1
|
Parvez MK, Al-Dosari MS, Sinha GP. Machine learning-based predictive models for identifying high active compounds against HIV-1 integrase. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2022; 33:387-402. [PMID: 35410555 DOI: 10.1080/1062936x.2022.2057588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
HIV-integrase is an important drug target because it catalyzes chromosomal integration of proviral DNA towards establishing latent infection. Computer-aided drug design has immensely contributed to identifying and developing novel antiviral drugs. We have developed various machine learning-based predictive models for identifying high activity compounds against HIV-integrase. Multiclass models were built using support vector machine with reasonable accuracy on the test and evaluation sets. The developed models were evaluated by rigorous validation approaches and the best features were selected by Boruta method. As compared to the model developed from all descriptors set, a slight improvement was observed among the selected descriptors. Validated models were further used for virtual screening of potential compounds from ChemBridge library. Of the six high active compounds predicted from selected models, compounds 9103124, 6642917 and 9082952 showed the most reasonable binding-affinity and stable-interaction with HIV-integrase active-site residues Asp64, Glu152 and Asn155. This was in agreement with previous reports on the essentiality of these residues against a wide range of inhibitors. We therefore highlight the rigorosity of validated classification models for accurate prediction and ranking of high active lead drugs against HIV-integrase.
Collapse
Affiliation(s)
- M K Parvez
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - M S Al-Dosari
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - G P Sinha
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Ha CHX, Lee NK, Rahman T, Hwang SS, Yam WK, Chee XW. Repurposing FDA-approved drugs as HIV-1 integrase inhibitors: an in silico investigation. J Biomol Struct Dyn 2022; 41:2146-2159. [PMID: 35067186 DOI: 10.1080/07391102.2022.2028677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Human Immunodeficiency Virus (HIV) infection is a global pandemic that has claimed 33 million lives to-date. One of the most efficacious treatments for naïve or pretreated HIV patients is the HIV integrase strand transfer inhibitors (INSTIs). However, given that HIV treatment is life-long, the emergence of HIV strains resistant to INSTIs is an imminent challenge. In this work, we showed two best regression QSAR models that were constructed using a boosted Random Forest algorithm (r2 = 0.998, q210CV = 0.721, q2external_test = 0.754) and a boosted K* algorithm (r2 = 0.987, q210CV = 0.721, q2external_test = 0.758) to predict the pIC50 values of INSTIs. Subsequently, the regression QSAR models were deployed against the Drugbank database for drug repositioning. The top-ranked compounds were further evaluated for their target engagement activity using molecular docking studies and accelerated Molecular Dynamics simulation. Lastly, their potential as INSTIs were also evaluated from our literature search. Our study offers the first example of a large-scale regression QSAR modelling effort for discovering highly active INSTIs to combat HIV infection.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Christopher Heng Xuan Ha
- Faculty of Engineering, Computing and Science, Swinburne University of Technology, Sarawak, Malaysia
| | - Nung Kion Lee
- Faculty of Cognitive Sciences and Human Development, Universiti Malaysia Sarawak, Sarawak, Malaysia
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Siaw San Hwang
- Faculty of Engineering, Computing and Science, Swinburne University of Technology, Sarawak, Malaysia
| | - Wai Keat Yam
- Centre for Bioinformatics, School of Data Sciences, Perdana University, Kuala Lumpur, Malaysia
| | - Xavier Wezen Chee
- Faculty of Engineering, Computing and Science, Swinburne University of Technology, Sarawak, Malaysia
| |
Collapse
|
3
|
Dong Y, Zhao C, Wang X, Xie M, Zhong X, Song R, Yu A, Wei J, Yao J, Shan D, Lv F, She G. Lvsiyujins A–G, new sesquiterpenoids, from Curcuma phaeocaulis Valeton root tuber and their preliminary pharmacological property assessment based on ADME evaluation, molecular docking and in vitro experiments. NEW J CHEM 2022. [DOI: 10.1039/d2nj00101b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Seven new sesquiterpenoids were isolated from the root tuber of C. phaeocaulis. A combination of calculations and experiments was used in structural analysis and biological activity exploration.
