75 research outputs found

    Molecular Basis for Drug Resistance in HIV-1 Protease

    Get PDF
    HIV-1 protease is one of the major antiviral targets in the treatment of patients infected with HIV-1. The nine FDA approved HIV-1 protease inhibitors were developed with extensive use of structure-based drug design, thus the atomic details of how the inhibitors bind are well characterized. From this structural understanding the molecular basis for drug resistance in HIV-1 protease can be elucidated. Selected mutations in response to therapy and diversity between clades in HIV-1 protease have altered the shape of the active site, potentially altered the dynamics and even altered the sequence of the cleavage sites in the Gag polyprotein. All of these interdependent changes act in synergy to confer drug resistance while simultaneously maintaining the fitness of the virus. New strategies, such as incorporation of the substrate envelope constraint to design robust inhibitors that incorporate details of HIV-1 protease’s function and decrease the probability of drug resistance, are necessary to continue to effectively target this key protein in HIV-1 life cycle

    Energetic and Dynamic Analysis of Inhibitor Binding to Drug-Resistant HIV-1 Proteases: A Dissertation

    Get PDF
    HIV-1 protease is a very important drug target for AIDS therapy. Nine protease inhibitors have been proved by FDA and used in AIDS treatment. Due to the high replication rate and the lack of fidelity of the HIV-1 reverse transcriptase, HIV-1 virus developed various drug-resistant variants. Although experimental methods such as crystallography and isothermal titration calorimetry provide structural and thermodynamic data on drug-resistant variants, they are unable to discern the mechanism by which the mutations confer resistance to inhibitors. Understanding the drug-resistance mechanism is crucial for developing new inhibitors more tolerant to the drug-resistant mutations. Computational methods such as free energy calculations and molecular dynamic simulations can provide insights to the drug resistance mechanism at an atomic level. In this thesis, I have focused on the elucidation of the energetic and dynamics of key drug-resistant variants of HIV-1 protease. Two multi-drug resistant variants, in comparison with wild-type HIV-1 protease were used for the comparisons: Flap+ (L10I, G48V, I54V, and V82A) which contains a combination of flap and active site mutations and ACT (V82T, I84V) that only contains active site mutations. In Chapter II, I applied free energy simulations and decomposition methods to study the differential mechanism of resistance to the two variants, Flap+ and ACT, to the recently FDA-approved protease inhibitor darunavir (DRV). In this study, the absolute and relative binding free energies of DRV with wild-type protease and the two protease variants were calculated with MM-PB/GBSA and thermodynamic integration methods, respectively. And the predicted results are in good agreement with the ITC experimental results. Free energy decomposition elucidates the mutations alter not only its own interaction with DRV but also other residues by changing the geometry of binding pocket. And the VdW interactions between the bis-THF group of DRV is predominant even in the drug-resistant variants. At the end of this chapter, I offer suggestions on developing new inhibitors that are based on DRV but might be less susceptible to drug-resistant mutations. In Chapter III, 20-ns MD simulations of the apo wildtype protease and the apo drug-resistant protease variant Flap+ are analyzed and compared. In these studies, these mutations have been found to decrease the protease flexibility in the apo form but increase the mobility when the protease is binding with inhibitor. In Chapter IV, more details of the free energy simulation and decomposition are discussed. NMR relaxation experiments were set up as a control for the MD simulation study of the dynamics of the Flap+ variant. The difficulty of finishing the NMR experiment is discussed and the solution and some preliminary results are shown. In summary, the scope of this thesis was to use computational methods to study drug-resistant protease variants’ thermodynamic and dynamic properties to illuminate the mechanism of protease drug resistance. This knowledge will contribute to rational design of new protease inhibitors which bind more tightly to the protease and hinder the development of drug-resistant mutations

    Computational ligand design and analysis in protein complexes using inverse methods, combinatorial search, and accurate solvation modeling

