201
|
Ranganathan A, Heine P, Rudling A, Plückthun A, Kummer L, Carlsson J. Ligand Discovery for a Peptide-Binding GPCR by Structure-Based Screening of Fragment- and Lead-Like Chemical Libraries. ACS Chem Biol 2017; 12:735-745. [PMID: 28032980 DOI: 10.1021/acschembio.6b00646] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Peptide-recognizing G protein-coupled receptors (GPCRs) are promising therapeutic targets but often resist drug discovery efforts. Determination of crystal structures for peptide-binding GPCRs has provided opportunities to explore structure-based methods in lead development. Molecular docking screens of two chemical libraries, containing either fragment- or lead-like compounds, against a neurotensin receptor 1 crystal structure allowed for a comparison between different drug development strategies for peptide-binding GPCRs. A total of 2.3 million molecules were screened computationally, and 25 fragments and 27 leads that were top-ranked in each library were selected for experimental evaluation. Of these, eight fragments and five leads were confirmed as ligands by surface plasmon resonance. The hit rate for the fragment screen (32%) was thus higher than for the lead-like library (19%), but the affinities of the fragments were ∼100-fold lower. Both screens returned unique scaffolds and demonstrated that a crystal structure of a stabilized peptide-binding GPCR can guide the discovery of small-molecule agonists. The complementary advantages of exploring fragment- and lead-like chemical space suggest that these strategies should be applied synergistically in structure-based screens against challenging GPCR targets.
Collapse
Affiliation(s)
- Anirudh Ranganathan
- Science
for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Philipp Heine
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Axel Rudling
- Science
for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Andreas Plückthun
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Lutz Kummer
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- G7 Therapeutics AG, Grabenstrasse
11a, 8952 Schlieren, Switzerland
| | - Jens Carlsson
- Science
for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, BMC,
Box 596, SE-751 24 Uppsala, Sweden
| |
Collapse
|
202
|
Hassan M, Abbas Q, Ashraf Z, Moustafa AA, Seo SY. Pharmacoinformatics exploration of polyphenol oxidases leading to novel inhibitors by virtual screening and molecular dynamic simulation study. Comput Biol Chem 2017; 68:131-142. [PMID: 28340400 DOI: 10.1016/j.compbiolchem.2017.02.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/23/2017] [Accepted: 02/21/2017] [Indexed: 01/30/2023]
Abstract
Polyphenol oxidases (PPOs)/tyrosinases are metal-dependent enzymes and known as important targets for melanogenesis. Although considerable attempts have been conducted to control the melanin-associated diseases by using various inhibitors. However, the exploration of the best anti-melanin inhibitor without side effect still remains a challenge in drug discovery. In present study, protein structure prediction, ligand-based pharmacophore modeling, virtual screening, molecular docking and dynamic simulation study were used to screen the strong novel inhibitor to cure melanogenesis. The 3D structures of PPO1 and PPO2 were built through homology modeling, while the 3D crystal structures of PPO3 and PPO4 were retrieved from PDB. Pharmacophore modeling was performed using LigandScout 3.1 software and top five models were selected to screen the libraries (2601 of Aurora and 727, 842 of ZINC). Top 10 hit compounds (C1-10) were short-listed having strong binding affinities for PPO1-4. Drug and synthetic accessibility (SA) scores along with absorption, distribution, metabolism, excretion and toxicity (ADMET) assessment were employed to scrutinize the best lead hit. C4 (name) hit showed the best predicted SA score (5.75), ADMET properties and drug-likeness behavior among the short-listed compounds. Furthermore, docking simulations were performed to check the binding affinity of C1-C10 compounds against target proteins (PPOs). The binding affinity values of complex between C4 and PPOs were higher than those of other complexes (-11.70, -12.1, -9.90 and -11.20kcal/mol with PPO1, PPO2, PPO3, or PPO4, respectively). From comparative docking energy and binding analyses, PPO2 may be considered as better target for melanogenesis than others. The potential binding modes of C4, C8 and C10 against PPO2 were explored using molecular dynamics simulations. The root mean square deviation and fluctuation (RMSD/RMSF) graphs results depict the significance of C4 over the other compounds. Overall, bioactivity and ligand efficiency profiles suggested that the proposed hit may be more effective inhibitors for melanogenesis.
Collapse
Affiliation(s)
- Mubashir Hassan
- Department of Biology, College of Natural Sciences, Kongju National University, Gongju 32588, Republic of Korea.
| | - Qamar Abbas
- Department of Biology, College of Natural Sciences, Kongju National University, Gongju 32588, Republic of Korea.
| | - Zaman Ashraf
- Department of Chemistry, Allama Iqbal Open University, Islamabad 44000, Pakistan.
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology, Westren Sydney University, Sydney, New South Wales, Australia; MARCS Institute for Brain and Behaviour, Westren Sydney University, Sydney, New South Wales, Australia.
| | - Sung-Yum Seo
- Department of Biology, College of Natural Sciences, Kongju National University, Gongju 32588, Republic of Korea.
| |
Collapse
|
203
|
New developments in probing and targeting protein acylation in malaria, leishmaniasis and African sleeping sickness. Parasitology 2017; 145:157-174. [PMID: 28270257 DOI: 10.1017/s0031182017000282] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Infections by protozoan parasites, such as Plasmodium falciparum or Leishmania donovani, have a significant health, social and economic impact and threaten billions of people living in tropical and sub-tropical regions of developing countries worldwide. The increasing range of parasite strains resistant to frontline therapeutics makes the identification of novel drug targets and the development of corresponding inhibitors vital. Post-translational modifications (PTMs) are important modulators of biology and inhibition of protein lipidation has emerged as a promising therapeutic strategy for treatment of parasitic diseases. In this review we summarize the latest insights into protein lipidation in protozoan parasites. We discuss how recent chemical proteomic approaches have delivered the first global overviews of protein lipidation in these organisms, contributing to our understanding of the role of this PTM in critical metabolic and cellular functions. Additionally, we highlight the development of new small molecule inhibitors to target parasite acyl transferases.
Collapse
|
204
|
Claveria-Gimeno R, Vega S, Abian O, Velazquez-Campoy A. A look at ligand binding thermodynamics in drug discovery. Expert Opin Drug Discov 2017; 12:363-377. [DOI: 10.1080/17460441.2017.1297418] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Rafael Claveria-Gimeno
- Institute of Biocomputation and Physics of Complex Systems (BIFI), IQFR-CSIC-BIFI and GBsC-CSIC-BIFI Joint Units, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), Zaragoza, Spain
| | - Sonia Vega
- Institute of Biocomputation and Physics of Complex Systems (BIFI), IQFR-CSIC-BIFI and GBsC-CSIC-BIFI Joint Units, Universidad de Zaragoza, Zaragoza, Spain
| | - Olga Abian
- Institute of Biocomputation and Physics of Complex Systems (BIFI), IQFR-CSIC-BIFI and GBsC-CSIC-BIFI Joint Units, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), Zaragoza, Spain
- Department of Biochemistry and Molecular and Cell Biology, Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Adrian Velazquez-Campoy
- Institute of Biocomputation and Physics of Complex Systems (BIFI), IQFR-CSIC-BIFI and GBsC-CSIC-BIFI Joint Units, Universidad de Zaragoza, Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), Zaragoza, Spain
- Department of Biochemistry and Molecular and Cell Biology, Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Fundación ARAID, Government of Aragon, Zaragoza, Spain
| |
Collapse
|
205
|
Pham-The H, Casañola-Martin G, Diéguez-Santana K, Nguyen-Hai N, Ngoc NT, Vu-Duc L, Le-Thi-Thu H. Quantitative structure-activity relationship analysis and virtual screening studies for identifying HDAC2 inhibitors from known HDAC bioactive chemical libraries. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2017; 28:199-220. [PMID: 28332438 DOI: 10.1080/1062936x.2017.1294198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/08/2017] [Indexed: 05/22/2023]
Abstract
Histone deacetylases (HDAC) are emerging as promising targets in cancer, neuronal diseases and immune disorders. Computational modelling approaches have been widely applied for the virtual screening and rational design of novel HDAC inhibitors. In this study, different machine learning (ML) techniques were applied for the development of models that accurately discriminate HDAC2 inhibitors form non-inhibitors. The obtained models showed encouraging results, with the global accuracy in the external set ranging from 0.83 to 0.90. Various aspects related to the comparison of modelling techniques, applicability domain and descriptor interpretations were discussed. Finally, consensus predictions of these models were used for screening HDAC2 inhibitors from four chemical libraries whose bioactivities against HDAC1, HDAC3, HDAC6 and HDAC8 have been known. According to the results of virtual screening assays, structures of some hits with pair-isoform-selective activity (between HDAC2 and other HDACs) were revealed. This study illustrates the power of ML-based QSAR approaches for the screening and discovery of potent, isoform-selective HDACIs.
