique characteristics including a plethora of target engagement modes, lower metabolism, and synthetic accessibility, among others. Phosphates are components of multiple drug molecules as well as clinical candidates, since they play a vital role in various biochemical functions, being components of nucleotides, energy currency- ATP as well as several enzyme cofactors. This review discusses the unique chemistry of boron functionalities as phosphate bioisosteres - "the boron-phosphorus elemental exchange strategy" as well as the superiority of boron groups over other commonly employed phosphate bioisosteres. Boron phosphate-mimetics have been utilized for the development of enzyme inhibitors as well as novel borononucleotides. Both the boron functionalities described in this review-boronic acids and benzoxaboroles-contain a boron connected to two oxygens and one carbon atom. The boron atom of these functional groups coordinates with a water molecule in the enzyme site forming a tetrahedral molecule which mimics the phosphate structure. Although boron phosphate-mimetic molecules - FDA-approved Crisaborole and phase II/III clinical candidate Acoziborole are products of the boron-phosphorus bioisosteric elemental exchange strategy, this technique is still in its infancy. The review aims to promote the use of this strategy in future medicinal chemistry projects.
Collapse
Affiliation(s)
- Namrashee V Mehta
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, 400019, Maharashtra, India.
| | - Arundhati Abhyankar
- Shri Vile Parle Kelavani Mandal's Dr Bhanuben Nanavati College of Pharmacy, Gate No.1, Mithibai College Campus, Vile Parle West, Mumbai, 400056, Maharashtra, India.
| | - Mariam S Degani
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga, Mumbai, 400019, Maharashtra, India.
| |
Collapse
|
2
|
Zhang C, Liu Y, Zhou Q, Fan H, Liu X, Hu J. Recent research advances in ATX inhibitors: An overview of primary literature. Bioorg Med Chem 2023; 90:117374. [PMID: 37354726 DOI: 10.1016/j.bmc.2023.117374] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/24/2023] [Accepted: 06/07/2023] [Indexed: 06/26/2023]
Abstract
The autoglobulin gene is the main enzyme for circulating LPA production and has lysophosphatidylcholine D activity, which catalyzes the production of lysophosphatidic acid and choline with lysophosphatidylcholine as substrate. A growing body of experimental evidence suggests that autoglobulin is involved in the pathogenesis of a variety of diseases. This review summarizes the different structural ATX inhibitors classified according to their binding mode to the ATX triple orientation site, and summarizes the conformational relationships and molecular docking of each type with ATX structure, hoping to contribute to the development of novel ATX inhibitors.
Collapse
Affiliation(s)
- Cheng Zhang
- Weifang Medical University, No. 7166 Baotong Road, Weifang 261053, PR China
| | - Yue Liu
- Weifang Medical University, No. 7166 Baotong Road, Weifang 261053, PR China
| | - Qinjiang Zhou
- Weifang Medical University, No. 7166 Baotong Road, Weifang 261053, PR China
| | - Hongze Fan
- Weifang Medical University, No. 7166 Baotong Road, Weifang 261053, PR China
| | - Xiaoxiao Liu
- Weifang Medical University, No. 7166 Baotong Road, Weifang 261053, PR China.
| | - Jinxing Hu
- Weifang Medical University, No. 7166 Baotong Road, Weifang 261053, PR China.
| |
Collapse
|
3
|
Vít O, Petrák J. Autotaxin and Lysophosphatidic Acid Signalling: the Pleiotropic Regulatory Network in Cancer. Folia Biol (Praha) 2023; 69:149-162. [PMID: 38583176 DOI: 10.14712/fb2023069050149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Autotaxin, also known as ecto-nucleotide pyrophosphatase/phosphodiesterase family member 2, is a secreted glycoprotein that plays multiple roles in human physiology and cancer pathology. This protein, by converting lysophosphatidylcholine into lysophosphatidic acid, initiates a complex signalling cascade with significant biological implications. The article outlines the autotaxin gene and protein structure, expression regulation and physiological functions, but focuses mainly on the role of autotaxin in cancer development and progression. Autotaxin and lysophosphatidic acid signalling influence several aspects of cancer, including cell proliferation, migration, metastasis, therapy resistance, and interactions with the immune system. The potential of autotaxin as a diagnostic biomarker and promising drug target is also examined.
Collapse
Affiliation(s)
- Ondřej Vít
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic.
| | - Jiří Petrák
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| |
Collapse
|
4
|
Banerjee S, Lee S, Norman DD, Tigyi GJ. Designing Dual Inhibitors of Autotaxin-LPAR GPCR Axis. Molecules 2022; 27:5487. [PMID: 36080255 PMCID: PMC9458164 DOI: 10.3390/molecules27175487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
The ATX-LPA-LPAR1 signaling pathway plays a universal role in stimulating diverse cellular responses, including cell proliferation, migration, survival, and invasion in almost every cell type. The ATX-LPAR1 axis is linked to several metabolic and inflammatory diseases including cancer, fibrosis, and rheumatoid arthritis. Numerous selective ATX or LPAR1 inhibitors have been developed and so far, their clinical efficacy has only been evaluated in idiopathic pulmonary fibrosis. None of the ATX and LPAR1 inhibitors have advanced to clinical trials for cancer and rheumatoid arthritis. Nonetheless, several research groups, including ours, have shown considerable benefit of simultaneous ATX and LPAR1 inhibition through combination therapy. Recent research suggests that dual-targeting therapies are superior to combination therapies that use two selective inhibitors. However, limited reports are available on ATX-LPAR1 dual inhibitors, potentially due to co-expression of multiple different LPARs with close structural similarities at the same target. In this review, we discuss rational design and future directions of dual ATX-LPAR1 inhibitors.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Chemistry, Middle Tennessee State University, 1301 E. Main Street, Murfreesboro, TN 37132, USA
- Molecular Biosciences Program, Middle Tennessee State University, 1301 E. Main Street, Murfreesboro, TN 37132, USA
| | - Suechin Lee
- Department of Physiology, University of Tennessee Health Science Center Memphis, 3 N. Dunlap Street, Memphis, TN 38163, USA
| | - Derek D. Norman
- Department of Physiology, University of Tennessee Health Science Center Memphis, 3 N. Dunlap Street, Memphis, TN 38163, USA
| | - Gabor J. Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, 3 N. Dunlap Street, Memphis, TN 38163, USA
| |
Collapse
|
5
|
Clark JM, Salgado-Polo F, Macdonald SJF, Barrett TN, Perrakis A, Jamieson C. Structure-Based Design of a Novel Class of Autotaxin Inhibitors Based on Endogenous Allosteric Modulators. J Med Chem 2022; 65:6338-6351. [PMID: 35440138 PMCID: PMC9059126 DOI: 10.1021/acs.jmedchem.2c00368] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Autotaxin (ATX) facilitates the hydrolysis of lysophosphatidylcholine to lysophosphatidic acid (LPA), a bioactive phospholipid, which facilitates a diverse range of cellular effects in multiple tissue types. Abnormal LPA expression can lead to the progression of diseases such as cancer and fibrosis. Previously, we identified a potent ATX steroid-derived hybrid (partially orthosteric and allosteric) inhibitor which did not form interactions with the catalytic site. Herein, we describe the design, synthesis, and biological evaluation of a focused library of novel steroid-derived analogues targeting the bimetallic catalytic site, representing an entirely unique class of ATX inhibitors of type V designation, which demonstrate significant pathway-relevant biochemical and phenotypic biological effects. The current compounds modulated LPA-mediated ATX allostery and achieved indirect blockage of LPA1 internalization, in line with the observed reduction in downstream signaling cascades and chemotaxis induction. These novel type V ATX inhibitors represent a promising tool to inactivate the ATX-LPA signaling axis.
Collapse
Affiliation(s)
- Jennifer M Clark
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| | - Fernando Salgado-Polo
- Oncode Institute and Division of Biochemistry, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Simon J F Macdonald
- Medicines Design, GlaxoSmithKline R&D, Stevenage, Hertfordshire SG1 2NY, United Kingdom
| | - Tim N Barrett
- Medicines Design, GlaxoSmithKline R&D, Stevenage, Hertfordshire SG1 2NY, United Kingdom
| | - Anastassis Perrakis
- Oncode Institute and Division of Biochemistry, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Craig Jamieson
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| |
Collapse
|
6
|
Messner K, Vuong B, Tranmer GK. The Boron Advantage: The Evolution and Diversification of Boron’s Applications in Medicinal Chemistry. Pharmaceuticals (Basel) 2022; 15:ph15030264. [PMID: 35337063 PMCID: PMC8948683 DOI: 10.3390/ph15030264] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/10/2022] [Accepted: 02/13/2022] [Indexed: 12/13/2022] Open
Abstract
In this review, the history of boron’s early use in drugs, and the history of the use of boron functional groups in medicinal chemistry applications are discussed. This includes diazaborines, boronic acids, benzoxaboroles, boron clusters, and carboranes. Furthermore, critical developments from these functional groups are highlighted along with recent developments, which exemplify potential prospects. Lastly, the application of boron in the form of a prodrug, softdrug, and as a nanocarrier are discussed to showcase boron’s emergence into new and exciting fields. Overall, we emphasize the evolution of organoboron therapeutic agents as privileged structures in medicinal chemistry and outline the impact that boron has had on drug discovery and development.
