1
|
Simeon S, Montanari D, Gleeson MP. Investigation of Factors Affecting the Performance of
in silico
Volume Distribution QSAR Models for Human, Rat, Mouse, Dog & Monkey. Mol Inform 2019; 38:e1900059. [DOI: 10.1002/minf.201900059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/03/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Saw Simeon
- Interdisciplinary Graduate Program in Bioscience, Faculty of ScienceKasetsart University Bangkok 10900 Thailand
- Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced StudiesKasetsart University Bangkok 10900 Thailand
| | - Dino Montanari
- DMPK and Bioanalysis, Aptuit Via Alessandro Fleming, 4 37135 Verona VR Italy
| | - Matthew Paul Gleeson
- Department of Chemistry, Faculty of ScienceKasetsart University Bangkok 10900 Thailand
- Department of Biomedical Engineering, Faculty of EngineeringKing Mongkut's Institute of Technology Ladkrabang Bangkok 10520 Thailand
| |
Collapse
|
2
|
Toma C, Gadaleta D, Roncaglioni A, Toropov A, Toropova A, Marzo M, Benfenati E. QSAR Development for Plasma Protein Binding: Influence of the Ionization State. Pharm Res 2018; 36:28. [PMID: 30591975 PMCID: PMC6308215 DOI: 10.1007/s11095-018-2561-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 12/17/2018] [Indexed: 01/05/2023]
Abstract
Purpose This study explored several strategies to improve the performance of literature QSAR models for plasma protein binding (PPB), such as a suitable endpoint transformation, a correct representation of chemicals, more consistency in the dataset, and a reliable definition of the applicability domain. Methods We retrieved human fraction unbound (Fu) data for 670 compounds from the literature and carefully checked them for consistency. Descriptors were calculated taking account of the ionization state of molecules at physiological pH (7.4), in order to better estimate the affinity of molecules to blood proteins. We used different algorithms and chemical descriptors to explore the most suitable strategy for modeling the endpoint. SMILES (simplified molecular input line entry system)-based string descriptors were also tested with the CORAL software (CORelation And Logic). We did an outlier analysis to establish the models to use (or not to use) in case of well recognized families. Results Internal validation of the selected models returned Q2 values close to 0.60. External validation also gave r2 values always greater than 0.60. The CORAL descriptor based model for √fu was the best, with r2 0.74 in external validation. Conclusions Performance in prediction confirmed the robustness of all the derived models and their suitability for real-life purposes, i.e. screening chemicals for their ADMET profiling. Optimization of descriptors can be useful in order to obtain the correct results with a ionized molecule. Electronic supplementary material The online version of this article (10.1007/s11095-018-2561-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cosimo Toma
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milano, Italy.
| | - Domenico Gadaleta
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milano, Italy
| | - Alessandra Roncaglioni
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milano, Italy
| | - Andrey Toropov
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milano, Italy
| | - Alla Toropova
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milano, Italy
| | - Marco Marzo
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milano, Italy
| | - Emilio Benfenati
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via la Masa 19, 20156, Milano, Italy
| |
Collapse
|
3
|
Cheminformatics in the Service of GPCR Drug Discovery. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2018; 1705:395-411. [PMID: 29188575 DOI: 10.1007/978-1-4939-7465-8_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cheminformatics is a broad discipline covering a wide range of computational approaches, including the characterization of molecular similarity, pattern recognition, and predictive modeling. The unifying theme that these apparently disparate methods have in common is the aim of extracting useable information from the increasing amounts of data that are associated with contemporary drug discovery projects. Both proprietary and publically available data can be exploited to help inform and improve the process of developing novel therapeutic molecules targeting the GPCR family of proteins.