    Get PDF
    Thesis (Ph. D.)--Massachusetts Institute of Technology, Dept. of Chemistry, 2006.Vita.Includes bibliographical references (p. 207-230).This thesis presents the development and application of several computational techniques to aid in the design and analysis of small molecules and peptides that bind to protein targets. First, an inverse small-molecule design algorithm is presented that can explore the space of ligands compatible with binding to a target protein using fast combinatorial search methods. The inverse design method was applied to design inhibitors of HIV-1 protease that should be less likely to induce resistance mutations because they fit inside a consensus substrate envelope. Fifteen designed inhibitors were chemically synthesized, and four of the tightest binding compounds to the wild-type protease exhibited broad specificity against a panel of drug resistance mutant proteases in experimental tests. Inverse protein design methods and charge optimization were also applied to improve the binding affinity of a substrate peptide for an inactivated mutant of HIV-1 protease, in an effort to learn more about the thermodynamics and mechanisms of peptide binding. A single mutant peptide calculated to have improved binding electrostatics exhibited greater than 10-fold improved affinity experimentally.(cont.) The second half of this thesis presents an accurate method for evaluating the electrostatic component of solvation and binding in molecular systems, based on curved boundary-element method solutions of the linearized Poisson-Boltzmann equation. Using the presented FFTSVD matrix compression algorithm and other techniques, a full linearized Poisson-Boltzmann equation solver is described that is capable of solving multi-region problems in molecular continuum electrostatics to high precision.Michael Darren Altman.Ph.D

    Crystallographic Analysis and Molecular Modeling Studies of HIV-1 Protease and Drug Resistant Mutants

    Get PDF
    HIV-1 protease (PR) is an effective target protein for drugs in anti-retroviral therapy (ART). Using PR inhibitors (PIs) in clinical therapy successfully reduces mortality of HIV infected patients. However, drug resistant variants are selected in AIDS patients because of the fast evolution of the viral genome. Structural, kinetic and MD simulations of PR variants with or without substrate or PIs were used to better understand the molecular basis of drug resistance. Information obtained from these extensive studies will benefit the design of more effective inhibitor in ART. Amprenavir (APV) inhibition of PRWT, and single mutants of PRV32I, PRI50V, PRI54M, PRI54V, PRI84V and PRL90M were studied and X-ray crystal structures of PR variants complexes with APV were solved at resolutions of 1.02-1.85 Ă… to identify structural alterations. Crystal structures of PRWT, PRV32I and PRI47V were solved at resolutions of 1.20-1.40 Ă…. Reaction intermediates were captured in the substrate binding cavity, which represent three consecutive steps in the catalytic reaction of HIV PR. HIV-1 PR20 variant is a multi-drug resistant variant from a clinical isolate and it is of utility to investigate the mechanisms of resistance. The crystal structures of PR20 with inactivating mutation D25N have been determined at 1.45-1.75 Ă… resolution, and three distinct flap conformations, open, twisted and tucked, were observed. These studies help understand molecular basis of drug resistance and provide clues for design of inhibitors to combat multi-drug resistant PR. The evaluation of electrostatic force in MD simulations is the computationally intensive work, which is of order theta(N2) with integration of all atom pairs. AMMP invokes Amortized FMM in summation of electrostatic force, which reduced work load to theta(N). A hybrid, CPU and GPU, parallel implementation of Amortized FMM was developed and improves the elapsed time of MD simulation 20 fold faster than CPU based parallelization

    Exploring Molecular Mechanisms of Drug Resistance in HIV-1 Protease through Biochemical and Biophysical Studies: A Dissertation

