51
|
Zhang J, Hsieh JH, Zhu H. Profiling animal toxicants by automatically mining public bioassay data: a big data approach for computational toxicology. PLoS One 2014; 9:e99863. [PMID: 24950175 PMCID: PMC4064997 DOI: 10.1371/journal.pone.0099863] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 05/16/2014] [Indexed: 01/31/2023] Open
Abstract
In vitro bioassays have been developed and are currently being evaluated as potential alternatives to traditional animal toxicity models. Already, the progress of high throughput screening techniques has resulted in an enormous amount of publicly available bioassay data having been generated for a large collection of compounds. When a compound is tested using a collection of various bioassays, all the testing results can be considered as providing a unique bio-profile for this compound, which records the responses induced when the compound interacts with different cellular systems or biological targets. Profiling compounds of environmental or pharmaceutical interest using useful toxicity bioassay data is a promising method to study complex animal toxicity. In this study, we developed an automatic virtual profiling tool to evaluate potential animal toxicants. First, we automatically acquired all PubChem bioassay data for a set of 4,841 compounds with publicly available rat acute toxicity results. Next, we developed a scoring system to evaluate the relevance between these extracted bioassays and animal acute toxicity. Finally, the top ranked bioassays were selected to profile the compounds of interest. The resulting response profiles proved to be useful to prioritize untested compounds for their animal toxicity potentials and form a potential in vitro toxicity testing panel. The protocol developed in this study could be combined with structure-activity approaches and used to explore additional publicly available bioassay datasets for modeling a broader range of animal toxicities.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Chemistry, Rutgers University, Camden, New Jersey, United States of America
- The Rutgers Center for Computational and Integrative Biology, Camden, New Jersey, United States of America
| | - Jui-Hua Hsieh
- Biomolecular Screening Branch, Division of National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Hao Zhu
- Department of Chemistry, Rutgers University, Camden, New Jersey, United States of America
- The Rutgers Center for Computational and Integrative Biology, Camden, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
52
|
Karthick V, Ramanathan K, Shanthi V, Rajasekaran R. Identification of potential inhibitors of H5N1 influenza A virus neuraminidase by ligand-based virtual screening approach. Cell Biochem Biophys 2014; 66:657-69. [PMID: 23306969 DOI: 10.1007/s12013-012-9510-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The neuraminidase (NA) of the influenza virus is the target of antiviral drug, oseltamivir. Recently, cases were reported that influenza virus becoming resistant to oseltamivir, necessitating the development of new long-acting antiviral compounds. In this report, a novel class of lead molecule with potential NA inhibitory activity was identified using a combination of virtual screening (VS), molecular docking, and molecular dynamic approach. The PubChem database was used to perform the VS analysis by employing oseltamivir as query. Subsequently, the data reduction was carried out by employing molecular docking study. Furthermore, the screened lead molecules were analyzed with respect to the Lipinski rule of five, drug-likeness, toxicity profiles, and other physico-chemical properties of drugs by suitable software program. Final screening was carried out by normal mode analysis and molecular dynamic simulation approach. The result indicates that CID 25145634, deuterium-enriched oseltamivir, become a promising lead compound and be effective in treating oseltamivir sensitive as well as resistant influenza virus strains.
