Abstract
Although highly active antiretroviral therapy (HAART) is effective in controlling the progression of AIDS, the emergence of drug-resistant strains increases the difficulty of successful treatment of patients with HIV infection. Increasing numbers of patients are facing the dilemma that comes with the running out of drug combinations for HAART. Computational methods play a key role in anti-HIV drug development. A substantial number of studies have been performed in anti-HIV drug development using various computational methods, such as virtual screening, QSAR, molecular docking, and homology modeling, etc. In this review, we summarize recent advances in the application of computational methods to anti-HIV drug development for five key targets as follows: reverse transcriptase, protease, integrase, CCR5, and CXCR4. We hope that this review will stimulate researchers from multiple disciplines to consider computational methods in the anti-HIV drug development process.
KEY WORDS: anti-HIV, computational methods, co-receptor, drug, enzyme
INTRODUCTION
In the absence of a successful preventive vaccine, the spread of human immunodeficiency virus (HIV) infection has caused a worldwide pandemic. Approximately 33 million people are currently living with HIV-1, approximately 2.1 million people die every year of acquired immunodeficiency syndrome (AIDS) and associated complications, and there are 2.5 million new infections every year (www.unaids.org). The first anti-HIV drug, azidothymidine (AZT), was approved in 1987 (1), and more than 30 anti-HIV drugs are currently used for clinical treatment (Table I, www.fda.org). Although highly active anti-retroviral therapy is effective in controlling the progression of AIDS, the combined use of multiple drugs is greatly hindered by the emergence of drug-resistant HIV strains. More and new anti-HIV drugs are therefore needed for clinical treatment.
Table I.
Drugs Used in the Treatment of HIV Infection (www.fda.gov)
| Brand name | Generic name | Manufacturer namea | Approval date | Type |
|---|---|---|---|---|
| Combivir | Lamivudine and zidovudine | GlaxoSmithKline | 27-Sep-97 | NRTI |
| Emtriva | Emtricitabine, FTC | Gilead Sciences | 02-Jul-03 | NRTI |
| Epivir | Lamivudine, 3TC | GlaxoSmithKline | 17-Nov-95 | NRTI |
| Epzicom | Abacavir and lamivudine | GlaxoSmithKline | 02-Aug-04 | NRTI |
| Hivid | Zalcitabine, dideoxycytidine, ddC (no longer marketed) | Hoffmann-La Roche | 19-Jun-92 | NRTI |
| Retrovir | Zidovudine, azidothymidine, AZT, ZDV | GlaxoSmithKline | 19-Mar-87 | NRTI |
| Trizivir | Abacavir,zidovudine, and lamivudine | GlaxoSmithKline | 14-Nov-00 | NRTI |
| Truvada | Tenofovir disoproxil fumarate and emtricitabine | Gilead Sciences, Inc. | 02-Aug-04 | NRTI |
| Videx EC | Enteric-coated didanosine, ddI EC | Bristol–Myers Squibb | 31-Oct-00 | NRTI |
| Videx | Didanosine, dideoxyinosine, ddI | Bristol–Myers Squibb | 9-Oct-91 | NRTI |
| Viread | Tenofovir disoproxil fumarate, TDF | Gilead | 26-Oct-01 | NRTI |
| Zerit | Stavudine, d4T | Bristol–Myers Squibb | 24-Jun-94 | NRTI |
| Ziagen | Abacavir sulfate, ABC | GlaxoSmithKline | 17-Dec-98 | NRTI |
| Edurant | Rilpivirine | Tibotec Therapeutics | 20-May-11 | NNRTI |
| Intelence | Etravirine | Tibotec Therapeutics | 18-Jan-08 | NNRTI |
| Rescriptor | Delavirdine, DLV | Pfizer | 4-Apr-97 | NNRTI |
| Sustiva | Efavirenz, EFV | Bristol–Myers Squibb | 17-Sep-98 | NNRTI |
| Viramune | Nevirapine, NVP | Boehringer Ingelheim | 21-Jun-96 | NNRTI |
| Viramune | Nevirapine, NVP | Boehringer Ingelheim | 25-Mar-11 | NNRTI |
| Agenerase | Amprenavir, APV (no longer marketed) | GlaxoSmithKline | 15-Apr-99 | PI |
| Aptivus | Tipranavir, TPV | Boehringer Ingelheim | 22-Jun-05 | PI |
| Crixivan | Indinavir, IDV, | Merck | 13-Mar-96 | PI |
| Fortovase | Saquinavir (no longer marketed) | Hoffmann-La Roche | 7-Nov-97 | PI |
| Invirase | Saquinavir mesylate, SQV | Hoffmann-La Roche | 6-Dec-95 | PI |
| Kaletra | Lopinavir and ritonavir, LPV/RTV | Abbott Laboratories | 15-Sep-00 | PI |
| Lexiva | Fosamprenavir calcium, FOS-APV | GlaxoSmithKline | 20-Oct-03 | PI |
| Norvir | Ritonavir, RTV | Abbott Laboratories | 1-Mar-96 | PI |
| Prezista | Darunavir | Tibotec, Inc. | 23-Jun-06 | PI |
| Reyataz | Atazanavir sulfate, ATV | Bristol–Myers Squibb | 20-Jun-03 | PI |
| Viracept | Nelfinavir mesylate, NFV | Agouron Pharmaceuticals | 14-Mar-97 | PI |
| Fuzeon | Enfuvirtide, T-20 | Hoffmann-La Roche & Trimeris | 13-Mar-03 | Fusion Inhibitor |
| Selzentry | Maraviroc | Pfizer | 06-August-07 | CCR5 inhibitor |
| Isentress | Raltegravir | Merck & Co., Inc. | 12-Oct-07 | INSTI |
| Stribild | Elvitegravir | Gilead Sciences | 27-Aug-12 | INSTI |
| Tivicay | Dolutegravir | GlaxoSmithKline | 13-Aug-13 | INSTI |
NRTI nucleoside reverse transcriptase inhibitors, NNRTI non-nucleoside reverse transcriptase inhibitor, PI protease inhibitor, CCR5 C-C chemokine receptor type 5, INSTI integrase strand transfer inhibitors, RTV ritonavir, NFV nelfinavir mesylate, ATV atazanavir sulfate, LPV lopinavir, FOS-APV fosamprenavir calcium, IDV indinavir, APV amprenavir, SQV saquinavir mesylate, NVP nevirapine, TPV tipranavir, EFV efavirenz, DLV delavirdine, TDF tenofovir disoproxil fumarate, ABC abacavir sulfate, d4T stavudine, ddI didanosine, dideoxyinosine, ddI EC enteric-coated didanosine, ZDV zidovudine, AZT azidothymidine, FTC emtricitabine, 3TC lamivudine, T-20 enfuvirtide
aManufacturer/sponsor at time of approval
Computational techniques are increasingly employed in drug discovery and optimization. Techniques applied to anti-HIV drug research are classified as (1) ligand methods based on known active compounds that can infer biological activity, such as classical quantitative structure–activity relationship (QSAR), (2) structure-based methods that rely on the 3D structure of protein receptors, such as molecular docking and molecular dynamics, and (3) universal methods, structure- or ligand-based, such as 3D QSAR or 3D pharmacophore elucidation (2). Homology modeling is usually useful when an experimental 3D structure of protein receptor is not available. A review has provided the theoretical introduction and detailed protocols of the computational methods used in anti-viral agent development (2). Although multiple methods are applied to anti-HIV drug development, receptor structure-based molecular docking and ligand-based QSAR are the most frequently used methods. The HIV life cycle has multiple stages, including entry, reverse transcription, integration, protein translation, assembly, and release. Throughout the entire process, many viral proteins and host receptors can be targeted for drug development. In this review, we summarize the recent progresses of anti-HIV drug development via computational methods applied to five main targets: three key viral enzymes (reverse transcriptase, protease, integrase) and two common co-receptors.
