1
|
Barresi E, Robello M, Baglini E, Poggetti V, Viviano M, Salerno S, Da Settimo F, Taliani S. Indol-3-ylglyoxylamide as Privileged Scaffold in Medicinal Chemistry. Pharmaceuticals (Basel) 2023; 16:997. [PMID: 37513909 PMCID: PMC10386336 DOI: 10.3390/ph16070997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
In recent years, indolylglyoxylamide-based derivatives have received much attention due to their application in drug design and discovery, leading to the development of a wide array of compounds that have shown a variety of pharmacological activities. Combining the indole nucleus, already validated as a "privileged structure," with the glyoxylamide function allowed for an excellent template to be obtained that is suitable to a great number of structural modifications aimed at permitting interaction with specific molecular targets and producing desirable therapeutic effects. The present review provides insight into how medicinal chemists have elegantly exploited the indolylglyoxylamide moiety to obtain potentially useful drugs, with a particular focus on compounds exhibiting activity in in vivo models or reaching clinical trials. All in all, this information provides exciting new perspectives on existing data that can be useful in further design of indolylglyoxylamide-based molecules with interesting pharmacological profiles. The aim of this report is to present an update of collection data dealing with the employment of this moiety in the rational design of compounds that are able to interact with a specific target, referring to the last 20 years.
Collapse
Affiliation(s)
- Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Marco Robello
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Monica Viviano
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
2
|
Shim KH, Sharma N, An SSA. Prion therapeutics: Lessons from the past. Prion 2022; 16:265-294. [PMID: 36515657 PMCID: PMC9754114 DOI: 10.1080/19336896.2022.2153551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 12/15/2022] Open
Abstract
Prion diseases are a group of incurable zoonotic neurodegenerative diseases (NDDs) in humans and other animals caused by the prion proteins. The abnormal folding and aggregation of the soluble cellular prion proteins (PrPC) into scrapie isoform (PrPSc) in the Central nervous system (CNS) resulted in brain damage and other neurological symptoms. Different therapeutic approaches, including stalling PrPC to PrPSc conversion, increasing PrPSc removal, and PrPC stabilization, for which a spectrum of compounds, ranging from organic compounds to antibodies, have been explored. Additionally, a non-PrP targeted drug strategy using serpin inhibitors has been discussed. Despite numerous scaffolds being screened for anti-prion activity in vitro, only a few were effective in vivo and unfortunately, almost none of them proved effective in the clinical studies, most likely due to toxicity and lack of permeability. Recently, encouraging results from a prion-protein monoclonal antibody, PRN100, were presented in the first human trial on CJD patients, which gives a hope for better future for the discovery of other new molecules to treat prion diseases. In this comprehensive review, we have re-visited the history and discussed various classes of anti-prion agents, their structure, mode of action, and toxicity. Understanding pathogenesis would be vital for developing future treatments for prion diseases. Based on the outcomes of existing therapies, new anti-prion agents could be identified/synthesized/designed with reduced toxicity and increased bioavailability, which could probably be effective in treating prion diseases.