Collapse
Affiliation(s)
- V Karthick
- Bioinformatics Division, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | | | | | | |
Collapse
|
53
|
Xie XQ, Wang L, Liu H, Ouyang Q, Fang C, Su W. Chemogenomics knowledgebased polypharmacology analyses of drug abuse related G-protein coupled receptors and their ligands. Front Pharmacol 2014; 5:3. [PMID: 24567719 PMCID: PMC3915241 DOI: 10.3389/fphar.2014.00003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 01/06/2014] [Indexed: 12/15/2022] Open
Abstract
Drug abuse (DA) and addiction is a complex illness, broadly viewed as a neurobiological impairment with genetic and environmental factors that influence its development and manifestation. Abused substances can disrupt the activity of neurons by interacting with many proteins, particularly G-protein coupled receptors (GPCRs). A few medicines that target the central nervous system (CNS) can also modulate DA related proteins, such as GPCRs, which can act in conjunction with the controlled psychoactive substance(s) and increase side effects. To fully explore the molecular interaction networks that underlie DA and to effectively modulate the GPCRs in these networks with small molecules for DA treatment, we built a drug-abuse domain specific chemogenomics knowledgebase (DA-KB) to centralize the reported chemogenomics research information related to DA and CNS disorders in an effort to benefit researchers across a broad range of disciplines. We then focus on the analysis of GPCRs as many of them are closely related with DA. Their distribution in human tissues was also analyzed for the study of side effects caused by abused drugs. We further implement our computational algorithms/tools to explore DA targets, DA mechanisms and pathways involved in polydrug addiction and to explore polypharmacological effects of the GPCR ligands. Finally, the polypharmacology effects of GPCRs-targeted medicines for DA treatment were investigated and such effects can be exploited for the development of drugs with polypharmacophore for DA intervention. The chemogenomics database and the analysis tools will help us better understand the mechanism of drugs abuse and facilitate to design new medications for system pharmacotherapy of DA.
Collapse
Affiliation(s)
- Xiang-Qun Xie
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh Pittsburgh, PA, USA ; Center for Chemical Methodologies and Library Development (UPCMLD) and Department of Chemistry, University of Pittsburgh Pittsburgh, PA, USA ; Drug Discovery Institute, University of Pittsburgh Pittsburgh, PA, USA ; Departments of Computational and Systems Biology, University of Pittsburgh Pittsburgh, PA, USA
| | - Lirong Wang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh Pittsburgh, PA, USA ; Center for Chemical Methodologies and Library Development (UPCMLD) and Department of Chemistry, University of Pittsburgh Pittsburgh, PA, USA ; Drug Discovery Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Haibin Liu
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh Pittsburgh, PA, USA ; Guangzhou Quality R&D Center of Traditional Chinese Medicine, School of Life Sciences, Sun Yat-Sen University Guangzhou, China
| | - Qin Ouyang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh Pittsburgh, PA, USA ; Center for Chemical Methodologies and Library Development (UPCMLD) and Department of Chemistry, University of Pittsburgh Pittsburgh, PA, USA ; Drug Discovery Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Cheng Fang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh Pittsburgh, PA, USA ; Center for Chemical Methodologies and Library Development (UPCMLD) and Department of Chemistry, University of Pittsburgh Pittsburgh, PA, USA ; Drug Discovery Institute, University of Pittsburgh Pittsburgh, PA, USA
| | - Weiwei Su
- Guangzhou Quality R&D Center of Traditional Chinese Medicine, School of Life Sciences, Sun Yat-Sen University Guangzhou, China
| |
Collapse
|
54
|
Blucher AS, McWeeney SK. Challenges in secondary analysis of high throughput screening data. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2014:114-124. [PMID: 24297539 PMCID: PMC3976302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Repurposing an existing drug for an alternative use is not only a cost effective method of development, but also a faster process due to the drug's previous clinical testing and established pharmokinetic profiles. A potentially rich resource for computational drug repositioning approaches is publically available high throughput screening data, available in databases such as PubChem Bioassay and ChemBank. We examine statistical and computational considerations for secondary analysis of publicly available high throughput screening (HTS) data with respect to metadata, data quality, and completeness. We discuss developing methods and best practices that can help to ameliorate these issues.
Collapse
Affiliation(s)
- Aurora S Blucher
- Division of Bioinformatics and Computational Biology, Oregon Health & Science University, Portland, OR 97203, USA.
