Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biomolecules ; 12(5)2022 05 12.
Article in English | MEDLINE | ID: mdl-35625621

ABSTRACT

The HIV-1 capsid (CA) protein has emerged as an attractive therapeutic target. However, all inhibitor designs and structural analyses for this essential HIV-1 protein have focused on the clade B HIV-1 (NL4-3) variant. This study creates, overproduces, purifies, and characterizes the CA proteins from clade A1, A2, B, C, and D isolates. These new CA constructs represent novel reagents that can be used in future CA-targeted inhibitor design and to investigate CA proteins' structural and biochemical properties from genetically diverse HIV-1 subtypes. Moreover, we used surface plasmon resonance (SPR) spectrometry and computational modeling to examine inter-clade differences in CA assembly and binding of PF-74, CPSF-6, and NUP-153. Interestingly, we found that HIV-1 CA from clade A1 does not bind to NUP-153, suggesting that the import of CA core structures through the nuclear pore complex may be altered for viruses from this clade. Overall, we have demonstrated that in silico generated models of the HIV-1 CA protein from clades other than the prototypically used clade B have utility in understanding and predicting biology and antiviral drug design and mechanism of action.


Subject(s)
HIV-1 , Antiviral Agents , Capsid Proteins/chemistry , HIV-1/genetics , HIV-1/metabolism
2.
Eur J Med Chem ; 227: 113903, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34653770

ABSTRACT

Further clinical development of PF74, a lead compound targeting HIV-1 capsid, is impeded by low antiviral activity and inferior metabolic stability. By modifying the benzene (region I) and indole of PF74, we identified two potent compounds (7m and 7u) with significantly improved metabolic stability. Compared to PF74, 7u displayed greater metabolic stability in human liver microsomes (HLMs) with half-life (t1/2) 109-fold that of PF74. Moreover, mechanism of action (MOA) studies demonstrated that 7m and 7u effectively mirrored the MOA of compounds that interact within the PF74 interprotomer pocket, showing direct and robust interactions with recombinant CA, and 7u displaying antiviral effects in both the early and late stages of HIV-1 replication. Furthermore, MD simulation corroborated that 7u was bound to the PF74 binding site, and the results of the online molinspiration software predicted that 7m and 7u had desirable physicochemical properties. Unexpectedly, this series of compounds exhibited better antiviral activity than PF74 against HIV-2, represented by compound 7m whose anti-HIV-2 activity was almost 5 times increased potency over PF74. Therefore, we have rationally redesigned the PF74 chemotype to inhibitors with novel structures and enhanced metabolic stability in this study. We hope that these new compounds can serve as a blueprint for developing a new generation of HIV treatment regimens.


Subject(s)
Anti-HIV Agents/pharmacology , Benzothiazoles/pharmacology , Capsid Proteins/antagonists & inhibitors , Drug Design , HIV-1/drug effects , Phenylalanine/pharmacology , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Benzothiazoles/chemistry , Benzothiazoles/metabolism , Capsid Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Phenylalanine/chemistry , Phenylalanine/metabolism , Structure-Activity Relationship , Virus Replication/drug effects
3.
Eur J Med Chem ; 226: 113848, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34592608

ABSTRACT

HIV-1 capsid (CA) plays indispensable and multiple roles in the life cycle of HIV-1, become an attractive target in antiviral therapy. Herein, we report the design, synthesis, and mechanism study of a novel series of dimerized phenylalanine derivatives as HIV-1 capsid inhibitors using 2-piperazineone or 2,5-piperazinedione as a linker. The structure-activity relationship (SAR) indicated that dimerized phenylalanines were more potent than monomers of the same chemotype. Further, the inclusion of fluorine substituted phenylalanine and methoxyl substituted aniline was found to be beneficial for antiviral activity. From the synthesized series, Q-c4 was found to be the most potent compound with an EC50 value of 0.57 µM, comparable to PF74. Interestingly, Q-c4 demonstrated a slightly higher affinity to the CA monomer than the CA hexamer, commensurate with its more significant effect in the late-stage of the HIV-1 lifecycle. Competitive SPR experiments with peptides from CPSF6 and NUP153 revealed that Q-c4 binds to the interprotomer pocket of hexameric CA as designed. Single-round infection assays showed that Q-c4 interferes with the HIV-1 life cycle in a dual-stage manner, affecting both pre-and post-integration. Stability assays in human plasma and human liver microsomes indicated that although Q-c4 has improved stability over PF74, this kind of inhibitor still requires further optimization. And the results of the online molinspiration software predicted that Q-c4 has desirable physicochemical properties but some properties still have some violation from the Lipinski rule of five. Overall, the dimerized phenylalanines are promising novel platforms for developing future HIV-1 CA inhibitors with considerable potential for optimization.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , Drug Design , HIV-1/drug effects , Phenylalanine/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Capsid Proteins/metabolism , Dimerization , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Molecular Structure , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Structure-Activity Relationship , Surface Plasmon Resonance
4.
Protein Sci ; 30(6): 1235-1246, 2021 06.
Article in English | MEDLINE | ID: mdl-33896065