Collapse
Affiliation(s)
- Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Niti Sharma
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| |
Collapse
|
3
|
Nocentini A, Capasso C, Supuran CT. Perspectives on the design and discovery of α-ketoamide inhibitors for the treatment of novel coronavirus: where do we stand and where do we go? Expert Opin Drug Discov 2022; 17:547-557. [DOI: 10.1080/17460441.2022.2052847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Alessio Nocentini
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Firenze, Italy
| | - Clemente Capasso
- Department of Biology, Agriculture and Food Sciences, Institute of Biosciences and Bioresources, Napoli, Italy
| | - Claudiu T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Firenze, Italy
| |
Collapse
|
4
|
|
5
|
Robinson RLM, Sarimveis H, Doganis P, Jia X, Kotzabasaki M, Gousiadou C, Harper SL, Wilkins T. Identifying diverse metal oxide nanomaterials with lethal effects on embryonic zebrafish using machine learning. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:1297-1325. [PMID: 34934606 PMCID: PMC8649207 DOI: 10.3762/bjnano.12.97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 10/28/2021] [Indexed: 06/14/2023]
Abstract
Manufacturers of nanomaterial-enabled products need models of endpoints that are relevant to human safety to support the "safe by design" paradigm and avoid late-stage attrition. Increasingly, embryonic zebrafish (Danio Rerio) are recognised as a key human safety relevant in vivo test system. Hence, machine learning models were developed for identifying metal oxide nanomaterials causing lethality to embryonic zebrafish up to 24 hours post-fertilisation, or excess lethality in the period of 24-120 hours post-fertilisation, at concentrations of 250 ppm or less. Models were developed using data from the Nanomaterial Biological-Interactions Knowledgebase for a dataset of 44 diverse, coated and uncoated metal or, in one case, metalloid oxide nanomaterials. Different modelling approaches were evaluated using nested cross-validation on this dataset. Models were initially developed for both lethality endpoints using multiple descriptors representing the composition of the core, shell and surface functional groups, as well as particle characteristics. However, interestingly, the 24 hours post-fertilisation data were found to be harder to predict, which could reflect different exposure routes. Hence, subsequent analysis focused on the prediction of excess lethality at 120 hours-post fertilisation. The use of two data augmentation approaches, applied for the first time in nano-QSAR research, was explored, yet both failed to boost predictive performance. Interestingly, it was found that comparable results to those originally obtained using multiple descriptors could be obtained using a model based upon a single, simple descriptor: the Pauling electronegativity of the metal atom. Since it is widely recognised that a variety of intrinsic and extrinsic nanomaterial characteristics contribute to their toxicological effects, this is a surprising finding. This may partly reflect the need to investigate more sophisticated descriptors in future studies. Future studies are also required to examine how robust these modelling results are on truly external data, which were not used to select the single descriptor model. This will require further laboratory work to generate comparable data to those studied herein.
Collapse
Affiliation(s)
| | - Haralambos Sarimveis
- School of Chemical Engineering, National Technical University of Athens, 9 Heroon Polytechniou str. Zografou Campus, 15780 Athens, Greece
| | - Philip Doganis
- School of Chemical Engineering, National Technical University of Athens, 9 Heroon Polytechniou str. Zografou Campus, 15780 Athens, Greece
| | - Xiaodong Jia
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Marianna Kotzabasaki
- School of Chemical Engineering, National Technical University of Athens, 9 Heroon Polytechniou str. Zografou Campus, 15780 Athens, Greece
| | - Christiana Gousiadou
- School of Chemical Engineering, National Technical University of Athens, 9 Heroon Polytechniou str. Zografou Campus, 15780 Athens, Greece
| | - Stacey Lynn Harper
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, Oregon, USA
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
- Oregon Nanoscience and Microtechnologies Institute, Eugene, Oregon, USA
| | - Terry Wilkins
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom
| |
Collapse
|
6
|
Robello M, Barresi E, Baglini E, Salerno S, Taliani S, Settimo FD. The Alpha Keto Amide Moiety as a Privileged Motif in Medicinal Chemistry: Current Insights and Emerging Opportunities. J Med Chem 2021; 64:3508-3545. [PMID: 33764065 PMCID: PMC8154582 DOI: 10.1021/acs.jmedchem.0c01808] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the years, researchers in drug discovery have taken advantage of the use of privileged structures to design innovative hit/lead molecules. The α-ketoamide motif is found in many natural products, and it has been widely exploited by medicinal chemists to develop compounds tailored to a vast range of biological targets, thus presenting clinical potential for a plethora of pathological conditions. The purpose of this perspective is to provide insights into the versatility of this chemical moiety as a privileged structure in drug discovery. After a brief analysis of its physical-chemical features and synthetic procedures to obtain it, α-ketoamide-based classes of compounds are reported according to the application of this motif as either a nonreactive or reactive moiety. The goal is to highlight those aspects that may be useful to understanding the perspectives of employing the α-ketoamide moiety in the rational design of compounds able to interact with a specific target.