| | | |
Collapse
|
55
|
Fourches D. Cheminformatics: At the Crossroad of Eras. CHALLENGES AND ADVANCES IN COMPUTATIONAL CHEMISTRY AND PHYSICS 2014. [DOI: 10.1007/978-94-017-9257-8_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
56
|
Hamon V, Bourgeas R, Ducrot P, Theret I, Xuereb L, Basse MJ, Brunel JM, Combes S, Morelli X, Roche P. 2P2I HUNTER: a tool for filtering orthosteric protein-protein interaction modulators via a dedicated support vector machine. J R Soc Interface 2013; 11:20130860. [PMID: 24196694 DOI: 10.1098/rsif.2013.0860] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Over the last 10 years, protein-protein interactions (PPIs) have shown increasing potential as new therapeutic targets. As a consequence, PPIs are today the most screened target class in high-throughput screening (HTS). The development of broad chemical libraries dedicated to these particular targets is essential; however, the chemical space associated with this 'high-hanging fruit' is still under debate. Here, we analyse the properties of 40 non-redundant small molecules present in the 2P2I database (http://2p2idb.cnrs-mrs.fr/) to define a general profile of orthosteric inhibitors and propose an original protocol to filter general screening libraries using a support vector machine (SVM) with 11 standard Dragon molecular descriptors. The filtering protocol has been validated using external datasets from PubChem BioAssay and results from in-house screening campaigns. This external blind validation demonstrated the ability of the SVM model to reduce the size of the filtered chemical library by eliminating up to 96% of the compounds as well as enhancing the proportion of active compounds by up to a factor of 8. We believe that the resulting chemical space identified in this paper will provide the scientific community with a concrete support to search for PPI inhibitors during HTS campaigns.
Collapse
Affiliation(s)
- Véronique Hamon
- Laboratory of integrative Structural and Chemical Biology (iSCB), Centre de Recherche en Cancérologie de Marseille (CRCM); CNRS UMR 7258, INSERM U 1068, Institut Paoli-Calmettes; and Aix-Marseille Universités, , Marseille 13009, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Melas IN, Kretsos K, Alexopoulos LG. Leveraging systems biology approaches in clinical pharmacology. Biopharm Drug Dispos 2013; 34:477-88. [PMID: 23983165 PMCID: PMC4034589 DOI: 10.1002/bdd.1859] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 08/12/2013] [Indexed: 01/15/2023]
Abstract
Computational modeling has been adopted in all aspects of drug research and development, from the early phases of target identification and drug discovery to the late-stage clinical trials. The different questions addressed during each stage of drug R&D has led to the emergence of different modeling methodologies. In the research phase, systems biology couples experimental data with elaborate computational modeling techniques to capture lifecycle and effector cellular functions (e.g. metabolism, signaling, transcription regulation, protein synthesis and interaction) and integrates them in quantitative models. These models are subsequently used in various ways, i.e. to identify new targets, generate testable hypotheses, gain insights on the drug's mode of action (MOA), translate preclinical findings, and assess the potential of clinical drug efficacy and toxicity. In the development phase, pharmacokinetic/pharmacodynamic (PK/PD) modeling is the established way to determine safe and efficacious doses for testing at increasingly larger, and more pertinent to the target indication, cohorts of subjects. First, the relationship between drug input and its concentration in plasma is established. Second, the relationship between this concentration and desired or undesired PD responses is ascertained. Recognizing that the interface of systems biology with PK/PD will facilitate drug development, systems pharmacology came into existence, combining methods from PK/PD modeling and systems engineering explicitly to account for the implicated mechanisms of the target system in the study of drug–target interactions. Herein, a number of popular system biology methodologies are discussed, which could be leveraged within a systems pharmacology framework to address major issues in drug development.
Collapse
Affiliation(s)
- Ioannis N Melas
- National Technical University of Athens, Athens, Greece; Protatonce Ltd, Athens, Greece
| | | | | |
Collapse
|
58
|
Chen NG, Golovlev V. Structural Key Bit Occurrence Frequencies and Dependencies in PubChem and Their Effect on Similarity Searches. Mol Inform 2013; 32:355-61. [PMID: 27481592 DOI: 10.1002/minf.201300006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/05/2013] [Indexed: 11/06/2022]
Abstract
Little published literature exists on the 881 bit structural keys used by PubChem for categorizing and comparing the compounds present in its database. We characterized these structural keys by examining their frequencies of occurrence within the PubChem compound database. In addition, bit dependencies, defined as the universal presence of a bit given the presence of another, were determined. We show that the vast majority of bits are rarely set and that substantial numbers of dependencies exist. A comparison of similarity searches with five United States Food and Drug Administration approved drugs as reference compounds using the full structural keys versus a variant in which all dependent bits were removed was performed using the Tanimoto coefficient. These bit dependencies not only affect similarity scores, but also alter the compounds returned in similarity searching. Judicious selection of bits is needed to maintain sufficient ability to differentiate related compounds.