REVERSE TRANSCRIPTASE
HIV is a retrovirus, and reverse transcriptase (RT) is its key enzyme; RT reverse transcribes the viral RNA into a provirus. RT plays a multifunctional role and is an essential component for HIV to complete the replication cycle. There are two types of reverse transcriptase inhibitors, namely non-nucleoside reverse transcriptase inhibitor (NNRTI) and nucleoside reverse transcriptase inhibitors (NRTI). As RT is the most important target for drug design, there are more than 240 crystal structures of HIV-1 RT and mutants available. Based on the vast number of crystal structures, numerous studies report the development of RT inhibitors using a computer-guided design. The structure-based molecular docking approach plays a key role in the computer-guided development of RT inhibitors. Although hundreds of HIV-1 RT structures were determined, only one structure was shown to contain an RNA/DNA hybrid before 2013. Recently, three structures of HIV-1 RT in complex with a non-nucleotide RT inhibitor (NVP) and an RNA/DNA hybrid were reported (3). These three structures differ from all previously reported RT–DNA complexes. These findings indicate that a RT–nucleic acid complex may adopt two structural states, one suited to DNA polymerization and the other suited to RNA degradation (3). Researchers also speculate that RT mutations that confer drug resistance, but that are distant from the inhibitor-binding sites, often map to the unique RT-hybrid interface that undergoes conformational changes between the two catalytic states (3). The structure–activity relationship (SAR) of three RT inhibitors of marine origin (THD, HDD, and ADD) was approached with molecular modeling (4). Molecular docking studies of THD into HIV-1 RT wildtype and 12 different mutants showed that mutations have little influence in the positioning and interactions of THD (4). Following a rational drug design approach, a modification of THD was suggested to improve its biological activity (4). Five docking programs (Glide, FlexX, Molegro Virtual Docker, AutoDock Vina, and Hyde) were evaluated for their ability to predict the relative biological activity of 111 known 1,2,4-triazole and 76 other azole type HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs) (5). The results show that after proper validation and optimization, molecular docking programs can help predict the relative biological activity of azole NNRTIs and facilitate the identification of novel triazole NNRTIs (5). Computational methods provide insights into the detailed interaction between compounds and targets, providing a comprehensive understanding of the pharmacological activities of compounds and information after modification of the drug. Computational methods are convenient, especially when large-scale experiments are difficult to conduct. Other studies have focused on the discovery of potential RT inhibitors via molecular docking. The unliganded HIV-1 RT (1DLO) was used for the virtual screening of 4-thiazolidinone and its derivatives (ChemBank database) by using AutoDock4 (6). One derivative, (5E)-3-(2-aminoethyl)-5-(2-thienylmethylene)-1,3-thiazolidine-2,4-dione (CID 3087795), was discovered to be a promising inhibitor for HIV-1 RT with a minimum energy score and the highest number of interactions with active site residues (6). Molecular docking is also widely used in SAR studies, as a way to evaluate the anti-viral activity of newly discovered or synthesized compounds (7–16).
PROTEASE
Protease (PR), as one of the three key enzymes, cleaves the viral polyprotein after its translation to release functionally mature proteins. After the protease is inactivated, the HIV virion becomes non-infectious. Two copies of 99 amino acid protein chains are non-covalently associated to form the long and symmetrical tunnel of the binding sites of HIV protease. Recently, a room-temperature joint X-ray/neutron structure of the HIV-1 protease in complex with the clinical drug amprenavir was reported, providing a direct determination of the hydrogen atom positions in the enzyme’s active site (17). This structure may provide insight for the design of improved protease inhibitors. Eleven PR inhibitors are FDA-approved at present (Table I). These PR inhibitors (PIs) are essential components of highly active antiretroviral therapy. Our review focuses on the discussion of recent innovative examples of PR inhibitor development via computer-guided drug design. Danuravir is the most potent HIV PR inhibitor that is known. A series of danuravir derivatives was evaluated with 3D-QSAR and docking methods (18), demonstrating that the ligand-based and receptor-based results were in agreement with the experimental results (18). Five QSAR models were generated on the basis of available experimental IC (50) values (HIV-I- and HIV-IIIB-infected MT4 and CEMSS cells and HIV-I-infected C8166 cells) (19). The sixth model was generated by combining the inhibitors of all five models, critically examining the role of conceptual DFT descriptors and docking scores on 156 HIV PR inhibitors (19). Previous molecular dynamics simulations revealed some opening pathways and conformations in the opening process of HIV PR (20). Recent studies evaluated the possibility of the existence of alternative opening pathways to the semi-open conformation, by performing a molecular dynamics simulation of the early opening process of an inhibitor-free HIV-1 protease with explicit solvent (20). An X-ray diffraction structure of HIV-1 protease was used in this study as a template. The mutant residues in 1D4L were restored to the original residues via model building (20). This made the results suitable for comparison with experimental findings. Consistent with former experimental studies, the closed form of the HIV-1 protease structure (inhibitor-free) converts to a semi-open conformation spontaneously (20). However, the opening motion to the semi-open conformation happens in a manner that differs from both the previously reported modes. In this simulation, the gap between the flap tips in the closed conformation was 15 Å thinner than in the semi-open conformation (20). Additionally, the alternative opening route discovered in this study was consistent with those observed by some nuclear magnetic resonance (NMR) or electron paramagnetic resonance (EPR) experiments that failed in identifying a large-amplitude curling motion as reported previously (20). Characterizing the dynamics of these opening and closing events led to a more comprehensive understanding of the binding of inhibitors. This is helpful for understanding the functional mechanism and substrate-/inhibitor-binding dynamics of PRs. Such findings provide new directions for the design of more effective inhibitors for the treatment of HIV infection.