ABSTRACT

Maltose binding protein (MBP) is used in recombinant protein expression as an affinity and solubility tag. The monoclonal antibody B48 binds MBP tightly and has no cross-reactivity to other proteins in an Escherichia coli lysate. This high level of specificity suggested that MBP contains an epitope that could prove useful as a purification and visualization tag for proteins expressed in E. coli. To discover the MBP epitope, a co-crystal structure was determined for MBP bound to its antibody and four amino acids of MBP were identified as critical for the binding interaction. Fusions of various fragments of MBP to the glutathione S-transferase protein were engineered in order to identify the smallest fragment still recognized by the α-MBP antibody. Stabilization of the epitope via mutational engineering resulted in a minimized 14 amino-acid tag.


Subject(s)
Epitopes/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Maltose-Binding Proteins/chemistry , Crystallography, X-Ray , Epitopes/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Maltose-Binding Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
5.
J Med Chem ; 64(7): 3747-3766, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33750123

ABSTRACT

Poor metabolic stability of the human immunodeficiency virus type-1 (HIV-1) capsid (CA) inhibitor PF-74 is a major concern in its development toward clinical use. To improve on the metabolic stability, we employed a novel multistep computationally driven workflow, which facilitated the rapid design of improved PF-74 analogs in an efficient manner. Using this workflow, we designed three compounds that interact specifically with the CA interprotomer pocket, inhibit HIV-1 infection, and demonstrate enantiomeric preference. Moreover, using this workflow, we were able to increase the metabolic stability 204-fold in comparison to PF-74 in only three analog steps. These results demonstrate our ability to rapidly design CA compounds using a novel computational workflow that has improved metabolic stability over the parental compound. This workflow can be further applied to the redesign of PF-74 and other promising inhibitors with a stability shortfall.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , HIV-1/drug effects , Indoles/pharmacology , Amino Acid Sequence , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Binding Sites , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Indoles/chemistry , Indoles/metabolism , Microbial Sensitivity Tests , Microsomes, Liver/metabolism , Molecular Docking Simulation , Protein Binding , Protein Stability , Stereoisomerism , Workflow
6.
Life (Basel) ; 11(2)2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33572761

ABSTRACT

The capsid (CA) protein of the human immunodeficiency virus type 1 (HIV-1) is an essential structural component of a virion and facilitates many crucial life cycle steps through interactions with host cell factors. Capsid shields the reverse transcription complex from restriction factors while it enables trafficking to the nucleus by hijacking various adaptor proteins, such as FEZ1 and BICD2. In addition, the capsid facilitates the import and localization of the viral complex in the nucleus through interaction with NUP153, NUP358, TNPO3, and CPSF-6. In the later stages of the HIV-1 life cycle, CA plays an essential role in the maturation step as a constituent of the Gag polyprotein. In the final phase of maturation, Gag is cleaved, and CA is released, allowing for the assembly of CA into a fullerene cone, known as the capsid core. The fullerene cone consists of ~250 CA hexamers and 12 CA pentamers and encloses the viral genome and other essential viral proteins for the next round of infection. As research continues to elucidate the role of CA in the HIV-1 life cycle and the importance of the capsid protein becomes more apparent, CA displays potential as a therapeutic target for the development of HIV-1 inhibitors.

