Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Biol Chem ; 290(16): 10504-17, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25724652

ABSTRACT

APOBEC3G (A3G) is a cellular cytidine deaminase that restricts HIV-1 replication by inducing G-to-A hypermutation in viral DNA and by deamination-independent mechanisms. HIV-1 Vif binds to A3G, resulting in its degradation via the 26 S proteasome. Therefore, this interaction represents a potential therapeutic target. To identify compounds that inhibit interaction between A3G and HIV-1 Vif in a high throughput format, we developed a homogeneous time-resolved fluorescence resonance energy transfer assay. A 307,520 compound library from the NIH Molecular Libraries Small Molecule Repository was screened. Secondary screens to evaluate dose-response performance and off-target effects, cell-based assays to identify compounds that attenuate Vif-dependent degradation of A3G, and assays testing antiviral activity in peripheral blood mononuclear cells and T cells were employed. One compound, N.41, showed potent antiviral activity in A3G(+) but not in A3G(-) T cells and had an IC50 as low as 8.4 µM and a TC50 of >100 µM when tested against HIV-1Ba-L replication in peripheral blood mononuclear cells. N.41 inhibited the Vif-A3G interaction and increased cellular A3G levels and incorporation of A3G into virions, thereby attenuating virus infectivity in a Vif-dependent manner. N.41 activity was also species- and Vif-dependent. Preliminary structure-activity relationship studies suggest that a hydroxyl moiety located at a phenylamino group is critical for N.41 anti-HIV activity and identified N.41 analogs with better potency (IC50 as low as 4.2 µM). These findings identify a new lead compound that attenuates HIV replication by liberating A3G from Vif regulation and increasing its innate antiviral activity.


Subject(s)
Anti-HIV Agents/pharmacology , Cytidine Deaminase/genetics , HIV-1/drug effects , Leukocytes, Mononuclear/drug effects , Small Molecule Libraries/pharmacology , T-Lymphocytes/drug effects , vif Gene Products, Human Immunodeficiency Virus/genetics , APOBEC-3G Deaminase , Anti-HIV Agents/chemistry , Biological Assay , Cell Line , Cytidine Deaminase/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression Regulation , HEK293 Cells , HIV-1/genetics , HIV-1/metabolism , Host-Pathogen Interactions , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Primary Cell Culture , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , Small Molecule Libraries/chemistry , Structure-Activity Relationship , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Replication/drug effects , vif Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , vif Gene Products, Human Immunodeficiency Virus/metabolism
2.
Antimicrob Agents Chemother ; 58(2): 687-97, 2014.
Article in English | MEDLINE | ID: mdl-24217696

ABSTRACT

PD 404,182 (PD) is a synthetic compound that was found to compromise HIV integrity via interaction with a nonenvelope protein viral structural component (A. M. Chamoun et al., Antimicrob. Agents Chemother. 56:672-681, 2012). The present study evaluates the potential of PD as an anti-HIV microbicide and establishes PD's virucidal activity toward another pathogen, herpes simplex virus (HSV). We show that the anti-HIV-1 50% inhibitory concentration (IC50) of PD, when diluted in seminal plasma, is ∼1 µM, similar to the IC50 determined in cell culture growth medium, and that PD retains full anti-HIV-1 activity after incubation in cervical fluid at 37°C for at least 24 h. In addition, PD is nontoxic toward vaginal commensal Lactobacillus species (50% cytotoxic concentration [CC50], >300 µM), freshly activated human peripheral blood mononuclear cells (CC50, ∼200 µM), and primary CD4(+) T cells, macrophages, and dendritic cells (CC50, >300 µM). PD also exhibited high stability in pH-adjusted Dulbecco's phosphate-buffered saline with little to no activity loss after 8 weeks at pH 4 and 42°C, indicating suitability for formulation for transportation and storage in developing countries. Finally, for the first time, we show that PD inactivates herpes simplex virus 1 (HSV-1) and HSV-2 at submicromolar concentrations. Due to the prevalence of HSV infection, the ability of PD to inactivate HSV may provide an additional incentive for use as a microbicide. The ability of PD to inactivate both HIV-1 and HSV, combined with its low toxicity and high stability, warrants additional studies for the evaluation of PD's microbicidal candidacy in animals and humans.