INTEGRASE
Integrase (IN) is the viral enzyme that catalyzes the reaction of integration. The integration of viral DNA into the host genome is critical for permanent infection by the virus, which is achieved through 3’-terminal processing (3'-P) and strand transfer (ST) (21). The first integrase inhibitor, raltegravir, was FDA-approved in 2007 for the clinical treatment of AIDS (22–24). Three integrase inhibitors have been FDA-approved for the treatment of HIV infection at present (Table I). Many studies were reported on the design and screening of new integrase inhibitors, targeting both strand transfer and lens epithelium-derived growth factor (IN-LEDGF)/p75 interaction via virtual screening, molecular dynamic simulation, and 3D-QSAR. Pharmacophore modeling and atom-based 3D-QSAR studies were performed for 3-hydroxypyrimidine-2,4-diones, of which three compounds were identified as potential HIV-1 IN strand transfer inhibitors (INSTIs) (25). Translocation of preintegration complex (PIC) (26) into the nucleus is a precondition for successful integration, which is one process involved in protein–protein interactions (PPI) (27). PIC is composed of integrase, viral DNA, and a bunch of host factors. Although studies of PIC are attractive to virologists, and significant progress has been made in recent years, the current understanding of PIC is incomplete. Lens epithelium-derived growth factor (LEDGF/p75) is the first reported cellular factor to interact with integrase (28, 29). The interaction details were disclosed with the report of the crystal structure of IN-LEDGF/p75 complex (30–32). Molecular dynamics simulations were also carried out to study the detailed interactions between IN and LEDGF/p75. Residues Gln168, Glu170, and Thr174 in chain A of IN, Thr125, and Trp131 in chain B of IN as well as Ile365, Asp366, Phe406, and Val408 in LEDGF/p75 were identified as hotspots at the binding interface (33). The crystal structure of the LEDGF/p75 integrase-binding domain (IBD) in complex with a dimer of the integrase catalytic core domain was reported (34). Disrupting the LEDGF/p75-integrase interaction is an attractive target for the development of novel integrase inhibitors. Against the LEDGF/p75 binding pocket of IN, 26 known drugs from DrugBank were screened via molecular docking, and eight drugs were identified as potential IN inhibitors that target IN-LEDGF/p75 interactions (35). A series of azaindole carboxylic acid derivatives, previously reported as promising HIV-1 integrase inhibitors, were studied through a three-dimensional quantitative structure–activity relationship (3D-QSAR) (36). Binding modes were explored by molecular docking based on the molecule with the highest activity (36). New azaindole carboxylic acid derivatives were designed, and their inhibitory activities on HIV-1 integrase were predicted based on the computational models constructed in these studies (36). Molecular docking is also used to simulate the interaction modes between IN and newly synthesized compounds. Halogen-substituted phenanthrene beta-diketo acids were synthesized and identified as new HIV-1 integrase inhibitors (37). The most active compound, 4-(6-chlorophenanthren-2-yl)-2,4-dioxobutanoic acid (18), inhibited both 3'-terminal processing and strand transfer (37). Two predominant binding modes that were distinct from the binding modes of raltegravir and elvitegravir were revealed via docking, and docking modes suggested a novel binding region in the IN active site (37). The rapid emergence of cross-resistance to strand transfer inhibitors is a serious issue in the therapy of AIDS. Molecular dynamics simulation is also used to study the detailed molecular mechanism of the cross-resistance mutation E138K/Q148K to raltegravir, elvitegravir, and dolutegravir, which are three important INSTIs (38). Via structure-based virtual screening, 38 compounds were selected from an in-house library containing 1,430 natural products and their derivatives. The virtual screening was performed using an induced fit model to discover inhibitors that target IN − LEDGF/p75 interactions (39). Eighteen hits were discovered through AlphaScreen bioassays. One compound, (E)-3-(2-chlorophenyl)-1-(2,4-dihydroxyphenyl)prop-2-en-1-one (NPD170), showed the highest anti-viral activity (EC50 = 1.81 μM) (39). Although the therapeutic index (TI) of NPD170 was not very high (4.88), this was a good case for the discovery of an anti-HIV compound via virtual screening.