Collapse
Affiliation(s)
- H Pham-The
- a Hanoi University of Pharmacy , Hanoi , Vietnam
| | - G Casañola-Martin
- b Department of Systems and Computer Engineering , Carleton University , Ottawa , ON , Canada
| | - K Diéguez-Santana
- c Faculty of Life Sciences , Amazonian State University , Puyo , Pastaza , Ecuador
| | - N Nguyen-Hai
- a Hanoi University of Pharmacy , Hanoi , Vietnam
| | - N T Ngoc
- a Hanoi University of Pharmacy , Hanoi , Vietnam
| | - L Vu-Duc
- d School of Medicine and Pharmacy, Vietnam National University , Hanoi , Vietnam
| | - H Le-Thi-Thu
- d School of Medicine and Pharmacy, Vietnam National University , Hanoi , Vietnam
| |
Collapse
|
206
|
Discovery of new nanomolar inhibitors of GPa: Extension of 2-oxo-1,2-dihydropyridinyl-3-yl amide-based GPa inhibitors. Eur J Med Chem 2017; 127:341-356. [PMID: 28076824 DOI: 10.1016/j.ejmech.2016.12.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 11/22/2022]
Abstract
Glycogen Phosphorylase (GP) is a functionally active dimeric enzyme, which is a target for inhibition of the conversion of glycogen to glucose-1-phosphate. In this study we report the design and synthesis of 14 new pyridone derivatives, and seek to extend the SAR analysis of these compounds. The SAR revealed the minor influence of the amide group, importance of the pyridone ring both spatially around the pyridine ring and for possible π-stacking, and confirmed a preference for inclusion of 3,4-dichlorobenzyl moieties, as bookends to the pyridone scaffold. Upon exploring a dimer strategy as part of the SAR analysis, the first extended 2-oxo-dihydropyridinyl-3-yl amide nanomolar based inhibitors of GPa (IC50 = 230 and 260 nM) were identified.
Collapse
|
207
|
Gyertyán I. Cognitive ‘Omics’: Pattern-Based Validation of Potential Drug Targets. Trends Pharmacol Sci 2017; 38:113-126. [DOI: 10.1016/j.tips.2016.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/11/2016] [Accepted: 10/18/2016] [Indexed: 01/01/2023]
|
208
|
Maia EHB, Campos VA, Dos Reis Santos B, Costa MS, Lima IG, Greco SJ, Ribeiro RIMA, Munayer FM, da Silva AM, Taranto AG. Octopus: a platform for the virtual high-throughput screening of a pool of compounds against a set of molecular targets. J Mol Model 2017; 23:26. [PMID: 28064377 DOI: 10.1007/s00894-016-3184-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 12/06/2016] [Indexed: 01/28/2023]
Abstract
Octopus is an automated workflow management tool that is scalable for virtual high-throughput screening (vHTS). It integrates MOPAC2016, MGLTools, PyMOL, and AutoDock Vina. In contrast to other platforms, Octopus can perform docking simulations of an unlimited number of compounds into a set of molecular targets. After generating the ligands in a drawing package in the Protein Data Bank (PDB) format, Octopus can carry out geometry refinement using the semi-empirical method PM7 implemented in MOPAC2016. Docking simulations can be performed using AutoDock Vina and can utilize the Our Own Molecular Targets (OOMT) databank. Finally, the proposed software compiles the best binding energies into a standard table. Here, we describe two successful case studies that were verified by biological assay. In the first case study, the vHTS process was carried out for 22 (phenylamino)urea derivatives. The vHTS process identified a metalloprotease with the PDB code 1GKC as a molecular target for derivative LE&007. In a biological assay, compound LE&007 was found to inhibit 80% of the activity of this enzyme. In the second case study, compound Tx001 was submitted to the Octopus routine, and the results suggested that Plasmodium falciparum ATP6 (PfATP6) as a molecular target for this compound. Following an antimalarial assay, Tx001 was found to have an inhibitory concentration (IC50) of 8.2 μM against PfATP6. These successful examples illustrate the utility of this software for finding appropriate molecular targets for compounds. Hits can then be identified and optimized as new antineoplastic and antimalarial drugs. Finally, Octopus has a friendly Linux-based user interface, and is available at www.drugdiscovery.com.br . Graphical Abstract Octopus: A platform for inverse virtual screening (IVS) to search new molecular targets for drugs.
Collapse
Affiliation(s)
- Eduardo Habib Bechelane Maia
- Universidade Federal de São João del Rei-Campus Centro-Oeste, Divinópolis, MG, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais-Campus Divinópolis, Divinópolis, MG, Brazil
| | - Vinícius Alves Campos
- Universidade Federal de São João del Rei-Campus Centro-Oeste, Divinópolis, MG, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais-Campus Divinópolis, Divinópolis, MG, Brazil
| | - Bianca Dos Reis Santos
- Universidade Federal de São João del Rei-Campus Centro-Oeste, Divinópolis, MG, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais-Campus Divinópolis, Divinópolis, MG, Brazil
| | - Marina Santos Costa
- Universidade Federal de São João del Rei-Campus Centro-Oeste, Divinópolis, MG, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais-Campus Divinópolis, Divinópolis, MG, Brazil
| | - Iann Gabriel Lima
- Universidade Federal de São João del Rei-Campus Centro-Oeste, Divinópolis, MG, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais-Campus Divinópolis, Divinópolis, MG, Brazil
| | - Sandro J Greco
- Universidade Federal do Espírito Santo-UFES, Vitória, ES, Brazil
| | - Rosy I M A Ribeiro
- Universidade Federal de São João del Rei-Campus Centro-Oeste, Divinópolis, MG, Brazil
| | - Felipe M Munayer
- Universidade Federal do Espírito Santo-UFES, Vitória, ES, Brazil
| | | | | |
Collapse
|
209
|
Recent Advances of Microfluidics Technologies in the Field of Medicinal Chemistry. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1016/bs.armc.2017.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
210
|
|
211
|
Chauhan J, Luthra T, Gundla R, Ferraro A, Holzgrabe U, Sen S. A diversity oriented synthesis of natural product inspired molecular libraries. Org Biomol Chem 2017; 15:9108-9120. [DOI: 10.1039/c7ob02230a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Diversity oriented synthesis of natural product inspired compounds from S-tryptophan methyl ester.
Collapse
Affiliation(s)
- Jyoti Chauhan
- Department of Chemistry
- School of Natural Sciences
- Shiv Nadar University
- GautamBudh Nagar
- India
| | - Tania Luthra
- Department of Chemistry
- School of Natural Sciences
- Shiv Nadar University
- GautamBudh Nagar
- India
| | - Rambabu Gundla
- Department of Chemistry
- Gitam Institute of Technology
- GITAM University
- Hyderabad
- India
| | - Antonio Ferraro
- Institute of Pharmacy and Food Chemistry
- University of Würzburg
- Am Hubland
- Germany
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry
- University of Würzburg
- Am Hubland
- Germany
| | - Subhabrata Sen
- Department of Chemistry
- School of Natural Sciences
- Shiv Nadar University
- GautamBudh Nagar
- India
| |
Collapse
|
212
|
Discovery of novel dual VEGFR2 and Src inhibitors using a multistep virtual screening approach. Future Med Chem 2016; 9:7-24. [PMID: 27995811 DOI: 10.4155/fmc-2016-0162] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
AIM Simultaneous inhibition of VEGFR2 and Src may enhance the efficacy of VEGFR2-targeted cancer therapeutics. Hence, development of dual inhibitors on VEGFR2 and Src can be a useful strategy for such treatments. MATERIALS & METHODS A multistep virtual screening protocol, comprising ligand-based support vector machines method, drug-likeness rules filter and structure-based molecular docking, was developed and employed to identify dual inhibitors of VEGFR2 and Src from a large commercial chemical library. Kinase inhibitory assays and cell viability assays were then used for experimental validation. RESULTS A set of compounds belonging to six different molecular scaffolds was identified and sent for biological evaluation. Compound 3c belonging to the 2-amino-3-cyanopyridine scaffold exhibited good antiproliferative effect and dual-target activities against VEGFR2 and Src. CONCLUSION This study demonstrated the ability of the multistep virtual screening approach to identify novel multitarget agents.
