1
|
Yuan Y, Chen S, Hu R, Wang X. MutualDTA: An Interpretable Drug-Target Affinity Prediction Model Leveraging Pretrained Models and Mutual Attention. J Chem Inf Model 2025. [PMID: 39878060 DOI: 10.1021/acs.jcim.4c01893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Efficient and accurate drug-target affinity (DTA) prediction can significantly accelerate the drug development process. Recently, deep learning models have been widely applied to DTA prediction and have achieved notable success. However, existing methods often encounter several common issues: first, the data representations lack sufficient information; second, the extracted features are not comprehensive; and third, most methods lack interpretability when modeling drug-target binding. To overcome the above-mentioned problems, we propose an interpretable deep learning model called MutualDTA for predicting DTA. MutualDTA leverages the power of pretrained models to obtain accurate representations of drugs and targets. It also employs well-designed modules to extract hidden features from these representations. Furthermore, the interpretability of MutualDTA is realized by the Mutual-Attention module, which (i) establishes relationships between drugs and proteins from the perspective of intermolecular interactions between drug atoms and protein amino acid residues and (ii) allows MutualDTA to capture the binding sites based on attention scores. The test results on two benchmark data sets show that MutualDTA achieves the best performance compared to the 12 state-of-the-art models. Attention visualization experiments show that MutualDTA can capture partial interaction sites, which not only helps drug developers reduce the search space for binding sites, but also demonstrates the interpretability of MutualDTA. Finally, the trained MutualDTA is applied to screen high-affinity drug screens targeting Alzheimer's disease (AD)-related proteins, and the screened drugs are partially present in the anti-AD drug library. These results demonstrate the reliability of MutualDTA in drug development.
Collapse
Affiliation(s)
- Yongna Yuan
- School of Information Science & Engineering, Lanzhou University, Lanzhou 730000, China
| | - Siming Chen
- School of Information Science & Engineering, Lanzhou University, Lanzhou 730000, China
| | - Rizhen Hu
- School of Information Science & Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xin Wang
- School of Information Science & Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
2
|
Gulzar M, Noor S, Hasan GM, Hassan MI. The role of serum and glucocorticoid-regulated kinase 1 in cellular signaling: Implications for drug development. Int J Biol Macromol 2024; 258:128725. [PMID: 38092114 DOI: 10.1016/j.ijbiomac.2023.128725] [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: 08/10/2023] [Revised: 11/23/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) is a ubiquitously expressed protein belonging to the Ser/Thr kinase family. It regulates diverse physiological processes, including epithelial sodium channel activity, hypertension, cell proliferation, and insulin sensitivity. Due to its significant role in the pathogenesis of numerous diseases, SGK1 can be exploited as a potential therapeutic target to address challenging health problems. SGK1 is associated with the development of obesity, and its overexpression enhances the sodium-glucose co-transporter 1 activity, which absorbs intestinal glucose. This review highlighted the detailed functional significance of SGK1 signaling and role in different diseases and subsequent therapeutic targeting. We aim to provide deeper mechanistic insights into understanding the pathogenesis and recent advancements in the SGK1 targeted drug development process. Small-molecule inhibitors are being developed with excellent binding affinity and improved SGK1 inhibition with desired selectivity. We have discussed small molecule inhibitors designed explicitly as potent SGK1 inhibitors and their therapeutic implications in various diseases. We further addressed the therapeutic potential and mechanism of action of these SGK1 inhibitors and provided a strong scientific foundation for developing effective therapeutics.
Collapse
Affiliation(s)
- Mehak Gulzar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
3
|
Maestro I, Madruga E, Boya P, Martínez A. Identification of a new structural family of SGK1 inhibitors as potential neuroprotective agents. J Enzyme Inhib Med Chem 2023; 38:2153841. [PMID: 36637025 PMCID: PMC9848319 DOI: 10.1080/14756366.2022.2153841] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
SGK1 is a serine/threonine kinase involved in several neurodegenerative-related pathways such as apoptosis, neuroinflammation, ionic channel regulation, and autophagy, among others. Despite its potential role as a pharmacological target against this kind of diseases, there are no reported inhibitors able to cross the BBB so far, being a field yet to be explored. In this context, a structure-based virtual screening against this kinase was performed, pointing out the deazapurine moiety as an interesting and easy-to-derivatize scaffold. Moreover, these inhibitors are able to i) exert neuroprotection in an in vitro model of AD and ii) block mitophagy in a PRKN-independent manner, reinforcing the hypothesis of SGK1 inhibitors as neuroprotective chemical tools.
Collapse
Affiliation(s)
- Ines Maestro
- Centro de Investigaciones, Biológicas Margarita Salas-CSIC, Madrid, Spain,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Enrique Madruga
- Centro de Investigaciones, Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Patricia Boya
- Centro de Investigaciones, Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciones, Biológicas Margarita Salas-CSIC, Madrid, Spain,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain,CONTACT Ana Martínez CIB-CSIC, Ramiro Maeztu 9, Madrid, 28040, Spain
| |
Collapse
|
4
|
Jang H, Park Y, Jang J. Serum and glucocorticoid-regulated kinase 1: Structure, biological functions, and its inhibitors. Front Pharmacol 2022; 13:1036844. [PMID: 36457711 PMCID: PMC9706101 DOI: 10.3389/fphar.2022.1036844] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/20/2022] [Indexed: 08/11/2023] Open
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) is a serine/threonine kinase belonging to the protein kinase A, G, and C (AGC) family. Upon initiation of the phosphoinositide 3-kinase (PI3K) signaling pathway, mammalian target of rapamycin complex 2 (mTORC2) and phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylate the hydrophobic motif and kinase domain of SGK1, respectively, inducing SGK1 activation. SGK1 modulates essential cellular processes such as proliferation, survival, and apoptosis. Hence, dysregulated SGK1 expression can result in multiple diseases, including hypertension, cancer, autoimmunity, and neurodegenerative disorders. This review provides a current understanding of SGK1, particularly in sodium transport, cancer progression, and autoimmunity. In addition, we summarize the developmental status of SGK1 inhibitors, their structures, and respective potencies evaluated in pre-clinical experimental settings. Collectively, this review highlights the significance of SGK1 and proposes SGK1 inhibitors as potential drugs for treatment of clinically relevant diseases.