CXCR4
HIV infection is initiated by the binding of viral gp120 to human CD4 on the cell surface. A conformational change of gp120 induced by binding exposes the co-receptor binding domain. The fusion of gp41 with the cell membrane after co-receptor binding is the final step of entry. CXCR4 (C-X-C chemokine receptor type 4) and CCR5 (C-C chemokine receptor type 5) are two major co-receptors of HIV, expressed on T cells and macrophages, respectively. CXCR4 is a seven trans-membrane G-protein-coupled receptor (GPCR) of stromal cell-derived factor-1alpha (SDF-1α). SDF-1α inhibits cell fusion and infection of syncytium-inducing phenotype HIV strains. CXCR4 is an appealing target for anti-viral drug development. AMD3100 was the first promising CXCR4 antagonist, but it proved to be unsuitable for the treatment of AIDS in later studies (40). Before 2011, the CXCR4 antagonist development depended on traditional chemical synthesis and drug design to mimic the available part of SDF-1α/CXCR4 structures. The first low molecular weight antagonist, AMD3100, is a representative example (40). T22 is a representative polypeptide antagonist of CXCR4, which was synthesized with chemical modification from the horseshoe crab hemocytic polypeptides (41–43). T134 is a small-sized analog of T22 with reduced positive charges, highly potent activity, and significantly less cytotoxicity (41). Two other CXCR4 antagonists, ALX40-4C and vMIP II, are active against the AMD3100 resistant strains (41, 44, 45). An excellent review on CXCR4 antagonist development before 2006 is available (46).
Lacking CXCR4 crystal structure, the usage of computational methods in antagonist development was limited. However, there may still be some potential research directions for future studies in this field. A three-dimensional model of human CXCR4 was constructed via homology modeling according to the high-resolution bovine rhodopsin structure (47). Interactions between the constructed CXCR4 and flavonoids were investigated with in silico docking (47). A similar homology modeling study was also performed to construct homology models of CXCR4 and CCR5. The models were used for virtually screening a library consisting of 602 known CXCR4 and CCR5 inhibitors and some 4,700 similar, presumed inactive, molecules for comparison of ligand-based shape-matching searches and receptor-based docking methods (48). Although these studies described valuable trials in developing CXCR4 antagonists through computational methods, the revealed CXCR4 crystal structures are much different from the structures constructed by homology modeling. Five independent crystal structures of CXCR4 bound to an antagonist small-molecule IT1t and a cyclic peptide CVX15 at 2.5–3.2 Å resolution were reported in 2010 (49). All structures reveal a consistent homodimer with an interface including helices V and VI, and this interface may be involved in regulating signaling (49). The ligand-binding sites are closer to the extracellular surface, the location and shape of which are much different from other G-protein-coupled receptors (49). The structures provide new clues about the interactions between CXCR4 and SDF-1α and with gp120 (49). Thereafter, a receptor-based virtual screening performance of the five crystallized CXCR4 structures along with a CXCR4 rhodopsin-based homology model was carried out using 248 known CXCR4 inhibitors from four different chemotype families and 4,696 different presumed inactives (50). The results showed that the 3OE6 structure achieved the highest docking-based performance and was proposed as the best for CXCR4 antagonist discovery via virtual screening (50). The 3OE6 structure of CXCR4 in complex with antagonist IT1t has been reported (49).
Recently, a highly specific and sensitive pharmacophore model was reported for identifying CXCR4 antagonists that could potentially serve as HIV entry inhibitors (51). It achieved the best performance when compared with docking and shape-matching virtual screening approaches using the 3OE6 CXCR4 crystal structure and high-affinity ligands as query molecules, respectively (51). Although no CXCR4 antagonist has been approved for the clinical treatment of HIV infection, numerous candidates are under investigation, and several are in clinical trials (41–43, 46, 52, 53). The disclosure of CXCR4 crystal structures will greatly improve the progress of development of HIV entry inhibitors through CXCR4.
CCR5
CCR5 is also a member of seven trans-membrane G-protein-coupled receptors. In contrast to CXCR4, CCR5 has three natural ligands: RANTES, MIP-1α, and MIP-1β (54, 55). Maraviroc is the first CCR5 co-receptor FDA-approved antagonist for the treatment of CCR5-tropic HIV infection (56, 57). The detailed usage of maraviroc in the treatment of HIV infection was reviewed by Perry (58). Vicriviroc is a small-molecule CCR5 antagonist that has entered phase III clinical trials (59). More information can be found in a review article on CCR5 antagonists, which also provides information on the integrase inhibitor and fusion inhibitor (60). Homology modeling was the main computational method for CCR5 antagonist development before 2013. Several studies were performed on the human CCR5 structure construction via homology modeling by using the X-ray structure of the bovine rhodopsin receptor (61–64). Additionally, a homology model of human CCR5 was developed based on the reported CXCR4 structure as a template (65). The 2.7 Å-resolution crystal structure of human CCR5 bound to the marketed HIV drug maraviroc was reported recently (66). The reported structure revealed a ligand-binding site that was distinct from the proposed major recognition sites for chemokines and gp120, providing insights into the mechanism of the allosteric inhibition of chemokine signaling and HIV entry (67). The high-resolution crystal structure of CCR5 enables structure-based drug discovery for the treatment of HIV-1 infection (67).
SUMMARY
This review emphasizes the advances of anti-HIV drug development via computational methods applied to five key targets. Although the above examples are representative, the computational methods described are also helpful in anti-viral drug development targeting other HIV proteins, such as gag (68), gp41 (69–71), and gp120. The interaction between viral gp120 and human CD4 not only initiates HIV infection but is also an important and attractive target for anti-AIDS drug development. Crystal structures of HIV gp120-CD4 complexes reveal a close interaction of the virus receptor with CD4 Phe43, which was embedded in a pocket of gp120 (72). Molecular docking is used widely in anti-AIDS drug studies targeting gp120-CD4 interactions (72–77). The high mutation rate of HIV leads to the emergence of drug-resistant strains. Computational methods play an important role in modern anti-HIV drug development. To characterize the step of virus mutation, virtual screening and QSAR significantly reduce the time needed for drug discovery. Considering the enormous number of currently available compounds from sources, such as plants, marine organisms, and bacteria, computational methods are clearly promising and low cost.
Acknowledgements
This work was partially supported by the National Natural Science Foundation of China (81360503), the United Foundation of Guizhou (Qiankehe J LKZ[2013]21), the Incubation Project for 2011 Collaborative Innovation Center for Tuberculosis Prevention and Cure in Guizhou Province, and the seed grant from Zunyi Medical University for newly recruited talents to Dr. Wan-Gang Gu.
Contributor Information
Wan-Gang Gu, Phone: +86-15-085592339, Email: oscar458@gmail.com.
Xuan Zhang, Email: snoopykm@126.com.