Collapse
|
213
|
Goncalves V, Brannigan JA, Laporte A, Bell AS, Roberts SM, Wilkinson AJ, Leatherbarrow RJ, Tate EW. Structure-guided optimization of quinoline inhibitors of Plasmodium N-myristoyltransferase. MEDCHEMCOMM 2016. [PMID: 28626547 PMCID: PMC5463734 DOI: 10.1039/c6md00531d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Quinolines with balanced activities against both Plasmodium vivax and Plasmodium falciparum N-myristoyltransferase were identified.
The parasite Plasmodium vivax is the most widely distributed cause of recurring malaria. N-Myristoyltransferase (NMT), an enzyme that catalyses the covalent attachment of myristate to the N-terminal glycine of substrate proteins, has been described as a potential target for the treatment of this disease. Herein, we report the synthesis and the structure-guided optimization of a series of quinolines with balanced activity against both Plasmodium vivax and Plasmodium falciparum N-myristoyltransferase (NMT).
Collapse
Affiliation(s)
- Victor Goncalves
- Department of Chemistry , Imperial College London , London SW7 2AZ , UK . ;
| | - James A Brannigan
- Structural Biology Laboratory , Department of Chemistry , University of York , York YO10 5DD , UK
| | - Alice Laporte
- Department of Chemistry , Imperial College London , London SW7 2AZ , UK . ;
| | - Andrew S Bell
- Department of Chemistry , Imperial College London , London SW7 2AZ , UK . ;
| | - Shirley M Roberts
- Structural Biology Laboratory , Department of Chemistry , University of York , York YO10 5DD , UK
| | - Anthony J Wilkinson
- Structural Biology Laboratory , Department of Chemistry , University of York , York YO10 5DD , UK
| | | | - Edward W Tate
- Department of Chemistry , Imperial College London , London SW7 2AZ , UK . ;
| |
Collapse
|
214
|
Li Z, Yang J, Wang X, Li H, Liu C, Wang N, Huang W, Qian H. Discovery of novel free fatty acid receptor 1 agonists bearing triazole core via click chemistry. Bioorg Med Chem 2016; 24:5449-5454. [DOI: 10.1016/j.bmc.2016.08.068] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 01/18/2023]
|
215
|
Bajusz D, Ferenczy GG, Keserű GM. Ensemble docking-based virtual screening yields novel spirocyclic JAK1 inhibitors. J Mol Graph Model 2016; 70:275-283. [PMID: 27771575 DOI: 10.1016/j.jmgm.2016.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 10/12/2016] [Accepted: 10/14/2016] [Indexed: 12/20/2022]
Abstract
Small molecule inhibition of Janus kinases (JAKs) has been demonstrated as a viable strategy for the treatment of various inflammatory conditions and continues to emerge in cancer-related indications. In this study, a large supplier database was screened to identify novel chemistry starting points for JAK1. The docking-based screening was followed up by testing ten hit compounds experimentally, out of which five have displayed single-digit micromolar and submicromolar IC50 values on JAK1. Thus, the study was concluded with the discovery of five novel JAK inhibitors from a tiny screening deck with a remarkable hitrate of 50%. The results have highlighted spirocyclic pyrrolopyrimidines with submicromolar JAK1 IC50 values and a preference for JAK1 over JAK2 as potential starting points in developing a novel class of JAK1 inhibitors.
Collapse
Affiliation(s)
- Dávid Bajusz
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary
| | - György G Ferenczy
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary.
| |
Collapse
|
216
|
Affiliation(s)
- Michael F. Rafferty
- Department of Medicinal Chemistry, University of Kansas, 4070 Malott Hall, Lawrence, Kansas 66045, United States
| |
Collapse
|
217
|
Deveau AP, Bentley VL, Berman JN. Using zebrafish models of leukemia to streamline drug screening and discovery. Exp Hematol 2016; 45:1-9. [PMID: 27720937 DOI: 10.1016/j.exphem.2016.09.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 10/20/2022]
Abstract
Current treatment strategies for acute leukemias largely rely on nonspecific cytotoxic drugs that result in high therapy-related morbidity and mortality. Cost-effective, pertinent animal models are needed to link in vitro studies with the development of new therapeutic agents in clinical trials on a high-throughput scale. However, targeted therapies have had limited success moving from bench to clinic, often due to unexpected off-target effects. The zebrafish has emerged as a reliable in vivo tool for modeling human leukemia. Zebrafish genetic and xenograft models of acute leukemia provide an unprecedented opportunity to conduct rapid, phenotype-based screens. This allows for the identification of relevant therapies while simultaneously evaluating drug toxicity, thus circumventing the limitations of target-centric approaches.
Collapse
Affiliation(s)
- Adam P Deveau
- Department of Pediatrics, IWK Health Centre, Halifax, Nova Scotia, Canada
| | - Victoria L Bentley
- Undergraduate Medical Program, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jason N Berman
- Department of Pediatrics, IWK Health Centre, Halifax, Nova Scotia, Canada; Departments of Microbiology and Immunology and Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
218
|
Tzvetkov NT, Neumann B, Stammler HG, Antonov L. A simple approach to multifunctionalized N1-alkylated 7-amino-6-azaoxindole derivatives using their in situ stabilized tautomer form. Tetrahedron 2016. [DOI: 10.1016/j.tet.2016.08.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
219
|
Borbás E, Sinkó B, Tsinman O, Tsinman K, Kiserdei É, Démuth B, Balogh A, Bodák B, Domokos A, Dargó G, Balogh GT, Nagy ZK. Investigation and Mathematical Description of the Real Driving Force of Passive Transport of Drug Molecules from Supersaturated Solutions. Mol Pharm 2016; 13:3816-3826. [DOI: 10.1021/acs.molpharmaceut.6b00613] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Enikő Borbás
- Department
of Organic Chemistry and Technology, Budapest University of Technology and Economics, Budapest 1111, Hungary
| | - Bálint Sinkó
- Pion Inc., Billerica, Massachusetts 01821, United States
| | - Oksana Tsinman
- Pion Inc., Billerica, Massachusetts 01821, United States
| | | | - Éva Kiserdei
- Department
of Organic Chemistry and Technology, Budapest University of Technology and Economics, Budapest 1111, Hungary
| | - Balázs Démuth
- Department
of Organic Chemistry and Technology, Budapest University of Technology and Economics, Budapest 1111, Hungary
| | - Attila Balogh
- Department
of Organic Chemistry and Technology, Budapest University of Technology and Economics, Budapest 1111, Hungary
| | - Brigitta Bodák
- Department
of Organic Chemistry and Technology, Budapest University of Technology and Economics, Budapest 1111, Hungary
| | - András Domokos
- Department
of Organic Chemistry and Technology, Budapest University of Technology and Economics, Budapest 1111, Hungary
| | - Gergő Dargó
- Department
of Organic Chemistry and Technology, Budapest University of Technology and Economics, Budapest 1111, Hungary
- Compound
Profiling Laboratory, Gedeon Richter Plc., Budapest 1103, Hungary
| | - György T. Balogh
- Compound
Profiling Laboratory, Gedeon Richter Plc., Budapest 1103, Hungary
| | - Zsombor K. Nagy
- Department
of Organic Chemistry and Technology, Budapest University of Technology and Economics, Budapest 1111, Hungary
| |
Collapse
|
220
|
A cell-based approach to characterize antimicrobial compounds through kinetic dose response. Bioorg Med Chem 2016; 24:6315-6319. [PMID: 27713016 DOI: 10.1016/j.bmc.2016.09.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/19/2016] [Accepted: 09/21/2016] [Indexed: 12/18/2022]
Abstract
The rapid spread of antibiotic resistance has created a pressing need for the development of novel drug screening platforms. Herein, we report on the use of cell-based kinetic dose response curves for small molecule characterization in antibiotic discovery efforts. Kinetically monitoring bacterial growth at sub-inhibitory concentrations of antimicrobial small molecules generates unique dose response profiles. We show that clustering of profiles by growth characteristics can classify antibiotics by mechanism of action. Furthermore, changes in growth kinetics have the potential to offer insight into the mechanistic action of novel molecules and can be used to predict off-target effects generated through structure-activity relationship studies. Kinetic dose response also allows for detection of unstable compounds early in the lead development process. We propose that this kinetic approach is a rapid and cost-effective means to gather critical information on antimicrobial small molecules during the hit selection and lead development pipeline.