Collapse
Affiliation(s)
- Katia Messner
- Rady Faculty of Health Science, College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (K.M.); (B.V.)
| | - Billy Vuong
- Rady Faculty of Health Science, College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (K.M.); (B.V.)
| | - Geoffrey K. Tranmer
- Rady Faculty of Health Science, College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada; (K.M.); (B.V.)
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Correspondence:
| |
Collapse
|
7
|
Xiao YC, Yu JL, Dai QQ, Li G, Li GB. Targeting Metalloenzymes by Boron-Containing Metal-Binding Pharmacophores. J Med Chem 2021; 64:17706-17727. [PMID: 34875836 DOI: 10.1021/acs.jmedchem.1c01691] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Metalloenzymes have critical roles in a wide range of biological processes and are directly involved in many human diseases; hence, they are considered as important targets for therapeutic intervention. The specific characteristics of metal ion(s)-containing active sites make exploitation of metal-binding pharmacophores (MBPs) critical to inhibitor development targeting metalloenzymes. This Perspective focuses on boron-containing MBPs, which display unique binding modes with metalloenzyme active sites, particularly via mimicking native substrates or tetrahedral transition states. The design concepts regarding boron-containing MBPs are highlighted through the case analyses on five distinct classes of clinically relevant nucleophilic metalloenzymes from medicinal chemistry perspectives. The challenges (e.g., selectivity) faced by some boron-containing MBPs and possible strategies (e.g., bioisosteres) for metalloenzyme inhibitor transformation are also discussed.
Collapse
Affiliation(s)
- You-Cai Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jun-Lin Yu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qing-Qing Dai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Gen Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guo-Bo Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Jia Y, Li Y, Xu XD, Tian Y, Shang H. Design and Development of Autotaxin Inhibitors. Pharmaceuticals (Basel) 2021; 14:ph14111203. [PMID: 34832985 PMCID: PMC8622848 DOI: 10.3390/ph14111203] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/18/2022] Open
Abstract
Autotaxin (ATX) is the only enzyme of the ecto-nucleotide pyrophosphatase/phosphodiesterase (ENPP2) family with lysophospholipase D (lysoPLD) activity, which is mainly responsible for the hydrolysis of extracellular lysophosphatidylcholine (LPC) into lysophosphatidic acid (LPA). LPA can induce various responses, such as cell proliferation, migration, and cytokine production, through six G protein-coupled receptors (LPA1-6). This signaling pathway is associated with metabolic and inflammatory disorder, and inhibiting this pathway has a positive effect on the treatment of related diseases, while ATX, as an important role in the production of LPA, has been shown to be associated with the occurrence and metastasis of tumors, fibrosis and cardiovascular diseases. From mimics of ATX natural lipid substrates to the rational design of small molecule inhibitors, ATX inhibitors have made rapid progress in structural diversity and design over the past 20 years, and three drugs, GLPG1690, BBT-877, and BLD-0409, have entered clinical trials. In this paper, we will review the structure of ATX inhibitors from the perspective of the transformation of design ideas, discuss the advantages and disadvantages of each inhibitor type, and put forward prospects for the development of ATX inhibitors in the future.
Collapse
Affiliation(s)
| | | | | | - Yu Tian
- Correspondence: (Y.T.); (H.S.)
| | | |
Collapse
|
9
|
Horgan C, O' Sullivan TP. Recent Developments in the Practical Application of Novel Carboxylic Acid Bioisosteres. Curr Med Chem 2021; 29:2203-2234. [PMID: 34420501 DOI: 10.2174/0929867328666210820112126] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/10/2021] [Accepted: 07/23/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The carboxylic acid is an important functional group which features in the pharmacophore of some 450 drugs. Unfortunately, some carboxylic acid-containing drugs have been withdrawn from market due to unforeseen toxicity issues. Other issues associated with the carboxylate moiety include reduced metabolic stability or limited passive diffusion across biological membranes. Medicinal chemists often turn to bioisosteres to circumvent such obstacles. OBJECTIVE The aim of this review is to provide a summary of the various applications of novel carboxylic acid bioisosteres which have appeared in the literature since 2013. RESULTS We have summarised the most recent developments in carboxylic acid bioisosterism. In particular, we focus on the changes in bioactivity, selectivity or physiochemical properties brought about by these substitutions, as well as the advantages and disadvantages of each isostere. CONCLUSION The topics discussed herein highlight the continued interest in carboxylate bioisosteres. The development of novel carboxylic acid substitutes which display improved pharmacological profiles is testament to the innovation and creativity required to overcome the challenges faced in modern drug design.
Collapse
Affiliation(s)
- Conor Horgan
- School of Chemistry, University College Cork, Cork. Ireland
| | | |
Collapse
|
10
|
Dobersalske C, Grundmann M, Timmermann A, Theisen L, Kölling F, Harris RC, Fuerstner C, Becker MS, Wunder F. Establishment of a novel, cell-based autotaxin assay. Anal Biochem 2021; 630:114322. [PMID: 34343482 DOI: 10.1016/j.ab.2021.114322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 11/29/2022]
Abstract
Autotaxin (ATX) plays an important role in (patho-)physiological lysophosphatidic acid (LPA) signaling. Here we describe the establishment of novel cell-based ATX assay formats. ATX-mediated LPA generation is detected by using a stable LPA receptor reporter cell line. In a first assay variant, ATX-mediated LPA generation is started in the absence of cells and the reaction mix is transferred to the reporter cells after stopping the reaction (two-tube assay). In a second assay variant, ATX is added to the reporter cells expressing the known autotaxin binding partners integrin β1, integrin β3 and the LPA receptor 1. LPA generation is started in the presence of cells and is detected in real-time (one-tube assay). Structurally diverse ATX inhibitors with different binding modes were characterized in both cell-based assay variants and were also tested in the well-established biochemical choline release assay. ATX inhibitors displayed similar potencies, regardless if the assay was performed in the absence or presence of cells, and comparable results were obtained in all three assay formats. In summary, our novel cell-based ATX assay formats are well-suited for sensitive detection of enzyme activity as well as for the characterization of ATX inhibitors in the presence and absence of cells.
Collapse
Affiliation(s)
- Celia Dobersalske
- Lead Discovery, Bayer AG, Pharma Research and Development Center, Aprather Weg 18a, D-42096, Wuppertal, Germany
| | - Manuel Grundmann
- Cardiovascular Research, Bayer AG, Pharma Research and Development Center, Aprather Weg 18a, D-42096, Wuppertal, Germany
| | - Andreas Timmermann
- Lead Discovery, Bayer AG, Pharma Research and Development Center, Aprather Weg 18a, D-42096, Wuppertal, Germany
| | - Laura Theisen
- Lead Discovery, Bayer AG, Pharma Research and Development Center, Aprather Weg 18a, D-42096, Wuppertal, Germany
| | - Florian Kölling
- Computational Molecular Design. Bayer AG, Pharma Research and Development Center, Aprather Weg 18a, D-42096, Wuppertal, Germany
| | | | - Chantal Fuerstner
- Medicinal Chemistry, Bayer AG, Pharma Research and Development Center, Aprather Weg 18a, D-42096, Wuppertal, Germany
| | - Michael S Becker
- Cardiovascular Research, Bayer AG, Pharma Research and Development Center, Aprather Weg 18a, D-42096, Wuppertal, Germany
| | - Frank Wunder
- Lead Discovery, Bayer AG, Pharma Research and Development Center, Aprather Weg 18a, D-42096, Wuppertal, Germany.
| |
Collapse
|
11
|
Patle R, Shinde S, Patel S, Maheshwari R, Jariyal H, Srivastava A, Chauhan N, Globisch C, Jain A, Tekade RK, Shard A. Discovery of boronic acid-based potent activators of tumor pyruvate kinase M2 and development of gastroretentive nanoformulation for oral dosing. Bioorg Med Chem Lett 2021; 42:128062. [PMID: 33901643 DOI: 10.1016/j.bmcl.2021.128062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023]
Abstract
Several studies have established that cancer cells explicitly over-express the less active isoform of pyruvate kinase M2 (PKM2) is critical for tumorigenesis. The activation of PKM2 towards tetramer formation may increase affinity towards phosphoenolpyruvate (PEP) and avoidance of the Warburg effect. Herein, we describe the design, synthesis, and development of boronic acid-based molecules as activators of PKM2. The designed molecules were inspired by existing anticancer scaffolds and several fragments were assembled in the derivatives. 6a-6d were synthesized using a multi-step synthetic strategy in 55-70% yields, starting from cheap and readily available materials. The compounds were selectively cytotoxic to kill the cancerous cells at 80 nM, while they were non-toxic to the normal cells. The kinetic studies established the compounds as novel activators of PKM2 and (E/Z)-(4-(3-(2-((4-chlorophenyl)amino)-4-(dimethylamino)thiazol-5-yl)-2-(ethoxycarbonyl)-3-oxoprop-1-en-1-yl) phenyl)boronic acid (6c) emerged as the most potent derivative. 6c was further evaluated using various in silico tools to understand the molecular mechanism of tetramer formation. Docking studies revealed that 6c binds to the PKM2 dimer at the dimeric interface. Further to ascertain the binding site and mechanism of action, rigorous MD (molecular dynamics) simulations were undertaken, which led to the conclusion that 6c stabilizes the center of the dimeric interface that possibly promotes tetramer formation. We further planned to make a tablet of the developed molecule for oral delivery, but it was seriously impeded owing to poor aqueous solubility of 6c. To improve aqueous solubility and retain 6c at the lower gastrointestinal tract, thiolated chitosan-based nanoparticles (TCNPs) were prepared and further developed as tablet dosage form to retain anticancer potency in the excised goat colon. Our findings may provide a valuable pharmacological mechanism for understanding metabolic underpinnings that may aid in the clinical development of new anticancer agents targeting PKM2.