Collapse
Affiliation(s)
- Hyunsoo Jang
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Youngjun Park
- Laboratory of Immune and Inflammatory Disease, College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, South Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, South Korea
| | - Jaebong Jang
- College of Pharmacy, Korea University, Sejong, South Korea
| |
Collapse
|
5
|
Wittmann C, Bacher F, Enyedy EA, Dömötör O, Spengler G, Madejski C, Reynisson J, Arion VB. Highly Antiproliferative Latonduine and Indolo[2,3- c]quinoline Derivatives: Complex Formation with Copper(II) Markedly Changes the Kinase Inhibitory Profile. J Med Chem 2022; 65:2238-2261. [PMID: 35104137 PMCID: PMC8842277 DOI: 10.1021/acs.jmedchem.1c01740] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
![]()
A series of latonduine
and indoloquinoline derivatives HL1–HL8 and their copper(II)
complexes (1–8) were synthesized and comprehensively
characterized. The structures of five compounds (HL6, [CuCl(L1)(DMF)]·DMF, [CuCl(L2)(CH3OH)], [CuCl(L3)]·0.5H2O, and [CuCl2(H2L5)]Cl·2DMF) were elucidated
by single crystal X-ray diffraction. The copper(II) complexes revealed
low micro- to sub-micromolar IC50 values with promising
selectivity toward human colon adenocarcinoma multidrug-resistant
Colo320 cancer cells as compared to the doxorubicin-sensitive Colo205
cell line. The lead compounds HL4 and 4 as well as HL8 and 8 induced apoptosis efficiently in Colo320 cells. In addition, the
copper(II) complexes had higher affinity to DNA than their metal-free
ligands. HL8 showed selective inhibition for
the PIM-1 enzyme, while 8 revealed strong inhibition
of five other enzymes, i.e., SGK-1, PKA, CaMK-1, GSK3β, and
MSK1, from a panel of 50 kinases. Furthermore, molecular modeling
of the ligands and complexes showed a good fit to the binding pockets
of these targets.
Collapse
Affiliation(s)
- Christopher Wittmann
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse, 42, Vienna A1090, Austria
| | - Felix Bacher
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse, 42, Vienna A1090, Austria
| | - Eva A Enyedy
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary.,MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Orsolya Dömötör
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary.,MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary.,Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, Szeged H-6725, Hungary
| | - Christian Madejski
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse, 42, Vienna A1090, Austria
| | - Jóhannes Reynisson
- School of Pharmacy and Bioengineering, Keele University, Hornbeam Building, Staffordshire ST5 5BG, United Kingdom
| | - Vladimir B Arion
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse, 42, Vienna A1090, Austria
| |
Collapse
|
6
|
Zhang S, Wang Y, Yu M, Shang Y, Chang Y, Zhao H, Kang Y, Zhao L, Xu L, Zhao X, Difrancesco D, Baruscotti M, Wang Y. Discovery of Herbacetin as a Novel SGK1 Inhibitor to Alleviate Myocardial Hypertrophy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2101485. [PMID: 34761560 PMCID: PMC8805583 DOI: 10.1002/advs.202101485] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 10/17/2021] [Indexed: 05/05/2023]
Abstract
Cardiac hypertrophy is a pivotal pathophysiological step of various cardiovascular diseases, which eventually leads to heart failure and death. Extracts of Rhodiola species (Ext.R), a class of commonly used medicinal herbs in Europe and East Asia, can attenuate cardiac hypertrophy both in vitro and in vivo. Serum/glucocorticoid regulated kinase 1 (SGK1) is identified as a potential target of Ext. R. By mass spectrometry-based kinase inhibitory assay, herbacetin (HBT) from Ext.R is identified as a novel SGK1 inhibitor with IC50 of 752 nmol. Thermal shift assay, KINOMEscan in vitro assay combined with molecular docking proves a direct binding between HBT and SGK1. Site-specific mutation of Asp177 in SGK1 completely ablates the inhibitory activity of HBT. The presence of OH groups at the C-3, C-8, C-4' positions of flavonoids is suggested to be favorable for the inhibition of SGK1 activity. Finally, HBT significantly suppresses cardiomyocyte hypertrophy in vitro and in vivo, reduces reactive oxygen species (ROS) synthesis and calcium accumulation. HBT decreases phosphorylation of SGK1 and regulates its downstream forkhead box protein O1 (FoxO1) signaling pathway. Taken together, the findings suggest that a panel of flavonoids structurally related to HBT may be novel leads for developing new therapeutics against cardiac hypertrophy.
Collapse
Affiliation(s)
- Shujing Zhang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Yingchao Wang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Min Yu
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Ye Shang
- State Key Laboratory of Component‐Based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Yanxu Chang
- State Key Laboratory of Component‐Based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Hong Zhao
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Yu Kang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Lu Zhao
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
| | - Lei Xu
- Institute of Bioinformatics and Medical EngineeringSchool of Electrical and Information EngineeringJiangsu University of TechnologyChangzhouJiangsu213001China
| | - Xiaoping Zhao
- School of Basic Medical SciencesZhejiang Chinese Medical UniversityHangzhou310053China
| | | | | | - Yi Wang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058China
- State Key Laboratory of Component‐Based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjin301617China
| |
Collapse
|
7
|
Halland N, Schmidt F, Weiss T, Li Z, Czech J, Saas J, Ding-Pfennigdorff D, Dreyer MK, Strübing C, Nazare M. Rational Design of Highly Potent, Selective, and Bioavailable SGK1 Protein Kinase Inhibitors for the Treatment of Osteoarthritis. J Med Chem 2021; 65:1567-1584. [PMID: 34931844 DOI: 10.1021/acs.jmedchem.1c01601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The serine/threonine kinase SGK1 is an activator of the β-catenin pathway and a powerful stimulator of cartilage degradation that is found to be upregulated under genomic control in diseased osteoarthritic cartilage. Today, no oral disease-modifying treatments are available and chronic treatment in this indication sets high requirements for the drug selectivity, pharmacokinetic, and safety profile. We describe the identification of a highly selective druglike 1H-pyrazolo[3,4-d]pyrimidine SGK1 inhibitor 17a that matches both safety and pharmacokinetic requirements for oral dosing. Rational compound design was facilitated by a novel hSGK1 co-crystal structure, and multiple ligand-based computer models were applied to guide the chemical optimization of the compound ADMET and selectivity profiles. Compounds were selected for subchronic proof of mechanism studies in the mouse femoral head cartilage explant model, and compound 17a emerged as a druglike SGK1 inhibitor, with a highly optimized profile suitable for oral dosing as a novel, potentially disease-modifying agent for osteoarthritis.