Collapse
Affiliation(s)
- Nelson G Chen
- Sci-Tec, Inc. 107 Canterbury Rd, Oak Ridge, TN 37830-7712, USA.
| | - Val Golovlev
- Sci-Tec, Inc. 107 Canterbury Rd, Oak Ridge, TN 37830-7712, USA
| |
Collapse
|
59
|
Wang L, Ma C, Wipf P, Liu H, Su W, Xie XQ. TargetHunter: an in silico target identification tool for predicting therapeutic potential of small organic molecules based on chemogenomic database. AAPS JOURNAL 2013; 15:395-406. [PMID: 23292636 DOI: 10.1208/s12248-012-9449-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 12/10/2012] [Indexed: 02/08/2023]
Abstract
Target identification of the known bioactive compounds and novel synthetic analogs is a very important research field in medicinal chemistry, biochemistry, and pharmacology. It is also a challenging and costly step towards chemical biology and phenotypic screening. In silico identification of potential biological targets for chemical compounds offers an alternative avenue for the exploration of ligand-target interactions and biochemical mechanisms, as well as for investigation of drug repurposing. Computational target fishing mines biologically annotated chemical databases and then maps compound structures into chemogenomical space in order to predict the biological targets. We summarize the recent advances and applications in computational target fishing, such as chemical similarity searching, data mining/machine learning, panel docking, and the bioactivity spectral analysis for target identification. We then described in detail a new web-based target prediction tool, TargetHunter (http://www.cbligand.org/TargetHunter). This web portal implements a novel in silico target prediction algorithm, the Targets Associated with its MOst SImilar Counterparts, by exploring the largest chemogenomical databases, ChEMBL. Prediction accuracy reached 91.1% from the top 3 guesses on a subset of high-potency compounds from the ChEMBL database, which outperformed a published algorithm, multiple-category models. TargetHunter also features an embedded geography tool, BioassayGeoMap, developed to allow the user easily to search for potential collaborators that can experimentally validate the predicted biological target(s) or off target(s). TargetHunter therefore provides a promising alternative to bridge the knowledge gap between biology and chemistry, and significantly boost the productivity of chemogenomics researchers for in silico drug design and discovery.
Collapse
Affiliation(s)
- Lirong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Computational Chemical Genomics Screening Center, Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
60
|
Kumar RB, Suresh MX. A computational perspective of molecular interactions through virtual screening, pharmacokinetic and dynamic prediction on ribosome toxin A chain and inhibitors of Ricinus communis. Pharmacognosy Res 2012; 4:2-10. [PMID: 22224054 PMCID: PMC3250034 DOI: 10.4103/0974-8490.91027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Revised: 09/04/2011] [Accepted: 12/22/2011] [Indexed: 11/28/2022] Open
Abstract
Background: Ricin is considered to be one of the most deadly toxins and gained its favor as a bioweapon that has a serious social and biological impact, due to its widespread nature and abundant availability. The hazardous effects of this toxin in human being are seen in almost all parts of the organ system. The severe consequences of the toxin necessitate the need for developing potential inhibitors that can effectively block its interaction with the host system. Materials and Methods: In order to identify potential inhibitors that can effectively block ricin, we employed various computational approaches. In this work, we computationally screened and analyzed 66 analogs and further tested their ADME/T profiles. From the kinetic and toxicity studies we selected six analogs that possessed appropriate pharmacokinetic and dynamic property. We have also performed a computational docking of these analogs with the target. Results: On the basis of the dock scores and hydrogen bond interactions we have identified analog 64 to be the best interacting molecule. Molecule 64 seems to have stable interaction with the residues Tyr80, Arg180, and Val81. The pharmacophore feature that describes the key functional features of a molecule was also studied and presented. Conclusion: The pharmacophore features of the drugs provided suggests the key functional groups that can aid in the design and synthesis of more potential inhibitors.