Collapse
Affiliation(s)
- Rajkumar Patle
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, India
| | - Shital Shinde
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, India
| | - Sagarkumar Patel
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, India
| | - Rahul Maheshwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad, India
| | - Heena Jariyal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, India
| | - Akshay Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, India
| | - Neelam Chauhan
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, India
| | | | - Alok Jain
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, India
| | - Rakesh K Tekade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad, India.
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, India.
| |
Collapse
|
12
|
Banerjee S, Norman DD, Deng S, Fakayode SO, Lee SC, Parrill AL, Li W, Miller DD, Tigyi GJ. Molecular modelling guided design, synthesis and QSAR analysis of new small molecule non-lipid autotaxin inhibitors. Bioorg Chem 2020; 103:104188. [PMID: 32890995 PMCID: PMC8163515 DOI: 10.1016/j.bioorg.2020.104188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/18/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023]
Abstract
The lysophospholipase D autotaxin (ATX) generates lysophosphatidic acid (LPA) that activates six cognate G-protein coupled receptors (GPCR) in cancerous cells, promoting their motility and invasion. Four novel compounds were generated aided by molecular docking guided design and synthesis techniques to obtain new dual inhibitors of ATX and the lysophosphatidic acid receptor subtype 1 (LPAR1). Biological evaluation of these compounds revealed two compounds, 10 and 11, as new ATX enzyme inhibitors with potencies in the range of 218-220 nM and water solubility (>100 µg/mL), but with no LPAR1 inhibitory activity. A QSAR model was generated that included four newly designed compounds and twenty-one additional compounds that we have reported previously. The QSAR model provided excellent predictability of the pharmacological activity and potency among structurally related drug candidates. This model will be highly useful in guiding the synthesis of new ATX inhibitors in the future.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Physical Sciences, University of Arkansas Fort Smith, Fort Smith, AR 72913, USA; Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Derek D Norman
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sayo O Fakayode
- Department of Physical Sciences, University of Arkansas Fort Smith, Fort Smith, AR 72913, USA
| | - Sue Chin Lee
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Abby L Parrill
- Department of Chemistry, Computational Research on Material Institute, The University of Memphis, Memphis, TN 38152, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Gabor J Tigyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
13
|
Silva MP, Saraiva L, Pinto M, Sousa ME. Boronic Acids and Their Derivatives in Medicinal Chemistry: Synthesis and Biological Applications. Molecules 2020; 25:E4323. [PMID: 32967170 PMCID: PMC7571202 DOI: 10.3390/molecules25184323] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/20/2022] Open
Abstract
Boron containing compounds have not been widely studied in Medicinal Chemistry, mainly due to the idea that this group could confer some toxicity. Nowadays, this concept has been demystified and, especially after the discovery of the drug bortezomib, the interest for these compounds, mainly boronic acids, has been growing. In this review, several activities of boronic acids, such as anticancer, antibacterial, antiviral activity, and even their application as sensors and delivery systems are addressed. The synthetic processes used to obtain these active compounds are also referred. Noteworthy, the molecular modification by the introduction of boronic acid group to bioactive molecules has shown to modify selectivity, physicochemical, and pharmacokinetic characteristics, with the improvement of the already existing activities. Besides, the preparation of compounds with this chemical group is relatively simple and well known. Taking into consideration these findings, this review reinforces the relevance of extending the studies with boronic acids in Medicinal Chemistry, in order to obtain new promising drugs shortly.
Collapse
Affiliation(s)
- Mariana Pereira Silva
- CIIMAR, Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, Portugal; (M.P.S.); (M.P.)
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, Portugal
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, Portugal
| | - Madalena Pinto
- CIIMAR, Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, Portugal; (M.P.S.); (M.P.)
| | - Maria Emília Sousa
- CIIMAR, Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira n° 228, 4050-313 Porto, Portugal; (M.P.S.); (M.P.)
| |
Collapse
|
14
|
Zagade AD, Shard A, Shinde S, Sahu AK, Sengupta P. Bioanalysis and Quadrupole-Time of Flight-Mass Spectrometry Driven In Vitro Metabolite Profiling of a New Boronic Acid-Based Anticancer Molecule. J Chromatogr Sci 2020; 58:796-803. [DOI: 10.1093/chromsci/bmaa044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 06/08/2020] [Accepted: 07/08/2020] [Indexed: 12/18/2022]
Abstract
Abstract
(E/Z)-(4-(3-(2-((4-chlorophenyl)amino)-4-(dimethylamino)thiazol-5-yl)-2-(ethoxy carbonyl)-3-oxoprop-1-en-1-yl)phenyl) boronic acid, a newly developed molecule having anticancer activity serves as a potential candidate for the further drug development process. In this study, to ascertain the anticancer potential of the molecule, we screened it against different cell lines and compared the activity against the standard drug doxorubicin. The molecule showed promising activity at a low concentration against almost all cell lines used in the study. Apart from that, the molecule was characterized for its pKa and a precise reverse phase high-performance liquid chromatography bioanalytical method has been developed. The method was validated according to the United States of Food and Drug Administration bioanalytical guideline and was found to produce linear response over the calibration range of 0.8–25 μg/mL. Inter- and intra-day accuracy were found to be in the range of 93.44–99.74%, whereas precision [% coefficient of variation (CV)] for inter- and intra-day was ranged between 1.63 and 5.79%, and 0.87 and 6.96%, respectively. The bioanalytical stability testing was carried out in different conditions including 8 h benchtop, 12 h autosampler and three freeze–thaw cycles. The analyte was stable in all the tested stability conditions. Finally, an in vitro metabolite identification study was conducted using quadrupole-time of flight-mass spectrometer, and two metabolites have been identified.
Collapse
Affiliation(s)
- Aarati Dilip Zagade
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar, Gujarat 382355, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar, Gujarat 382355, India
| | - Shital Shinde
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar, Gujarat 382355, India
| | - Amit Kumar Sahu
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar, Gujarat 382355, India
| | - Pinaki Sengupta
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
15
|
Design and discovery of boronic acid drugs. Eur J Med Chem 2020; 195:112270. [DOI: 10.1016/j.ejmech.2020.112270] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/22/2020] [Accepted: 03/22/2020] [Indexed: 12/15/2022]
|
16
|
Gerokonstantis DT, Nikolaou A, Magkrioti C, Afantitis A, Aidinis V, Kokotos G, Moutevelis-Minakakis P. Synthesis of novel 2-pyrrolidinone and pyrrolidine derivatives and study of their inhibitory activity against autotaxin enzyme. Bioorg Med Chem 2019; 28:115216. [PMID: 31864778 DOI: 10.1016/j.bmc.2019.115216] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
Autotaxin (ATX), a glycoprotein (~125 kDa) isolated as an autocrine motility factor from melanoma cells, belongs to a seven-membered family of ectonucleotide pyrophosphatase/phosphodiesterase (ENPP), and exhibits lysophospholipase D activity. ATX is responsible for the hydrolysis of lysophosphatidylcholine (LPC) to produce the bioactive lipid lysophosphatidic acid (LPA), which is upregulated in a variety of pathological inflammatory conditions, including fibrosis, cancer, liver toxicity and thrombosis. Given its role in human disease, the ATX-LPA axis is an interesting target for therapy, and the development of novel potent ATX inhibitors is of great importance. In the present work a novel class of ATX inhibitors, optically active derivatives of 2-pyrrolidinone and pyrrolidine heterocycles were synthesized. Some of them exhibited interesting in vitro activity, namely the hydroxamic acid 16 (IC50 700 nM) and the carboxylic acid 40b (IC50 800 nM), while the boronic acid derivatives 3k (IC50 50 nM), 3l (IC50 120 nM), 3 m (IC50 180 nM) and 21 (IC50 35 nM) were found to be potent inhibitors of ATX.
Collapse
Affiliation(s)
| | - Aikaterini Nikolaou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Christiana Magkrioti
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens 16672, Greece
| | | | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens 16672, Greece
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece
| | - Panagiota Moutevelis-Minakakis
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| |
Collapse
|
17
|
Do HH, Ullah S, Villinger A, Lecka J, Sévigny J, Ehlers P, Iqbal J, Langer P. Palladium-catalyzed synthesis and nucleotide pyrophosphatase inhibition of benzo[4,5]furo[3,2- b]indoles. Beilstein J Org Chem 2019; 15:2830-2839. [PMID: 31807218 PMCID: PMC6880817 DOI: 10.3762/bjoc.15.276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/12/2019] [Indexed: 01/22/2023] Open
Abstract
A two-step palladium-catalyzed procedure based on Suzuki–Miyaura cross coupling, followed by a double Buchwald–Hartwig reaction, allows for the synthesis of pharmaceutically relevant benzo[4,5]furo[3,2-b]indoles in moderate to very good yield. The synthesized compounds have been analyzed with regard to their inhibitory activity (IC50) of nucleotide pyrophosphatases h-NPP1 and h-NPP3. The activity lies in the nanomolar range. The results were rationalized based on docking studies.