Jun-Fa Yuan, Email: jfyuan@mail.hzau.edu.cn.
References
- 1.Brook I. Approval of zidovudine (AZT) for acquired immunodeficiency syndrome. A challenge to the medical and pharmaceutical communities. JAMA. 1987;258(11):1517. doi: 10.1001/jama.1987.03400110099035. [DOI] [PubMed] [Google Scholar]
- 2.Kirchmair J, Distinto S, Liedl KR, Markt P, Rollinger JM, Schuster D, et al. Development of anti-viral agents using molecular modeling and virtual screening techniques. Infect Disord Drug Targets. 2010. [DOI] [PubMed]
- 3.Lapkouski M, Tian L, Miller JT, Le Grice SF, Yang W. Complexes of HIV-1 RT, NNRTI and RNA/DNA hybrid reveal a structure compatible with RNA degradation. Nat Struct Mol Biol. 2013;20(2):230–6. doi: 10.1038/nsmb.2485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Miceli LA, Teixeira VL, Castro HC, Rodrigues CR, Mello JF, Albuquerque MG, et al. Molecular docking studies of marine diterpenes as inhibitors of wild-type and mutants HIV-1 reverse transcriptase. Mar Drugs. 2013;11(11):4127–43. doi: 10.3390/md11114127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Fraczek T, Siwek A, Paneth P. Assessing molecular docking tools for relative biological activity prediction: a case study of triazole HIV-1 NNRTIs. J Chem Inf Model. 2013. doi: 10.1021/ci400427a. [DOI] [PubMed]
- 6.Seniya C, Yadav A, Uchadia K, Kumar S, Sagar N, Shrivastava P, et al. Molecular docking of (5E)-3-(2-aminoethyl)-5-(2-thienylmethylene)-1,3-thiazolidine-2,4-dione on HIV-1 reverse transcriptase: novel drug acting on enzyme. Bioinformation. 2012;8(14):678–83. doi: 10.6026/97320630008678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mokale SN, Lokwani D, Shinde DB. Synthesis, biological activity and docking study of imidazol-5-one as novel non-nucleoside HIV-1 reverse transcriptase inhibitors. Bioorg Med Chem. 2012;20(9):3119–27. doi: 10.1016/j.bmc.2012.02.037. [DOI] [PubMed] [Google Scholar]
- 8.Ma XD, He QQ, Zhang X, Yang SQ, Yang LM, Gu SX, et al. Synthesis, structure-activity relationships, and docking studies of N-phenylarylformamide derivatives (PAFAs) as non-nucleoside HIV reverse transcriptase inhibitors. Eur J Med Chem. 2012;58:504–12. doi: 10.1016/j.ejmech.2012.03.032. [DOI] [PubMed] [Google Scholar]
- 9.Jain SV, Ghate M, Bhadoriya KS, Bari SB, Chaudhari A, Borse JS. 2D, 3D-QSAR and docking studies of 1,2,3-thiadiazole thioacetanilides analogues as potent HIV-1 non-nucleoside reverse transcriptase inhibitors. Org Med Chem Lett. 2012;2(1):22. doi: 10.1186/2191-2858-2-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Seal A, Aykkal R, Babu RO, Ghosh M. Docking study of HIV-1 reverse transcriptase with phytochemicals. Bioinformation. 2011;5(10):430–9. doi: 10.6026/97320630005430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mao Y, Li Y, Hao M, Zhang S, Ai C. Docking, molecular dynamics and quantitative structure-activity relationship studies for HEPTs and DABOs as HIV-1 reverse transcriptase inhibitors. J Mol Model. 2011;18(5):2185–98. doi: 10.1007/s00894-011-1236-8. [DOI] [PubMed] [Google Scholar]
- 12.Latha RS, Vijayaraj R, Singam ER, Chitra K, Subramanian V. 3D-QSAR and docking studies on the HEPT derivatives of HIV-1 reverse transcriptase. Chem Biol Drug Des. 2011;78(3):418–26. doi: 10.1111/j.1747-0285.2011.01162.x. [DOI] [PubMed] [Google Scholar]
- 13.Huang M, Grant GH, Richards WG. Binding modes of diketo-acid inhibitors of HIV-1 integrase: a comparative molecular dynamics simulation study. J Mol Graph Model. 2011;29(7):956–64. doi: 10.1016/j.jmgm.2011.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Garcia-Sosa AT, Sild S, Takkis K, Maran U. Combined approach using ligand efficiency, cross-docking, and antitarget hits for wild-type and drug-resistant Y181C HIV-1 reverse transcriptase. J Chem Inf Model. 2011;51(10):2595–611. doi: 10.1021/ci200203h. [DOI] [PubMed] [Google Scholar]
- 15.Cichero E, Fossa P. Docking-based 3D-QSAR analyses of pyrazole derivatives as HIV-1 non-nucleoside reverse transcriptase inhibitors. J Mol Model. 2011;18(4):1573–82. doi: 10.1007/s00894-011-1190-5. [DOI] [PubMed] [Google Scholar]
- 16.Bollini M, Domaoal RA, Thakur VV, Gallardo-Macias R, Spasov KA, Anderson KS, et al. Computationally-guided optimization of a docking hit to yield catechol diethers as potent anti-HIV agents. J Med Chem. 2011;54(24):8582–91. doi: 10.1021/jm201134m. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Weber IT, Waltman MJ, Mustyakimov M, Blakeley MP, Keen DA, Ghosh AK, et al. Joint X-ray/neutron crystallographic study of HIV-1 protease with clinical inhibitor amprenavir: insights for drug design. J Med Chem. 2013;56(13):5631–5. doi: 10.1021/jm400684f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ul-Haq Z, Usmani S, Shamshad H, Mahmood U, Halim SA. A combined 3D-QSAR and docking studies for the in-silico prediction of HIV-protease inhibitors. Chem Cent J. 2013;7(1):88. doi: 10.1186/1752-153X-7-88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Srivastava HK, Choudhury C, Sastry GN. The efficacy of conceptual DFT descriptors and docking scores on the QSAR models of HIV protease inhibitors. Med Chem. 2012;8(5):811–25. doi: 10.2174/157340612802084351. [DOI] [PubMed] [Google Scholar]