Collapse
Affiliation(s)
- Ying Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Chongjun Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Xiuhuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Meng Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Xiangjian Zhong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Ruolan Song
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Axiang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Jing Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Jianling Yao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Dongjie Shan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Fang Lv
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| | - Gaimei She
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, P. R. China
| |
Collapse
|
4
|
Melo R, Lemos A, Preto AJ, Bueschbell B, Matos-Filipe P, Barreto C, Almeida JG, Silva RDM, Correia JDG, Moreira IS. An Overview of Antiretroviral Agents for Treating HIV Infection in Paediatric Population. Curr Med Chem 2020; 27:760-794. [PMID: 30182840 DOI: 10.2174/0929867325666180904123549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/11/2018] [Accepted: 07/11/2018] [Indexed: 12/19/2022]
Abstract
Paediatric Acquired ImmunoDeficiency Syndrome (AIDS) is a life-threatening and infectious disease in which the Human Immunodeficiency Virus (HIV) is mainly transmitted through Mother-To- Child Transmission (MTCT) during pregnancy, labour and delivery, or breastfeeding. This review provides an overview of the distinct therapeutic alternatives to abolish the systemic viral replication in paediatric HIV-1 infection. Numerous classes of antiretroviral agents have emerged as therapeutic tools for downregulation of different steps in the HIV replication process. These classes encompass Non- Nucleoside Analogue Reverse Transcriptase Inhibitors (NNRTIs), Nucleoside/Nucleotide Analogue Reverse Transcriptase Inhibitors (NRTIs/NtRTIs), INtegrase Inhibitors (INIs), Protease Inhibitors (PIs), and Entry Inhibitors (EIs). Co-administration of certain antiretroviral drugs with Pharmacokinetic Enhancers (PEs) may boost the effectiveness of the primary therapeutic agent. The combination of multiple antiretroviral drug regimens (Highly Active AntiRetroviral Therapy - HAART) is currently the standard therapeutic approach for HIV infection. So far, the use of HAART offers the best opportunity for prolonged and maximal viral suppression, and preservation of the immune system upon HIV infection. Still, the frequent administration of high doses of multiple drugs, their inefficient ability to reach the viral reservoirs in adequate doses, the development of drug resistance, and the lack of patient compliance compromise the complete HIV elimination. The development of nanotechnology-based drug delivery systems may enable targeted delivery of antiretroviral agents to inaccessible viral reservoir sites at therapeutic concentrations. In addition, the application of Computer-Aided Drug Design (CADD) approaches has provided valuable tools for the development of anti-HIV drug candidates with favourable pharmacodynamics and pharmacokinetic properties.