Collapse
Affiliation(s)
- Hoang Huy Do
- Institut für Chemie, Universität Rostock, Albert Einstein Str. 3a, 18059 Rostock, Germany.,Faculty of Chemistry, VNU Hanoi University of Science, 19 Le Thanh Tong, Hoan Kiem, Hanoi, 110403, Vietnam
| | - Saif Ullah
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad-22060, Pakistan
| | - Alexander Villinger
- Institut für Chemie, Universität Rostock, Albert Einstein Str. 3a, 18059 Rostock, Germany
| | - Joanna Lecka
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, QC, G1V 0A6, Canada.,Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, G1V 4G2, Canada
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, QC, G1V 0A6, Canada.,Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, G1V 4G2, Canada
| | - Peter Ehlers
- Institut für Chemie, Universität Rostock, Albert Einstein Str. 3a, 18059 Rostock, Germany.,Leibniz-Institut für Katalyse an der Universität Rostock e.V., Albert Einstein Str. 29a, 18059 Rostock, Germany
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad-22060, Pakistan
| | - Peter Langer
- Institut für Chemie, Universität Rostock, Albert Einstein Str. 3a, 18059 Rostock, Germany.,Leibniz-Institut für Katalyse an der Universität Rostock e.V., Albert Einstein Str. 29a, 18059 Rostock, Germany
| |
Collapse
|
18
|
Wang W, Zhao F, Zhao Y, Pan W, Cao P, Wu L, Wang Z, Zhao X, Zhao Y, Wang H. Design, Synthesis, and Preliminary Bioactivity Evaluation of 2,7-Substituted Carbazole Derivatives as Potent Autotaxin Inhibitors and Antitumor Agents†. Anticancer Agents Med Chem 2019; 19:256-264. [PMID: 30173652 DOI: 10.2174/1871520618666180830161821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/18/2018] [Accepted: 08/03/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Autotaxin-LPA signaling has been implicated in cancer progression, and targeted for the discovery of cancer therapeutic agents. OBJECTIVE Potential ATX inhibitors were synthesized to develop novel leading compounds and effective anticancer agents. METHODS The present work designs and synthesizes a series of 2,7-subsitituted carbazole derivatives with different terminal groups R [R = -Cl (I), -COOH (II), -B(OH)2 (III), or -PO(OH)2 (I-IV)]. The inhibition of these compounds on the enzymatic activity of ATX was measured using FS-3 and Bis-pNpp as substrates, and the cytotoxicity of these compounds was evaluated using SW620, SW480, PANC-1, and SKOV-3 human carcinoma cells. Furthermore, the binding of leading compound with ATX was analyzed by molecular docking. RESULTS Compound III was shown to be a promising antitumor candidate by demonstrating both good inhibition of ATX enzymatic activity and high cytotoxicity against human cancer cell lines. Molecular docking study shows that compound III is located in a pocket, which mainly comprises amino acids 209 to 316 in domain 2 of ATX, and binds with these residues of ATX through van der Waals, conventional hydrogen bonds, and hydrophobic interactions. CONCLUSION Compound III with the terminal group R = -B(OH)2 has the most potent inhibitory effect with the greatest cytotoxicity to cancer cells. Moreover, the docking model provides a structural basis for the future optimization of promising antitumor compounds.
Collapse
Affiliation(s)
- Wenming Wang
- Biology Institute of Shanxi, Shanxi, Taiyuan 030006, China.,Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Shanxi, Taiyuan 030006, China
| | - Fengmei Zhao
- Biology Institute of Shanxi, Shanxi, Taiyuan 030006, China
| | - Yarui Zhao
- Biology Institute of Shanxi, Shanxi, Taiyuan 030006, China
| | - Weiwei Pan
- Biology Institute of Shanxi, Shanxi, Taiyuan 030006, China
| | - Pengcheng Cao
- Biology Institute of Shanxi, Shanxi, Taiyuan 030006, China
| | - Lintao Wu
- Department of Chemistry, Changzhi University, Shanxi, Changzhi 046011, China
| | - Zhijun Wang
- Department of Chemistry, Changzhi University, Shanxi, Changzhi 046011, China
| | - Xuan Zhao
- Department of Chemistry, University of Memphis, Memphis, TN 38152, United States
| | - Yi Zhao
- Biology Institute of Shanxi, Shanxi, Taiyuan 030006, China
| | - Hongfei Wang
- Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Shanxi, Taiyuan 030006, China
| |
Collapse
|
19
|
Examining barbiturate scaffold for the synthesis of new agents with biological interest. Future Med Chem 2019; 11:2063-2079. [DOI: 10.4155/fmc-2018-0541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: Barbiturates have a long history of being used as drugs presenting wide varieties of biological activities (antimicrobial, anti-urease and antioxidant). Reactive oxygen species are associated with inflammation implicated in cancer, atherosclerosis and autoimmune diseases. Multitarget agents represent a powerful approach to the therapy of complicated inflammatory diseases. Results: A novel series of barbiturates has been synthesized and evaluated in several in vitro assays. Compound 16b (lipoxygenases inhibitor, 55.0 μM) was found to be a cyclooxygenase-2 inhibitor (27.5 μM). Compound 8b was profiled as a drug-like candidate. Conclusion: The barbiturate core represents a new scaffold for lipoxygenases inhibition, and the undertaken derivatives show promise as multiple-target agents to combat inflammatory diseases.
Collapse
|
20
|
Semreen MH, El-Gamal MI, Ullah S, Jalil S, Zaib S, Anbar HS, Lecka J, Sévigny J, Iqbal J. Synthesis, biological evaluation, and molecular docking study of sulfonate derivatives as nucleotide pyrophosphatase/phosphodiesterase (NPP) inhibitors. Bioorg Med Chem 2019; 27:2741-2752. [PMID: 31088715 DOI: 10.1016/j.bmc.2019.04.031] [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] [Received: 04/08/2019] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Abstract
A new series of sulfonate derivatives 1a-zk were synthesized and evaluated as inhibitors of nucleotide pyrophosphatases. Most of the compounds exhibited good to moderate inhibition towards NPP1, NPP2, and NPP3 isozymes. Compound 1m was a potent and selective inhibitor of NPP1 with an IC50 value of 0.387 ± 0.007 µM. However, the most potent inhibitor of NPP3 was found as 1x with an IC50 value of 0.214 ± 0.012 µM. In addition, compound 1e was the most active inhibitor of NPP2 with an IC50 value of 0.659 ± 0.007 µM. Docking studies of the most potent compounds were carried out, and the computational results supported the in vitro results.
Collapse
Affiliation(s)
- Mohammad H Semreen
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt.
| | - Saif Ullah
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Saquib Jalil
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Sumera Zaib
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Hanan S Anbar
- Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt
| | - Joanna Lecka
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; Centre de Recherche du CHU de Québec - Université Laval, Québec, QC G1V 4G2, Canada
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada; Centre de Recherche du CHU de Québec - Université Laval, Québec, QC G1V 4G2, Canada
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan.
| |
Collapse
|
21
|
Matralis AN, Afantitis A, Aidinis V. Development and therapeutic potential of autotaxin small molecule inhibitors: From bench to advanced clinical trials. Med Res Rev 2018; 39:976-1013. [PMID: 30462853 DOI: 10.1002/med.21551] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/21/2018] [Accepted: 10/19/2018] [Indexed: 12/11/2022]
Abstract
Several years after its isolation from melanoma cells, an increasing body of experimental evidence has established the involvement of Autotaxin (ATX) in the pathogenesis of several diseases. ATX, an extracellular enzyme responsible for the hydrolysis of lysophosphatidylcholine (LPC) into the bioactive lipid lysophosphatidic acid (LPA), is overexpressed in a variety of human metastatic cancers and is strongly implicated in chronic inflammation and liver toxicity, fibrotic diseases, and thrombosis. Accordingly, the ATX-LPA signaling pathway is considered a tractable target for therapeutic intervention substantiated by the multitude of research campaigns that have been successful in identifying ATX inhibitors by both academia and industry. Furthermore, from a therapeutic standpoint, the entry and the so far promising results of the first ATX inhibitor in advanced clinical trials against idiopathic pulmonary fibrosis (IPF) lends support to the viability of this approach, bringing it to the forefront of drug discovery efforts. The present review article aims to provide a comprehensive overview of the most important series of ATX inhibitors developed so far. Special weight is lent to the design, structure activity relationship and mode of binding studies carried out, leading to the identification of advanced leads. The most significant in vitro and in vivo pharmacological results of these advanced leads are also summarized. Lastly, the development of the first ATX inhibitor entered in clinical trials accompanied by its phase 1 and 2a clinical trial data is disclosed.