Collapse
|
221
|
Xiao D, Hu JJ, Zhu JY, Wang SB, Zhuo RX, Zhang XZ. A redox-responsive mesoporous silica nanoparticle with a therapeutic peptide shell for tumor targeting synergistic therapy. NANOSCALE 2016; 8:16702-16709. [PMID: 27714082 DOI: 10.1039/c6nr04784j] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
In this study, we report a novel redox-responsive mesoporous silica nanoparticle (MSN)-based nanocarrier, capping with a therapeutic peptide ((RGDWWW)2KC) containing a RGD target motif, for tumor targeting synergistic therapy, which is designated as TTSTMSN. The MSN was decorated with a tumor-targeting therapeutic peptide as a potential gatekeeper. The two branched peptides containing rich tryptophans allowed the pores to be blocked via π-π stacking and hydrophobic interactions. Once the drug loaded nanoparticles were taken up by the cancer cells through integrin-mediated endocytosis, the therapeutic peptide capping shells on the surface of MSNs were released, inducing the loaded drug to diffuse into the cytoplasm after breaking of the disulfide bonds, triggered by the high concentration of glutathione (GSH) in cancer cells. At the same time, the falling therapeutic rich tryptophans in the branched chains interacted with DNA due to the indole rings, leading to disturbance of the DNA structure through the strong π interactions and causing cell apoptosis. There is no such report on capping of drug loaded porous silica with a therapeutic peptide shell, co-delivering an anticancer drug and therapeutic agent for tumor targeting synergistic therapy, which will have great potential in developing multifunctional nanocarriers based on therapeutic peptides for synergistic treatment.
Collapse
Affiliation(s)
- Dong Xiao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Jing-Jing Hu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Jing-Yi Zhu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Shi-Bo Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| |
Collapse
|
222
|
Huang YW, Frontier AJ. Nazarov Cyclization/Internal Redox Cyclization Sequence for the Synthesis of N-Heterocyclic Bridged Ring Systems. Org Lett 2016; 18:4896-4899. [DOI: 10.1021/acs.orglett.6b02369] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yu-Wen Huang
- Department of Chemistry, University of Rochester, Rochester, New York 14627-0216, United States
| | - Alison J. Frontier
- Department of Chemistry, University of Rochester, Rochester, New York 14627-0216, United States
| |
Collapse
|
223
|
Plate-based diversity subset screening generation 2: an improved paradigm for high-throughput screening of large compound files. Mol Divers 2016; 20:789-803. [PMID: 27631533 PMCID: PMC5055576 DOI: 10.1007/s11030-016-9692-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/29/2016] [Indexed: 01/07/2023]
Abstract
High-throughput screening (HTS) is an effective method for lead and probe discovery that is widely used in industry and academia to identify novel chemical matter and to initiate the drug discovery process. However, HTS can be time consuming and costly and the use of subsets as an efficient alternative to screening entire compound collections has been investigated. Subsets may be selected on the basis of chemical diversity, molecular properties, biological activity diversity or biological target focus. Previously, we described a novel form of subset screening: plate-based diversity subset (PBDS) screening, in which the screening subset is constructed by plate selection (rather than individual compound cherry-picking), using algorithms that select for compound quality and chemical diversity on a plate basis. In this paper, we describe a second-generation approach to the construction of an updated subset: PBDS2, using both plate and individual compound selection, that has an improved coverage of the chemical space of the screening file, whilst only selecting the same number of plates for screening. We describe the validation of PBDS2 and its successful use in hit and lead discovery. PBDS2 screening became the default mode of singleton (one compound per well) HTS for lead discovery in Pfizer.
Collapse
|
224
|
Elvang PA, Hinna AH, Brouwers J, Hens B, Augustijns P, Brandl M. Bile Salt Micelles and Phospholipid Vesicles Present in Simulated and Human Intestinal Fluids: Structural Analysis by Flow Field–Flow Fractionation/Multiangle Laser Light Scattering. J Pharm Sci 2016; 105:2832-2839. [DOI: 10.1016/j.xphs.2016.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 02/04/2016] [Accepted: 03/03/2016] [Indexed: 12/11/2022]
|
225
|
Silva T, Mohamed T, Shakeri A, Rao PPN, Martínez-González L, Pérez DI, Martínez A, Valente MJ, Garrido J, Uriarte E, Serrão P, Soares-da-Silva P, Remião F, Borges F. Development of Blood-Brain Barrier Permeable Nitrocatechol-Based Catechol O-Methyltransferase Inhibitors with Reduced Potential for Hepatotoxicity. J Med Chem 2016; 59:7584-97. [PMID: 27463695 DOI: 10.1021/acs.jmedchem.6b00666] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Recent efforts have been focused on the development of centrally active COMT inhibitors, which can be valuable assets for neurological disorders such as Parkinson's disease, due to the severe hepatotoxicity risk associated with tolcapone. New nitrocatechol COMT inhibitors based on naturally occurring caffeic acid and caffeic acid phenethyl ester were developed. All nitrocatechol derivatives displayed potent inhibition of peripheral and cerebral COMT within the nanomolar range. Druglike derivatives 13, 15, and 16 were predicted to cross the blood-brain barrier in vitro and were significantly less toxic than tolcapone and entacapone when incubated at 50 μM with rat primary hepatocytes. Moreover, their unique acidity and electrochemical properties decreased the chances of formation of reactive quinone-imines and, as such, the potential for hepatotoxicity. The binding mode of 16 confirmed that the major interactions with COMT were established via the nitrocatechol ring, allowing derivatization of the side chain for future lead optimization efforts.
Collapse
Affiliation(s)
- Tiago Silva
- CIQ/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Tarek Mohamed
- School of Pharmacy, Health Sciences Campus, University of Waterloo , 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Arash Shakeri
- School of Pharmacy, Health Sciences Campus, University of Waterloo , 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Praveen P N Rao
- School of Pharmacy, Health Sciences Campus, University of Waterloo , 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | | | - Daniel I Pérez
- Centro de Investigaciónes Biológicas, CSIC, C/Ramiro de Maeztu, 28040 Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciónes Biológicas, CSIC, C/Ramiro de Maeztu, 28040 Madrid, Spain
| | - Maria João Valente
- Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, UCIBIO-REQUIMTE , Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Jorge Garrido
- Departamento de Engenharia Química, Instituto Superior de Engenharia do Porto (ISEP), Instituto Politécnico Porto , 4200-072 Porto, Portugal
| | - Eugenio Uriarte
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela , 15782 Santiago de Compostela, Spain
| | - Paula Serrão
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto , 4200-319 Porto, Portugal.,MedInUP-Center for Drug Discovery and Innovative Medicines, University of Porto, 4200-319, Porto, Portugal
| | - Patrício Soares-da-Silva
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto , 4200-319 Porto, Portugal.,MedInUP-Center for Drug Discovery and Innovative Medicines, University of Porto, 4200-319, Porto, Portugal
| | - Fernando Remião
- Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, UCIBIO-REQUIMTE , Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Fernanda Borges
- CIQ/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto , Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| |
Collapse
|
226
|
Hoagland DT, Liu J, Lee RB, Lee RE. New agents for the treatment of drug-resistant Mycobacterium tuberculosis. Adv Drug Deliv Rev 2016; 102:55-72. [PMID: 27151308 PMCID: PMC4903924 DOI: 10.1016/j.addr.2016.04.026] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 02/06/2023]
Abstract
Inadequate dosing and incomplete treatment regimens, coupled with the ability of the tuberculosis bacilli to cause latent infections that are tolerant of currently used drugs, have fueled the rise of multidrug-resistant tuberculosis (MDR-TB). Treatment of MDR-TB infections is a major clinical challenge that has few viable or effective solutions; therefore patients face a poor prognosis and years of treatment. This review focuses on emerging drug classes that have the potential for treating MDR-TB and highlights their particular strengths as leads including their mode of action, in vivo efficacy, and key medicinal chemistry properties. Examples include the newly approved drugs bedaquiline and delaminid, and other agents in clinical and late preclinical development pipeline for the treatment of MDR-TB. Herein, we discuss the challenges to developing drugs to treat tuberculosis and how the field has adapted to these difficulties, with an emphasis on drug discovery approaches that might produce more effective agents and treatment regimens.