- 20.Lu T, Chen Y, Li XY. An insight into the opening path to semi-open conformation of HIV-1 protease by molecular dynamics simulation. AIDS. 2010;24(8):1121–5. doi: 10.1097/QAD.0b013e3283383c4c. [DOI] [PubMed] [Google Scholar]
- 21.Savarino A. In-Silico docking of HIV-1 integrase inhibitors reveals a novel drug type acting on an enzyme/DNA reaction intermediate. Retrovirology. 2007;4:21. doi: 10.1186/1742-4690-4-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Gandhi RT. Raltegravir, a new HIV integrase inhibitor. AIDS Clin Care. 2007;19(12):105. [PubMed] [Google Scholar]
- 23.Evering TH, Markowitz M. Raltegravir (MK-0518): an integrase inhibitor for the treatment of HIV-1. Drugs Today (Barc) 2007;43(12):865–77. doi: 10.1358/dot.2007.43.12.1146063. [DOI] [PubMed] [Google Scholar]
- 24.Cahn P, Sued O. Raltegravir: a new antiretroviral class for salvage therapy. Lancet. 2007;369(9569):1235–6. doi: 10.1016/S0140-6736(07)60571-6. [DOI] [PubMed] [Google Scholar]
- 25.Reddy KK, Singh SK, Tripathi SK, Selvaraj C. Identification of potential HIV-1 integrase strand transfer inhibitors: in silico virtual screening and QM/MM docking studies. SAR QSAR Environ Res. 2013;24(7):581–95. doi: 10.1080/1062936X.2013.772919. [DOI] [PubMed] [Google Scholar]
- 26.Picton AC, Paximadis M, Tiemessen CT. Genetic variation within the gene encoding the HIV-1 CCR5 coreceptor in two South African populations. Infect Genet Evol. 2010;10(4):487–94. doi: 10.1016/j.meegid.2010.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Piller SC, Caly L, Jans DA. Nuclear import of the pre-integration complex (PIC): the Achilles heel of HIV? Curr Drug Targets. 2003;4(5):409–29. doi: 10.2174/1389450033490984. [DOI] [PubMed] [Google Scholar]
- 28.Llano M, Delgado S, Vanegas M, Poeschla EM. Lens epithelium-derived growth factor/p75 prevents proteasomal degradation of HIV-1 integrase. J Biol Chem. 2004;279(53):55570–7. doi: 10.1074/jbc.M408508200. [DOI] [PubMed] [Google Scholar]
- 29.Cherepanov P, Devroe E, Silver PA, Engelman A. Identification of an evolutionarily conserved domain in human lens epithelium-derived growth factor/transcriptional co-activator p75 (LEDGF/p75) that binds HIV-1 integrase. J Biol Chem. 2004;279(47):48883–92. doi: 10.1074/jbc.M406307200. [DOI] [PubMed] [Google Scholar]
- 30.Chen JC, Krucinski J, Miercke LJ, Finer-Moore JS, Tang AH, Leavitt AD, et al. Crystal structure of the HIV-1 integrase catalytic core and C-terminal domains: a model for viral DNA binding. Proc Natl Acad Sci U S A. 2000;97(15):8233–8. doi: 10.1073/pnas.150220297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hare S, Shun MC, Gupta SS, Valkov E, Engelman A, Cherepanov P. A novel co-crystal structure affords the design of gain-of-function lentiviral integrase mutants in the presence of modified PSIP1/LEDGF/p75. PLoS Pathog. 2009;5(1):e1000259. doi: 10.1371/journal.ppat.1000259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rhodes DI, Peat TS, Vandegraaff N, Jeevarajah D, Newman J, Martyn J, et al. Crystal structures of novel allosteric peptide inhibitors of HIV integrase identify new interactions at the LEDGF binding site. Chembiochem. 2011;12(15):2311–5. doi: 10.1002/cbic.201100350. [DOI] [PubMed] [Google Scholar]
- 33.Zhao Y, Li W, Zeng J, Liu G, Tang Y. Insights into the interactions between HIV-1 integrase and human LEDGF/p75 by molecular dynamics simulation and free energy calculation. Proteins. 2008;72(2):635–45. doi: 10.1002/prot.21955. [DOI] [PubMed] [Google Scholar]
- 34.Cherepanov P, Ambrosio AL, Rahman S, Ellenberger T, Engelman A. Structural basis for the recognition between HIV-1 integrase and transcriptional coactivator p75. Proc Natl Acad Sci U S A. 2005;102(48):17308–13. doi: 10.1073/pnas.0506924102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hu G, Li X, Sun X, Lu W, Liu G, Huang J, et al. Identification of old drugs as potential inhibitors of HIV-1 integrase-human LEDGF/p75 interaction via molecular docking. J Mol Model. 2012;18(12):4995–5003. doi: 10.1007/s00894-012-1494-0. [DOI] [PubMed] [Google Scholar]
- 36.Yu S, Wang P, Li Y, Liu Y, Zhao G. Docking-based CoMFA and CoMSIA study of azaindole carboxylic acid derivatives as promising HIV-1 integrase inhibitors. SAR QSAR Environ Res. 2013;24(10):819–39. doi: 10.1080/1062936X.2013.820792. [DOI] [PubMed] [Google Scholar]
- 37.Sharma H, Sanchez TW, Neamati N, Detorio M, Schinazi RF, Cheng X, et al. Synthesis, docking, and biological studies of phenanthrene beta-diketo acids as novel HIV-1 integrase inhibitors. Bioorg Med Chem Lett. 2013;23(22):6146–51. doi: 10.1016/j.bmcl.2013.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Xue W, Jin X, Ning L, Wang M, Liu H, Yao X. Exploring the molecular mechanism of cross-resistance to HIV-1 integrase strand transfer inhibitors by molecular dynamics simulation and residue interaction network analysis. J Chem Inf Model. 2012;53(1):210–22. doi: 10.1021/ci300541c. [DOI] [PubMed] [Google Scholar]