Collapse
|
4
|
Automatically updating predictive modeling workflows support decision-making in drug design. Future Med Chem 2016; 8:1779-96. [DOI: 10.4155/fmc-2016-0070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Using predictive models for early decision-making in drug discovery has become standard practice. We suggest that model building needs to be automated with minimum input and low technical maintenance requirements. Models perform best when tailored to answering specific compound optimization related questions. If qualitative answers are required, 2-bin classification models are preferred. Integrating predictive modeling results with structural information stimulates better decision making. For in silico models supporting rapid structure–activity relationship cycles the performance deteriorates within weeks. Frequent automated updates of predictive models ensure best predictions. Consensus between multiple modeling approaches increases the prediction confidence. Combining qualified and nonqualified data optimally uses all available information. Dose predictions provide a holistic alternative to multiple individual property predictions for reaching complex decisions.
Collapse
|
5
|
Gedeck P, Lu Y, Skolnik S, Rodde S, Dollinger G, Jia W, Berellini G, Vianello R, Faller B, Lombardo F. Benefit of Retraining pKa Models Studied Using Internally Measured Data. J Chem Inf Model 2015; 55:1449-59. [DOI: 10.1021/acs.jcim.5b00172] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Peter Gedeck
- Novartis Institute for Tropical Diseases Pte. Ltd., 10 Biopolis Road, #05-01 Chromos, Singapore 138670, Singapore
| | - Yipin Lu
- Novartis Institute for Biomedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Suzanne Skolnik
- Novartis Institute for Biomedical Research, 250 Massachusetts Ave, Cambridge, Massachusetts 02139, United States
| | - Stephane Rodde
- Novartis Institute for Biomedical Research, Postfach, CH-4002 Basel, Switzerland
| | - Gavin Dollinger
- Novartis Institute for Biomedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Weiping Jia
- Novartis Institute for Biomedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Giuliano Berellini
- Novartis Institute for Biomedical Research, 250 Massachusetts Ave, Cambridge, Massachusetts 02139, United States
| | - Riccardo Vianello
- Novartis Institute for Biomedical Research, Postfach, CH-4002 Basel, Switzerland
| | - Bernard Faller
- Novartis Institute for Biomedical Research, Postfach, CH-4002 Basel, Switzerland
| | - Franco Lombardo
- Novartis Institute for Biomedical Research, 250 Massachusetts Ave, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
6
|
Lambrinidis G, Vallianatou T, Tsantili-Kakoulidou A. In vitro, in silico and integrated strategies for the estimation of plasma protein binding. A review. Adv Drug Deliv Rev 2015; 86:27-45. [PMID: 25819487 DOI: 10.1016/j.addr.2015.03.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 02/11/2015] [Accepted: 03/20/2015] [Indexed: 12/28/2022]
Abstract
Plasma protein binding (PPB) strongly affects drug distribution and pharmacokinetic behavior with consequences in overall pharmacological action. Extended plasma protein binding may be associated with drug safety issues and several adverse effects, like low clearance, low brain penetration, drug-drug interactions, loss of efficacy, while influencing the fate of enantiomers and diastereoisomers by stereoselective binding within the body. Therefore in holistic drug design approaches, where ADME(T) properties are considered in parallel with target affinity, considerable efforts are focused in early estimation of PPB mainly in regard to human serum albumin (HSA), which is the most abundant and most important plasma protein. The second critical serum protein α1-acid glycoprotein (AGP), although often underscored, plays also an important and complicated role in clinical therapy and thus the last years it has been studied thoroughly too. In the present review, after an overview of the principles of HSA and AGP binding as well as the structure topology of the proteins, the current trends and perspectives in the field of PPB predictions are presented and discussed considering both HSA and AGP binding. Since however for the latter protein systematic studies have started only the last years, the review focuses mainly to HSA. One part of the review highlights the challenge to develop rapid techniques for HSA and AGP binding simulation and their performance in assessment of PPB. The second part focuses on in silico approaches to predict HSA and AGP binding, analyzing and evaluating structure-based and ligand-based methods, as well as combination of both methods in the aim to exploit the different information and overcome the limitations of each individual approach. Ligand-based methods use the Quantitative Structure-Activity Relationships (QSAR) methodology to establish quantitate models for the prediction of binding constants from molecular descriptors, while they provide only indirect information on binding mechanism. Efforts for the establishment of global models, automated workflows and web-based platforms for PPB predictions are presented and discussed. Structure-based methods relying on the crystal structures of drug-protein complexes provide detailed information on the underlying mechanism but are usually restricted to specific compounds. They are useful to identify the specific binding site while they may be important in investigating drug-drug interactions, related to PPB. Moreover, chemometrics or structure-based modeling may be supported by experimental data a promising integrated alternative strategy for ADME(T) properties optimization. In the case of PPB the use of molecular modeling combined with bioanalytical techniques is frequently used for the investigation of AGP binding.