Collapse
Affiliation(s)
- Nis Halland
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Friedemann Schmidt
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Tilo Weiss
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Ziyu Li
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Jörg Czech
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Joachim Saas
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | | | - Matthias K Dreyer
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Carsten Strübing
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Marc Nazare
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
| |
Collapse
|
8
|
Lautens MJ, Tan JH, Serrat X, Del Borrello S, Schertzberg MR, Fraser AG. Identification of enzymes that have helminth-specific active sites and are required for Rhodoquinone-dependent metabolism as targets for new anthelmintics. PLoS Negl Trop Dis 2021; 15:e0009991. [PMID: 34843467 PMCID: PMC8659336 DOI: 10.1371/journal.pntd.0009991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/09/2021] [Accepted: 11/11/2021] [Indexed: 11/18/2022] Open
Abstract
Soil transmitted helminths (STHs) are major human pathogens that infect over a billion people. Resistance to current anthelmintics is rising and new drugs are needed. Here we combine multiple approaches to find druggable targets in the anaerobic metabolic pathways STHs need to survive in their mammalian host. These require rhodoquinone (RQ), an electron carrier used by STHs and not their hosts. We identified 25 genes predicted to act in RQ-dependent metabolism including sensing hypoxia and RQ synthesis and found 9 are required. Since all 9 have mammalian orthologues, we used comparative genomics and structural modeling to identify those with active sites that differ between host and parasite. Together, we found 4 genes that are required for RQ-dependent metabolism and have different active sites. Finding these high confidence targets can open up in silico screens to identify species selective inhibitors of these enzymes as new anthelmintics.
Collapse
Affiliation(s)
- Margot J. Lautens
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - June H. Tan
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Xènia Serrat
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Andrew G. Fraser
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
9
|
Chen Q, Tu Y, Mak S, Chen J, Lu J, Chen C, Yang X, Wang S, Wen S, Ma S, Li M, Han Y, Wah-Keung Tsim K, Pi R. Discovery of a novel small molecule PT109 with multi-targeted effects against Alzheimer's disease in vitro and in vivo. Eur J Pharmacol 2020; 883:173361. [PMID: 32673674 DOI: 10.1016/j.ejphar.2020.173361] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 11/27/2022]
Abstract
Alzheimer's disease (AD), which is characterized by impairment of cognitive functions, is a chronic neurodegenerative disease that mainly affects the elderly. Currently available anti-AD drugs can only offer limited symptom-relieving effects. "One-compound-Multitargeted Strategy" have been recognized as the promising way to win the war against AD. Herein we report a potential anti-AD agent PT109 with multi-functions. First, an 81-kinase screening was carried out and results showed that PT109 potently inhibited c-Jun N-terminal kinases and Serum and glucocorticoid-inducible kinase 1, which are the important signaling molecules involved in neurogenesis, neuroprotection and neuroinflammation and mildly inhibit glycogen synthase kinase-3β as well as protein kinase C gamma, both are involved in AD pathological processes. In addition, invitro studies of immunofluorescent staining and Western blot showed that PT109 might promote the neurogenesis of C17.2 cells and induce synaptogenesis in primary cultured rat hippocampal neurons. We detected and confirmed the neuroprotective effect of PT109 in cultured HT22 cells by MTT assay, dehydrogenase assay, glutathione assay and reactive oxygen species assay. Furthermore, the results of Western blot, ELISA assay and immunofluorescent staining indicated that PT109 attenuated lipopolysaccharide-induced inflammation in BV2 cells and primary astrocytes. The results of Morris water maze and Step-through test indicated that PT109 improved the spatial learning ability in APP/PS1 mice. More importantly, the invivo pharmacokinetic parameters indicated that PT109 had better medicinal properties. Taken together, our findings suggest that PT109 may be a promising candidate for treating AD through multiple targets although further studies are ought to be conducted.
Collapse
Affiliation(s)
- Qiuhe Chen
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, 510006, China
| | - Yalin Tu
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, 510006, China
| | - Shinghung Mak
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong
| | - Jingkao Chen
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, 510006, China
| | - Junfeng Lu
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, 510006, China
| | - Chen Chen
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, 510006, China
| | - Xiaohong Yang
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, 510006, China
| | - Shengnan Wang
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, 510006, China
| | - Shijun Wen
- Cancer Center of South China, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shanshan Ma
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mingtao Li
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong
| | - Karl Wah-Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong
| | - Rongbiao Pi
- Department of Pharmacology, School of Medicine, Sun Yat-sen University, Guangzhou, 510006, China; Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou, 510006, China.