    Get PDF
    The human immunodeficiency virus type-1 (HIV-1) is the leading cause of acquired immunodeficiency syndrome (AIDS) in the world. As there is no cure currently available to treat HIV-1 infections or AIDS, the major focus of drug development efforts has been to target viral replication in an effort to slow down the progression of the infection to AIDS. The aspartyl protease of HIV-1 is an important component in the viral replication cycle and thus, has been an important anti-HIV-1 drug target. Currently there are nine protease inhibitors (PIs) that are being used successfully as a part of highly active antiretroviral therapy (HAART). However, as is with all HIV-1 drug targets, the emergence of drug resistance substitutions within protease is a major obstacle in the use of PIs. Understanding how amino acid substitutions within protease confer drug resistance is key to develop new PIs that are not influenced by resistance mutations. Thus, the primary focus of my dissertation research was to understand the molecular basis for drug resistance caused by some of these resistance substitutions. Until recently, the genetic diversity of the HIV-1 genome was not considered to be important in formulating treatment strategies. However, as the prevalence of HIV-1 continues, the variability of the HIV-1 genome has now been identified as an important factor in how the virus spreads as well as how fast the infection progresses to AIDS. Clinical studies have also revealed that the pathway to protease inhibitor resistance can vary between HIV-1 clades. Therefore, in studying the molecular basis of drug resistance in HIV-1 protease, I have also attempted to understand how genetic variability in HIV-1 protease contributes to PI resistance. In Chapters II, III and Appendix 1, I have examined how clade specific amino acid variations within HIV-1 CRF01_AE and clade C protease affect enzyme structure and activity. Furthermore, I have examined how these sequence variations, which are predominantly outside the active site, contribute to inhibitor resistance in comparison to clade B protease. With the results presented in Chapter II, I was able to show that sequence variations within CRF01_AE protease resulted in structural changes within the protease that might influence enzyme activity. In Chapter III, I focused on how sequence variations in CRF01_AE influence protease activity and inhibitor binding in comparison to clade B protease. Enzyme kinetics data showed that the CRF01-AE had reduced catalytic turnover rates when compared to clade B protease. Binding data also indicated that CRF01_AE protease had an inherent weaker affinity for the PIs nelfinavir (NFV) and darunavir (DRV). In work described in Chapter III, I have also examined the different pathways to NFV resistance seen in CRF01_AE and clade B protease. Using x-ray crystallographic studies I have shown the molecular mechanism by which the two different pathways confer NFV resistance. Furthermore, I provide a rational for why different resistance pathways might emerge in the two clades. In Appendix I, I present results from a parallel study carried out on clade C protease. In Chapter IV, I have examined the role of residue 50 in HIV-1 protease in modulating inhibitor binding. Patients failing amprevavir (APV) and DRV therapy often develop the I50V substitution while the I50L substitution is often observed in patients failing atazanavir (ATV) therapy. This indicates that by making subtle changes at residue 50 the protease is able to confer differential PI resistance. With binding data presented in this chapter I have shown that substitutions at residue 50 change the susceptibility profiles of APV, DRV and ATV. Furthermore, from analyses of protease-inhibitor complexes, I have described structural insights into how substitutions at residue 50 can modulate inhibitor binding. This thesis presents results that reveal mechanistic insights into how a number of resistance substitutions within protease confer drug resistance. The results on non-B clade proteases demonstrate that clade specific sequence variations play a role in modulating enzyme activity and influence the pathway taken to confer PI resistance. Furthermore, the results provide structural insights into how amino acid substitutions outside the active site effectively alter inhibitor binding

    Interactive molecular dynamics in virtual reality for accurate flexible protein-ligand docking

    Get PDF
    Simulating drug binding and unbinding is a challenge, as the rugged energy landscapes that separate bound and unbound states require extensive sampling that consumes significant computational resources. Here, we describe the use of interactive molecular dynamics in virtual reality (iMD-VR) as an accurate low-cost strategy for flexible protein-ligand docking. We outline an experimental protocol which enables expert iMD-VR users to guide ligands into and out of the binding pockets of trypsin, neuraminidase, and HIV-1 protease, and recreate their respective crystallographic protein-ligand binding poses within 5 - 10 minutes. Following a brief training phase, our studies shown that iMD-VR novices were able to generate unbinding and rebinding pathways on similar timescales as iMD-VR experts, with the majority able to recover binding poses within 2.15 Angstrom RMSD of the crystallographic binding pose. These results indicate that iMD-VR affords sufficient control for users to carry out the detailed atomic manipulations required to dock flexible ligands into dynamic enzyme active sites and recover crystallographic poses, offering an interesting new approach for simulating drug docking and generating binding hypotheses.Comment: PLOS ON