Collapse
Affiliation(s)
- Marco Robello
- Synthetic Bioactive Molecules Section, LBC, NIDDK, NIH, 8 Center Drive, Room 404, Bethesda, Maryland 20892, United States
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
7
|
|
8
|
Wu C, Zhou J, He G, Li H, Yang Q, Wang R, Zhou Y, Liu H. Ruthenium(ii)-catalyzed selective C–H bond activation of imidamides and coupling with sulfoxonium ylides: an efficient approach for the synthesis of highly functional 3-ketoindoles. Org Chem Front 2019. [DOI: 10.1039/c9qo00048h] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ruthenium-catalyzed selective C–H bond activation of imidamides and annulation of sulfoxonium ylides were achieved, which afforded a series of 3-ketoindole derivatives in good yields, with good functional group compatibility.
Collapse
Affiliation(s)
- Chenglin Wu
- School of Pharmacy
- China Pharmaceutical University
- Nanjing 210009
- China
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
| | - Jianhui Zhou
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Guoxue He
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Huihui Li
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Qiaolan Yang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Run Wang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Yu Zhou
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Hong Liu
- School of Pharmacy
- China Pharmaceutical University
- Nanjing 210009
- China
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
| |
Collapse
|
9
|
Zhang X, Bian J, Li X, Wu X, Dong Y, You Q. 2-Substituted 3,7,8-trimethylnaphtho[1,2- b ]furan-4,5-diones as specific L-shaped NQO1-mediated redox modulators for the treatment of non-small cell lung cancer. Eur J Med Chem 2017; 138:616-629. [DOI: 10.1016/j.ejmech.2017.06.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/10/2017] [Accepted: 06/14/2017] [Indexed: 02/03/2023]
|
10
|
Abbas Q, Ashraf Z, Hassan M, Nadeem H, Latif M, Afzal S, Seo SY. Development of highly potent melanogenesis inhibitor by in vitro, in vivo and computational studies. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:2029-2046. [PMID: 28740364 PMCID: PMC5503496 DOI: 10.2147/dddt.s137550] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The present work describes the synthesis of few hydroxylated amide derivatives as melanogenesis inhibitors. In vitro, in vivo and computational studies proved that compound 6d is a highly potent melanogenesis inhibitor compared to standard kojic acid. The title amides 4a–e and 6a–e were synthesized following simple reaction routes with excellent yields. Most of the synthesized compounds exhibited good mushroom tyrosinase inhibitory activity, but compound 6d showed excellent activity (IC50 0.15 µM) compared to standard kojic acid (IC50 16.69 µM). Lineweaver–Burk plots were used for the determination of kinetic mechanism, and it was found that compounds 4c and 6d showed non-competitive inhibition while 6a and 6b showed mixed-type inhibition. The kinetic mechanism further revealed that compound 6d formed irreversible complex with the target enzyme tyrosinase. The Ki values determined for compounds 4c, 6a, 6b and 6d are 0.188, 0.84, 2.20 and 0.217 µM respectively. Results of human tyrosinase inhibitory activity in A375 human melanoma cells showed that compound 6d exhibited 91.9% inhibi-tory activity at a concentration of 50 µg/mL. In vivo cytotoxicity evaluation of compound 6d in zebrafish embryos showed that it is non-toxic to zebrafish. Melanin depigmentation assay performed in zebrafish indicated that compound 6d possessed greater potential in decreasing melanin contents compared to kojic acid at the same concentration. Computational studies also supported the wet lab findings as compound 6d showed a highest binding affinity with the target protein (PDBID: 2Y9X) with a binding energy value of −7.90 kcal/mol. Molecular dynamic simulation studies also proved that amide 6d formed the most stable complex with tyrosinase. Based upon our in vitro, in vivo and computational studies, we propose that compound 6d is a promising candidate for the development of safe cosmetic agent.