| |
Collapse
|
10
|
Maestro I, Boya P, Martinez A. Serum- and glucocorticoid-induced kinase 1, a new therapeutic target for autophagy modulation in chronic diseases. Expert Opin Ther Targets 2020; 24:231-243. [PMID: 32067528 DOI: 10.1080/14728222.2020.1730328] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Autophagy, a basic cellular degradation pathway essential for survival, is altered both in aging and in many chronic human diseases, including infections, cancer, heart disease, and neurodegeneration. Identifying new therapeutic targets for the control and modulation of autophagy events is therefore of utmost importance in drug discovery. Serum and glucocorticoid activated kinase 1 (SGK1), known for decades for its role in ion channel modulation, is now known to act as a switch for autophagy homeostasis, and has emerged as a novel and important therapeutic target likely to attract considerable research attention in the coming years.Areas covered: In this general review of SGK1 we describe the kinase's structure and its roles in physiological and pathological contexts. We also discuss small-molecule modulators of SGK1 activity. These modulators are of particular interest to medicinal chemists and pharmacists seeking to develop more potent and selective drug candidates for SGK1, which, despite its key role in autophagy, remains relatively understudied.Expert opinion: The main future challenges in this area are (i) deciphering the role of SGK1 in selective autophagy processes (e.g. mitophagy, lipophagy, and aggrephagy); (ii) identifying selective allosteric modulators of SGK1 with specific biological functions; and (iii) conducting first-in-man clinical studies.
Collapse
Affiliation(s)
- Inés Maestro
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Patricia Boya
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
11
|
Zhang Z, Xu Q, Song C, Mi B, Zhang H, Kang H, Liu H, Sun Y, Wang J, Lei Z, Guan H, Li F. Serum- and Glucocorticoid-inducible Kinase 1 is Essential for Osteoclastogenesis and Promotes Breast Cancer Bone Metastasis. Mol Cancer Ther 2020; 19:650-660. [PMID: 31694887 DOI: 10.1158/1535-7163.mct-18-0783] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/30/2018] [Accepted: 10/31/2019] [Indexed: 11/16/2022]
Abstract
Bone metastasis is a severe complication associated with various carcinomas. It causes debilitating pain and pathologic fractures and dramatically impairs patients' quality of life. Drugs aimed at osteoclast formation significantly reduce the incidence of skeletal complications and are currently the standard treatment for patients with bone metastases. Here, we reported that serum- and glucocorticoid-inducible kinase 1 (SGK1) plays a pivotal role in the formation and function of osteoclasts by regulating the Ca2+ release-activated Ca2+ channel Orai1. We showed that SGK1 inhibition represses osteoclastogenesis in vitro and prevents bone loss in vivo Furthermore, we validated the effect of SGK1 on bone metastasis by using an intracardiac injection model in mice. Inhibition of SGK1 resulted in a significant reduction in bone metastasis. Subsequently, the Oncomine and the OncoLnc database were employed to verify the differential expression and the association with clinical outcome of SGK1 gene in patients with breast cancer. Our data mechanistically demonstrated the regulation of the SGK1 in the process of osteoclastogenesis and revealed SGK1 as a valuable target for curing bone metastasis diseases.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Xu
- Department of Hematology, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao Song
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Baoguo Mi
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China
| | - Honghua Zhang
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Honglei Kang
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huiyong Liu
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunlong Sun
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jia Wang
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhuowei Lei
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hanfeng Guan
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Feng Li
- Department of Orthopedics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
12
|
Schoene J, Gazzi T, Lindemann P, Christmann M, Volkamer A, Nazaré M. Probing 2
H
‐Indazoles as Templates for SGK1, Tie2, and SRC Kinase Inhibitors. ChemMedChem 2019; 14:1514-1527. [DOI: 10.1002/cmdc.201900328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/26/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Jens Schoene
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Thais Gazzi
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Peter Lindemann
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
| | - Mathias Christmann
- Organische ChemieInstitut für Chemie und BiochemieFreie Universität Berlin Takustrasse. 3 14195 Berlin Germany
| | - Andrea Volkamer
- In silico Toxicology and Structural Bioinformatics Group, Institute of PhysiologyCharité—Universitätsmedizin Berlin Charitéplatz 1 10117 Berlin Germany
| | - Marc Nazaré
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Campus BerlinBuch Robert-Roessle-Str. 10 13125 Berlin Germany
- Anna-Louisa-Karsch-Str. 2 10178 Berlin Germany
| |
Collapse
|
13
|
Drießen D, Stuhldreier F, Frank A, Stark H, Wesselborg S, Stork B, Müller TJJ. Novel meriolin derivatives as rapid apoptosis inducers. Bioorg Med Chem 2019; 27:3463-3468. [PMID: 31248707 DOI: 10.1016/j.bmc.2019.06.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/28/2019] [Accepted: 06/18/2019] [Indexed: 12/14/2022]
Abstract
3-(Hetero)aryl substituted 7-azaindoles possessing multikinase inhibitor activity are readily accessed in a one-pot Masuda borylation-Suzuki coupling sequence. Several promising derivatives were identified as apoptosis inducers and, emphasizing the multikinase inhibition potential, as sphingosine kinase 2 inhibitors. Our measurements provide additional insights into the structure-activity relationship of meriolin derivatives, suggesting derivatives bearing a pyridine moiety with amino groups in 2-position as most active anticancer compounds and thus as highly promising candidates for future in vivo studies.
Collapse
Affiliation(s)
- Daniel Drießen
- Institut für Organische Chemie und Makromolekulare Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Fabian Stuhldreier
- Institut für Molekulare Medizin I, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Annika Frank
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße1, D-40225 Düsseldorf, Germany
| | - Holger Stark
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße1, D-40225 Düsseldorf, Germany
| | - Sebastian Wesselborg
- Institut für Molekulare Medizin I, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Björn Stork
- Institut für Molekulare Medizin I, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany
| | - Thomas J J Müller
- Institut für Organische Chemie und Makromolekulare Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, D-40225 Düsseldorf, Germany.