Collapse
Affiliation(s)
- Alexios N Matralis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Antreas Afantitis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece.,NovaMechanics Ltd Cheminformatics Company, Nicosia, Cyprus
| | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| |
Collapse
|
22
|
Fisher N, Edwards MG, Hemming R, Allin SM, Wallis JD, Bulman Page PC, Mckenzie MJ, Jones SM, Elsegood MRJ, King-Underwood J, Richardson A. Synthesis and Activity of a Novel Autotaxin Inhibitor-Icodextrin Conjugate. J Med Chem 2018; 61:7942-7951. [PMID: 30059212 DOI: 10.1021/acs.jmedchem.8b00935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Autotaxin is an extracellular phospholipase D that catalyzes the hydrolysis of lysophosphatidyl choline (LPC) to generate the bioactive lipid lysophosphatidic acid (LPA). Autotaxin has been implicated in many pathological processes relevant to cancer. Intraperitoneal administration of an autotaxin inhibitor may benefit patients with ovarian cancer; however, low molecular mass compounds are known to be rapidly cleared from the peritoneal cavity. Icodextrin is a polymer that is already in clinical use because it is slowly eliminated from the peritoneal cavity. Herein we report conjugation of the autotaxin inhibitor HA155 to icodextrin. The conjugate inhibits autotaxin activity (IC50 = 0.86 ± 0.13 μg mL-1) and reduces cell migration. Conjugation of the inhibitor increased its solubility, decreased its membrane permeability, and improved its intraperitoneal retention in mice. These observations demonstrate the first application of icodextrin as a covalently-bonded drug delivery platform with potential use in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Natalie Fisher
- School of Pharmacy and Institute for Science and Technology in Medicine , Keele University , Keele ST5 5BG , U.K.,Keele Molecular Chemistry Group, Lennard-Jones Laboratories, School of Chemical and Physical Sciences , Keele University , Keele ST5 5BG , U.K
| | - Michael G Edwards
- Keele Molecular Chemistry Group, Lennard-Jones Laboratories, School of Chemical and Physical Sciences , Keele University , Keele ST5 5BG , U.K
| | - Ryan Hemming
- School of Science and Technology , Nottingham Trent University , Nottingham NG11 8NS , U.K
| | - Steven M Allin
- School of Science and Technology , Nottingham Trent University , Nottingham NG11 8NS , U.K
| | - John D Wallis
- School of Science and Technology , Nottingham Trent University , Nottingham NG11 8NS , U.K
| | | | - Michael J Mckenzie
- Charnwood Molecular Ltd. , The Heritage Building, Prince William Road , Loughborough LE11 5DA , U.K
| | - Stefanie M Jones
- School of Pharmacy and Institute for Science and Technology in Medicine , Keele University , Keele ST5 5BG , U.K
| | - Mark R J Elsegood
- Department of Chemistry , Loughborough University , Loughborough LE11 3TU , U.K
| | - John King-Underwood
- Computational Chemistry Resource , Old Cottage Hospital , Ledbury HR8 1ED , U.K
| | - Alan Richardson
- School of Pharmacy and Institute for Science and Technology in Medicine , Keele University , Keele ST5 5BG , U.K
| |
Collapse
|
23
|
Nikolaou A, Ninou I, Kokotou MG, Kaffe E, Afantitis A, Aidinis V, Kokotos G. Hydroxamic Acids Constitute a Novel Class of Autotaxin Inhibitors that Exhibit in Vivo Efficacy in a Pulmonary Fibrosis Model. J Med Chem 2018; 61:3697-3711. [PMID: 29620892 DOI: 10.1021/acs.jmedchem.8b00232] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autotaxin (ATX) catalyzes the hydrolysis of lysophosphatidylcholine (LPC) generating the lipid mediator lysophosphatidic acid (LPA). Both ATX and LPA are involved in various pathological inflammatory conditions, including fibrosis and cancer, and have attracted great interest as medicinal targets over the past decade. Thus, the development of novel potent ATX inhibitors is of great importance. We have developed a novel class of ATX inhibitors containing the zinc binding functionality of hydroxamic acid. Such novel hydroxamic acids that incorporate a non-natural δ-amino acid residue exhibit high in vitro inhibitory potency over ATX (IC50 values 50-60 nM). Inhibitor 32, based on δ-norleucine, was tested for its efficacy in a mouse model of pulmonary inflammation and fibrosis induced by bleomycin and exhibited promising efficacy. The novel hydroxamic ATX inhibitors provide excellent tools for the study of the role of the enzyme and could contribute to the development of novel therapeutic agents for the treatment of fibrosis and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Aikaterini Nikolaou
- Laboratory of Organic Chemistry, Department of Chemistry , National and Kapodistrian University of Athens , Panepistimiopolis , Athens 15771 , Greece
| | - Ioanna Ninou
- Division of Immunology , Biomedical Sciences Research Center "Alexander Fleming" , Athens 16672 , Greece
| | - Maroula G Kokotou
- Laboratory of Organic Chemistry, Department of Chemistry , National and Kapodistrian University of Athens , Panepistimiopolis , Athens 15771 , Greece
| | - Eleanna Kaffe
- Division of Immunology , Biomedical Sciences Research Center "Alexander Fleming" , Athens 16672 , Greece
| | | | - Vassilis Aidinis
- Division of Immunology , Biomedical Sciences Research Center "Alexander Fleming" , Athens 16672 , Greece
| | - George Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry , National and Kapodistrian University of Athens , Panepistimiopolis , Athens 15771 , Greece
| |
Collapse
|
24
|
Kuttruff CA, Ferrara M, Bretschneider T, Hoerer S, Handschuh S, Nosse B, Romig H, Nicklin P, Roth GJ. Discovery of BI-2545: A Novel Autotaxin Inhibitor That Significantly Reduces LPA Levels in Vivo. ACS Med Chem Lett 2017; 8:1252-1257. [PMID: 29259743 DOI: 10.1021/acsmedchemlett.7b00312] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 11/08/2017] [Indexed: 01/01/2023] Open
Abstract
In an effort to find new therapeutic interventions addressing the unmet medical need of patients with idiopathic pulmonary fibrosis, we initiated a program to identify new autotaxin (ATX) inhibitors. Starting from a recently published compound (PF-8380), we identified several highly potent ATX inhibitors with improved pharmacokinetic and safety profiles. Further optimization efforts resulted in the identification of a single-digit nanomolar lead compound (BI-2545) that shows substantial lowering of LPA in vivo and is therefore considered a valuable tool for further studies.
Collapse
Affiliation(s)
| | - Marco Ferrara
- Boehringer Ingelheim
Research Italia S.a.s. di BI IT S.r.l., Via G. Lorenzini 8, 20139 Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Pantsar T, Singha P, Nevalainen TJ, Koshevoy I, Leppänen J, Poso A, Niskanen JM, Pasonen-Seppänen S, Savinainen JR, Laitinen T, Laitinen JT. Design, synthesis, and biological evaluation of 2,4-dihydropyrano[2,3-c]pyrazole derivatives as autotaxin inhibitors. Eur J Pharm Sci 2017; 107:97-111. [DOI: 10.1016/j.ejps.2017.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/28/2017] [Accepted: 07/03/2017] [Indexed: 01/19/2023]
|
26
|
Nikolaou A, Kokotou MG, Limnios D, Psarra A, Kokotos G. Autotaxin inhibitors: a patent review (2012-2016). Expert Opin Ther Pat 2017; 27:815-829. [DOI: 10.1080/13543776.2017.1323331] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Aikaterini Nikolaou
- Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Maroula G. Kokotou
- Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Limnios
- Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Psarra
- Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
27
|
Zhang Y, Borrel A, Ghemtio L, Regad L, Boije af Gennäs G, Camproux AC, Yli-Kauhaluoma J, Xhaard H. Structural Isosteres of Phosphate Groups in the Protein Data Bank. J Chem Inf Model 2017; 57:499-516. [DOI: 10.1021/acs.jcim.6b00519] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
| | - Alexandre Borrel
- Laboratoire
Molécules Thérapeutiques in silico (MTi), UMRS-973, Université Paris Diderot, Sorbonne Paris Cité, INSERM, F-75013 Paris, France
| | | | - Leslie Regad
- Laboratoire
Molécules Thérapeutiques in silico (MTi), UMRS-973, Université Paris Diderot, Sorbonne Paris Cité, INSERM, F-75013 Paris, France
| | | | - Anne-Claude Camproux
- Laboratoire
Molécules Thérapeutiques in silico (MTi), UMRS-973, Université Paris Diderot, Sorbonne Paris Cité, INSERM, F-75013 Paris, France
| | | | | |
Collapse
|
28
|
Banerjee S, Norman DD, Lee SC, Parrill AL, Pham TCT, Baker DL, Tigyi GG, Miller DD. Highly Potent Non-Carboxylic Acid Autotaxin Inhibitors Reduce Melanoma Metastasis and Chemotherapeutic Resistance of Breast Cancer Stem Cells. J Med Chem 2017; 60:1309-1324. [PMID: 28112925 PMCID: PMC7938327 DOI: 10.1021/acs.jmedchem.6b01270] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Autotaxin (ATX, aka. ENPP2) is the main source of the lipid mediator lysophosphatidic acid (LPA) in biological fluids. This study reports on inhibitors of ATX derived by lead optimization of the benzene-sulfonamide in silico hit compound 3. The new analogues provide a comprehensive structure-activity relationship of the benzene-sulfonamide scaffold that yielded a series of highly potent ATX inhibitors. The three most potent analogues (3a, IC50 ∼ 32 nM; 3b, IC50 ∼ 9 nM; and 14, IC50 ∼ 35 nM) inhibit ATX-dependent invasion of A2058 human melanoma cells in vitro. Two of the most potent compounds, 3b and 3f (IC50 ∼ 84 nM), lack inhibitory action on ENPP6 and ENPP7 but possess weak antagonist action specific to the LPA1 G protein-coupled receptor. In particular, compound 3b potently reduced in vitro chemotherapeutic resistance of 4T1 breast cancer stem-like cells to paclitaxel and significantly reduced B16 melanoma metastasis in vivo.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Derek D. Norman
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Sue Chin Lee
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Abby L. Parrill
- Department of Chemistry, The University of Memphis, Memphis, Tennessee 38152, United States
- Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152, United States
| | - Truc Chi T. Pham
- Department of Chemistry, The University of Memphis, Memphis, Tennessee 38152, United States
| | - Daniel L. Baker
- Department of Chemistry, The University of Memphis, Memphis, Tennessee 38152, United States
| | - Gabor G. Tigyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D. Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
29
|
A rhodium(III)-based inhibitor of autotaxin with antiproliferative activity. Biochim Biophys Acta Gen Subj 2017; 1861:256-263. [DOI: 10.1016/j.bbagen.2016.11.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/09/2016] [Accepted: 11/21/2016] [Indexed: 12/17/2022]
|
30
|
Miller LM, Keune WJ, Castagna D, Young LC, Duffy EL, Potjewyd F, Salgado-Polo F, Engel García P, Semaan D, Pritchard JM, Perrakis A, Macdonald SJF, Jamieson C, Watson AJB. Structure-Activity Relationships of Small Molecule Autotaxin Inhibitors with a Discrete Binding Mode. J Med Chem 2017; 60:722-748. [PMID: 27982588 DOI: 10.1021/acs.jmedchem.6b01597] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autotaxin (ATX) is a secreted enzyme responsible for the hydrolysis of lysophosphatidylcholine (LPC) to the bioactive lysophosphatidic acid (LPA) and choline. The ATX-LPA signaling pathway is implicated in cell survival, migration, and proliferation; thus, the inhibition of ATX is a recognized therapeutic target for a number of diseases including fibrotic diseases, cancer, and inflammation, among others. Many of the developed synthetic inhibitors for ATX have resembled the lipid chemotype of the native ligand; however, a small number of inhibitors have been described that deviate from this common scaffold. Herein, we report the structure-activity relationships (SAR) of a previously reported small molecule ATX inhibitor. We show through enzyme kinetics studies that analogues of this chemotype are noncompetitive inhibitors, and by using a crystal structure with ATX we confirm the discrete binding mode.