Collapse
|
7
|
Varadharajan S, Winiwarter S, Carlsson L, Engkvist O, Anantha A, Kogej T, Fridén M, Stålring J, Chen H. Exploring In Silico Prediction of the Unbound Brain-to-Plasma Drug Concentration Ratio: Model Validation, Renewal, and Interpretation. J Pharm Sci 2015; 104:1197-206. [DOI: 10.1002/jps.24301] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/14/2014] [Accepted: 11/18/2014] [Indexed: 01/13/2023]
|
8
|
Leahy DE, Sykora V. Automation of decision making in drug design. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 10:e437-41. [PMID: 24179997 DOI: 10.1016/j.ddtec.2013.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
9
|
Vallianatou T, Lambrinidis G, Tsantili-Kakoulidou A. In silicoprediction of human serum albumin binding for drug leads. Expert Opin Drug Discov 2013; 8:583-95. [DOI: 10.1517/17460441.2013.777424] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
10
|
Gleeson MP, Montanari D. Strategies for the generation, validation and application of in silico ADMET models in lead generation and optimization. Expert Opin Drug Metab Toxicol 2012; 8:1435-46. [PMID: 22849616 DOI: 10.1517/17425255.2012.711317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The most desirable chemical starting point in drug discovery is a hit or lead with a good overall profile, and where there may be issues; a clear SAR strategy should be identifiable to minimize the issue. Filtering based on drug-likeness concepts are a first step, but more accurate theoretical methods are needed to i) estimate the biological profile of molecule in question and ii) based on the underlying structure-activity relationships used by the model, estimate whether it is likely that the molecule in question can be altered to remove these liabilities. AREAS COVERED In this paper, the authors discuss the generation of ADMET models and their practical use in decision making. They discuss the issues surrounding data collation, experimental errors, the model assessment and validation steps, as well as the different types of descriptors and statistical models that can be used. This is followed by a discussion on how the model accuracy will dictate when and where it can be used in the drug discovery process. The authors also discuss how models can be developed to more effectively enable multiple parameter optimization. EXPERT OPINION Models can be applied in lead generation and lead optimization steps to i) rank order a collection of hits, ii) prioritize the experimental assays needed for different hit series, iii) assess the likelihood of resolving a problem that might be present in a particular series in lead optimization and iv) screen a virtual library based on a hit or lead series to assess the impact of diverse structural changes on the predicted properties.
Collapse
Affiliation(s)
- Matthew Paul Gleeson
- Kasetsart University, Faculty of Science, Department of Chemistry, 50 Phaholyothin Rd, Chatuchak, Bangkok 10900, Thailand.