| |
Collapse
|
14
|
Identification, structure modification, and characterization of potential small-molecule SGK3 inhibitors with novel scaffolds. Acta Pharmacol Sin 2018; 39:1902-1912. [PMID: 30038340 DOI: 10.1038/s41401-018-0087-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/10/2018] [Indexed: 12/16/2022] Open
Abstract
The serum and glucocorticoid-regulated kinase (SGK) family has been implicated in the regulation of many cellular processes downstream of the PI3K pathway. It plays a crucial role in PI3K-mediated tumorigenesis, making it a potential therapeutic target for cancer. SGK family consists of three isoforms (SGK1, SGK2, and SGK3), which have high sequence homology in the kinase domain and similar substrate specificity with the AKT family. In order to identify novel compounds capable of inhibiting SGK3 activity, a high-throughput screening campaign against 50,400 small molecules was conducted using a fluorescence-based kinase assay that has a Z' factor above 0.5. It identified 15 hits (including nitrogen-containing aromatic, flavone, hydrazone, and naphthalene derivatives) with IC50 values in the low micromolar to sub-micromolar range. Four compounds with a similar scaffold (i.e., a hydrazone core) were selected for structural modification and 18 derivatives were synthesized. Molecular modeling was then used to investigate the structure-activity relationship (SAR) and potential protein-ligand interactions. As a result, a series of SGK inhibitors that are active against both SGK1 and SGK3 were developed and important functional groups that control their inhibitory activity identified.
Collapse
|
15
|
Serum and glucocorticoid inducible protein kinases (SGKs): a potential target for cancer intervention. Acta Pharm Sin B 2018; 8:767-771. [PMID: 30245963 PMCID: PMC6146383 DOI: 10.1016/j.apsb.2018.07.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/03/2018] [Accepted: 06/14/2018] [Indexed: 12/11/2022] Open
Abstract
The serum and glucocorticoid inducible protein kinase (SGK) family members share similar structure, substrate specificity and function with AKT and signal downstream of the phosphatidylinositol 3-kinase (PI3K) signalling pathway. They regulate a range of fundamental cellular processes such as cell proliferation and survival, thereby playing an important role in cancer development. This perspective intends to give an overview on the involvement of SGKs (particularly SGK3) in cancer progression, and compares the actions of SGK3 and AKT in cell cycle regulation, oncogenic signalling, and the potential as a therapeutic target for cancer.
Collapse
|
16
|
Asquith CRM, Godoi PH, Couñago RM, Laitinen T, Scott JW, Langendorf CG, Oakhill JS, Drewry DH, Zuercher WJ, Koutentis PA, Willson TM, Kalogirou AS. 1,2,6-Thiadiazinones as Novel Narrow Spectrum Calcium/Calmodulin-Dependent Protein Kinase Kinase 2 (CaMKK2) Inhibitors. Molecules 2018; 23:molecules23051221. [PMID: 29783765 PMCID: PMC6019134 DOI: 10.3390/molecules23051221] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/31/2022] Open
Abstract
We demonstrate for the first time that 4H-1,2,6-thiadiazin-4-one (TDZ) can function as a chemotype for the design of ATP-competitive kinase inhibitors. Using insights from a co-crystal structure of a 3,5-bis(arylamino)-4H-1,2,6-thiadiazin-4-one bound to calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2), several analogues were identified with micromolar activity through targeted displacement of bound water molecules in the active site. Since the TDZ analogues showed reduced promiscuity compared to their 2,4-dianilinopyrimidine counter parts, they represent starting points for development of highly selective kinase inhibitors.
Collapse
Affiliation(s)
- Christopher R M Asquith
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill, NC 27599, USA.
| | - Paulo H Godoi
- Structural Genomics Consortium, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo 13083-886, Brazil.
| | - Rafael M Couñago
- Structural Genomics Consortium, Universidade Estadual de Campinas-UNICAMP, Campinas, São Paulo 13083-886, Brazil.
- Center for Molecular and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Avenida Cândido Rondon 400, P. O. Box 6010, 13083-875 Campinas, São Paulo 13083-886, Brazil.
| | - Tuomo Laitinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland.
| | - John W Scott
- St Vincent's Institute and Department of Medicine, University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia.
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia.
- The Florey Institute of Neuroscience and Mental Health, Parkville 3052, Australia.
| | - Christopher G Langendorf
- St Vincent's Institute and Department of Medicine, University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia.
| | - Jonathan S Oakhill
- St Vincent's Institute and Department of Medicine, University of Melbourne, 41 Victoria Parade, Fitzroy 3065, Australia.
- Mary MacKillop Institute for Health Research, Australian Catholic University, 215 Spring Street, Melbourne 3000, Australia.
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - William J Zuercher
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | | - Timothy M Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Andreas S Kalogirou
- Department of Chemistry, University of Cyprus, P. O. Box 20537, 1678 Nicosia, Cyprus.
- Department of Life Sciences, School of Sciences, European University Cyprus, 6 Diogenis Str., Engomi, P. O. Box 22006, 1516 Nicosia, Cyprus.
| |
Collapse
|
17
|
Nam S, Ware DC, Brothers PJ. Campestarenes: new building blocks with 5-fold symmetry. Org Biomol Chem 2018; 16:6460-6469. [DOI: 10.1039/c8ob00957k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
New campestarene derivatives bear functional groups designed to facilitate the formation of supramolecular assemblies of these 5-fold symmetric building blocks.
Collapse
Affiliation(s)
- Seong Nam
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - David C. Ware
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
| | - Penelope J. Brothers
- School of Chemical Sciences
- University of Auckland
- Auckland 1142
- New Zealand
- MacDiarmid Institute for Advanced Materials and Nanotechnology
| |
Collapse
|
18
|
Feneyrolles C, Guiet L, Singer M, Van Hijfte N, Daydé-Cazals B, Fauvel B, Chevé G, Yasri A. Discovering novel 7-azaindole-based series as potent AXL kinase inhibitors. Bioorg Med Chem Lett 2017; 27:862-866. [PMID: 28094183 DOI: 10.1016/j.bmcl.2017.01.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 12/14/2016] [Accepted: 01/06/2017] [Indexed: 12/27/2022]
Abstract
AXL is a receptor tyrosine kinase that plays a key role in tumor growth and proliferation. The scientific community has validated AXL as therapeutic target in the treatment of cancers for several years now, and several AXL inhibitors have been developed but none of them are approved. In this context, we started to design new kinase inhibitors targeting AXL from the 7-azaindole scaffold well known to interact with the ATP binding site of the kinase. Focused screening and chemical diversification around 7-azaindole scaffold were developed, based on modeling studies and medicinal chemistry rational, leading to the discovery of a new family of hits with potent inhibitory activity against AXL.