7.
J Med Chem ; 63(9): 4790-4810, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32298111

ABSTRACT

The HIV-1 CA protein has gained remarkable attention as a promising therapeutic target for the development of new antivirals, due to its pivotal roles in HIV-1 replication (structural and regulatory). Herein, we report the design and synthesis of three series of benzenesulfonamide-containing phenylalanine derivatives obtained by further structural modifications of PF-74 to aid in the discovery of more potent and drug-like HIV-1 CA inhibitors. Structure-activity relationship studies of these compounds led to the identification of new phenylalanine derivatives with a piperazinone moiety, represented by compound 11l, which exhibited anti-HIV-1NL4-3 activity 5.78-fold better than PF-74. Interestingly, 11l also showed anti-HIV-2ROD activity (EC50 = 31 nM), with almost 120 times increased potency over PF-74. However, due to the higher significance of HIV-1 as compared to HIV-2 for the human population, this manuscript focuses on the mechanism of action of our compounds in the context of HIV-1. SPR studies on representative compounds confirmed CA as the binding target. The action stage determination assay demonstrated that these inhibitors exhibited antiviral activities with a dual-stage inhibition profile. The early-stage inhibitory activity of compound 11l was 6.25 times more potent as compared to PF-74 but appeared to work via the accelerating capsid core assembly rather than stabilization. However, the mechanism by which they exert their antiviral activity in the late stage appears to be the same as PF-74 with less infectious HIV-1 virions produced in their presence, as judged p24 content studies. MD simulations provided the key rationale for the promising antiviral potency of 11l. Additionally, 11l exhibited a modest increase in HLM and human plasma metabolic stabilities as compared to PF-74, as well as a moderately improved pharmacokinetic profile, favorable oral bioavailability, and no acute toxicity. These studies provide insights and serve as a starting point for subsequent medicinal chemistry efforts in optimizing these promising HIV inhibitors.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , HIV-1/drug effects , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Sulfonamides/pharmacology , Animals , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/toxicity , Cell Line, Tumor , Drug Design , Female , HIV-1/chemistry , HIV-2/chemistry , HIV-2/drug effects , Humans , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Phenylalanine/pharmacokinetics , Phenylalanine/toxicity , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity , Virus Replication/drug effects
8.
Molecules ; 25(6)2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32245167

ABSTRACT

Fostemsavir/temsavir is an investigational HIV-1 entry inhibitor currently in late-stage clinical trials. Although it holds promise to be a first-in-class Env-targeted entry inhibitor for the clinic, issues with bioavailability relegate its use to salvage therapies only. As such, the development of a small molecule HIV-1 entry inhibitor that can be used in standard combination antiretroviral therapy (cART) remains a longstanding goal for the field. We previously demonstrated the ability of extending the chemotypes available to this class of inhibitor as the first step towards this overarching goal. In addition to poor solubility, metabolic stability is a crucial determinant of bioavailability. Therefore, in this short communication, we assess the metabolic stabilities of five of our novel chemotype entry inhibitors. We found that changing the piperazine core region of temsavir alters the stability of the compound in human liver microsome assays. Moreover, we identified an entry inhibitor with more than twice the metabolic stability of temsavir and demonstrated that the orientation of the core replacement is critical for this increase. This work further demonstrates the feasibility of our long-term goal-to design an entry inhibitor with improved drug-like qualities-and warrants expanded studies to achieve this.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Organophosphates/chemistry , Piperazines/chemistry , Triazoles/metabolism , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/chemistry , Azabicyclo Compounds/pharmacology , Azetidines/chemical synthesis , Azetidines/chemistry , Azetidines/pharmacology , Chromatography, Liquid , HEK293 Cells , HIV Core Protein p24/chemistry , HIV Core Protein p24/metabolism , HIV Fusion Inhibitors/metabolism , HIV Infections/virology , Humans , Microsomes, Liver/virology , Organophosphates/pharmacology , Piperazines/pharmacology , Protein Binding , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , Tandem Mass Spectrometry , Triazoles/chemical synthesis , Triazoles/pharmacology , Triazoles/toxicity
9.
Eur J Med Chem ; 190: 112085, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-32066010