Subject(s)
Antiviral Agents/pharmacology , HIV-1/drug effects , Herpesvirus 1, Human/drug effects , Imines/pharmacology , Thiazines/pharmacology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Dendritic Cells/cytology , Dendritic Cells/drug effects , Drug Stability , Extracellular Fluid/chemistry , Female , HEK293 Cells , HIV-1/growth & development , Herpesvirus 1, Human/growth & development , Humans , Inhibitory Concentration 50 , Macrophages/cytology , Macrophages/drug effects , Male , Primary Cell Culture , Semen/chemistry , Temperature , Vero Cells
3.
Antimicrob Agents Chemother ; 58(12): 7056-71, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224013

ABSTRACT

In the absence of universally available antiretroviral (ARV) drugs or a vaccine against HIV-1, microbicides may offer the most immediate hope for controlling the AIDS pandemic. The most advanced and clinically effective microbicides are based on ARV agents that interfere with the earliest stages of HIV-1 replication. Our objective was to identify and characterize novel ARV-like inhibitors, as well as demonstrate their efficacy at blocking HIV-1 transmission. Abasic phosphorothioate 2' deoxyribose backbone (PDB) oligomers were evaluated in a variety of mechanistic assays and for their ability to inhibit HIV-1 infection and virus transmission through primary human cervical mucosa. Cellular and biochemical assays were used to elucidate the antiviral mechanisms of action of PDB oligomers against both lab-adapted and primary CCR5- and CXCR4-utilizing HIV-1 strains, including a multidrug-resistant isolate. A polarized cervical organ culture was used to test the ability of PDB compounds to block HIV-1 transmission to primary immune cell populations across ectocervical tissue. The antiviral activity and mechanisms of action of PDB-based compounds were dependent on oligomer size, with smaller molecules preventing reverse transcription and larger oligomers blocking viral entry. Importantly, irrespective of molecular size, PDBs potently inhibited virus infection and transmission within genital tissue samples. Furthermore, the PDB inhibitors exhibited excellent toxicity and stability profiles and were found to be safe for vaginal application in vivo. These results, coupled with the previously reported intrinsic anti-inflammatory properties of PDBs, support further investigations in the development of PDB-based topical microbicides for preventing the global spread of HIV-1.


Subject(s)
Cervix Uteri/drug effects , HIV-1/drug effects , Phosphorothioate Oligonucleotides/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcription/drug effects , Virus Internalization/drug effects , Animals , Cervix Uteri/virology , Deoxyribose/chemistry , Epithelial Cells/drug effects , Epithelial Cells/virology , Female , Gene Expression , HIV-1/enzymology , HIV-1/genetics , HIV-1/growth & development , Humans , Male , Mice , Mice, Inbred C57BL , Mucous Membrane/drug effects , Mucous Membrane/virology , Organ Culture Techniques , Phosphorothioate Oligonucleotides/chemical synthesis , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Receptors, CXCR4/antagonists & inhibitors , Reverse Transcriptase Inhibitors/chemical synthesis , Sperm Motility/drug effects , Structure-Activity Relationship , Vagina/drug effects , Vagina/virology
4.
Bioorg Med Chem Lett ; 24(23): 5439-45, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25454268

ABSTRACT

With the emergence of drug-resistant strains and the cumulative toxicities associated with current therapies, demand remains for new inhibitors of HIV-1 replication. The inhibition of HIV-1 entry is an attractive, yet underexploited therapeutic approach with implications for salvage and preexposure prophylactic regimens, as well as topical microbicides. Using the combination of a field-derived bioactive conformation template to perform virtual screening and iterative bioisosteric replacements, coupled with in silico predictions of absorption, distribution, metabolism, and excretion, we have identified new leads for HIV-1 entry inhibitors.


Subject(s)
HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Drug Discovery , Humans , Molecular Conformation
5.
J Chem Inf Model ; 54(11): 3080-90, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25302989

ABSTRACT

Despite the considerable successes of highly active antiretroviral therapy (HAART) for the treatment of HIV/AIDS, cumulative drug toxicities and the development of multidrug-resistant virus necessitate the search for new classes of antiretroviral agents with novel modes of action. The HIV-1 capsid (CA) protein has been structurally and functionally characterized as a druggable target. We have recently designed a novel small molecule inhibitor I-XW-053 using the hybrid structure based method to block the interface between CA N-terminal domains (NTD-NTD interface) with micromolar affinity. In an effort to optimize and improve the efficacy of I-XW-053, we have developed the structure activity relationship of I-XW-053 compound series using ligand efficiency methods. Fifty-six analogues of I-XW-053 were designed that could be subclassified into four different core domains based on their ligand efficiency values computed as the ratio of binding efficiency (BEI) and surface efficiency (SEI) indices. Compound 34 belonging to subcore-3 showed an 11-fold improvement over I-XW-053 in blocking HIV-1 replication in primary human peripheral blood mononuclear cells (PBMCs). Surface plasmon resonance experiments confirmed the binding of compound 34 to purified HIV-1 CA protein. Molecular docking studies on compound 34 and I-XW-053 to HIV-1 CA protein suggested that they both bind to NTD-NTD interface region but with different binding modes, which was further validated using site-directed mutagenesis studies.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , HIV-1/drug effects , HIV-1/physiology , Virus Replication/drug effects , Humans , Structure-Activity Relationship
6.
J Virol ; 86(16): 8472-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22647699