Collapse
Affiliation(s)
- Clémence Feneyrolles
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, 34189 Montpellier Cedex 4, France
| | - Léa Guiet
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, 34189 Montpellier Cedex 4, France
| | - Mathilde Singer
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, 34189 Montpellier Cedex 4, France
| | - Nathalie Van Hijfte
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, 34189 Montpellier Cedex 4, France
| | - Bénédicte Daydé-Cazals
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, 34189 Montpellier Cedex 4, France
| | - Bénédicte Fauvel
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, 34189 Montpellier Cedex 4, France
| | - Gwénaël Chevé
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, 34189 Montpellier Cedex 4, France
| | - Abdelaziz Yasri
- OriBase Pharma, Cap Gamma, Parc Euromédecine, 1682 rue de la Valsière, CS 17383, 34189 Montpellier Cedex 4, France.
| |
Collapse
|
19
|
Sequentially Palladium-Catalyzed Processes in One-Pot Syntheses of Heterocycles. APPLIED SCIENCES-BASEL 2015. [DOI: 10.3390/app5041803] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Llona-Minguez S, Desroses M, Ghassemian A, Jacques SA, Eriksson L, Isacksson R, Koolmeister T, Stenmark P, Scobie M, Helleday T. Vinylic MIDA Boronates: New Building Blocks for the Synthesis of Aza-Heterocycles. Chemistry 2015; 21:7394-8. [PMID: 25809883 DOI: 10.1002/chem.201406549] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Indexed: 01/23/2023]
Abstract
A two-step synthesis of structurally diverse pyrrole-containing bicyclic systems is reported. ortho-Nitro-haloarenes coupled with vinylic N-methyliminodiacetic acid (MIDA) boronates generate ortho-vinyl-nitroarenes, which undergo a "metal-free" nitrene insertion, resulting in a new pyrrole ring. This novel synthetic approach has a wide substrate tolerance and it is applicable in the preparation of more complex "drug-like" molecules. Interestingly, an ortho-nitro-allylarene derivative furnished a cyclic β-aminophosphonate motif.
Collapse
Affiliation(s)
- Sabin Llona-Minguez
- Science for Life Laboratory, Division of Translational, Medicine & Chemical Biology, Department of Medical Biochemistry & Biophysics, Karolinska Institutet, Stockholm, 171 21 (Sweden) http://www.helleday.org.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hassan S, Müller TJJ. Multicomponent Syntheses based upon Copper-Catalyzed Alkyne-Azide Cycloaddition. Adv Synth Catal 2015. [DOI: 10.1002/adsc.201400904] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
Halland N, Schmidt F, Weiss T, Saas J, Li Z, Czech J, Dreyer M, Hofmeister A, Mertsch K, Dietz U, Strübing C, Nazare M. Discovery of N-[4-(1H-Pyrazolo[3,4-b]pyrazin-6-yl)-phenyl]-sulfonamides as Highly Active and Selective SGK1 Inhibitors. ACS Med Chem Lett 2015; 6:73-8. [PMID: 25589934 DOI: 10.1021/ml5003376] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/17/2014] [Indexed: 12/11/2022] Open
Abstract
From a virtual screening starting point, inhibitors of the serum and glucocorticoid regulated kinase 1 were developed through a combination of classical medicinal chemistry and library approaches. This resulted in highly active small molecules with nanomolar activity and a good overall in vitro and ADME profile. Furthermore, the compounds exhibited unusually high kinase and off-target selectivity due to their rigid structure.
Collapse
Affiliation(s)
- Nis Halland
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Friedemann Schmidt
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Tilo Weiss
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Joachim Saas
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Ziyu Li
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Jörg Czech
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Matthias Dreyer
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Armin Hofmeister
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Katharina Mertsch
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Uwe Dietz
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Carsten Strübing
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Marc Nazare
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
| |
Collapse
|
23
|
Ortuso F, Amato R, Artese A, D'antona L, Costa G, Talarico C, Gigliotti F, Bianco C, Trapasso F, Schenone S, Musumeci F, Botta L, Perrotti N, Alcaro S. In silico identification and biological evaluation of novel selective serum/glucocorticoid-inducible kinase 1 inhibitors based on the pyrazolo-pyrimidine scaffold. J Chem Inf Model 2014; 54:1828-32. [PMID: 24896223 DOI: 10.1021/ci500235f] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The serum/glucocorticoid-inducible kinase 1 (Sgk1) has demonstrated antiapoptotic function and the capability to regulate cell survival, proliferation, and differentiation. A pivotal role of Sgk1 in carcinogenesis and in resistance to anticancer therapy has been suggested. With the aim of identifying new Sgk1 modulators, 322 pyrazolo-pyrimidine derivatives have been virtually screened with respect to a crystallographic model of Sgk1. The top five ranked compounds have been evaluated demonstrating Sgk1 inhibition in vitro and selectivity compared to RAC-alpha serine/threonine-protein kinase (Akt1).