Collapse
Affiliation(s)
- Rita Melo
- Centro de Ciencias e Tecnologias Nucleares, Instituto Superior Tecnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), Bobadela LRS 2695-066, Portugal.,CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1ºandar, Universidade de Coimbra, Coimbra 3004-517, Portugal
| | - Agostinho Lemos
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1ºandar, Universidade de Coimbra, Coimbra 3004-517, Portugal.,GIGA Cyclotron Research Centre In Vivo Imaging, University of Liège, Liège 4000, Belgium
| | - António J Preto
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1ºandar, Universidade de Coimbra, Coimbra 3004-517, Portugal
| | - Beatriz Bueschbell
- Pharmaceutical Chemistry I, PharmaCenter, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | - Pedro Matos-Filipe
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1ºandar, Universidade de Coimbra, Coimbra 3004-517, Portugal
| | - Carlos Barreto
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1ºandar, Universidade de Coimbra, Coimbra 3004-517, Portugal
| | - José G Almeida
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1ºandar, Universidade de Coimbra, Coimbra 3004-517, Portugal
| | - Rúben D M Silva
- Centro de Ciencias e Tecnologias Nucleares, Instituto Superior Tecnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), Bobadela LRS 2695-066, Portugal
| | - João D G Correia
- Centro de Ciencias e Tecnologias Nucleares, Instituto Superior Tecnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), Bobadela LRS 2695-066, Portugal
| | - Irina S Moreira
- CNC - Center for Neuroscience and Cell Biology; Rua Larga, FMUC, Polo I, 1ºandar, Universidade de Coimbra, Coimbra 3004-517, Portugal.,Bijvoet Center for Biomolecular Research, Faculty of Science - Chemistry, Utrecht University, Utrecht 3584CH, Netherland
| |
Collapse
|
5
|
Sharma VK, Bharatam PV. Identification of Selective Inhibitors of LdDHFR Enzyme Using Pharmacoinformatic Methods. J Comput Biol 2020; 28:43-59. [PMID: 32207987 DOI: 10.1089/cmb.2019.0332] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dihydrofolate reductase (DHFR) is a well-known enzyme of the folate metabolic pathway and it is a validated drug target for leishmaniasis. However, only a few leads are reported against Leishmania donovani DHFR (LdDHFR), and thus, there is a need to identify new inhibitors. In this article, pharmacoinformatic tools such as molecular docking, virtual screening, absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiling, and molecular dynamics (MD) simulations were utilized to identify potential LdDHFR inhibitors. Initially, a natural DHFR substrate (dihydrofolate), a classical DHFR inhibitor (methotrexate), and a potent LdDHFR inhibitor, that is, "5-(3-(octyloxy)benzyl)pyrimidine-2,4-diamine" (LEAD) were docked in the active site of the LdDHFR and MD simulated to understand the binding mode characteristics of the substrates/inhibitors in the LdDHFR. The shape of the LEAD molecule was used as a query for shape-based virtual screening, while the three-dimensional structure of LdDHFR was utilized for docking-based virtual screening. In silico ADMET factors were also considered during virtual screening. These two screening processes yielded 25 suitable hits, which were further validated for their selectivity toward LdDHFR using molecular docking and prime molecular mechanics/generalized born surface area analysis in the human DHFR (HsDHFR). Best six hits, which were selective and energetically favorable for the LdDHFR, were chosen for MD simulations. The MD analysis showed that four of the hits exhibited very good binding affinity for LdDHFR with respect to HsDHFR, and two hits were found to be more selective than the reported potent LdDHFR inhibitor. The present study thus identifies hits that can be further designed and modified as potent LdDHFR inhibitors.
Collapse
Affiliation(s)
- Vishnu Kumar Sharma
- Department of Pharmacoinformatics and National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, India
| | - Prasad V Bharatam
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, India
| |
Collapse
|
6
|
Sharma S, Gupta J, Prabhakar PK, Gupta P, Solanki P, Rajput A. Phytochemical Repurposing of Natural Molecule: Sabinene for Identification of Novel Therapeutic Benefits UsingIn SilicoandIn VitroApproaches. Assay Drug Dev Technol 2019; 17:339-351. [DOI: 10.1089/adt.2019.939] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Shamini Sharma
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Jeena Gupta
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Pranav Kumar Prabhakar
- Department of Medical Laboratory Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Pawan Gupta
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Department of Research Facilitation, Division of Research and Development, Lovely Professional University, Phagwara, Punjab, India
| | - Preeti Solanki
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Anuradha Rajput
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
7
|
Samorlu AS, Yelekçi K, Ibrahim Uba A. The design of potent HIV-1 integrase inhibitors by a combined approach of structure-based virtual screening and molecular dynamics simulation. J Biomol Struct Dyn 2019; 37:4644-4650. [DOI: 10.1080/07391102.2018.1557559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Augustine S. Samorlu
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Fatih, Istanbul, Turkey
| | - Kemal Yelekçi
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Fatih, Istanbul, Turkey
| | - Abdullahi Ibrahim Uba