Collapse
Affiliation(s)
- Qamar Abbas
- Department of Biology, College of Natural Sciences, Kongju National University, Gongju, Republic of Korea
| | - Zaman Ashraf
- Department of Chemistry, Allama Iqbal Open University, Islamabad
| | - Mubashir Hassan
- Department of Biology, College of Natural Sciences, Kongju National University, Gongju, Republic of Korea
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Muhammad Latif
- Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunawwarah, Kingdom of Saudi Arabia
| | - Samina Afzal
- Faculty of Pharmacy, Bahauddin Zakria University, Multan, Pakistan
| | - Sung-Yum Seo
- Department of Biology, College of Natural Sciences, Kongju National University, Gongju, Republic of Korea
| |
Collapse
|
11
|
Pagadala NS, Syed K, Bhat R. In silico strategies on prion pathogenic conversion and inhibition from PrPC–PrPSc. Expert Opin Drug Discov 2017; 12:241-248. [DOI: 10.1080/17460441.2017.1287171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Nataraj S. Pagadala
- Department of Medical Microbiology and Immunology, 6-020 Katz Group Centre, University of Alberta, Edmonton, Canada
| | - Khajamohiddin Syed
- Unit for Drug Discovery Research, Department of Health Sciences, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, South Africa
| | - Rakesh Bhat
- Department of Medical Microbiology and Immunology, 6-020 Katz Group Centre, University of Alberta, Edmonton, Canada
| |
Collapse
|
12
|
Aldilla VR, Nizalapur S, Martin A, Marjo CE, Rich A, Yee E, Suwannakot P, Black DS, Thordarson P, Kumar N. Design, synthesis, and characterisation of glyoxylamide-based short peptides as self-assembled gels. NEW J CHEM 2017. [DOI: 10.1039/c7nj02248d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
First example of glyoxylamide-based short peptides which can encapsulate organic solvents and water at relatively low concentrations.
Collapse
Affiliation(s)
| | | | - Adam Martin
- School of Chemistry
- UNSW Australia
- Sydney
- Australia
| | - Chris E. Marjo
- Mark Wainwright Analytical Centre
- UNSW Australia
- Sydney NSW 2052
- Australia
| | - Anne Rich
- Mark Wainwright Analytical Centre
- UNSW Australia
- Sydney NSW 2052
- Australia
| | - Eugene Yee
- School of Chemistry
- UNSW Australia
- Sydney
- Australia
| | | | | | | | - Naresh Kumar
- School of Chemistry
- UNSW Australia
- Sydney
- Australia
| |
Collapse
|
13
|
Hu HY, Yu XD, Wang F, Lin CR, Zeng JZ, Qiu YK, Fang MJ, Wu Z. Novel N-Substituted 2-(2-(Adamantan-1-yl)-1H-Indol-3-yl)-2-Oxoacetamide Derivatives: Synthesis and Biological Evaluation. Molecules 2016; 21:E530. [PMID: 27164070 PMCID: PMC6273615 DOI: 10.3390/molecules21050530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/03/2016] [Accepted: 04/16/2016] [Indexed: 11/16/2022] Open
Abstract
In this study, a series of novel N-substituted 2-(2-(adamantan-1-yl)-1H-indol-3-yl)-2-oxoacetamide derivatives were synthesized, and evaluated for their cytotoxicity in human cell lines including Hela (cervical cancer), MCF7 (breast cancer ) and HepG2 (liver cancer). Several compounds were found to have potent anti-proliferative activity against those human cancer cell lines and compound 5r showed the most potent biological activity against HepG2 cells with an IC50 value of 10.56 ± 1.14 μΜ. In addition, bioassays showed that compound 5r induced time-dependent and dose-dependent cleavage of poly ADP-ribose polymerase (PARP), and also induced a dose-dependent increase in caspase-3 and caspase-8 activity, but had little effect on caspase-9 protease activity in HepG2 cells. These results provide evidence that 5r-induced apoptosis in HepG2 cell is caspase-8-dependent.