Collapse
Affiliation(s)
- Lisa M Miller
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde , Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K
| | - Willem-Jan Keune
- Division of Biochemistry, Netherlands Cancer Institute/Antoni van Leeuwenhoek , Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands
| | - Diana Castagna
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde , Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K
| | - Louise C Young
- Strathclyde Institute of Pharmacy and Biomolecular Science, University of Strathclyde , John Arbuthnott Building (Hamnet Wing), 161 Cathedral Street, Glasgow G1 1XL, U.K
| | - Emma L Duffy
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde , Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K
| | - Frances Potjewyd
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde , Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K
| | - Fernando Salgado-Polo
- Division of Biochemistry, Netherlands Cancer Institute/Antoni van Leeuwenhoek , Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands
| | - Paloma Engel García
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde , Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K
| | - Dima Semaan
- Division of Biochemistry, Netherlands Cancer Institute/Antoni van Leeuwenhoek , Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands
| | - John M Pritchard
- Fibrosis Discovery Performance Unit, GlaxoSmithKline, Medicines Research Centre , Gunnels Wood Road, Stevenage SG1 2NY, U.K
| | - Anastassis Perrakis
- Division of Biochemistry, Netherlands Cancer Institute/Antoni van Leeuwenhoek , Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands
| | - Simon J F Macdonald
- Fibrosis Discovery Performance Unit, GlaxoSmithKline, Medicines Research Centre , Gunnels Wood Road, Stevenage SG1 2NY, U.K
| | - Craig Jamieson
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde , Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K
| | - Allan J B Watson
- WestCHEM, Department of Pure and Applied Chemistry, University of Strathclyde , Thomas Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K
| |
Collapse
|
31
|
Shah P, Cheasty A, Foxton C, Raynham T, Farooq M, Gutierrez IF, Lejeune A, Pritchard M, Turnbull A, Pang L, Owen P, Boyd S, Stowell A, Jordan A, Hamilton NM, Hitchin JR, Stockley M, MacDonald E, Quesada MJ, Trivier E, Skeete J, Ovaa H, Moolenaar WH, Ryder H. Discovery of potent inhibitors of the lysophospholipase autotaxin. Bioorg Med Chem Lett 2016; 26:5403-5410. [PMID: 27780639 DOI: 10.1016/j.bmcl.2016.10.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 12/15/2022]
Abstract
The autotaxin-lysophosphatidic acid (ATX-LPA) axis has been implicated in several disease conditions including inflammation, fibrosis and cancer. This makes ATX an attractive drug target and its inhibition may lead to useful therapeutic agents. Through a high throughput screen (HTS) we identified a series of small molecule inhibitors of ATX which have subsequently been optimized for potency, selectivity and developability properties. This has delivered drug-like compounds such as 9v (CRT0273750) which modulate LPA levels in plasma and are suitable for in vivo studies. X-ray crystallography has revealed that these compounds have an unexpected binding mode in that they do not interact with the active site zinc ions but instead occupy the hydrophobic LPC pocket extending from the active site of ATX together with occupying the LPA 'exit' channel.
Collapse
Affiliation(s)
- Pritom Shah
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Anne Cheasty
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Caroline Foxton
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Tony Raynham
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Muddasar Farooq
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Irene Farre Gutierrez
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Aurore Lejeune
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Michelle Pritchard
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Andrew Turnbull
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Leon Pang
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Paul Owen
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Susan Boyd
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Alexandra Stowell
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Allan Jordan
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Niall M Hamilton
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - James R Hitchin
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Martin Stockley
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Ellen MacDonald
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Mar Jimenez Quesada
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Elisabeth Trivier
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Jana Skeete
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Huib Ovaa
- Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Wouter H Moolenaar
- Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Hamish Ryder
- Cancer Research Technology, Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| |
Collapse
|
32
|
Katsamakas S, Papadopoulos AG, Hadjipavlou-Litina D. Boronic Acid Group: A Cumbersome False Negative Case in the Process of Drug Design. Molecules 2016; 21:molecules21091185. [PMID: 27617984 PMCID: PMC6273881 DOI: 10.3390/molecules21091185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 08/23/2016] [Accepted: 09/02/2016] [Indexed: 11/26/2022] Open
Abstract
Herein we present, an exhaustive docking analysis considering the case of autotaxin (ATX). HA155, a small molecule inhibitor of ATX, is co-crystallized. In order to further extract conclusions on the nature of the bond formed between the ligands and the amino acid residues of the active site, density functional theory (DFT) calculations were undertaken. However, docking does not provide reproducible results when screening boronic acid derivatives and their binding orientations to protein drug targets. Based on natural bond orbital (NBO) calculations, the formed bond between Ser/Thr residues is characterized more accurately as a polar covalent bond instead of a simple nonpolar covalent one. The presented results are acceptable and could be used in screening as an active negative filter for boron compounds. The hydroxyl groups of amino acids are bonded with the inhibitor’s boron atom, converting its hybridization to sp3.
Collapse
Affiliation(s)
- Sotirios Katsamakas
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece.
| | - Anastasios G Papadopoulos
- Laboratory of Applied Quantum Chemistry, School of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece.
| | - Dimitra Hadjipavlou-Litina
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece.
| |
Collapse
|
33
|
Discovery and synthetic optimization of a novel scaffold for hydrophobic tunnel-targeted autotaxin inhibition. Bioorg Med Chem 2016; 24:4660-4674. [PMID: 27544588 DOI: 10.1016/j.bmc.2016.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/01/2016] [Accepted: 08/03/2016] [Indexed: 12/11/2022]
Abstract
Autotaxin (ATX) is a ubiquitous ectoenzyme that hydrolyzes lysophosphatidylcholine (LPC) to form the bioactive lipid mediator lysophosphatidic acid (LPA). LPA activates specific G-protein coupled receptors to elicit downstream effects leading to cellular motility, survival, and invasion. Through these pathways, upregulation of ATX is linked to diseases such as cancer and cardiovascular disease. Recent crystal structures confirm that the catalytic domain of ATX contains multiple binding regions including a polar active site, hydrophobic tunnel, and a hydrophobic pocket. This finding is consistent with the promiscuous nature of ATX hydrolysis of multiple and diverse substrates and prior investigations of inhibitor impacts on ATX enzyme kinetics. The current study used virtual screening methods to guide experimental identification and characterization of inhibitors targeting the hydrophobic region of ATX. An initially discovered inhibitor, GRI392104 (IC50 4μM) was used as a lead for synthetic optimization. In total twelve newly synthesized inhibitors of ATX were more potent than GRI392104 and were selective for ATX as they had no effect on other LPC-specific NPP family members or on LPA1-5 GPCR.