Collapse
Affiliation(s)
- Francesco Ortuso
- Departments of "Scienze della Salute" and ‡"Medicina Sperimentale e Clinica", University "Magna Græcia" of Catanzaro , Viale Europa Loc. Germaneto, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Glucocorticoid receptor activity contributes to resistance to androgen-targeted therapy in prostate cancer. Discov Oncol 2014; 5:72-89. [PMID: 24615402 DOI: 10.1007/s12672-014-0173-2] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/18/2014] [Indexed: 10/25/2022] Open
Abstract
Despite new treatments for castrate-resistant prostate cancer (CRPC), the prognosis of patients with CRPC remains bleak due to acquired resistance to androgen receptor (AR)-directed therapy. The glucocorticoid receptor (GR) and AR share several transcriptional targets, including the anti-apoptotic genes serum and glucocorticoid-regulated kinase 1 (SGK1) and Map kinase phosphatase 1 (MKP1)/dual specificity phosphatase 1 (DUSP1). Because GR expression increases in a subset of primary prostate cancer (PC) following androgen deprivation therapy, we sought to determine whether GR activation can contribute to resistance to AR-directed therapy. We studied CWR-22Rv1 and LAPC4 AR/GR-expressing PC cell lines following treatment with combinations of the androgen R1881, AR antagonist MDV3100, GR agonist dexamethasone, GR antagonists mifepristone and CORT 122928, or the SGK1 inhibitor GSK650394. Cell lines stably expressing GR (NR3C1)-targeted shRNA or ectopic SGK1-Flag were also studied in vivo. GR activation diminished the effects of the AR antagonist MDV3100 on tumor cell viability. In addition, GR activation increased prostate-specific antigen (PSA) secretion and induced SGKI and MKP1/DUSP gene expression. Glucocorticoid-mediated cell viability was diminished by a GR antagonist or by co-treatment with the SGK1 inhibitor GSK650394. In vivo, GR depletion delayed castrate-resistant tumor formation, while SGK1-Flag-overexpressing PC xenografts displayed accelerated castrate-resistant tumor initiation, supporting a role for SGK1 in GR-mediated CRPC progression. We studied several PC models before and following treatment with androgen blockade and found that increased GR expression and activity contributed to tumor-promoting PC cell viability. Increased GR-regulated SGK1 expression appears, at least in part, to mediate enhanced PC cell survival. Therefore, GR and/or SGK1 inhibition may be useful adjuncts to AR blockade for treating CRPC.
Collapse
|
25
|
Gourdain S, Dairou J, Denhez C, Bui LC, Rodrigues-Lima F, Janel N, Delabar JM, Cariou K, Dodd RH. Development of DANDYs, new 3,5-diaryl-7-azaindoles demonstrating potent DYRK1A kinase inhibitory activity. J Med Chem 2013; 56:9569-85. [PMID: 24188002 DOI: 10.1021/jm401049v] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A series of 3,5-diaryl-1H-pyrrolo[2,3-b]pyridines were synthesized and evaluated for inhibition of DYRKIA kinase in vitro. Derivatives having hydroxy groups on the aryl moieties (2c, 2j-l) demonstrated high inhibitory potencies with Kis in the low nanomolar range. Their methoxy analogues were up to 100 times less active. Docking studies at the ATP binding site suggested that these compounds bind tightly to this site via a network of multiple H-bonds with the peptide backbone. None of the active compounds were cytotoxic to KB cells at 10(-6) M. Kinase profiling revealed that compound 2j showed 2-fold selectivity for DYRK1A with respect to DYRK2 and DYRK3.
Collapse
Affiliation(s)
- Stéphanie Gourdain
- Institut de Chimie des Substances Naturelles, Centre de Recherche de Gif, UPR 2301, CNRS , Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lang F, Voelkl J. Therapeutic potential of serum and glucocorticoid inducible kinase inhibition. Expert Opin Investig Drugs 2013; 22:701-14. [PMID: 23506284 DOI: 10.1517/13543784.2013.778971] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Expression of serum-and-glucocorticoid-inducible kinase-1 (SGK1) is low in most cells, but dramatically increases under certain pathophysiological conditions, such as glucocorticoid or mineralocorticoid excess, inflammation with TGFβ release, hyperglycemia, cell shrinkage and ischemia. SGK1 is activated by insulin and growth factors via phosphatidylinositide-3-kinase, 3-phosphoinositide-dependent kinase and mammalian target of rapamycin. SGK1 sensitive functions include activation of ion channels (including epithelial Na(+) channel ENaC, voltage gated Na(+) channel SCN5A transient receptor potential channels TRPV4 - 6, Ca(2+) release activated Ca(2+) channel Orai1/STIM1, renal outer medullary K(+) channel ROMK, voltage gated K(+) channels KCNE1/KCNQ1, kainate receptor GluR6, cystic fibrosis transmembrane regulator CFTR), carriers (including Na(+),Cl(-) symport NCC, Na(+),K(+),2Cl(-) symport NKCC, Na(+)/H(+) exchangers NHE1 and NHE3, Na(+), glucose symport SGLT1, several amino acid transporters), and Na(+)/K(+)-ATPase. SGK1 regulates several enzymes (e.g., glycogen synthase kinase-3, ubiquitin-ligase Nedd4-2) and transcription factors (e.g., forkhead transcription factor 3a, β-catenin, nuclear factor kappa B). AREAS COVERED The phenotype of SGK1 knockout mice is mild and SGK1 is apparently dispensible for basic functions. Excessive SGK1 expression and activity, however, contributes to the pathophysiology of several disorders, including hypertension, obesity, diabetes, thrombosis, stroke, fibrosing disease, infertility and tumor growth. A SGK1 gene variant (prevalence ∼ 3 - 5% in Caucasians and ∼ 10% in Africans) is associated with hypertension, stroke, obesity and type 2 diabetes. SGK1 inhibitors have been developed and shown to reduce blood pressure of hyperinsulinemic mice and to counteract tumor cell survival. EXPERT OPINION Targeting SGK1 may be a therapeutic option in several clinical conditions, including metabolic syndrome and tumor growth.