Collapse
Affiliation(s)
- Hong-Yu Hu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Xu-Dong Yu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Fei Wang
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Chun-Rong Lin
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Jin-Zhang Zeng
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Ying-Kun Qiu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Mei-Juan Fang
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Zhen Wu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| |
Collapse
|
14
|
Colley HE, Muthana M, Danson SJ, Jackson LV, Brett ML, Harrison J, Coole SF, Mason DP, Jennings LR, Wong M, Tulasi V, Norman D, Lockey PM, Williams L, Dossetter AG, Griffen EJ, Thompson MJ. An Orally Bioavailable, Indole-3-glyoxylamide Based Series of Tubulin Polymerization Inhibitors Showing Tumor Growth Inhibition in a Mouse Xenograft Model of Head and Neck Cancer. J Med Chem 2015; 58:9309-33. [DOI: 10.1021/acs.jmedchem.5b01312] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Helen E. Colley
- School
of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield S10 2TA, U.K
| | - Munitta Muthana
- Department
of Oncology, The University of Sheffield, Medical School, Beech
Hill Road, Sheffield S10
2RX, U.K
| | - Sarah J. Danson
- Academic
Unit of Clinical Oncology and Sheffield Experimental Medicine Centre, Weston Park Hospital, Whitham Road, Sheffield S10 2SJ, U.K
| | - Lucinda V. Jackson
- Department
of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Matthew L. Brett
- Department
of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Joanne Harrison
- Department
of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Sean F. Coole
- Department
of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Daniel P. Mason
- Department
of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| | - Luke R. Jennings
- School
of Clinical Dentistry, University of Sheffield, 19 Claremont Crescent, Sheffield S10 2TA, U.K
| | - Melanie Wong
- Charles River, 8−9 Spire
Green Centre, Harlow, Harlow, Essex CM19 5TR, U.K
| | - Vamshi Tulasi
- Charles River, 8−9 Spire
Green Centre, Harlow, Harlow, Essex CM19 5TR, U.K
| | - Dennis Norman
- Charles River, 8−9 Spire
Green Centre, Harlow, Harlow, Essex CM19 5TR, U.K
| | - Peter M. Lockey
- Charles River, 8−9 Spire
Green Centre, Harlow, Harlow, Essex CM19 5TR, U.K
| | - Lynne Williams
- Department
of Oncology, The University of Sheffield, Medical School, Beech
Hill Road, Sheffield S10
2RX, U.K
| | - Alexander G. Dossetter
- MedChemica Limited, Ebenezer House,
Ryecroft, Newcastle-Under-Lyme, Staffordshire ST5 2BE, U.K
| | - Edward J. Griffen
- MedChemica Limited, Ebenezer House,
Ryecroft, Newcastle-Under-Lyme, Staffordshire ST5 2BE, U.K
| | - Mark J. Thompson
- Department
of Chemistry, University of Sheffield, Brook Hill, Sheffield S3 7HF, U.K
| |
Collapse
|
15
|
Wu C, Zhao F, Shu S, Wang J, Liu H. Palladium-catalyzed intramolecular addition of C–N bond to alkynes: a novel approach to 3-diketoindoles. RSC Adv 2015. [DOI: 10.1039/c5ra18813j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Palladium-catalyzed intramolecular addition of C–N bond to alkynes to synthesize highly functional 3-diketoindoles has been achieved.
Collapse
Affiliation(s)
- Chenglin Wu
- Nano Science and Technology Institute
- University of Science and Technology of China
- Suzhou 215123
- People's Republic of China
- CAS Key Laboratory of Receptor Research
| | - Fei Zhao
- Sichuan Industrial Institute of Antibiotics
- Chengdu University
- People's Republic of China
| | - Shuangjie Shu
- CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- People's Republic of China
| | - Jiang Wang
- CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- People's Republic of China
| | - Hong Liu
- Nano Science and Technology Institute
- University of Science and Technology of China
- Suzhou 215123
- People's Republic of China
- CAS Key Laboratory of Receptor Research
| |
Collapse
|
16
|
Abstract
The zebrafish (Danio rerio) is a small, tropical, freshwater fish that has emerged as a powerful vertebrate model organism for studying genetics and development. Its small size, transparency, cost-effectiveness, close genome homology to humans compared with invertebrates, and capacity for genetic manipulation are all valuable attributes for an excellent animal model. There are additional advantages for using zebrafish specifically in drug discovery, including ease of exposure to chemicals in water. In effect, zebrafish can bridge a gap between in vitro and mammalian work, reducing the use of larger animals and attrition rates. In the drug-discovery process, zebrafish can be used at many stages, including target identification and validation, identification of lead compounds, studying structure-activity relationships and drug safety profiling. In this review, we highlight the potential for the zebrafish model to make the drug-discovery process simpler, more effective and cost-efficient.