ABSTRACT

HIV-1 CA is involved in different stages of the viral replication cycle, performing essential roles in both early (uncoating, reverse transcription, nuclear import, integration) and late events (assembly). Recent efforts have demonstrated HIV-1 CA protein as a prospective therapeutic target for the development of new antivirals. The most extensively studied CA inhibitor, PF-3450074 (PF-74, discovered by Pfizer), that targets an inter-protomer pocket within the CA hexamer. Herein we reported the design, synthesis, and biological evaluation of a series of 4-phenyl-1H-1,2,3-triazole phenylalanine derivatives as HIV-1 CA inhibitors based on PF-74 scaffold. Most of the analogues demonstrated potent antiviral activities, among them, the anti-HIV-1 activity of 6a-9 (EC50 = 3.13 µM) is particularly prominent. The SPR binding assay of selected compounds (6a-9, 6a-10, 5b) suggested direct and effective interaction with recombinant CA proteins. The mechanism of action studies also demonstrated that 6a-9 displays the effects in both the early and late stages of HIV-1 replication. To explore the potential binding mode of the here presented analogues, 6a-9 was analyzed by MD simulation to predict its binding to the active site of HIV-1 CA monomer. In conclusion, this novel series of antivirals can serve as a starting point for the development of a new generation of HIV-1 treatment regimen and highlights the potentiality of CA as a therapeutic target.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , HIV-1/chemistry , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Triazoles/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Cell Line, Tumor , Drug Design , Humans , Microbial Sensitivity Tests , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Molecular Structure , Phenylalanine/chemical synthesis , Phenylalanine/metabolism , Protein Binding , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/metabolism , Virus Replication/drug effects
10.
Molecules ; 24(8)2019 Apr 22.
Article in English | MEDLINE | ID: mdl-31013646

ABSTRACT

Small-molecule HIV-1 entry inhibitors are an extremely attractive therapeutic modality. We have previously demonstrated that the entry inhibitor class can be optimized by using computational means to identify and extend the chemotypes available. Here we demonstrate unique and differential effects of previously published antiviral compounds on the gross structure of the HIV-1 Env complex, with an azabicyclohexane scaffolded inhibitor having a positive effect on glycoprotein thermostability. We demonstrate that modification of the methyltriazole-azaindole headgroup of these entry inhibitors directly effects the potency of the compounds, and substitution of the methyltriazole with an amine-oxadiazole increases the affinity of the compound 1000-fold over parental by improving the on-rate kinetic parameter. These findings support the continuing exploration of compounds that shift the conformational equilibrium of HIV-1 Env as a novel strategy to improve future inhibitor and vaccine design efforts.


Subject(s)
Anti-HIV Agents , HIV Envelope Protein gp120/metabolism , HIV-1/metabolism , Virus Internalization/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HEK293 Cells , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/pathology , Humans , Molecular Docking Simulation , Structure-Activity Relationship
11.
RSC Adv ; 9(50): 28961-28986, 2019.
Article in English | MEDLINE | ID: mdl-32089839

ABSTRACT

The HIV-1 capsid (CA) protein plays crucial roles in both early and late stages of the viral life cycle, which has intrigued researchers to target it to develop anti-HIV drugs. Accordingly, in this research, we report the design, synthesis and biological evaluation of a series of novel phenylalanine derivatives as HIV-1 CA protein inhibitors using the Cu(I)-catalyzed azide and alkyne 1,3-dipolar cycloaddition (CuAAC) reaction. Among this series of inhibitors, compound II-10c displayed a remarkable anti-HIV activity (EC50 = 2.13 µM, CC50 > 35.49 µM). Furthermore, surface plasmon resonance (SPR) binding assays showed that compounds II-10c and PF-74 (lead compound) have similar affinities to HIV-1 CA monomer. Further investigation showed that the weak permeability and water solubility of representative compounds were probably the important factors that restricted their cell-based activity. Preliminary structure-activity relationships (SARs) were inferred based on the activities of these compounds, and their known structure. The most promising new compound was studied with molecular dynamics simulation (MD) to determine the preferred interactions with the drug target. Finally, the activities of members of this series of inhibitors were deeply inspected to find the potential reasons for their anti-HIV-1 activity from various perspectives. This highlights the important factors required to design compounds with improved potency.