ABSTRACT

The HIV-1 capsid (CA) protein plays essential roles in both early and late stages of virl replication and has emerged as a novel drug target. We report hybrid structure-based virtual screening to identify small molecules with the potential to interact with the N-terminal domain (NTD) of HIV-1 CA and disrupt early, preintegration steps of the HIV-1 replication cycle. The small molecule 4,4'-[dibenzo[b,d]furan-2,8-diylbis(5-phenyl-1H-imidazole-4,2-diyl)]dibenzoic acid (CK026), which had anti-HIV-1 activity in single- and multiple-round infections but failed to inhibit viral replication in peripheral blood mononuclear cells (PBMCs), was identified. Three analogues of CK026 with reduced size and better drug-like properties were synthesized and assessed. Compound I-XW-053 (4-(4,5-diphenyl-1H-imidazol-2-yl)benzoic acid) retained all of the antiviral activity of the parental compound and inhibited the replication of a diverse panel of primary HIV-1 isolates in PBMCs, while displaying no appreciable cytotoxicity. This antiviral activity was specific to HIV-1, as I-XW-053 displayed no effect on the replication of SIV or against a panel of nonretroviruses. Direct interaction of I-XW-053 was quantified with wild-type and mutant CA protein using surface plasmon resonance and isothermal titration calorimetry. Mutation of Ile37 and Arg173, which are required for interaction with compound I-XW-053, crippled the virus at an early, preintegration step. Using quantitative PCR, we demonstrated that treatment with I-XW-053 inhibited HIV-1 reverse transcription in multiple cell types, indirectly pointing to dysfunction in the uncoating process. In summary, we have identified a CA-specific compound that targets and inhibits a novel region in the NTD-NTD interface, affects uncoating, and possesses broad-spectrum anti-HIV-1 activity.


Subject(s)
Anti-HIV Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , HIV-1/drug effects , HIV-1/physiology , Virus Uncoating/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/toxicity , Calorimetry , Cell Line , Humans , Microbial Sensitivity Tests , Protein Binding , Real-Time Polymerase Chain Reaction , Reverse Transcription/drug effects , Simian Immunodeficiency Virus/drug effects , Surface Plasmon Resonance , Virus Internalization/drug effects , Virus Replication/drug effects
7.
Bioorg Med Chem Lett ; 23(4): 1132-5, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23305922

ABSTRACT

Due to the emergence of drug-resistant strains and the cumulative toxicities associated with current therapies, demand remains for new inhibitors of HIV-1 replication. The HIV-1 matrix (MA) protein is an essential viral component with established roles in the assembly of the virus. Using virtual and surface plasmon resonance (SPR)-based screening, we describe the identification of the first small molecule to bind to the HIV-1 MA protein and to possess broad range anti-HIV properties.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , Humans , Models, Molecular , Molecular Targeted Therapy , Surface Plasmon Resonance
8.
J Biol Chem ; 286(32): 28370-81, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21659523

ABSTRACT

Blocking HIV-1 cell entry has long been a major goal of anti-HIV drug development. Here, we report a successful design of two highly potent chimeric HIV entry inhibitors composed of one CCR5-targeting RANTES (regulated on activation normal T cell expressed and secreted) variant (5P12-RANTES or 5P14-RANTES (Gaertner, H., Cerini, F., Escola, J. M., Kuenzi, G., Melotti, A., Offord, R., Rossitto-Borlat, I., Nedellec, R., Salkowitz, J., Gorochov, G., Mosier, D., and Hartley, O. (2008) Proc. Natl. Acad. Sci. U.S.A. 105, 17706-17711)) linked to a gp41 fusion inhibitor, C37. Chimeric inhibitors 5P12-linker-C37 and 5P14-linker-C37 showed extremely high antiviral potency in single cycle and replication-competent viral assays against R5-tropic viruses, with IC(50) values as low as 0.004 nm. This inhibition was somewhat strain-dependent and was up to 100-fold better than the RANTES variant alone or in combination with unlinked C37. The chimeric inhibitors also fully retained the antiviral activity of C37 against X4-tropic viruses, and this inhibition can be further enhanced significantly if the target cell co-expresses CCR5 receptor. On human peripheral blood mononuclear cells, the inhibitors showed very strong inhibition against R5-tropic Ba-L strain and X4-tropic IIIB strain, with IC(50) values as low as 0.015 and 0.44 nm, which are 45- and 16-fold better than the parent inhibitors, respectively. A clear delivery mechanism requiring a covalent linkage between the two segments of the chimera was observed and characterized. Furthermore, the two chimeric inhibitors are fully recombinant and are easily produced at low cost. These attributes make them excellent candidates for anti-HIV microbicides. The results of this study also suggest a potent approach for optimizing existing HIV entry inhibitors or designing new inhibitors.