Collapse
|
17
|
Antiprion compounds that reduce PrP(Sc) levels in dividing and stationary-phase cells. Bioorg Med Chem 2013; 21:7999-8012. [PMID: 24183589 DOI: 10.1016/j.bmc.2013.09.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 08/30/2013] [Accepted: 09/07/2013] [Indexed: 11/20/2022]
Abstract
During prion diseases, a normally benign, host protein, denoted PrP(C), undergoes alternative folding into the aberrant isoform, PrP(Sc). We used ELISA to identify and confirm hits in order to develop leads that reduce PrP(Sc) in prion-infected dividing and stationary-phase mouse neuroblastoma (ScN2a-cl3) cells. We tested 52,830 diverse small molecules in dividing cells and 49,430 in stationary-phase cells. This led to 3100 HTS and 970 single point confirmed (SPC) hits in dividing cells, 331 HTS and 55 confirmed SPC hits in stationary-phase cells as well as 36 confirmed SPC hits active in both. Fourteen chemical leads were identified from confirmed SPC hits in dividing cells and three in stationary-phase cells. From more than 682 compounds tested in concentration-effect relationships in dividing cells to determine potency (EC50), 102 had EC50 values between 1 and 10 μM and 50 had EC50 values of <1 μM; none affected cell viability. We observed an excellent correlation between EC50 values determined by ELISA and Western immunoblotting for 28 representative compounds in dividing cells (R(2)=0.75; p <0.0001). Of the 55 confirmed SPC hits in stationary-phase cells, 23 were piperazine, indole, or urea leads. The EC50 values of one indole in stationary-phase and dividing ScN2a-cl3 cells were 7.5 and 1.6 μM, respectively. Unexpectedly, the number of hits in stationary-phase cells was ~10% of that in dividing cells. The explanation for this difference remains to be determined.
Collapse
|
18
|
Li Z, Rao S, Gever JR, Widjaja K, Prusiner SB, Michael Silber B. Optimization of Arylamides as Novel, Potent and Brain-penetrant Antiprion Lead Compounds. ACS Med Chem Lett 2013; 4:647-650. [PMID: 23977416 DOI: 10.1021/ml300454k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The prion diseases caused by PrPSc, an alternatively folded form of the cellular prion protein (PrPC), are rapidly progressive, fatal, and untreatable neurodegenerative syndromes. We employed HTS ELISA assays to identify compounds that lower the level of PrPSc in prion-infected mouse neuroblastoma (ScN2a-cl3) cells and identified a series of arylamides. SAR studies indicated that small amides with one aromatic, or heteroaromatic ring, on each side of the amide bond are of modest potency. Of note, benzamide (7), with an EC50 of 2200 nM, was one of only a few arylamide hits with a piperazine group on its aniline moiety. The basic piperazine nitrogen can be protonated at physiologic pH, improving solubility, and therefore we wanted to exploit this feature in our search for a drug candidate. An SAR campaign resulted in several key analogs, including a set with biaryl groups introduced on the carbonyl side for improved potency. Several of these biaryl analogs have submicromolar potency, with the most potent analog 17 having an EC50 = 22 nM. More importantly, 17 and several biarylamides (20, 24, 26, 27) were able to traverse the BBB and displayed excellent drug levels in the brains of mice following oral dosing. These biarylamides may represent good starting points for further lead optimization for the identification of potential drug candidates for the treatment of prion diseases.