Collapse
Affiliation(s)
- Daniel T Hoagland
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Pharmaceutical Sciences Graduate Program, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jiuyu Liu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Robin B Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
227
|
In vitro effects of a new fused azaisocytosine-like congener on relative cell proliferation, necrosis and cell cycle in cancer and normal cell cultures. Mol Cell Biochem 2016; 418:179-88. [DOI: 10.1007/s11010-016-2744-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/15/2016] [Indexed: 01/31/2023]
|
228
|
Stotani S, Lorenz C, Winkler M, Medda F, Picazo E, Ortega Martinez R, Karawajczyk A, Sanchez-Quesada J, Giordanetto F. Design and Synthesis of Fsp(3)-Rich, Bis-Spirocyclic-Based Compound Libraries for Biological Screening. ACS COMBINATORIAL SCIENCE 2016; 18:330-6. [PMID: 27163646 DOI: 10.1021/acscombsci.6b00005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The exploration of innovative chemical space is a critical step in the early phases of drug discovery. Bis-spirocyclic frameworks occur in natural products and other biologically relevant metabolites and show attractive features, such as molecular compactness, structural complexity, and three-dimensional character. A concise approach to the synthesis of bis-spirocyclic-based compound libraries starting from readily available commercial reagents and robust chemical transformations has been developed. A number of novel bis-spirocyclic scaffold examples, as implemented in the European Lead Factory project, is presented.
Collapse
Affiliation(s)
- Silvia Stotani
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| | - Christoph Lorenz
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| | - Matthias Winkler
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| | - Federico Medda
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| | - Edwige Picazo
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| | - Raquel Ortega Martinez
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| | - Anna Karawajczyk
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| | - Jorge Sanchez-Quesada
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| | - Fabrizio Giordanetto
- Medicinal Chemistry, Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, 44227 Dortmund, Germany
| |
Collapse
|
229
|
Bergström CAS, Charman WN, Porter CJH. Computational prediction of formulation strategies for beyond-rule-of-5 compounds. Adv Drug Deliv Rev 2016; 101:6-21. [PMID: 26928657 DOI: 10.1016/j.addr.2016.02.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/11/2016] [Accepted: 02/17/2016] [Indexed: 12/12/2022]
Abstract
The physicochemical properties of some contemporary drug candidates are moving towards higher molecular weight, and coincidentally also higher lipophilicity in the quest for biological selectivity and specificity. These physicochemical properties move the compounds towards beyond rule-of-5 (B-r-o-5) chemical space and often result in lower water solubility. For such B-r-o-5 compounds non-traditional delivery strategies (i.e. those other than conventional tablet and capsule formulations) typically are required to achieve adequate exposure after oral administration. In this review, we present the current status of computational tools for prediction of intestinal drug absorption, models for prediction of the most suitable formulation strategies for B-r-o-5 compounds and models to obtain an enhanced understanding of the interplay between drug, formulation and physiological environment. In silico models are able to identify the likely molecular basis for low solubility in physiologically relevant fluids such as gastric and intestinal fluids. With this baseline information, a formulation scientist can, at an early stage, evaluate different orally administered, enabling formulation strategies. Recent computational models have emerged that predict glass-forming ability and crystallisation tendency and therefore the potential utility of amorphous solid dispersion formulations. Further, computational models of loading capacity in lipids, and therefore the potential for formulation as a lipid-based formulation, are now available. Whilst such tools are useful for rapid identification of suitable formulation strategies, they do not reveal drug localisation and molecular interaction patterns between drug and excipients. For the latter, Molecular Dynamics simulations provide an insight into the interplay between drug, formulation and intestinal fluid. These different computational approaches are reviewed. Additionally, we analyse the molecular requirements of different targets, since these can provide an early signal that enabling formulation strategies will be required. Based on the analysis we conclude that computational biopharmaceutical profiling can be used to identify where non-conventional gateways, such as prediction of 'formulate-ability' during lead optimisation and early development stages, are important and may ultimately increase the number of orally tractable contemporary targets.
Collapse
Affiliation(s)
- Christel A S Bergström
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia; Department of Pharmacy, Uppsala University, Uppsala Biomedical Center, P.O. Box 580, SE-751 23 Uppsala, Sweden.
| | - William N Charman
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia; ARC Centre of Excellence in Convergent Nano-Bio Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
230
|
Leeson PD. Molecular inflation, attrition and the rule of five. Adv Drug Deliv Rev 2016; 101:22-33. [PMID: 26836397 DOI: 10.1016/j.addr.2016.01.018] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/12/2016] [Accepted: 01/18/2016] [Indexed: 12/18/2022]
Abstract
Physicochemical properties underlie all aspects of drug action and are critical for solubility, permeability and successful formulation. Specific physicochemical properties shown to be relevant to oral drugs are size, lipophilicity, ionisation, hydrogen bonding, polarity, aromaticity and shape. The rule of 5 (Ro5) and subsequent studies have raised awareness of the importance of compound quality amongst bioactive molecules. Lipophilicity, probably the most important physical property of oral drugs, has on average changed little over time in oral drugs, until increases in drugs published after 1990. In contrast other molecular properties such as average size have increased significantly. Factors influencing property inflation include the targets pursued, where antivirals frequently violate the Ro5, risk/benefit considerations, and variable drug discovery practices. The compounds published in patents from the pharmaceutical industry are on average larger, more lipophilic and less complex than marketed oral drugs. The variation between individual companies' patented compounds is due to different practices and not to the targets pursued. Overall, there is demonstrable physical property attrition in moving from patents to candidate drugs to marketed drugs. The pharmaceutical industry's recent poor productivity has been due, in part, to progression of molecules that are unable to unambiguously test clinical efficacy, and attrition can therefore be improved by ensuring candidate drug quality is 'fit for purpose.' The combined ligand efficiency (LE) and lipophilic ligand efficiency (LLE) values of many marketed drugs are optimised relative to other molecules acting at the same target. Application of LLE in optimisation can help identify improved leads, even with challenging targets that seem to require lipophilic ligands. Because of their targets, some projects may need to pursue 'beyond Ro5' physicochemical space; such projects will require non-standard lead generation and optimisation and should not dominate in a well-balanced portfolio. Compound quality is controllable by lead selection and optimisation and should not be a cause of clinical failure.
Collapse
Affiliation(s)
- Paul D Leeson
- Paul Leeson Consulting Ltd, The Malt House, Main Street, Congerstone, Nuneaton, Warks CV13 6LZ, UK.
| |
Collapse
|
231
|
Li Z, Qiu Q, Geng X, Yang J, Huang W, Qian H. Free fatty acid receptor agonists for the treatment of type 2 diabetes: drugs in preclinical to phase II clinical development. Expert Opin Investig Drugs 2016; 25:871-90. [PMID: 27171154 DOI: 10.1080/13543784.2016.1189530] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The alarming prevalence of type 2 diabetes mellitus (T2DM) stimulated the exploitation of new antidiabetic drugs with extended durability and enhanced safety. In this regard, the free fatty acid receptor 1 (FFA1) and FFA4 have emerged as attractive targets in the last decade. FFA1 has prominent advantages in promoting insulin and incretin secretion while FFA4 shows great potential in incretin secretion, insulin sensitization and anti-inflammatory effects. AREA COVERED Herein, the authors focus specifically on FFA1 and FFA4 agonists in clinical trials and preclinical development. LY2922470, P11187 and SHR0534 are currently active in clinical trials while the CNX-011-67, SAR1, DS-1558 and BMS-986118 are in preclinical phase. The information for this review is retrieved from Integrity, Scifinder, Espacenet and clinicaltrials.gov databases. EXPERT OPINION Current proof-of-concept in clinical trials suggests that FFA1 agonists have a significant improvement for T2DM without the risk of hypoglycemia. However, there are still several challenging problems including the mechanism of the receptor and the efficacy and safety of the ligands.