- Department of Bioinformatics and Genetics, Faculty of Engineering and Natural Sciences, Kadir Has University, Fatih, Istanbul, Turkey
- Centre for Biotechnology Research, Bayero University Kano, Kano, Nigeria
| |
Collapse
|
8
|
Investigation of indirubin derivatives: a combination of 3D-QSAR, molecular docking, and ADMET towards the design of new DRAK2 inhibitors. Struct Chem 2018. [DOI: 10.1007/s11224-018-1134-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
9
|
Chen N, Chen J, Yao B, Li Z. QSAR Study on Antioxidant Tripeptides and the Antioxidant Activity of the Designed Tripeptides in Free Radical Systems. Molecules 2018; 23:molecules23061407. [PMID: 29890782 PMCID: PMC6100293 DOI: 10.3390/molecules23061407] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022] Open
Abstract
In this study, quantitative structure-activity relationship (QSAR) models were determined based on 91 antioxidant tripeptides. We firstly adopted the stepwise regression (SWR) method for selecting key variables without autocorrelation and then utilized multiple linear regression (MLR), support vector machine (SVM), random forest (RF), and partial least square regression (PLS) to develop predictive QSAR models based on the screened variables. The results demonstrated that all the established models have good reliability (R²train > 0.86, Q²train > 0.70) and relatively good predictability (R²test > 0.88). The contribution of amino acid residues was calculated from the stepwise regression combined with multiple linear regression (SWR-MLR) method model that shows Trp, Tyr, or Cys at C-terminus is favorable for antioxidant activity of tripeptides. Nineteen antioxidant tripeptides were designed based on SWR-MLR models, and the antioxidant activity of these tripeptides were evaluated using three antioxidant assays in free radical systems (1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging capacity, trolox equivalent antioxidant capacity assay, and the ferric reducing antioxidant power assay). The experimental antioxidant activities of these tripeptides were higher than the calculated/predicted activity values of the QSAR models. The QSAR models established can be used to identify and screen novel antioxidant tripeptides with high activity.
Collapse
Affiliation(s)
- Nan Chen
- School of Life Science, Chongqing University, Chongqing 401331, China.
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
- Chongqing Key Laboratory of Industrial Fermentation Microorganism, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Ji Chen
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
- Chongqing Key Laboratory of Industrial Fermentation Microorganism, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Bo Yao
- College of Chemistry and Chemical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
- Chongqing Key Laboratory of Industrial Fermentation Microorganism, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Zhengguo Li
- School of Life Science, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
10
|
Aouidate A, Ghaleb A, Ghamali M, Chtita S, Ousaa A, Choukrad M, Sbai A, Bouachrine M, Lakhlifi T. Structural basis of pyrazolopyrimidine derivatives as CAMKIIδ kinase inhibitors: insights from 3D QSAR, docking studies and in silico ADMET evaluation. CHEMICAL PAPERS 2018. [DOI: 10.1007/s11696-018-0510-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
11
|
Aouidate A, Ghaleb A, Ghamali M, Ousaa A, Choukrad M, Sbai A, Bouachrine M, Lakhlifi T. 3D QSAR studies, molecular docking and ADMET evaluation, using thiazolidine derivatives as template to obtain new inhibitors of PIM1 kinase. Comput Biol Chem 2018; 74:201-211. [PMID: 29635214 DOI: 10.1016/j.compbiolchem.2018.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/03/2018] [Accepted: 03/10/2018] [Indexed: 02/04/2023]
Abstract
Proviral Integration site for Moloney murine leukemia virus-1 (PIM1) belongs to the serine/threonine kinase family of Ca2+-calmodulin-dependent protein kinase (CAMK) group, which is involved in cell survival and proliferation as well as a number of other signal transduction pathways. Thus, PIM1 is regarded as a promising target for treatment of cancers. In the present paper, a three-dimensional quantitative structure activity relationship (3D-QSAR) and molecular docking were performed to investigate the binding between PIM1 and thiazolidine inhibitors in order to design potent inhibitors. The comparative molecular similarity indices analysis (CoMSIA) was developed using twenty-six molecules having pIC50 ranging from 8.854 to 6.011 (IC50 in nM). The best CoMSIA model gave significant statistical quality. The determination coefficient (R2) and Leave-One-Out cross-validation coefficient (Q2) are 0.85 and 0.58, respectively. Furthermore, the predictive ability of this model was evaluated by external validation((n = 11, R2test = 0.72, and MAE = 0.170 log units). The graphical contour maps could provide structural features to improve inhibitory activity. Furthermore, a good consistency between contour maps and molecular docking strongly demonstrates that the molecular modeling is reliable. Based on these satisfactory results, we designed several new potent PIM1 inhibitors and their inhibitory activities were predicted by the molecular models. Additionally, those newly designed inhibitors, showed promising results in the preliminary in silico ADMET evaluations, compared to the best inhibitor from the studied dataset. The results expand our understanding of thiazolidines as inhibitors of PIM1 and could be of great help in lead optimization for early drug discovery of highly potent inhibitors.