    Kinetic and Crystallographic Studies of Drug-Resistant Mutants of HIV-1 Protease: Insights into the Drug Resistance Mechanisms

    Get PDF
    HIV-1 protease (PR) inhibitors (PIs) are important anti-HIV drugs for the treatment of AIDS and have shown great success in reducing mortality and prolonging the life of HIV-infected individuals. However, the rapid development of drug resistance is one of the major factors causing the reduced effectiveness of PIs. Consequently, various drug resistant mutants of HIV-1 PR have been extensively studied to gain insight into the mechanisms of drug resistance. In this study, the crystal structures, dimer stabilities, and kinetics data have been analyzed for wild type PR and over 10 resistant mutants including PRL24I, PRI32V, PRM46L, PRG48V, PRI50V, PRF53L, PRI54V, PRI54M, PRG73S and PRL90M. These mutations lie in varied structural regions of PR: adjacent to the active site, in the inhibitor binding site, the flap or at protein surface. The enzymatic activity and inhibition were altered in mutant PR to various degrees. Crystal structures of the mutants complexed with a substrate analog inhibitor or drugs indinavir, saquinavir and darunavir were determined at resolutions of 0.84 – 1.50 Å. Each mutant revealed distinct structural changes, which are usually located at the mutated residue, the flap and inhibitor binding sites. Moreover, darunavir was shown to bind to PR at a new site on the flap surface in PRI32V and PRM46L. The existence of this additional inhibitor binding site may explain the high effectiveness of darunavir on drug resistant mutants. Moreover, the unliganded structure PRF53L had a wider separation at the tips of the flaps than in unliganded wild type PR. The absence of flap interactions in PRF53L suggests a novel mechanism for drug resistance. Therefore, this study enhanced our understanding of the role of individual residues in the development of drug resistance and the structural basis of drug resistance mechanisms. Atomic resolution crystal structures are valuable for the design of more potent protease inhibitors to overcome the drug resistance problem

    Raltegravir, elvitegravir, and metoogravir: the birth of "me-too" HIV-1 integrase inhibitors

    Get PDF
    Merck's MK-0518, known as raltegravir, has recently become the first FDA-approved HIV-1 integrase (IN) inhibitor and has since risen to blockbuster drug status. Much research has in turn been conducted over the last few years aimed at recreating but optimizing the compound's interactions with the protein. Resulting me-too drugs have shown favorable pharmacokinetic properties and appear drug-like but, as expected, most have a highly similar interaction with IN to that of raltegravir. We propose that, based upon conclusions drawn from our docking studies illustrated herein, most of these me-too MK-0518 analogues may experience a low success rate against raltegravir-resistant HIV strains. As HIV has a very high mutational competence, the development of drugs with new mechanisms of inhibitory action and/or new active substituents may be a more successful route to take in the development of second- and third-generation IN inhibitors

    Computational studies of mutations associated to resistance in HIV-1 macromolecular targets and implications in rational design of novel antiviral agents