Collapse
Affiliation(s)
- Florian Lang
- University of Tuebingen, Department of Physiology, Tuebingen, Germany.
| | | |
Collapse
|
27
|
Cardiovascular effects of a novel selective Rho kinase inhibitor, 2-(1H-indazole-5-yl)amino-4-methoxy-6-piperazino triazine (DW1865). Eur J Pharmacol 2013; 702:218-26. [PMID: 23376156 DOI: 10.1016/j.ejphar.2013.01.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 01/08/2013] [Accepted: 01/16/2013] [Indexed: 11/23/2022]
Abstract
The arising critical implications of Rho kinase signaling in cardiovascular diseases have been attracting attention in the pharmacological potential of Rho kinase inhibitors. We identified a novel inhibitor of Rho kinase (2-(1H-indazole-5-yl)amino-4-methoxy-6-piperazino triazine; DW 1865) and characterized its effects in biochemical, cellular, tissue and animal based assays. DW 1865 potently inhibited the kinase activity of both Rho kinase 1 and Rho kinase 2 in vitro, and behaved as an ATP-competitive inhibitor. Interestingly, DW1865 was 10 times more potent in inhibiting Rho kinase activities than fasudil as a selective Rho kinase inhibitor. The activity of DW1865 was shown to be highly selective for Rho kinase in the panel assay of 13 other kinases. In the isolated vascular tissue study, DW1865 exerted vasorelaxation in phenylephrine- or 5-hydroxytriptamine-induced contraction in a concentration-dependent manner manner. In spontaneously hypertensive rats, administration of DW1865 caused a significant and dose-related reduction in blood pressure. Furthermore, DW1865 blocked angiotensin II-induced stress fiber formation and cellular hypertrophy in rat heart-derived H9c2 cells. Taken together, these results suggest that DW1865 is a highly selective and potent Rho kinase inhibitor that will alleviate the pathophysiological actions of Rho kinase such as stress fiber formation, cellular hypertrophy, and hypertension.
Collapse
|
28
|
Iaroshenko VO, Vilches-Herrera M, Gevorgyan A, Mkrtchyan S, Arakelyan K, Ostrovskyi D, Abbasi MS, Supe L, Hakobyan A, Villinger A, Volochnyuk DM, Tolmachev A. Design, synthesis and transformation of some heteroannulated 3-aminopyridines—purine isosteres with exocyclic nitrogen atom. Tetrahedron 2013. [DOI: 10.1016/j.tet.2012.11.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
29
|
Geldenhuys WJ, Talasila PK, Sadana P. Identification of a novel serum and glucocorticoid regulated kinase-1 (SGK1) ligand from virtual screening. Bioorg Med Chem Lett 2012; 22:5675-8. [PMID: 22858098 DOI: 10.1016/j.bmcl.2012.06.096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 06/25/2012] [Accepted: 06/27/2012] [Indexed: 10/28/2022]
Abstract
The serum and glucocorticoid regulated kinase-1 (SGK1) is part of the serine/threonine kinase family and has therapeutic potential in several neurodegenerative diseases such as ischemic stroke and Parkinson's disease. Here we use structure-based virtual screening to identify a novel ligand which inhibits SGK1 activity. The data presented here can be used for future scaffold hopping and possible drug development efforts.
Collapse
Affiliation(s)
- Werner J Geldenhuys
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | | | | |
Collapse
|
30
|
Wenglowsky S, Ahrendt KA, Buckmelter AJ, Feng B, Gloor SL, Gradl S, Grina J, Hansen JD, Laird ER, Lunghofer P, Mathieu S, Moreno D, Newhouse B, Ren L, Risom T, Rudolph J, Seo J, Sturgis HL, Voegtli WC, Wen Z. Pyrazolopyridine inhibitors of B-RafV600E. Part 2: structure-activity relationships. Bioorg Med Chem Lett 2011; 21:5533-7. [PMID: 21802293 DOI: 10.1016/j.bmcl.2011.06.097] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 06/21/2011] [Indexed: 10/18/2022]
Abstract
Structure-activity relationships around a novel series of B-Raf(V600E) inhibitors are reported. The enzymatic and cellular potencies of inhibitors derived from two related hinge-binding groups were compared and3-methoxypyrazolopyridine proved to be superior. The 3-alkoxy group of lead B-Raf(V600E) inhibitor 1 was extended and minimally affected potency. The propyl sulfonamide tail of compound 1, which occupies the small lipophilic pocket formed by an outward shift of the αC-helix, was expanded to a series of arylsulfonamides. X-ray crystallography revealed that this lipophilic pocket unexpectedly enlarges to accommodate the bulkier aryl group.
Collapse
Affiliation(s)
- Steve Wenglowsky
- ArrayBioPharma, 3200 Walnut Street, Boulder, CO 80301, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bamborough P, Brown MJ, Christopher JA, Chung CW, Mellor GW. Selectivity of kinase inhibitor fragments. J Med Chem 2011; 54:5131-43. [PMID: 21699136 DOI: 10.1021/jm200349b] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A kinase-focused screening set of fragments has been assembled and has proved successful for the discovery of ligand-efficient hits against many targets. Here we present some of our general conclusions from this exercise. Notably, we present the first profiling results for literature fragments that have previously been used as starting points for optimization against individual kinases. We consider the importance of screening format and the extent to which selectivity is helpful in selecting fragments for progression. Results are also outlined for fragments targeting the DFG-out conformation and for atypical kinases such as PIM1 and lipid kinases.
Collapse
Affiliation(s)
- Paul Bamborough
- GlaxoSmithKline R&D, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK.
| | | | | | | | | |
Collapse
|
32
|
Merkul E, Klukas F, Dorsch D, Grädler U, Greiner HE, Müller TJJ. Rapid preparation of triazolyl substituted NH-heterocyclic kinase inhibitors via one-pot Sonogashira coupling–TMS-deprotection–CuAAC sequence. Org Biomol Chem 2011; 9:5129-36. [DOI: 10.1039/c1ob05586k] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
33
|
Hong S, Lee S, Kim B, Lee H, Hong SS, Hong S. Discovery of new azaindole-based PI3Kα inhibitors: Apoptotic and antiangiogenic effect on cancer cells. Bioorg Med Chem Lett 2010; 20:7212-5. [DOI: 10.1016/j.bmcl.2010.10.108] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 10/20/2010] [Accepted: 10/21/2010] [Indexed: 12/22/2022]
|