Collapse
Affiliation(s)
- Adnane Aouidate
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - Adib Ghaleb
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - Mounir Ghamali
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - Abdellah Ousaa
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - M'barek Choukrad
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | - Abdelouahid Sbai
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| | | | - Tahar Lakhlifi
- MCNSL, School of Sciences, Moulay Ismail University, Meknes, Morocco.
| |
Collapse
|
12
|
A structure-based design approach to advance the allyltyrosine-based series of HIV integrase inhibitors. Tetrahedron 2018. [DOI: 10.1016/j.tet.2017.11.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
13
|
Furanone derivatives as new inhibitors of CDC7 kinase: development of structure activity relationship model using 3D QSAR, molecular docking, and in silico ADMET. Struct Chem 2018. [DOI: 10.1007/s11224-018-1086-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
14
|
Sachithanandham J, Konda Reddy K, Solomon K, David S, Kumar Singh S, Vadhini Ramalingam V, Alexander Pulimood S, Cherian Abraham O, Rupali P, Sridharan G, Kannangai R. Effect of HIV-1 Subtype C integrase mutations implied using molecular modeling and docking data. Bioinformation 2016; 12:221-230. [PMID: 28149058 PMCID: PMC5267967 DOI: 10.6026/97320630012221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 01/12/2023] Open
Abstract
The degree of sequence variation in HIV-1 integrase genes among infected patients and their impact on clinical response to Anti retroviral therapy (ART) is of interest. Therefore, we collected plasma samples from 161 HIV-1 infected individuals for subsequent integrase gene amplification (1087 bp). Thus, 102 complete integrase gene sequences identified as HIV-1 subtype-C was assembled. This sequence data was further used for sequence analysis and multiple sequence alignment (MSA) to assess position specific frequency of mutations within pol gene among infected individuals. We also used biophysical geometric optimization technique based molecular modeling and docking (Schrodinger suite) methods to infer differential function caused by position specific sequence mutations towards improved inhibitor selection. We thus identified accessory mutations (usually reduce susceptibility) leading to the resistance of some known integrase inhibitors in 14% of sequences in this data set. The Stanford HIV-1 drug resistance database provided complementary information on integrase resistance mutations to deduce molecular basis for such observation. Modeling and docking analysis show reduced binding by mutants for known compounds. The predicted binding values further reduced for models with combination of mutations among subtype C clinical strains. Thus, the molecular basis implied for the consequence of mutations in different variants of integrase genes of HIV-1 subtype C clinical strains from South India is reported. This data finds utility in the design, modification and development of a representative yet an improved inhibitor for HIV-1 integrase.