Collapse
Affiliation(s)
- R Barani Kumar
- Department of Bioinformatics, Sathyabama University, Chennai, Tamil Nadu, India
| | | |
Collapse
|
61
|
Mahasenan KV, Li C. Novel inhibitor discovery through virtual screening against multiple protein conformations generated via ligand-directed modeling: a maternal embryonic leucine zipper kinase example. J Chem Inf Model 2012; 52:1345-55. [PMID: 22540736 DOI: 10.1021/ci300040c] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Kinase targets have been demonstrated to undergo major conformational reorganization upon ligand binding. Such protein conformational plasticity remains a significant challenge in structure-based virtual screening methodology and may be approximated by screening against an ensemble of diverse protein conformations. Maternal embryonic leucine zipper kinase (MELK), a member of serine-threonine kinase family, has been recently found to be involved in the tumerogenic state of glioblastoma, breast, ovarian, and colon cancers. We therefore modeled several conformers of MELK utilizing the available chemogenomic and crystallographic data of homologous kinases. We carried out docking pose prediction and virtual screening enrichment studies with these conformers. The performances of the ensembles were evaluated by their ability to reproduce known inhibitor bioactive conformations and to efficiently recover known active compounds early in the virtual screen when seeded with decoy sets. A few of the individual MELK conformers performed satisfactorily in reproducing the native protein-ligand pharmacophoric interactions up to 50% of the cases. By selecting an ensemble of a few representative conformational states, most of the known inhibitor binding poses could be rationalized. For example, a four conformer ensemble is able to recover 95% of the studied actives, especially with imperfect scoring function(s). The virtual screening enrichment varied considerably among different MELK conformers. Enrichment appears to improve by selection of a proper protein conformation. For example, several holo and unliganded active conformations are better to accommodate diverse chemotypes than ATP-bound conformer. These results prove that using an ensemble of diverse conformations could give a better performance. Applying this approach, we were able to screen a commercially available library of half a million compounds against three conformers to discover three novel inhibitors of MELK, one from each template. Among the three compounds validated via experimental enzyme inhibition assays, one is relatively potent (15; K(d) = 0.37 μM), one moderately active (12; K(d) = 3.2 μM), and one weak but very selective (9; K(d) = 18 μM). These novel hits may be utilized to assist in the development of small molecule therapeutic agents useful in diseases caused by deregulated MELK, and perhaps more importantly, the approach demonstrates the advantages of choosing an appropriate ensemble of a few conformers in pursuing compound potency, selectivity, and novel chemotypes over using single target conformation for structure-based drug design in general.
Collapse
Affiliation(s)
- Kiran V Mahasenan
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | | |
Collapse
|
62
|
Christopher F, Thangam EB, Suresh MX. A Bioinformatics Search for Selective Histamine H4 Receptor Antagonists Through Structure-Based Virtual Screening Strategies. Chem Biol Drug Des 2012; 79:749-59. [DOI: 10.1111/j.1747-0285.2012.01336.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
63
|
Wang L, Ma C, Wipf P, Xie XQ. Linear and Nonlinear Support Vector Machine for the Classification of Human 5-HT1A Ligand Functionality. Mol Inform 2012; 31:85-95. [PMID: 27478180 DOI: 10.1002/minf.201100126] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 12/11/2011] [Indexed: 11/06/2022]
Abstract
Upon binding to a receptor, agonists and antagonists can induce distinct biological functions and thus lead to significantly different pharmacological responses. Thus, in silico prediction or in vitro characterization of ligand agonistic or antagonistic functionalities is an important step toward identifying specific pharmacological therapeutics. In this study, we investigated the molecular properties of agonists and antagonists of human 5-hydroxytryptamine receptor subtype 1A (5-HT1A ). Subsequently, intrinsic functions of these ligands (agonists/antagonists) were modeled by support vector machine (SVM), using five 2D molecular fingerprints and the 3D Topomer distance. Five kernel functions, including linear, polynomial, RBF, Tanimoto and a novel Topomer kernel based on Topomer 3D similarity were used to develop linear and nonlinear classifiers. These classifiers were validated through cross-validation, yielding a classification accuracy ranging from 80.4 % to 92.3 %. The performance of different kernels and fingerprints was analyzed and discussed. Linear and nonlinear models were further interpreted through the illustration of underlying classification mechanism. The computation protocol has been automated and demonstrated through our online service. This study expands the scope and applicability of similarity-based methods in cheminformatics, which are typically used for the identification of active molecules against a target protein. Our findings provide a good starting point for further systematic classifications of other GPCR ligands and for the data mining of large chemical libraries.