- 39.Hu G, Li X, Zhang X, Li Y, Ma L, Yang LM, et al. Discovery of inhibitors to block interactions of HIV-1 integrase with human LEDGF/p75 via structure-based virtual screening and bioassays. J Med Chem. 2012;55(22):10108–17. doi: 10.1021/jm301226a. [DOI] [PubMed] [Google Scholar]
- 40.Schols D, Este JA, Henson G, De Clercq E. Bicyclams, a class of potent anti-HIV agents, are targeted at the HIV coreceptor fusin/CXCR-4. Antivir Res. 1997;35(3):147–56. doi: 10.1016/S0166-3542(97)00025-9. [DOI] [PubMed] [Google Scholar]
- 41.Kanbara K, Fujii N, Nakashima H. [A study of anti-HIV compounds which interfere the virus entry via coreceptor CXCR4] Kansenshogaku Zasshi. 2000;74(3):237–44. doi: 10.11150/kansenshogakuzasshi1970.74.237. [DOI] [PubMed] [Google Scholar]
- 42.Tamamura H, Imai M, Ishihara T, Masuda M, Funakoshi H, Oyake H, et al. Pharmacophore identification of a chemokine receptor (CXCR4) antagonist, T22 ([Tyr(5,12), Lys7]-polyphemusin II), which specifically blocks T cell-line-tropic HIV-1 infection. Bioorg Med Chem. 1998;6(7):1033–41. doi: 10.1016/S0968-0896(98)00061-3. [DOI] [PubMed] [Google Scholar]
- 43.Murakami T, Zhang TY, Koyanagi Y, Tanaka Y, Kim J, Suzuki Y, et al. Inhibitory mechanism of the CXCR4 antagonist T22 against human immunodeficiency virus type 1 infection. J Virol. 1999;73(9):7489–96. doi: 10.1128/jvi.73.9.7489-7496.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Doranz BJ, Grovit-Ferbas K, Sharron MP, Mao SH, Goetz MB, Daar ES, et al. A small-molecule inhibitor directed against the chemokine receptor CXCR4 prevents its use as an HIV-1 coreceptor. J Exp Med. 1997;186(8):1395–400. doi: 10.1084/jem.186.8.1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhou N, Luo Z, Luo J, Hall JW, Huang Z. A novel peptide antagonist of CXCR4 derived from the N-terminus of viral chemokine vMIP-II. Biochemistry. 2000;39(13):3782–7. doi: 10.1021/bi992750v. [DOI] [PubMed] [Google Scholar]
- 46.Schols D. HIV coreceptor CXCR4 antagonists. Curr Opin HIV AIDS. 2006;1(5):361–6. doi: 10.1097/01.COH.0000239847.13205.ce. [DOI] [PubMed] [Google Scholar]
- 47.Singh S, Malik BK, Sharma DK. Targeting HIV-1 through molecular modeling and docking studies of CXCR4: leads for therapeutic development. Chem Biol Drug Des. 2007;69(3):191–203. doi: 10.1111/j.1747-0285.2007.00478.x. [DOI] [PubMed] [Google Scholar]
- 48.Perez-Nueno VI, Ritchie DW, Rabal O, Pascual R, Borrell JI, Teixido J. Comparison of ligand-based and receptor-based virtual screening of HIV entry inhibitors for the CXCR4 and CCR5 receptors using 3D ligand shape matching and ligand-receptor docking. J Chem Inf Model. 2008;48(3):509–33. doi: 10.1021/ci700415g. [DOI] [PubMed] [Google Scholar]
- 49.Wu B, Chien EY, Mol CD, Fenalti G, Liu W, Katritch V, et al. Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists. Science. 2010;330(6007):1066–71. doi: 10.1126/science.1194396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Planesas JM, Perez-Nueno VI, Borrell JI, Teixido J. Impact of the CXCR4 structure on docking-based virtual screening of HIV entry inhibitors. J Mol Graph Model. 2012;38:123–36. doi: 10.1016/j.jmgm.2012.06.010. [DOI] [PubMed] [Google Scholar]
- 51.Karaboga AS, Planesas JM, Petronin F, Teixido J, Souchet M, Perez-Nueno VI. Highly specific and sensitive pharmacophore model for identifying CXCR4 antagonists. Comparison with docking and shape-matching virtual screening performance. J Chem Inf Model. 2013;53(5):1043–56. doi: 10.1021/ci400037y. [DOI] [PubMed] [Google Scholar]
- 52.Feng Z, Dubyak GR, Lederman MM, Weinberg A. Cutting edge: human beta defensin 3—a novel antagonist of the HIV-1 coreceptor CXCR4. J Immunol. 2006;177(2):782–6. doi: 10.4049/jimmunol.177.2.782. [DOI] [PubMed] [Google Scholar]
- 53.Princen K, Hatse S, Vermeire K, Bridger GJ, Skerlj RT, De Clercq E, et al. The antiviral activity of the CXCR4 antagonist AMD3100 is independent of the cytokine-induced CXCR4/HIV coreceptor expression level. AIDS Res Hum Retrovir. 2003;19(12):1135–9. doi: 10.1089/088922203771881239. [DOI] [PubMed] [Google Scholar]
- 54.Dorner BG, Scheffold A, Rolph MS, Huser MB, Kaufmann SH, Radbruch A, et al. MIP-1alpha, MIP-1beta, RANTES, and ATAC/lymphotactin function together with IFN-gamma as type 1 cytokines. Proc Natl Acad Sci U S A. 2002;99(9):6181–6. doi: 10.1073/pnas.092141999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, et al. CC CKR5: a RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science. 1996;272(5270):1955–8. doi: 10.1126/science.272.5270.1955. [DOI] [PubMed] [Google Scholar]
- 56.Sax PE. FDA approval: maraviroc. AIDS Clin Care. 2007;19(9):75. [PubMed] [Google Scholar]
- 57.Yost R, Pasquale TR, Sahloff EG. Maraviroc: a coreceptor CCR5 antagonist for management of HIV infection. Am J Health Syst Pharm. 2009;66(8):715–26. doi: 10.2146/ajhp080206. [DOI] [PubMed] [Google Scholar]