Collapse
|
34
|
Castagna D, Budd DC, Macdonald SJF, Jamieson C, Watson AJB. Development of Autotaxin Inhibitors: An Overview of the Patent and Primary Literature. J Med Chem 2016; 59:5604-21. [DOI: 10.1021/acs.jmedchem.5b01599] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Diana Castagna
- WestCHEM, Department of Pure and Applied
Chemistry, University of Strathclyde, Glasgow, G1 1XL, U.K
| | - David C. Budd
- Medicines Research Centre, GlaxoSmithKline, Gunnel
Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| | - Simon J. F. Macdonald
- Medicines Research Centre, GlaxoSmithKline, Gunnel
Wood Road, Stevenage, Hertfordshire, SG1 2NY, U.K
| | - Craig Jamieson
- WestCHEM, Department of Pure and Applied
Chemistry, University of Strathclyde, Glasgow, G1 1XL, U.K
| | - Allan J. B. Watson
- WestCHEM, Department of Pure and Applied
Chemistry, University of Strathclyde, Glasgow, G1 1XL, U.K
| |
Collapse
|
35
|
Federico L, Jeong KJ, Vellano CP, Mills GB. Autotaxin, a lysophospholipase D with pleomorphic effects in oncogenesis and cancer progression. J Lipid Res 2016; 57:25-35. [PMID: 25977291 PMCID: PMC4689343 DOI: 10.1194/jlr.r060020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/07/2015] [Indexed: 12/18/2022] Open
Abstract
The ectonucleotide pyrophosphatase/phosphodiesterase type 2, more commonly known as autotaxin (ATX), is an ecto-lysophospholipase D encoded by the human ENNP2 gene. ATX is expressed in multiple tissues and participates in numerous key physiologic and pathologic processes, including neural development, obesity, inflammation, and oncogenesis, through the generation of the bioactive lipid, lysophosphatidic acid. Overwhelming evidence indicates that altered ATX activity leads to oncogenesis and cancer progression through the modulation of multiple hallmarks of cancer pathobiology. Here, we review the structural and catalytic characteristics of the ectoenzyme, how its expression and maturation processes are regulated, and how the systemic integration of its pleomorphic effects on cells and tissues may contribute to cancer initiation, progression, and therapy. Additionally, the up-to-date spectrum of the most frequent ATX genomic alterations from The Cancer Genome Atlas project is reported for a subset of cancers.
Collapse
Affiliation(s)
- Lorenzo Federico
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Kang Jin Jeong
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Christopher P Vellano
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| | - Gordon B Mills
- Department of Systems Biology, University of Texas M. D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
36
|
Stein AJ, Bain G, Prodanovich P, Santini AM, Darlington J, Stelzer NMP, Sidhu RS, Schaub J, Goulet L, Lonergan D, Calderon I, Evans JF, Hutchinson JH. Structural Basis for Inhibition of Human Autotaxin by Four Potent Compounds with Distinct Modes of Binding. Mol Pharmacol 2015; 88:982-92. [PMID: 26371182 DOI: 10.1124/mol.115.100404] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 09/11/2015] [Indexed: 12/17/2022] Open
Abstract
Autotaxin (ATX) is a secreted enzyme that hydrolyzes lysophosphatidylcholine to lysophosphatidic acid (LPA). LPA is a bioactive phospholipid that regulates diverse biological processes, including cell proliferation, migration, and survival/apoptosis, through the activation of a family of G protein-coupled receptors. The ATX-LPA pathway has been implicated in many pathologic conditions, including cancer, fibrosis, inflammation, cholestatic pruritus, and pain. Therefore, ATX inhibitors represent an attractive strategy for the development of therapeutics to treat a variety of diseases. Mouse and rat ATX have been crystallized previously with LPA or small-molecule inhibitors bound. Here, we present the crystal structures of human ATX in complex with four previously unpublished, structurally distinct ATX inhibitors. We demonstrate that the mechanism of inhibition of each compound reflects its unique interactions with human ATX. Our studies may provide a basis for the rational design of novel ATX inhibitors.
Collapse
Affiliation(s)
- Adam J Stein
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Gretchen Bain
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Pat Prodanovich
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Angelina M Santini
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Janice Darlington
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Nina M P Stelzer
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Ranjinder S Sidhu
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Jeffrey Schaub
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Lance Goulet
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Dave Lonergan
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Imelda Calderon
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - Jilly F Evans
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| | - John H Hutchinson
- Cayman Chemical Company, Ann Arbor, Michigan (A.J.S., N.M.P.S., R.S.S., J.S.); and PharmAkea, San Diego, California (G.B., P.P., A.M.S., J.D., L.G., D.L., I.C., J.F.E., J.H.H.)
| |
Collapse
|
37
|
Barbayianni E, Kaffe E, Aidinis V, Kokotos G. Autotaxin, a secreted lysophospholipase D, as a promising therapeutic target in chronic inflammation and cancer. Prog Lipid Res 2015; 58:76-96. [DOI: 10.1016/j.plipres.2015.02.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 01/20/2015] [Accepted: 02/12/2015] [Indexed: 02/07/2023]
|
38
|
Mortikov VY, Rodinovskaya LA, Fedorov AE, Shestopalov AM, Belyakov PA. Synthesis of heterocyclic compounds from 4-formylpyrazoles. Russ Chem Bull 2014. [DOI: 10.1007/s11172-014-0451-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
39
|
Bordessa A, Colin-Cassin C, Grillier-Vuissoz I, Kuntz S, Mazerbourg S, Husson G, Vo M, Flament S, Martin H, Chapleur Y, Boisbrun M. Optimization of troglitazone derivatives as potent anti-proliferative agents: towards more active and less toxic compounds. Eur J Med Chem 2014; 83:129-40. [PMID: 24953030 DOI: 10.1016/j.ejmech.2014.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 05/16/2014] [Accepted: 06/09/2014] [Indexed: 11/25/2022]
Abstract
Δ2-Troglitazone derivatives were shown to exhibit anti-proliferative activity in a PPARγ-independent manner. We prepared various compounds in order to increase their potency and decrease their toxicity towards non-malignant primary cultured hepatocytes. Many compounds induced viabilities less than 20% at 10 μM on various cancer cell lines. Furthermore, five of them showed hepatocyte viability of 80% or more at 200 μM. In addition, compounds 17 and 18 exhibited promising maximum tolerated doses on a murine model, enabling future investigations.
Collapse
Affiliation(s)
- Andrea Bordessa
- Université de Lorraine, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France; CNRS, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France
| | - Christelle Colin-Cassin
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Isabelle Grillier-Vuissoz
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Sandra Kuntz
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Sabine Mazerbourg
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Gauthier Husson
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Myriam Vo
- Université de Lorraine, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France; CNRS, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France
| | - Stéphane Flament
- Université de Lorraine, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France; CNRS, CRAN, UMR 7039, BP 70239, F-54506 Vandœuvre-lès-Nancy Cedex, France
| | - Hélène Martin
- Université de Franche-Comté, Laboratoire de Toxicologie Cellulaire, EA 4267, 25030 Besançon Cedex, France
| | - Yves Chapleur
- Université de Lorraine, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France; CNRS, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France
| | - Michel Boisbrun
- Université de Lorraine, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France; CNRS, SRSMC, UMR 7565, BP 70239, F-54506 Vandœuvre-lès-Nancy, France.
| |
Collapse
|
40
|
Albers HMHG, Kuijl C, Bakker J, Hendrickx L, Wekker S, Farhou N, Liu N, Blasco-Moreno B, Scanu T, den Hertog J, Celie P, Ovaa H, Neefjes J. Integrating chemical and genetic silencing strategies to identify host kinase-phosphatase inhibitor networks that control bacterial infection. ACS Chem Biol 2014; 9:414-22. [PMID: 24274083 PMCID: PMC3934374 DOI: 10.1021/cb400421a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
![]()
Every
year three million people die as a result of bacterial infections,
and this number may further increase due to resistance to current
antibiotics. These antibiotics target almost all essential bacterial
processes, leaving only a few new targets for manipulation. The host
proteome has many more potential targets for manipulation in order
to control bacterial infection, as exemplified by the observation
that inhibiting the host kinase Akt supports the elimination of different
intracellular bacteria including Salmonella and M. tuberculosis. If host kinases are involved in the control
of bacterial infections, phosphatases could be as well. Here we present
an integrated small interference RNA and small molecule screen to
identify host phosphatase-inhibitor combinations that control bacterial
infection. We define host phosphatases inhibiting intracellular growth
of Salmonella and identify corresponding inhibitors
for the dual specificity phosphatases DUSP11 and 27. Pathway analysis
places many kinases and phosphatases controlling bacterial infection
in an integrated pathway centered around Akt. This network controls
host cell metabolism, survival, and growth and bacterial survival
and reflect a natural host cell response to bacterial infection. Inhibiting
two enzyme classes with opposite activities–kinases and phosphatases–may
be a new strategy to overcome infections by antibiotic-resistant bacteria.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bernat Blasco-Moreno
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Doctor Aiguader, 88, 08003 Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
41
|
Fells JI, Lee SC, Norman DD, Tsukahara R, Kirby JR, Nelson S, Seibel W, Papoian R, Patil R, Miller DD, Parrill AL, Pham TC, Baker DL, Bittman R, Tigyi G. Targeting the hydrophobic pocket of autotaxin with virtual screening of inhibitors identifies a common aromatic sulfonamide structural motif. FEBS J 2014; 281:1017-28. [PMID: 24314137 DOI: 10.1111/febs.12674] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 10/31/2013] [Accepted: 11/27/2013] [Indexed: 12/20/2022]
Abstract
Modulation of autotaxin (ATX), the lysophospholipase D enzyme that produces lysophosphatidic acid, with small-molecule inhibitors is a promising strategy for blocking the ATX-lysophosphatidic acid signaling axis. Although discovery campaigns have been successful in identifying ATX inhibitors, many of the reported inhibitors target the catalytic cleft of ATX. A recent study provided evidence for an additional inhibitory surface in the hydrophobic binding pocket of ATX, confirming prior studies that relied on enzyme kinetics and differential inhibition of substrates varying in size. Multiple hits from previous high-throughput screening for ATX inhibitors were obtained with aromatic sulfonamide derivatives interacting with the hydrophobic pocket. Here, we describe the development of a ligand-based strategy and its application in virtual screening, which yielded novel high-potency inhibitors that target the hydrophobic pocket of ATX. Characterization of the structure-activity relationship of these new inhibitors forms the foundation of a new pharmacophore model of the hydrophobic pocket of ATX.