Collapse
Affiliation(s)
| | - Karnati Konda Reddy
- SNHRC Vellore and Computer-Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics Alagappa University, Karaikudi, Tamil Nadu, India
| | - King Solomon
- Departments of Clinical Virology Alagappa University, Karaikudi, Tamil Nadu, India
| | - Shoba David
- Departments of Clinical Virology Alagappa University, Karaikudi, Tamil Nadu, India
| | - Sanjeev Kumar Singh
- SNHRC Vellore and Computer-Aided Drug Design and Molecular Modeling Lab, Department of Bioinformatics Alagappa University, Karaikudi, Tamil Nadu, India
| | | | | | | | - Pricilla Rupali
- Departments of Internal Medicine, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Gopalan Sridharan
- Christian Medical College, Vellore, Sri Sakthi Amma Institute of Biomedical Research Institute
| | - Rajesh Kannangai
- Departments of Clinical Virology Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
15
|
Su M, Tan J, Lin CY. Development of HIV-1 integrase inhibitors: recent molecular modeling perspectives. Drug Discov Today 2015. [PMID: 26220090 DOI: 10.1016/j.drudis.2015.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Of the three viral enzymes essential to HIV replication, HIV-1 integrase (IN) is gaining popularity as a target for the antiviral therapy of AIDS. Substantial work focusing on IN has been done over the past three decades, which has facilitated and led to the approval of three drugs. Here, we discuss in detail the development of IN inhibitors between January 2012 and May 2014, with a particular focus on molecular simulation. We highlight controversial aspects of computational drug design and refer to alternative practices where appropriate. The analysis of these computational approaches provides some useful clues to the possible future discovery of novel IN inhibitors.
Collapse
Affiliation(s)
- Min Su
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Jianjun Tan
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, China.
| | - Chun-Yuan Lin
- Department of Computer Science and Information Engineering, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
16
|
Rawat S, Gupta P, Kumar A, Garg P, Suri CR, Sahoo DK. Molecular Mechanism of Poly(vinyl alcohol) Mediated Prevention of Aggregation and Stabilization of Insulin in Nanoparticles. Mol Pharm 2015; 12:1018-30. [DOI: 10.1021/mp5003653] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sanjay Rawat
- CSIR−Institute of Microbial Technology, Sector 39-A, Chandigarh 160036, India
| | - Pawan Gupta
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Sector 67, Mohali 160062, India
| | - Anil Kumar
- CSIR−Institute of Microbial Technology, Sector 39-A, Chandigarh 160036, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, Sector 67, Mohali 160062, India
| | - C. Raman Suri
- CSIR−Institute of Microbial Technology, Sector 39-A, Chandigarh 160036, India
| | - Debendra K. Sahoo
- CSIR−Institute of Microbial Technology, Sector 39-A, Chandigarh 160036, India
| |
Collapse
|
17
|
Guan J, Zhao Y, Zhu H, An Z, Yu Y, Li R, Yu Z. A rapid and sensitive UHPLC–MS/MS method for quantification of 2-(2-hydroxypropanamido) benzoic acid in rat plasma: Application to a pharmacokinetic study. J Pharm Biomed Anal 2014; 95:20-5. [DOI: 10.1016/j.jpba.2014.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 02/08/2023]
|
18
|
Maganti L, Ghoshal N. 3D-QSAR studies and shape based virtual screening for identification of novel hits to inhibit MbtA inMycobacterium tuberculosis. J Biomol Struct Dyn 2014; 33:344-64. [DOI: 10.1080/07391102.2013.872052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
19
|
Wu L, Wu Y, Shen H, Gong P, Cao L, Wang G, Hao H. Quantitative structure–ion intensity relationship strategy to the prediction of absolute levels without authentic standards. Anal Chim Acta 2013; 794:67-75. [DOI: 10.1016/j.aca.2013.07.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Revised: 07/12/2013] [Accepted: 07/13/2013] [Indexed: 10/26/2022]
|
20
|
In silico screening for identification of novel HIV-1 integrase inhibitors using QSAR and docking methodologies. Med Chem Res 2013. [DOI: 10.1007/s00044-013-0490-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|