Collapse
Affiliation(s)
- Zhe Li
- Institute
for Neurodegenerative Diseases, ‡Department of Neurology, §Department of Bioengineering and
Therapeutic Sciences, University of California, San Francisco, California 94143, United States
| | - Satish Rao
- Institute
for Neurodegenerative Diseases, ‡Department of Neurology, §Department of Bioengineering and
Therapeutic Sciences, University of California, San Francisco, California 94143, United States
| | - Joel R. Gever
- Institute
for Neurodegenerative Diseases, ‡Department of Neurology, §Department of Bioengineering and
Therapeutic Sciences, University of California, San Francisco, California 94143, United States
| | - Kartika Widjaja
- Institute
for Neurodegenerative Diseases, ‡Department of Neurology, §Department of Bioengineering and
Therapeutic Sciences, University of California, San Francisco, California 94143, United States
| | - Stanley B. Prusiner
- Institute
for Neurodegenerative Diseases, ‡Department of Neurology, §Department of Bioengineering and
Therapeutic Sciences, University of California, San Francisco, California 94143, United States
| | - B. Michael Silber
- Institute
for Neurodegenerative Diseases, ‡Department of Neurology, §Department of Bioengineering and
Therapeutic Sciences, University of California, San Francisco, California 94143, United States
| |
Collapse
|
19
|
Wagner J, Ryazanov S, Leonov A, Levin J, Shi S, Schmidt F, Prix C, Pan-Montojo F, Bertsch U, Mitteregger-Kretzschmar G, Geissen M, Eiden M, Leidel F, Hirschberger T, Deeg AA, Krauth JJ, Zinth W, Tavan P, Pilger J, Zweckstetter M, Frank T, Bähr M, Weishaupt JH, Uhr M, Urlaub H, Teichmann U, Samwer M, Bötzel K, Groschup M, Kretzschmar H, Griesinger C, Giese A. Anle138b: a novel oligomer modulator for disease-modifying therapy of neurodegenerative diseases such as prion and Parkinson's disease. Acta Neuropathol 2013; 125:795-813. [PMID: 23604588 PMCID: PMC3661926 DOI: 10.1007/s00401-013-1114-9] [Citation(s) in RCA: 283] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/01/2013] [Accepted: 04/02/2013] [Indexed: 01/10/2023]
Abstract
In neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD) and prion diseases, deposits of aggregated disease-specific proteins are found. Oligomeric aggregates are presumed to be the key neurotoxic agent. Here we describe the novel oligomer modulator anle138b [3-(1,3-benzodioxol-5-yl)-5-(3-bromophenyl)-1H-pyrazole], an aggregation inhibitor we developed based on a systematic high-throughput screening campaign combined with medicinal chemistry optimization. In vitro, anle138b blocked the formation of pathological aggregates of prion protein (PrPSc) and of α-synuclein (α-syn), which is deposited in PD and other synucleinopathies such as dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Notably, anle138b strongly inhibited all prion strains tested including BSE-derived and human prions. Anle138b showed structure-dependent binding to pathological aggregates and strongly inhibited formation of pathological oligomers in vitro and in vivo both for prion protein and α-synuclein. Both in mouse models of prion disease and in three different PD mouse models, anle138b strongly inhibited oligomer accumulation, neuronal degeneration, and disease progression in vivo. Anle138b had no detectable toxicity at therapeutic doses and an excellent oral bioavailability and blood–brain-barrier penetration. Our findings indicate that oligomer modulators provide a new approach for disease-modifying therapy in these diseases, for which only symptomatic treatment is available so far. Moreover, our findings suggest that pathological oligomers in neurodegenerative diseases share structural features, although the main protein component is disease-specific, indicating that compounds such as anle138b that modulate oligomer formation by targeting structure-dependent epitopes can have a broad spectrum of activity in the treatment of different protein aggregation diseases.