Collapse
Affiliation(s)
- Zheng Li
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China
| | - Qianqian Qiu
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China
| | - Xinqian Geng
- b Department of Endocrinology and Metabolism , Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes , Shanghai , PR China
| | - Jianyong Yang
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China
| | - Wenlong Huang
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China.,c Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease , China Pharmaceutical University , Nanjing , PR China
| | - Hai Qian
- a Center of Drug Discovery, State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing , PR China.,c Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease , China Pharmaceutical University , Nanjing , PR China
| |
Collapse
|
232
|
|
233
|
|
234
|
Design, synthesis and Structure–activity relationship studies of new thiazole-based free fatty acid receptor 1 agonists for the treatment of type 2 diabetes. Eur J Med Chem 2016; 113:246-57. [DOI: 10.1016/j.ejmech.2016.02.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 02/14/2016] [Accepted: 02/15/2016] [Indexed: 01/03/2023]
|
235
|
Li Z, Pan M, Su X, Dai Y, Fu M, Cai X, Shi W, Huang W, Qian H. Discovery of novel pyrrole-based scaffold as potent and orally bioavailable free fatty acid receptor 1 agonists for the treatment of type 2 diabetes. Bioorg Med Chem 2016; 24:1981-7. [DOI: 10.1016/j.bmc.2016.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/04/2016] [Accepted: 03/05/2016] [Indexed: 11/26/2022]
|
236
|
Mansour NR, Paveley R, Gardner JMF, Bell AS, Parkinson T, Bickle Q. High Throughput Screening Identifies Novel Lead Compounds with Activity against Larval, Juvenile and Adult Schistosoma mansoni. PLoS Negl Trop Dis 2016; 10:e0004659. [PMID: 27128493 PMCID: PMC4851381 DOI: 10.1371/journal.pntd.0004659] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 04/01/2016] [Indexed: 11/18/2022] Open
Abstract
An estimated 600 million people are affected by the helminth disease schistosomiasis caused by parasites of the genus Schistosoma. There is currently only one drug recommended for treating schistosomiasis, praziquantel (PZQ), which is effective against adult worms but not against the juvenile stage. In an attempt to identify improved drugs for treating the disease, we have carried out high throughput screening of a number of small molecule libraries with the aim of identifying lead compounds with balanced activity against all life stages of Schistosoma. A total of almost 300,000 compounds were screened using a high throughput assay based on motility of worm larvae and image analysis of assay plates. Hits were screened against juvenile and adult worms to identify broadly active compounds and against a mammalian cell line to assess cytotoxicity. A number of compounds were identified as promising leads for further chemical optimization. Schistosomiasis is a parasitic infection that affects an estimated 600 million people in developing countries. Treatment is currently dependent on a single drug which is only active against the adult form of the disease. Treatment of infected patients eliminates adult worms but leaves any juveniles to survive and develop, thus continuing the infection. A drug acting equally against adult and juvenile worms is likely to be more effective at eliminating the disease. We have identified quality chemical leads with balanced activity against all life stages of the parasite and with a good window over cytotoxicity, which could form the basis of an exciting new drug discovery project.
Collapse
Affiliation(s)
- Nuha R. Mansour
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ross Paveley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - J. Mark F. Gardner
- Salvensis, Discovery Park House, Discovery Park, Sandwich, Kent, United Kingdom
| | - Andrew S. Bell
- Salvensis, Discovery Park House, Discovery Park, Sandwich, Kent, United Kingdom
| | - Tanya Parkinson
- Salvensis, Discovery Park House, Discovery Park, Sandwich, Kent, United Kingdom
- * E-mail:
| | - Quentin Bickle
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
237
|
Sun H, Pan P, Tian S, Xu L, Kong X, Li Y, Dan Li, Hou T. Constructing and Validating High-Performance MIEC-SVM Models in Virtual Screening for Kinases: A Better Way for Actives Discovery. Sci Rep 2016; 6:24817. [PMID: 27102549 PMCID: PMC4840416 DOI: 10.1038/srep24817] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/06/2016] [Indexed: 01/23/2023] Open
Abstract
The MIEC-SVM approach, which combines molecular interaction energy components (MIEC) derived from free energy decomposition and support vector machine (SVM), has been found effective in capturing the energetic patterns of protein-peptide recognition. However, the performance of this approach in identifying small molecule inhibitors of drug targets has not been well assessed and validated by experiments. Thereafter, by combining different model construction protocols, the issues related to developing best MIEC-SVM models were firstly discussed upon three kinase targets (ABL, ALK, and BRAF). As for the investigated targets, the optimized MIEC-SVM models performed much better than the models based on the default SVM parameters and Autodock for the tested datasets. Then, the proposed strategy was utilized to screen the Specs database for discovering potential inhibitors of the ALK kinase. The experimental results showed that the optimized MIEC-SVM model, which identified 7 actives with IC50 < 10 μM from 50 purchased compounds (namely hit rate of 14%, and 4 in nM level) and performed much better than Autodock (3 actives with IC50 < 10 μM from 50 purchased compounds, namely hit rate of 6%, and 2 in nM level), suggesting that the proposed strategy is a powerful tool in structure-based virtual screening.
Collapse
Affiliation(s)
- Huiyong Sun
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
- State Key Lab of CAD&CG, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Peichen Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Sheng Tian
- Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Lei Xu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Xiaotian Kong
- Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Youyong Li
- Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, P. R. China
| | - Dan Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
- State Key Lab of CAD&CG, Zhejiang University, Hangzhou, Zhejiang 310058, P. R. China
| |
Collapse
|
238
|
Liu N, Wei L, Huang L, Yu F, Zheng W, Qin B, Zhu DQ, Morris-Natschke SL, Jiang S, Chen CH, Lee KH, Xie L. Novel HIV-1 Non-nucleoside Reverse Transcriptase Inhibitor Agents: Optimization of Diarylanilines with High Potency against Wild-Type and Rilpivirine-Resistant E138K Mutant Virus. J Med Chem 2016; 59:3689-704. [PMID: 27070547 DOI: 10.1021/acs.jmedchem.5b01827] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Three series (6, 13, and 14) of new diarylaniline (DAAN) analogues were designed, synthesized, and evaluated for anti-HIV potency, especially against the E138K viral strain with a major mutation conferring resistance to the new-generation non-nucleoside reverse transcriptase inhibitor drug rilpivirine (1b). Promising new compounds were then assessed for physicochemical and associated pharmaceutical properties, including aqueous solubility, log P value, and metabolic stability, as well as predicted lipophilic parameters of ligand efficiency, ligand lipophilic efficiency, and ligand efficiency-dependent lipophilicity indices, which are associated with ADME property profiles. Compounds 6a, 14c, and 14d showed high potency against the 1b-resistant E138K mutated viral strain as well as good balance between anti-HIV-1 activity and desirable druglike properties. From the perspective of optimizing future NNRTI compounds as clinical trial candidates, computational modeling results provided valuable information about how the R(1) group might provide greater efficacy against the E138K mutant.
Collapse
Affiliation(s)
- Na Liu
- Beijing Institute of Pharmacology & Toxicology , 27 Tai-Ping Road, Beijing 100850, China
| | - Lei Wei
- Beijing Institute of Pharmacology & Toxicology , 27 Tai-Ping Road, Beijing 100850, China
| | - Li Huang
- Surgical Oncology Research Facility, Duke University Medical Center , Box 2926, Durham, North Carolina 27710, United States
| | - Fei Yu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University , Shanghai 200032, China.,Lindsley F. Kimball Research Institute, New York Blood Center , New York, New York 10065, United States
| | - Weifan Zheng
- Department of Pharmaceutical Sciences & BRITE Institute, North Carolina Central University , Durham, North Carolina 27707, United States
| | - Bingjie Qin
- Beijing Institute of Pharmacology & Toxicology , 27 Tai-Ping Road, Beijing 100850, China
| | - Dong-Qin Zhu
- Beijing Institute of Pharmacology & Toxicology , 27 Tai-Ping Road, Beijing 100850, China
| | - Susan L Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599-7568, United States
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University , Shanghai 200032, China.,Lindsley F. Kimball Research Institute, New York Blood Center , New York, New York 10065, United States
| | - Chin-Ho Chen
- Surgical Oncology Research Facility, Duke University Medical Center , Box 2926, Durham, North Carolina 27710, United States
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599-7568, United States.,Chinese Medicine Research and Development Center, China Medical University and Hospital , Taichung, Taiwan
| | - Lan Xie
- Beijing Institute of Pharmacology & Toxicology , 27 Tai-Ping Road, Beijing 100850, China.,Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599-7568, United States
| |
Collapse
|
239
|
Mignani S, Huber S, Tomás H, Rodrigues J, Majoral JP. Compound high-quality criteria: a new vision to guide the development of drugs, current situation. Drug Discov Today 2016; 21:573-84. [PMID: 26802700 DOI: 10.1016/j.drudis.2016.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/12/2016] [Accepted: 01/13/2016] [Indexed: 02/08/2023]
Abstract
For several decades, the pharmaceutical industry has suffered due to major issues such as reductions of the number of FDA approved drugs and biologics. Several analyses have been highlighted that the 'druglikeness' is one of the strategies to improve succeed rates of screening such as, for instance, high-throughput screening (HTS), and then hits (as starting point), leads and clinical candidates. It is clear that the improvement of compound quality accelerates the drug discovery projects. The monitoring of several indices to avoid 'molecular obesity' (ADMET problems) of final drugs from good-quality 'low-fat' starting points represents today a powerful strategy of optimization process. The development of the new guides to find drugs highlighting attempts at improving the attrition rate from hits to final medicines by focusing on how to improve the druggability of hits, leads and drugs during the drug discovery process represents a key approach to design next better generation of medicines.