Collapse
Affiliation(s)
- Lirong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Center for Chemical Methodologies & Library Development (UP-CMLD), Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA tel.: +1-412-383-5276; fax: +1-412-383-7436
| | - Chao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, Center for Chemical Methodologies & Library Development (UP-CMLD), Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA tel.: +1-412-383-5276; fax: +1-412-383-7436.,Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Peter Wipf
- Department of Pharmaceutical Sciences, School of Pharmacy, Center for Chemical Methodologies & Library Development (UP-CMLD), Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA tel.: +1-412-383-5276; fax: +1-412-383-7436.,Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, School of Pharmacy, Center for Chemical Methodologies & Library Development (UP-CMLD), Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA tel.: +1-412-383-5276; fax: +1-412-383-7436. .,Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
64
|
Ma C, Wang L, Xie XQ. GPU accelerated chemical similarity calculation for compound library comparison. J Chem Inf Model 2011; 51:1521-7. [PMID: 21692447 DOI: 10.1021/ci1004948] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chemical similarity calculation plays an important role in compound library design, virtual screening, and "lead" optimization. In this manuscript, we present a novel GPU-accelerated algorithm for all-vs-all Tanimoto matrix calculation and nearest neighbor search. By taking advantage of multicore GPU architecture and CUDA parallel programming technology, the algorithm is up to 39 times superior to the existing commercial software that runs on CPUs. Because of the utilization of intrinsic GPU instructions, this approach is nearly 10 times faster than existing GPU-accelerated sparse vector algorithm, when Unity fingerprints are used for Tanimoto calculation. The GPU program that implements this new method takes about 20 min to complete the calculation of Tanimoto coefficients between 32 M PubChem compounds and 10K Active Probes compounds, i.e., 324G Tanimoto coefficients, on a 128-CUDA-core GPU.
Collapse
Affiliation(s)
- Chao Ma
- Department of Computational and Systems Biology, Joint Pitt/CMU Computational Biology Program, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | | | | |
Collapse
|
65
|
Ma C, Lazo JS, Xie XQ. Compound acquisition and prioritization algorithm for constructing structurally diverse compound libraries. ACS COMBINATORIAL SCIENCE 2011; 13:223-31. [PMID: 21480665 DOI: 10.1021/co100033m] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In the present study, we report a compound acquisition and prioritization algorithm established for rational chemical library purchasing or compound synthesis to increase the diversity of an existing compound collection. This method was established based on a chemistry-space calculation using BCUT (Burden CAS University of Texas) descriptors. To identify the acquisition of compounds from candidate collections into the existing collection, a derived distance-based selection rule was applied, and the results were well supported by pairwise similarity calculations and cell-partition statistics in chemistry space. The correlation between chemistry-space distance and Tanimoto similarity index was also studied to justify the compound acquisition strategy through weighted linear regression. As a rational approach for library design, the distance-based selection rule exhibits certain advantages in prioritizing compound selection to enhance the overall structural diversity of an existing in-house compound collection or virtual combinatorial library for in silico screening, diversity oriented synthesis, and high-throughput screening.
Collapse
Affiliation(s)
- Chao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, ‡Department of Computational Biology, §Drug Discovery Institute, ∥Department of Pharmacology and Chemical Biology, and ⊥Pittsburgh Center for Chemical Methodologies and Library Development, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - John S. Lazo
- Department of Pharmaceutical Sciences, School of Pharmacy, ‡Department of Computational Biology, §Drug Discovery Institute, ∥Department of Pharmacology and Chemical Biology, and ⊥Pittsburgh Center for Chemical Methodologies and Library Development, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, School of Pharmacy, ‡Department of Computational Biology, §Drug Discovery Institute, ∥Department of Pharmacology and Chemical Biology, and ⊥Pittsburgh Center for Chemical Methodologies and Library Development, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|