Collapse
Affiliation(s)
- James I Fells
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Scott SA, Mathews TP, Ivanova PT, Lindsley CW, Brown HA. Chemical modulation of glycerolipid signaling and metabolic pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:1060-84. [PMID: 24440821 DOI: 10.1016/j.bbalip.2014.01.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 01/04/2023]
Abstract
Thirty years ago, glycerolipids captured the attention of biochemical researchers as novel cellular signaling entities. We now recognize that these biomolecules occupy signaling nodes critical to a number of physiological and pathological processes. Thus, glycerolipid-metabolizing enzymes present attractive targets for new therapies. A number of fields-ranging from neuroscience and cancer to diabetes and obesity-have elucidated the signaling properties of glycerolipids. The biochemical literature teems with newly emerging small molecule inhibitors capable of manipulating glycerolipid metabolism and signaling. This ever-expanding pool of chemical modulators appears daunting to those interested in exploiting glycerolipid-signaling pathways in their model system of choice. This review distills the current body of literature surrounding glycerolipid metabolism into a more approachable format, facilitating the application of small molecule inhibitors to novel systems. This article is part of a Special Issue entitled Tools to study lipid functions.
Collapse
Affiliation(s)
- Sarah A Scott
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Thomas P Mathews
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Pavlina T Ivanova
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - H Alex Brown
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
43
|
Kawaguchi M, Okabe T, Okudaira S, Nishimasu H, Ishitani R, Kojima H, Nureki O, Aoki J, Nagano T. Screening and X-ray crystal structure-based optimization of autotaxin (ENPP2) inhibitors, using a newly developed fluorescence probe. ACS Chem Biol 2013; 8:1713-21. [PMID: 23688339 DOI: 10.1021/cb400150c] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Autotaxin (ATX), also known as ectonucleotide pyrophosphatase/phosphodiesterase 2 (ENPP2), was originally identified as a tumor cell autocrine motility factor and was found to be identical to plasma lysophospholipase D, which is the predominant contributor to lysophosphatidic acid (LPA) production from lysophospholipids. ATX is therefore considered to regulate the physiological and pathological roles of LPA, including angiogenesis, lymphocyte trafficking, tissue fibrosis, and cancer cell invasion and metastasis. Thus, it is a potential therapeutic target. Here, we first developed a sensitive and specific ATX fluorescence probe, TG-mTMP, and used it to screen ATX inhibitors in a large chemical library. This probe, which is superior to previously available probes FS-3 and CPF4 in terms of sensitivity or specificity, enabled us to identify several novel ATX inhibitor scaffolds. We solved the crystal structures of ATX complexes with the hit compounds at high resolution (1.75-1.95 Å) and used this information to guide optimization of the structure of a selected inhibitor. The optimized compounds, 3BoA and its derivatives, exhibited potent ATX-inhibitory activity both in vitro and in vivo. These inhibitors are expected to be useful tools to understand the roles of ATX in vitro and in vivo and may also be candidate anti-ATX therapeutic agents.
Collapse
Affiliation(s)
| | - Takayoshi Okabe
- Open Innovation
Center for Drug
Discovery, The University of Tokyo, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shinichi Okudaira
- Graduate School of Pharmaceutical
Sciences, Tohoku University, 6-3 Aoba,
Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | | | | | - Hirotatsu Kojima
- Open Innovation
Center for Drug
Discovery, The University of Tokyo, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | - Junken Aoki
- Graduate School of Pharmaceutical
Sciences, Tohoku University, 6-3 Aoba,
Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Tetsuo Nagano
- Open Innovation
Center for Drug
Discovery, The University of Tokyo, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
44
|
Norman DD, Ibezim A, Scott WE, White S, Parrill AL, Baker DL. Autotaxin inhibition: development and application of computational tools to identify site-selective lead compounds. Bioorg Med Chem 2013; 21:5548-60. [PMID: 23816044 DOI: 10.1016/j.bmc.2013.05.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 05/20/2013] [Accepted: 05/28/2013] [Indexed: 01/29/2023]
Abstract
Autotaxin (ATX) catalyzes the conversion of lysophosphatidyl choline (LPC) to lysophosphatidic acid (LPA). Both ATX and LPA have been linked to pathophysiologies ranging from cancer to neuropathic pain. Inhibition of LPA production by ATX is therefore of therapeutic interest. Here we report the application of previously-developed, subsite-targeted pharmacophore models in a screening workflow that involves either docking or binary QSAR as secondary filters to identify ATX inhibitors from previously unreported structural types, four of which have sub-micromolar inhibition constants. Cell-based assays demonstrate that ATX inhibition and cytotoxicity structure-activity-relationships (SAR) exhibit selectivity cliffs, characterized by structurally similar compounds exhibiting similar biological activities with respect to ATX inhibition but very different biological activities with respect to cytotoxicity. Thus, general cytotoxicity should not be used as an early filter to eliminate candidate ATX inhibitor scaffolds from further SAR studies. Assays using two substrates of vastly different sizes demonstrate that the tools developed to identify compounds binding outside the central core of the active site did identify compounds acting at an allosteric site. In contrast, tools developed to identify active-site directed compounds did not identify active-site directed compounds. The stronger volume overlap imposed when selecting screening candidates expected to bind outside the active site is likely responsible for the stronger match between intended and actual target site.
Collapse
Affiliation(s)
- Derek D Norman
- Department of Chemistry, The University of Memphis, Memphis, TN 38152, United States
| | | | | | | | | | | |
Collapse
|
45
|
Barbayianni E, Magrioti V, Moutevelis-Minakakis P, Kokotos G. Autotaxin inhibitors: a patent review. Expert Opin Ther Pat 2013; 23:1123-32. [PMID: 23641951 DOI: 10.1517/13543776.2013.796364] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Autotaxin (ATX) is a lysophospholipase D enzyme that hydrolyzes lysophosphatidylcholine to lysophosphatidic acid (LPA) and choline. LPA is a bioactive lipid mediator that activates several transduction pathways, and is involved in migration, proliferation and survival of various cells. Thus, ATX is an attractive medicinal target. AREAS COVERED The aim of this review is to summarize ATX inhibitors, reported in patents from 2006 up to now, describing their discovery and biological evaluation. EXPERT OPINION ATX has been implicated in various pathological conditions, such as cancer, chronic inflammation, neuropathic pain, fibrotic diseases, etc. Although there is an intensive effort on the discovery of potent and selective ATX inhibitors in order to identify novel medicinal agents, up to now, no ATX inhibitor has reached clinical trials. However, the use of ATX inhibitors seems an attractive strategy for the development of novel medicinal agents, for example anticancer therapeutics.
Collapse
Affiliation(s)
- Efrosini Barbayianni
- University of Athens, Department of Chemistry, Laboratory of Organic Chemistry, Panepistimiopolis, Athens 15771, Greece
| | | | | | | |
Collapse
|
46
|
Jiménez-Aligaga K, Bermejo-Bescós P, Martín-Aragón S, Csákÿ AG. Discovery of alkenylboronic acids as neuroprotective agents affecting multiple biological targets involved in Alzheimer's disease. Bioorg Med Chem Lett 2012; 23:426-9. [PMID: 23219701 DOI: 10.1016/j.bmcl.2012.11.068] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 11/15/2012] [Accepted: 11/18/2012] [Indexed: 12/30/2022]
Abstract
Alkenylboronic acids have shown important biological activities that contribute to neuroprotection. We have determined their influence on the β-amyloid (βA) aggregation process, β-secretase and acethylcholinesterase activities on cell-free systems, on the redox and lipid peroxidation status, and on the vulnerability to apoptotic death in an APPswe neuroblastoma cell line, before and after hydrogen peroxide treatment. We have discovered that 2-arylvinylboronic acids and some of their esters possess a set of properties which makes them highly useful as neuroprotective agents affecting multiple biological targets involved in AD. These properties are not paralleled by the related 2-arylboronic acids.
Collapse
Affiliation(s)
- Karim Jiménez-Aligaga
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | | | | | | |
Collapse
|
47
|
Abstract
Lysophosphatidic acid (LPA; monoacyl-glycerol-3-phosphate) is a lipid mediator that functions as a mitogen and motility factor for many cell types. LPA signals through six specific G protein-coupled receptors, named LPA(1-6), which trigger both overlapping and distinct signaling pathways. LPA is produced from extracellular lysophosphatidylcholine by a secreted lysophospholipase D, named autotaxin (ATX), originally identified as an "autocrine motility factor" for tumor cells. ATX-LPA signaling is vital for embryonic development and promotes tumor formation, angiogenesis, and experimental metastasis in mice. Elevated expression of ATX and/or aberrant expression of LPA receptors are found in several human malignancies, while loss of LPA(6) function has been implicated in bladder cancer. In this review, we summarize our present understanding of ATX and LPA receptor signaling in cancer.
Collapse
Affiliation(s)
- Anna J S Houben
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
48
|
Nishimasu H, Ishitani R, Aoki J, Nureki O. A 3D view of autotaxin. Trends Pharmacol Sci 2012; 33:138-45. [DOI: 10.1016/j.tips.2011.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 12/16/2011] [Accepted: 12/23/2011] [Indexed: 12/26/2022]
|
49
|
Affiliation(s)
- Harald M H G Albers
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
50
|
Elliott TS, Slowey A, Ye Y, Conway SJ. The use of phosphate bioisosteres in medicinal chemistry and chemical biology. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20079a] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|