Collapse
Affiliation(s)
- Serge Mignani
- Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, 45, rue des Saints Pères, 75006 Paris, France.
| | - Scot Huber
- SCYNEXIS, Inc., P.O. Box 12878, Research Triangle Park, NC 27709, USA
| | - Helena Tomás
- CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9000-390 Funchal, Portugal
| | - João Rodrigues
- CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9000-390 Funchal, Portugal.
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, BP 44099, 31077 Toulouse Cedex 4, France; Université de Toulouse, UPS, INPT, 31077 Toulouse Cedex 4, France.
| |
Collapse
|
240
|
Navarrete-Vázquez G, Austrich-Olivares A, Godínez-Chaparro B, Hidalgo-Figueroa S, Estrada-Soto S, Hernández-Núñez E, Torres-Gómez H, Schepmann D, Wünsch B. Discovery of 2-(3,4-dichlorophenoxy)-N-(2-morpholin-4-ylethyl)acetamide: A selective σ1 receptor ligand with antinociceptive effect. Biomed Pharmacother 2016; 79:284-93. [PMID: 27044839 DOI: 10.1016/j.biopha.2016.02.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 02/07/2023] Open
Abstract
Compound 2-(3,4-dichlorophenoxy)-N-(2-morpholin-4-ylethyl)acetamide (1) was designed, prepared and the in vitro binding evaluation against σ1 and σ2 receptors was measured. Compound 1 showed high σ1 receptor affinity (Ki=42 nM) and it was 36-times more selective for σ1 than σ2 receptor. Also, it was performed a molecular docking of compound 1 into the ligand binding pocket homology model of σ1 receptor, showing a salt bridge between the ionized morpholine ring and Asp126, as well as important short contacts with residues Tyr120, His154 and Trp164. Ligand efficiency indexes and predicted toxicity analysis revealed an excellent intrinsic quality of 1. The antinociceptive effect of compound 1 was determined using the formalin test. The ipsilateral local peripheral (10-300 μg/paw) and intrathecal (100 μg/rat) administration of 1 produced a reduction in formalin-induced nociception. The in vivo results indicated that 1 may be effective in treating inflammatory pain.
Collapse
Affiliation(s)
- Gabriel Navarrete-Vázquez
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico, Mexico.
| | - Amaya Austrich-Olivares
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico, Mexico
| | - Beatriz Godínez-Chaparro
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, México D.F., 04960, Mexico, Mexico
| | - Sergio Hidalgo-Figueroa
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico, Mexico
| | - Samuel Estrada-Soto
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos 62209, Mexico, Mexico
| | - Emanuel Hernández-Núñez
- Cátedra CONACyT, Departamento de Recursos del Mar, Centro de Investigación y de Estudios Avanzados del IPN, Unidad Mérida, 97310 Yucatán, Mexico, Mexico
| | - Héctor Torres-Gómez
- Institute for Chemistry and Chemical Biology, Zürich University of Applied Sciences, 8820 Wädenswil, Switzerland, Switzerland; Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms-Universität Münster, D-48149 Münster, Germany, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms-Universität Münster, D-48149 Münster, Germany, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der Westfälischen Wilhelms-Universität Münster, D-48149 Münster, Germany, Germany
| |
Collapse
|
241
|
Meanwell NA. Improving Drug Design: An Update on Recent Applications of Efficiency Metrics, Strategies for Replacing Problematic Elements, and Compounds in Nontraditional Drug Space. Chem Res Toxicol 2016; 29:564-616. [DOI: 10.1021/acs.chemrestox.6b00043] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nicholas A. Meanwell
- Department of Discovery Chemistry, Bristol-Myers Squibb Research & Development, Wallingford, Connecticut 06492, United States
| |
Collapse
|
242
|
Loughlin WA, Jenkins ID, Karis ND, Schweiker SS, Healy PC. 2-Oxo-1,2-dihydropyridinyl-3-yl amide-based GPa inhibitors: Design, synthesis and structure-activity relationship study. Eur J Med Chem 2016; 111:1-14. [DOI: 10.1016/j.ejmech.2016.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 01/18/2016] [Accepted: 01/18/2016] [Indexed: 02/08/2023]
|
243
|
Affiliation(s)
- Raphael M. Franzini
- Department
of Medicinal Chemistry,
College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, Utah 84112, United States
| | - Cassie Randolph
- Department
of Medicinal Chemistry,
College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, Utah 84112, United States
| |
Collapse
|
244
|
Huang S, O’Donnell KP, Keen JM, Rickard MA, McGinity JW, Williams RO. A New Extrudable Form of Hypromellose: AFFINISOL™ HPMC HME. AAPS PharmSciTech 2016; 17:106-19. [PMID: 26335416 DOI: 10.1208/s12249-015-0395-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/08/2015] [Indexed: 11/30/2022] Open
Abstract
Hypromellose is a hydrophilic polymer widely used in immediate- and modified-release oral pharmaceutical dosage forms. However, currently available grades of hypromellose are difficult, if not impossible, to process by hot melt extrusion (HME) because of their high glass transition temperature, high melt viscosity, and low degradation temperature. To overcome these challenges, a modified grade of hypromellose, AFFINISOL™ HPMC HME, was recently introduced. It has a significantly lower glass transition temperature and melt viscosity as compared to other available grades of hypromellose. The objective of this paper is to assess the extrudability and performance of AFFINISOL™ HPMC HME (100LV and 4M) as compared to other widely used polymers in HME, including HPMC 2910 100cP (the currently available hypromellose), Soluplus®, Kollidon® VA 64, and EUDRAGIT® E PO. Formulations containing polymer and carbamazepine (CBZ) were extruded on a co-rotating 16-mm twin-screw extruder, and the effect of temperature, screw speed, and feed rate was investigated. The performance of the solid dispersions was evaluated based on Flory-Huggins modeling and characterized by differential scanning calorimetry (DSC), X-ray powder diffraction (XRD), Raman spectroscopy, Fourier-transform infrared (FTIR) spectroscopy, and dissolution. All formulations extruded well except for HPMC 2910 100cP, which resulted in over-torqueing the extruder (machine overloading because the motor cannot provide efficient energy to rotate the shaft). Among the HME extrudates, only the EUDRAGIT® E PO formulation was crystalline as confirmed by DSC, XRD, and Raman, which agreed with predictions from Flory-Huggins modeling. Dissolution testing was conducted under both sink and non-sink conditions. Sink dissolution testing in neutral media revealed that amorphous CBZ in the HME extrudates completely dissolved within 15 min, which was much more rapid than the time for complete dissolution of bulk CBZ (60 min) and EUDRAGIT® E PO solid dispersion (more than 6 h). Non-sink dissolution in acidic media testing revealed that only CBZ contained in the AFFINISOL™ HPMC HME, and EUDRAGIT® E PO solid dispersions rapidly supersaturated after 15 min, reaching a twofold drug concentration compared to the CBZ equilibrium solubility. In summary, AFFINISOL™ HPMC HME 100LV and AFFINISOL™ HPMC HME 4M are useful in the pharmaceutical HME process to increase wetting and dissolution properties of poorly water-soluble drugs like CBZ.
Collapse
|
245
|
Li S, Tian Y, Jones DS, Andrews GP. Optimising Drug Solubilisation in Amorphous Polymer Dispersions: Rational Selection of Hot-melt Extrusion Processing Parameters. AAPS PharmSciTech 2016; 17:200-13. [PMID: 26729536 DOI: 10.1208/s12249-015-0450-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/21/2015] [Indexed: 11/30/2022] Open
Abstract
The aim of this article was to construct a T-ϕ phase diagram for a model drug (FD) and amorphous polymer (Eudragit® EPO) and to use this information to understand the impact of how temperature-composition coordinates influenced the final properties of the extrudate. Defining process boundaries and understanding drug solubility in polymeric carriers is of utmost importance and will help in the successful manufacture of new delivery platforms for BCS class II drugs. Physically mixed felodipine (FD)-Eudragit(®) EPO (EPO) binary mixtures with pre-determined weight fractions were analysed using DSC to measure the endset of melting and glass transition temperature. Extrudates of 10 wt% FD-EPO were processed using temperatures (110°C, 126°C, 140°C and 150°C) selected from the temperature-composition (T-ϕ) phase diagrams and processing screw speed of 20, 100 and 200rpm. Extrudates were characterised using powder X-ray diffraction (PXRD), optical, polarised light and Raman microscopy. To ensure formation of a binary amorphous drug dispersion (ADD) at a specific composition, HME processing temperatures should at least be equal to, or exceed, the corresponding temperature value on the liquid-solid curve in a F-H T-ϕ phase diagram. If extruded between the spinodal and liquid-solid curve, the lack of thermodynamic forces to attain complete drug amorphisation may be compensated for through the use of an increased screw speed. Constructing F-H T-ϕ phase diagrams are valuable not only in the understanding drug-polymer miscibility behaviour but also in rationalising the selection of important processing parameters for HME to ensure miscibility of drug and polymer.