- 58.Perry CM. Maraviroc: a review of its use in the management of CCR5-tropic HIV-1 infection. Drugs. 2010;70(9):1189–213. doi: 10.2165/11203940-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 59.Kummerle T, Lehmann C, Hartmann P, Wyen C, Fatkenheuer G. Vicriviroc: a CCR5 antagonist for treatment-experienced patients with HIV-1 infection. Expert Opin Investig Drugs. 2009;18(11):1773–85. doi: 10.1517/13543780903357478. [DOI] [PubMed] [Google Scholar]
- 60.Gatanaga H. [Integrase inhibitor, CCR5 antagonist, fusion inhibitor] Nihon Rinsho. 2010;68(3):520–4. [PubMed] [Google Scholar]
- 61.Arumugam K, Crouzy S, Chevigne A, Seguin-Devaux C, Schmit JC. Structure prediction of GPCRs using piecewise homologs and application to the human CCR5 chemokine receptor: validation through agonist and antagonist docking. J Biomol Struct Dyn. 2013. doi: 10.1080/07391102.2013.817952. [DOI] [PubMed]
- 62.Shahlaei M, Madadkar-Sobhani A, Mahnam K, Fassihi A, Saghaie L, Mansourian M. Homology modeling of human CCR5 and analysis of its binding properties through molecular docking and molecular dynamics simulation. Biochim Biophys Acta. 2010;1808(3):802–17. doi: 10.1016/j.bbamem.2010.12.004. [DOI] [PubMed] [Google Scholar]
- 63.Fano A, Ritchie DW, Carrieri A. Modeling the structural basis of human CCR5 chemokine receptor function: from homology model building and molecular dynamics validation to agonist and antagonist docking. J Chem Inf Model. 2006;46(3):1223–35. doi: 10.1021/ci050490k. [DOI] [PubMed] [Google Scholar]
- 64.Li G, Haney KM, Kellogg GE, Zhang Y. Comparative docking study of anibamine as the first natural product CCR5 antagonist in CCR5 homology models. J Chem Inf Model. 2009;49(1):120–32. doi: 10.1021/ci800356a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Gadhe CG, Kothandan G, Cho SJ. Computational modeling of human coreceptor CCR5 antagonist as a HIV-1 entry inhibitor: using an integrated homology modeling, docking, and membrane molecular dynamics simulation analysis approach. J Biomol Struct Dyn. 2012;31(11):1251–76. doi: 10.1080/07391102.2012.732342. [DOI] [PubMed] [Google Scholar]
- 66.von Bubnoff A. Researchers solve high-resolution structure of HIV co-receptor CCR5. IAVI Rep. 2013;17(3):15. [PubMed] [Google Scholar]
- 67.Tan Q, Zhu Y, Li J, Chen Z, Han GW, Kufareva I, et al. Structure of the CCR5 chemokine receptor-HIV entry inhibitor maraviroc complex. Science. 2013;341(6152):1387–90. doi: 10.1126/science.1241475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Cui M, Huang X, Luo X, Briggs JM, Ji R, Chen K, et al. Molecular docking and 3D-QSAR studies on gag peptide analogue inhibitors interacting with human cyclophilin A. J Med Chem. 2002;45(24):5249–59. doi: 10.1021/jm020082x. [DOI] [PubMed] [Google Scholar]
- 69.Chang DK, Hsu CS. Biophysical evidence of two docking sites of the carboxyl heptad repeat region within the amino heptad repeat region of gp41 of human immunodeficiency virus type 1. Antivir Res. 2007;74(1):51–8. doi: 10.1016/j.antiviral.2006.12.006. [DOI] [PubMed] [Google Scholar]
- 70.Dennison SM, Stewart SM, Stempel KC, Liao HX, Haynes BF, Alam SM. Stable docking of neutralizing human immunodeficiency virus type 1 gp41 membrane-proximal external region monoclonal antibodies 2F5 and 4E10 is dependent on the membrane immersion depth of their epitope regions. J Virol. 2009;83(19):10211–23. doi: 10.1128/JVI.00571-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Gochin M, Zhou G, Phillips AH. Paramagnetic relaxation assisted docking of a small indole compound in the HIV-1 gp41 hydrophobic pocket. ACS Chem Biol. 2010;6(3):267–74. doi: 10.1021/cb100368d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Chong Teoh T, Heidelberg T, Rizman-Idid M. Systematic protein-protein docking and molecular dynamics studies of HIV-1 gp120 and CD4: insights for new drug development. Daru. 2011;19(6):469–75. [PMC free article] [PubMed] [Google Scholar]
- 73.Kong R, Tan JJ, Ma XH, Chen WZ, Wang CX. Prediction of the binding mode between BMS-378806 and HIV-1 gp120 by docking and molecular dynamics simulation. Biochim Biophys Acta. 2006;1764(4):766–72. doi: 10.1016/j.bbapap.2005.12.017. [DOI] [PubMed] [Google Scholar]
- 74.Berchanski A, Lapidot A. Prediction of HIV-1 entry inhibitors neomycin-arginine conjugates interaction with the CD4-gp120 binding site by molecular modeling and multistep docking procedure. Biochim Biophys Acta. 2007;1768(9):2107–19. doi: 10.1016/j.bbamem.2007.04.017. [DOI] [PubMed] [Google Scholar]
- 75.Teixeira C, Serradji N, Maurel F, Barbault F. Docking and 3D-QSAR studies of BMS-806 analogs as HIV-1 gp120 entry inhibitors. Eur J Med Chem. 2009;44(9):3524–32. doi: 10.1016/j.ejmech.2009.03.028. [DOI] [PubMed] [Google Scholar]
- 76.Li L, Chen H, Zhao RN, Han JG. The investigations on HIV-1 gp120 bound with BMS-488043 by using docking and molecular dynamics simulations. J Mol Model. 2012;19(2):905–17. doi: 10.1007/s00894-012-1619-5. [DOI] [PubMed] [Google Scholar]
- 77.Sivan SK, Vangala R, Manga V. Molecular docking guided structure based design of symmetrical N, N'-disubstituted urea/thiourea as HIV-1 gp120–CD4 binding inhibitors. Bioorg Med Chem. 2013;21(15):4591–9. doi: 10.1016/j.bmc.2013.05.038. [DOI] [PubMed] [Google Scholar]