12.
J Drug Des Res ; 5(2)2018.
Article in English | MEDLINE | ID: mdl-30393786

ABSTRACT

Recent efforts by both academic and pharmaceutical researchers have focused on the HIV-1 capsid (CA) protein as a new therapeutic target. An interprotomer pocket within the hexamer configuration of the CA, which is also a binding site for key host dependency factors, is the target of the most widely studied CA inhibitor compound PF-3450074 (PF-74). Despite its popularity, PF-74 suffers from properties that limit its usefulness as a lead, most notably it's extremely poor metabolic stability. To minimize unfavorable qualities, we investigated bioisosteric modification of the PF-74 scaffold as a first step in redeveloping this compound. Using a field-based bioisostere identification method, coupled with biochemical and biological assessment, we have created four new compounds that inhibit HIV-1 infection and that bind to the assembled CA hexamer. Detailed mechanism of action studies indicates that the modifications alter the manner in which these new compounds affect HIV-1 capsid core stability, as compared to the parental compound. Further investigations are underway to redevelop these compounds to optimize potency and drug-like characteristics and to deeply define the mechanism of action.

13.
Eur J Med Chem ; 158: 478-492, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30243152

ABSTRACT

The HIV-1 capsid (CA) protein plays essential roles in both early and late stages of HIV-1 replication and is considered an important, clinically unexploited therapeutic target. As such, small drug-like molecules that inhibit this critical HIV-1 protein have become a priority for several groups. Therefore, in this study we explore small molecule targeting of the CA protein, and in particular a very attractive inter-protomer pocket. We report the design, parallel synthesis, and anti-HIV-1 activity evaluation of a series of novel phenylalanine derivatives as HIV-1 CA protein inhibitors synthesized via Cu(I)-catalyzed alkyne-azide 1,3-dipolar cycloaddition (CuAAC) reaction. We demonstrate robust inhibitory activity over a range of potencies against the HIV-1 NL4-3 reference strain. In particular, compound 13m exhibited the greatest potency and lowest toxicity within this new series with an EC50 value of 4.33 µM and CC50 value of >57.74 µM (SI > 13.33). These values are very similar to the lead compound PF-74 (EC50 = 5.95 µM, CC50 > 70.50 µM, SI > 11.85) in our assay, despite significant structural difference. Furthermore, we demonstrate via surface plasmon resonance (SPR) binding assays that 13m interacts robustly with recombinant HIV-1 CA and exhibits antiviral activity in both the early and late stages of HIV-1 replication. Overall, the novel parallel synthesis and structure-activity relationships (SARs) identified within this study set the foundation for further rational optimization and discovery of CA-targeting compounds with improved potency.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Capsid/drug effects , HIV-1/drug effects , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Anti-HIV Agents/chemical synthesis , Click Chemistry , HIV Infections/drug therapy , HIV Infections/virology , Humans , Molecular Dynamics Simulation , Phenylalanine/chemical synthesis , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
14.
Molecules ; 23(8)2018 Aug 03.
Article in English | MEDLINE | ID: mdl-30081466

ABSTRACT

The entry of HIV-1 into permissible cells remains an extremely attractive and underexploited therapeutic intervention point. We have previously demonstrated the ability to extend the chemotypes available for optimization in the entry inhibitor class using computational means. Here, we continue this effort, designing and testing three novel compounds with the ability to inhibit HIV-1 entry. We demonstrate that alteration of the core moiety of these entry inhibitors directly influences the potency of the compounds, despite common proximal and distal groups. Moreover, by establishing for the first time a surface plasmon resonance (SPR)-based interaction assay with soluble recombinant SOSIP Env trimers, we demonstrate that the off-rate (kd) parameter shows the strongest correlation with potency in an antiviral assay. Finally, we establish an underappreciated relationship between the potency of a ligand and its degree of electrostatic complementarity (EC) with its target, the Env complex. These findings not only broaden the chemical space in this inhibitor class, but also establish a rapid and simple assay to evaluate future HIV-1 entry inhibitors.


Subject(s)
Anti-HIV Agents/chemistry , HIV-1/drug effects , Piperazines/chemistry , Pyrroles/chemistry , Triazoles/chemistry , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , Cell Line , Cell Survival/drug effects , Drug Design , HIV Envelope Protein gp120/metabolism , HIV-1/physiology , Humans , Kinetics , Molecular Docking Simulation , Piperazines/chemical synthesis , Piperazines/pharmacology , Protein Binding , Protein Multimerization , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Structure-Activity Relationship , Surface Plasmon Resonance , Triazoles/chemical synthesis , Triazoles/pharmacology , Virus Internalization/drug effects , env Gene Products, Human Immunodeficiency Virus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...