Collapse
|
246
|
Egieyeh SA, Syce J, Malan SF, Christoffels A. Prioritization of anti-malarial hits from nature: chemo-informatic profiling of natural products with in vitro antiplasmodial activities and currently registered anti-malarial drugs. Malar J 2016; 15:50. [PMID: 26823078 PMCID: PMC4731946 DOI: 10.1186/s12936-016-1087-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 01/09/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND A large number of natural products have shown in vitro antiplasmodial activities. Early identification and prioritization of these natural products with potential for novel mechanism of action, desirable pharmacokinetics and likelihood for development into drugs is advantageous. Chemo-informatic profiling of these natural products were conducted and compared to currently registered anti-malarial drugs (CRAD). METHODS Natural products with in vitro antiplasmodial activities (NAA) were compiled from various sources. These natural products were sub-divided into four groups based on inhibitory concentration (IC50). Key molecular descriptors and physicochemical properties were computed for these compounds and analysis of variance used to assess statistical significance amongst the sets of compounds. Molecular similarity analysis, estimation of drug-likeness, in silico pharmacokinetic profiling, and exploration of structure-activity landscape were also carried out on these sets of compounds. RESULTS A total of 1040 natural products were selected and a total of 13 molecular descriptors were analysed. Significant differences were observed among the sub-groups of NAA and CRAD for at least 11 of the molecular descriptors, including number of hydrogen bond donors and acceptors, molecular weight, polar and hydrophobic surface areas, chiral centres, oxygen and nitrogen atoms, and shape index. The remaining molecular descriptors, including clogP, number of rotatable bonds and number of aromatic rings, did not show any significant difference when comparing the two compound sets. Molecular similarity and chemical space analysis identified natural products that were structurally diverse from CRAD. Prediction of the pharmacokinetic properties and drug-likeness of these natural products identified over 50% with desirable drug-like properties. Nearly 70% of all natural products were identified as potentially promiscuous compounds. Structure-activity landscape analysis highlighted compound pairs that form 'activity cliffs'. In all, prioritization strategies for the NAA were proposed. CONCLUSIONS Chemo-informatic profiling of NAA and CRAD have produced a wealth of information that may guide decisions and facilitate anti-malarial drug development from natural products. Articulation of the information provided within an interactive data-mining environment led to a prioritized list of NAA.
Collapse
Affiliation(s)
- Samuel Ayodele Egieyeh
- South African Medial Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, Cape Town, South Africa. .,School of Pharmacy, University of the Western Cape, Bellville, Cape Town, South Africa.
| | - James Syce
- School of Pharmacy, University of the Western Cape, Bellville, Cape Town, South Africa.
| | - Sarel F Malan
- School of Pharmacy, University of the Western Cape, Bellville, Cape Town, South Africa.
| | - Alan Christoffels
- South African Medial Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, Cape Town, South Africa.
| |
Collapse
|
247
|
An Intriguing Correlation Based on the Superimposition of Residue Pairs with Inhibitors that Target Protein-Protein Interfaces. Sci Rep 2016; 6:18543. [PMID: 26730437 PMCID: PMC4698585 DOI: 10.1038/srep18543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/19/2015] [Indexed: 11/26/2022] Open
Abstract
Druggable sites on protein-protein interfaces are difficult to predict. To survey inhibitor-binding sites onto which residues are superimposed at protein-protein interfaces, we analyzed publicly available information for 39 inhibitors that target the protein-protein interfaces of 8 drug targets. By focusing on the differences between residues that were superimposed with inhibitors and non-superimposed residues, we observed clear differences in the distances and changes in the solvent-accessible surface areas (∆SASA). Based on the observation that two or more residues were superimposed onto inhibitors in 37 (95%) of 39 protein-inhibitor complexes, we focused on the two-residue relationships. Application of a cross-validation procedure confirmed a linear negative correlation between the absolute value of the dihedral angle and the sum of the ∆SASAs of the residues. Finally, we applied the regression equation of this correlation to four inhibitors that bind to new sites not bound by the 39 inhibitors as well as additional inhibitors of different targets. Our results shed light on the two-residue correlation between the absolute value of the dihedral angle and the sum of the ∆SASA, which may be a useful relationship for identifying the key two-residues as potential targets of protein-protein interfaces.
Collapse
|
248
|
Colomer I, Empson CJ, Craven P, Owen Z, Doveston RG, Churcher I, Marsden SP, Nelson A. A divergent synthetic approach to diverse molecular scaffolds: assessment of lead-likeness using LLAMA, an open-access computational tool. Chem Commun (Camb) 2016; 52:7209-12. [DOI: 10.1039/c6cc03244c] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
LLAMA was used to assess the lead-likeness of scaffolds prepared via complementary cyclisations of hex-2-ene-1,6-diamine derivatives.
Collapse
Affiliation(s)
| | - Christopher J. Empson
- School of Chemistry
- University of Leeds
- Leeds
- UK
- Astbury Centre for Structural Molecular Biology
| | - Philip Craven
- School of Chemistry
- University of Leeds
- Leeds
- UK
- Astbury Centre for Structural Molecular Biology
| | | | - Richard G. Doveston
- School of Chemistry
- University of Leeds
- Leeds
- UK
- Astbury Centre for Structural Molecular Biology
| | - Ian Churcher
- GlaxoSmithKline Medicines Research Centre
- Stevenage
- UK
| | | | - Adam Nelson
- School of Chemistry
- University of Leeds
- Leeds
- UK
- Astbury Centre for Structural Molecular Biology
| |
Collapse
|
249
|
Bajusz D, Ferenczy GG, Keserű GM. Discovery of Subtype Selective Janus Kinase (JAK) Inhibitors by Structure-Based Virtual Screening. J Chem Inf Model 2015; 56:234-47. [DOI: 10.1021/acs.jcim.5b00634] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Dávid Bajusz
- Medicinal Chemistry Research
Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary
| | - György G. Ferenczy
- Medicinal Chemistry Research
Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary
| | - György M. Keserű
- Medicinal Chemistry Research
Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., Budapest 1117, Hungary
| |
Collapse
|
250
|
Noor Z, Afzal N, Rashid S. Exploration of Novel Inhibitors for Class I Histone Deacetylase Isoforms by QSAR Modeling and Molecular Dynamics Simulation Assays. PLoS One 2015; 10:e0139588. [PMID: 26431201 PMCID: PMC4592208 DOI: 10.1371/journal.pone.0139588] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/15/2015] [Indexed: 12/20/2022] Open
Abstract
Histone deacetylases (HDAC) are metal-dependent enzymes and considered as important targets for cell functioning. Particularly, higher expression of class I HDACs is common in the onset of multiple malignancies which results in deregulation of many target genes involved in cell growth, differentiation and survival. Although substantial attempts have been made to control the irregular functioning of HDACs by employing various inhibitors with high sensitivity towards transformed cells, limited success has been achieved in epigenetic cancer therapy. Here in this study, we used ligand-based pharmacophore and 2-dimensional quantitative structure activity relationship (QSAR) modeling approaches for targeting class I HDAC isoforms. Pharmacophore models were generated by taking into account the known IC50 values and experimental energy scores with extensive validations. The QSAR model having an external R2 value of 0.93 was employed for virtual screening of compound libraries. 10 potential lead compounds (C1-C10) were short-listed having strong binding affinities for HDACs, out of which 2 compounds (C8 and C9) were able to interact with all members of class I HDACs. The potential binding modes of HDAC2 and HDAC8 to C8 were explored through molecular dynamics simulations. Overall, bioactivity and ligand efficiency (binding energy/non-hydrogen atoms) profiles suggested that proposed hits may be more effective inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Zainab Noor
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| | - Noreen Afzal
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| | - Sajid Rashid
- National Center for Bioinformatics, Quaid I Azam University, Islamabad, Pakistan
| |
Collapse
|