| | | |
Collapse
|
11
|
Yang Y, Engkvist O, Llinàs A, Chen H. Beyond Size, Ionization State, and Lipophilicity: Influence of Molecular Topology on Absorption, Distribution, Metabolism, Excretion, and Toxicity for Druglike Compounds. J Med Chem 2012; 55:3667-77. [DOI: 10.1021/jm201548z] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yidong Yang
- Discovery
Sciences, Computational Sciences, Computational Chemistry, and ‡R&I iMED, In Vitro & In Vivo ADME, AstraZeneca R&D Mölndal,
SE-431 83 Mölndal, Sweden
| | - Ola Engkvist
- Discovery
Sciences, Computational Sciences, Computational Chemistry, and ‡R&I iMED, In Vitro & In Vivo ADME, AstraZeneca R&D Mölndal,
SE-431 83 Mölndal, Sweden
| | - Antonio Llinàs
- Discovery
Sciences, Computational Sciences, Computational Chemistry, and ‡R&I iMED, In Vitro & In Vivo ADME, AstraZeneca R&D Mölndal,
SE-431 83 Mölndal, Sweden
| | - Hongming Chen
- Discovery
Sciences, Computational Sciences, Computational Chemistry, and ‡R&I iMED, In Vitro & In Vivo ADME, AstraZeneca R&D Mölndal,
SE-431 83 Mölndal, Sweden
| |
Collapse
|
12
|
Sherer EC, Verras A, Madeira M, Hagmann WK, Sheridan RP, Roberts D, Bleasby K, Cornell WD. QSAR Prediction of Passive Permeability in the LLC-PK1 Cell Line: Trends in Molecular Properties and Cross-Prediction of Caco-2 Permeabilities. Mol Inform 2012; 31:231-45. [DOI: 10.1002/minf.201100157] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/06/2012] [Indexed: 01/16/2023]
|
13
|
Wood DJ, Buttar D, Cumming JG, Davis AM, Norinder U, Rodgers SL. Automated QSAR with a Hierarchy of Global and Local Models. Mol Inform 2011; 30:960-72. [DOI: 10.1002/minf.201100107] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 10/13/2011] [Indexed: 11/06/2022]
|
14
|
Rodgers SL, Davis AM, Tomkinson NP, van de Waterbeemd H. Predictivity of Simulated ADME AutoQSAR Models over Time. Mol Inform 2011; 30:256-66. [DOI: 10.1002/minf.201000160] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Accepted: 01/24/2011] [Indexed: 11/08/2022]
|
15
|
Gedeck P, Kramer C, Ertl P. Computational analysis of structure-activity relationships. PROGRESS IN MEDICINAL CHEMISTRY 2010; 49:113-60. [PMID: 20855040 DOI: 10.1016/s0079-6468(10)49004-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Peter Gedeck
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Forum 1, Novartis Campus, CH-4056 Basel, Switzerland
| | | | | |
Collapse
|
16
|
Madden JC. In Silico Approaches for Predicting Adme Properties. CHALLENGES AND ADVANCES IN COMPUTATIONAL CHEMISTRY AND PHYSICS 2010. [DOI: 10.1007/978-1-4020-9783-6_10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
17
|
The importance of the domain of applicability in QSAR modeling. J Mol Graph Model 2008; 26:1315-26. [DOI: 10.1016/j.jmgm.2008.01.002] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Revised: 01/11/2008] [Accepted: 01/11/2008] [Indexed: 11/19/2022]
|
18
|
Generation of in-silico cytochrome P450 1A2, 2C9, 2C19, 2D6, and 3A4 inhibition QSAR models. J Comput Aided Mol Des 2007; 21:559-73. [DOI: 10.1007/s10822-007-9139-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Accepted: 10/04/2007] [Indexed: 01/22/2023]
|
19
|
|
20
|
Rodgers SL, Davis AM, Tomkinson NP, van de Waterbeemd H. QSAR Modeling Using Automatically Updating Correction Libraries: Application to a Human Plasma Protein Binding Model. J Chem Inf Model 2007; 47:2401-7. [PMID: 17887744 DOI: 10.1021/ci700197x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It is assumed that compounds occupying the same region of model space will be subject to similar errors in prediction, and hence, where these errors are known, they can be applied to predictions. Thus, any available measured data can be used to refine predictions of query compounds. This study describes the application of a correction library to a human plasma protein binding model. Compounds that have been measured since the model was built are entered into the library to improve predictions of current compounds. Time-series simulations were conducted to measure the time dependence of the correction library. This study demonstrates significant improvements in predictions where a library is applied, compared with both a static model and an updating model that includes recently measured data.
Collapse
Affiliation(s)
- Sarah L Rodgers
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, Leicestershire, United Kingdom.
| | | | | | | |
Collapse
|