Collapse
Affiliation(s)
- Jens Wagner
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Sergey Ryazanov
- NMR based structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Andrei Leonov
- NMR based structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Johannes Levin
- Neurologische Klinik, Klinikum der Ludwig-Maximilians-Universität München, Marchioninistr. 15, 81377 Munich, Germany
| | - Song Shi
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Felix Schmidt
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 23, 81377 Munich, Germany
- Neurologische Klinik, Klinikum der Ludwig-Maximilians-Universität München, Marchioninistr. 15, 81377 Munich, Germany
| | - Catharina Prix
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | | | - Uwe Bertsch
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 23, 81377 Munich, Germany
- Present Address: Institut für Immunologie, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Gerda Mitteregger-Kretzschmar
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Markus Geissen
- Friedrich-Loeffler-Institut, Bundesforschungsinstitut für Tiergesundheit, Greifswald-Insel Riems, Germany
- Present Address: Department of Vascular Medicine, UKE, Hamburg, Germany
| | - Martin Eiden
- Friedrich-Loeffler-Institut, Bundesforschungsinstitut für Tiergesundheit, Greifswald-Insel Riems, Germany
| | - Fabienne Leidel
- Friedrich-Loeffler-Institut, Bundesforschungsinstitut für Tiergesundheit, Greifswald-Insel Riems, Germany
| | | | - Andreas A. Deeg
- BioMolekulare Optik, Ludwig-Maximilians-Universität, Munich, Germany
| | - Julian J. Krauth
- BioMolekulare Optik, Ludwig-Maximilians-Universität, Munich, Germany
| | - Wolfgang Zinth
- BioMolekulare Optik, Ludwig-Maximilians-Universität, Munich, Germany
| | - Paul Tavan
- BioMolekulare Optik, Ludwig-Maximilians-Universität, Munich, Germany
| | - Jens Pilger
- NMR based structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Markus Zweckstetter
- NMR based structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Tobias Frank
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- Neurologie, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Mathias Bähr
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- Neurologie, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Jochen H. Weishaupt
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- Neurologie, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Manfred Uhr
- Labor für Pharmakokinetik, Max-Planck-Institut für Psychiatrie, Munich, Germany
| | - Henning Urlaub
- Bioanalytische Massenspektrometrie, Max-Planck-Institut für biophysikalische Chemie, Göttingen, Germany
- Bioanalytics, Department of Clinical Chemistry, University Medical Center, Göttingen, Germany
| | - Ulrike Teichmann
- Tierhaltung, Max-Planck-Institut für biophysikalische Chemie, Göttingen, Germany
| | - Matthias Samwer
- Zelluläre Logistik, Max-Planck-Institut für biophysikalische Chemie, Göttingen, Germany
| | - Kai Bötzel
- Neurologische Klinik, Klinikum der Ludwig-Maximilians-Universität München, Marchioninistr. 15, 81377 Munich, Germany
| | - Martin Groschup
- Friedrich-Loeffler-Institut, Bundesforschungsinstitut für Tiergesundheit, Greifswald-Insel Riems, Germany
| | - Hans Kretzschmar
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Christian Griesinger
- NMR based structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Armin Giese
- Zentrum für Neuropathologie und Prionforschung, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| |
Collapse
|
20
|
|
21
|
Li Z, Silber BM, Rao S, Gever JR, Bryant C, Gallardo-Godoy A, Dolghih E, Widjaja K, Elepano M, Jacobson MP, Prusiner SB, Renslo AR. 2-Aminothiazoles with improved pharmacotherapeutic properties for treatment of prion disease. ChemMedChem 2013; 8:847-57. [PMID: 23509039 DOI: 10.1002/cmdc.201300007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Indexed: 11/09/2022]
Abstract
Recently, we described the aminothiazole lead (4-biphenyl-4-ylthiazol-2-yl)-(6-methylpyridin-2-yl)-amine (1), which exhibits many desirable properties, including excellent stability in liver microsomes, oral bioavailability of ∼40 %, and high exposure in the brains of mice. Despite its good pharmacokinetic properties, compound 1 exhibited only modest potency in mouse neuroblastoma cells overexpressing the disease-causing prion protein PrP(Sc) . Accordingly, we sought to identify analogues of 1 with improved antiprion potency in ScN2a-cl3 cells while retaining similar or superior properties. Herein we report the discovery of improved lead compounds such as (6-methylpyridin-2-yl)-[4-(4-pyridin-3-yl-phenyl)thiazol-2-yl]amine and cyclopropanecarboxylic acid (4-biphenylthiazol-2-yl)amide, which exhibit brain exposure/EC50 ratios at least tenfold greater than that of compound 1.
Collapse
Affiliation(s)
- Zhe Li
- Institute for Neurodegenerative Disease, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|