    Get PDF
    Al fine di identificare nuovi farmaci anti-HIV capaci di superare i problemi legati alla resistenza, è stato condotto uno studio teorico combinando l’analisi strutturale sui modelli cristallografici della trascrittasi inversa (RT), i dati clinici relativi ai residui conservati dell’RT ed un’innovativa metodica computazionale basata sulle mappe di GRID. Tale analisi ha permesso di riprodurre i risultati clinici e di evidenziare le conseguenze delle mutazioni nella fase di ricognizione. Inoltre l’approccio computazionale ha portato all’identificazione di un modello farmacoforico utile per la progettazione di nuovi inibitori dell’RT. E’ stato riscontrato che la presenza del polimorfismo I135T nei pazienti NNRTI-naïve correlasse in modo significativo con la mutazione K103N nei casi di fallimento agli NNRTI, suggerendo così che la sostituzione I135T rappresenti un punto cruciale per l’evoluzione della resistenza agli NNRTI. Le simulazioni di dinamica molecolare (MD) hanno mostrato che la mutazione I135T contribuisce alla stabilizzazione della chiusura della tasca di legame degli NNRTI indotta dalla K103N in seguito alla riduzione della distanza ed all’aumento del numero di legami idrogeno tra l’Asn103 e la Tyr188. Inoltre è stata valutata l’influenza di due mutazioni associate a resistenza, L33F e L76V, presenti a livello della proteasi (PR) di HIV-1 rispetto alla ricognizione molecolare del Lopinavir (LPV) e dell’Atazanavir (ATV). L’analisi delle energie di interazione ottenute in seguito alla MD ha rivelato che la mutazione L33F determina una riduzione delle interazioni tra il ligando ed il recettore, dell’affinità di legame e della stabilità del dimero per entrambi gli inibitori della PR. In presenza della mutazione L76V, il LPV ha mostrato una minore affinità di legame ed un ridotto network di legami idrogeno, mentre i complessi con l’ATV hanno rivelato una migliore affinità, un effetto stabilizzante a livello dell’interfaccia del dimero e più efficaci interazioni ligando-recettore, in accordo con i dati di ipersuscettibilità. Al fine di valutare la stabilità del 6-helix bundle, sono state studiate le proprietà conformazionali della glicoproteina gp41 in presenza delle mutazioni associate a resistenza all’enfuvirtide V38A ed N140I. Le simulazioni di MD hanno mostrato che la copresenza delle mutazioni V38A+N140I è in grado di abolire l’interazione stabilita tra i residui 38 e 145, che risulta fondamentale per la stabilizzazione del 6-helix bundle.In order to discover novel selective anti-HIV resistance-evading drugs, a theoretical study was carried out combining structural analysis of RT crystallographic models, clinical data about RT conserved residues and an innovative computational method based on GRID maps. Such analysis allowed to reproduce clinical results and to highlight the consequences of the mutations in the recognition step. Moreover the computational approach generated a pharmacophore model useful for the design of novel RT inhibitors. The presence of the I135T polymorphism in NNRTI-naive patients significantly correlated with the appearance of K103N in cases of NNRTI failure, suggesting that I135T may represent a crucial determinant of NNRTI resistance evolution. Molecular Dynamics simulations (MD) showed that I135T can contribute to the stabilization of the K103N-induced closure of the NNRTI binding pocket by reducing the distance and increasing the number of hydrogen bonds between 103N and 188Y. In addition the influence of two drug resistance-associated mutations, L33F and L76V, of HIV-1 PR has been evaluated with respect to lopinavir (LPV) and atazanavir (ATV) molecular recognition. The evaluation of the interaction energies after the MD revealed that L33F substitution is related to reduced host/guest interactions, decreased affinity and to a dimer destabilizing effect for both PR inhibitors. In presence of L76V mutation, LPV showed a lowered binding affinity and a reduced hydrogen bonding network, while ATV complexes revealed a more productive binding affinity, increased host/guest interactions and dimer stabilizing effects, in agreement with hyper susceptibility data. With the aim to estimate the stability of its 6-helix bundle, the gp41 conformational properties were investigated in presence of V38A and N140I, known enfuvirtide resistance-associated mutations. MD showed that the co-presence of V38A+N140I abolished the interaction between residue 38 and 145 important for the 6-helix-bundle stabilization
    • …
    corecore