Subject(s)
HIV Fusion Inhibitors/pharmacology , HIV Infections/drug therapy , HIV-1/physiology , Peptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Virus Internalization/drug effects , HIV Fusion Inhibitors/chemistry , HIV Infections/metabolism , HeLa Cells , Humans , Peptides/chemistry , Receptors, CCR5/metabolism , Recombinant Fusion Proteins/chemistry , Viral Tropism/drug effects
9.
Antimicrob Agents Chemother ; 55(1): 264-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20956603

ABSTRACT

The development of an anti-HIV microbicide is critical in the fight against the spread of HIV. It is shown here that the covalent linking of compounds that bind gp120 with compounds that bind gp41 can inhibit HIV entry even more potently than individual inhibitors or noncovalent combinations. The most striking example involves griffithsin, a potent HIV inhibitor that binds to the surface of HIV gp120. While griffithsin inhibits HIV Env-mediated fusion in a CCR5-tropic cell-cell fusion assay with a 50% inhibitory concentration (IC(50)) of 1.31 ± 0.87 nM and the gp41-binding peptide C37 shows an IC(50) of 18.2 ± 7.6 nM, the covalently linked combination of griffithsin with C37 (Griff37) has an IC(50) of 0.15 ± 0.05 nM, exhibiting a potency 8.7-fold greater than that of griffithsin alone. Similarly, in CXCR4-tropic cell-cell fusion assays, Griff37 is 5.2-fold more potent than griffithsin alone. In viral assays, both griffithsin and Griff37 inhibit HIV replication at midpicomolar levels, but the linked compound Griff37 is severalfold more potent than griffithsin alone against both CCR5- and CXCR4-tropic virus strains. Another example of this strategy is the covalently linked combination of peptide C37 with a variant of the gp120-binding peptide CD4M33 (L. Martin et al., Nat. Biotechnol. 21:71-76, 2003). Also, nuclear magnetic resonance (NMR) spectra for several of these compounds are shown, including, to our knowledge, the first published NMR spectrum for griffithsin.


Subject(s)
Algal Proteins/chemistry , Algal Proteins/pharmacology , Antiviral Agents/pharmacology , HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp41/metabolism , HIV-1/drug effects , HIV-1/metabolism , Lectins/chemistry , Lectins/pharmacology , Peptides/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cell Line , Cell Line, Tumor , HeLa Cells , Humans , Peptides/chemical synthesis , Peptides/chemistry , Plant Lectins , Protein Binding
10.
Antimicrob Agents Chemother ; 54(7): 2901-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20439609

ABSTRACT

CMX157 is a lipid (1-0-hexadecyloxypropyl) conjugate of the acyclic nucleotide analog tenofovir (TFV) with activity against both wild-type and antiretroviral drug-resistant HIV strains, including multidrug nucleoside/nucleotide analog-resistant viruses. CMX157 was consistently >300-fold more active than tenofovir against multiple viruses in several different cell systems. CMX157 was active against all major subtypes of HIV-1 and HIV-2 in fresh human peripheral blood mononuclear cells (PBMCs) and against all HIV-1 strains evaluated in monocyte-derived macrophages, with 50% effective concentrations (EC(50)s) ranging between 0.20 and 7.2 nM. The lower CMX157 EC(50)s can be attributed to better cellular uptake of CMX157, resulting in higher intracellular levels of the active antiviral anabolite, TFV-diphosphate (TFV-PP), inside target cells. CMX157 produced >30-fold higher levels of TFV-PP in human PBMCs exposed to physiologically relevant concentrations of the compounds than did TFV. Unlike conventional prodrugs, including TFV disoproxil fumarate (Viread), CMX157 remains intact in plasma, facilitating uptake by target cells and decreasing relative systemic exposure to TFV. There was no detectable antagonism with CMX157 in combination with any marketed antiretroviral drug, and it possessed an excellent in vitro cytotoxicity profile. CMX157 is a promising clinical candidate to treat wild-type and antiretroviral drug-resistant HIV, including strains that fail to respond to all currently available nucleoside/nucleotide reverse transcriptase inhibitors.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/virology , Nucleosides/therapeutic use , Nucleotides/therapeutic use , Organophosphonates/therapeutic use , Adenine/adverse effects , Adenine/pharmacology , Adenine/therapeutic use , Anti-HIV Agents/adverse effects , Cell Survival/drug effects , Cells, Cultured , Drug Resistance, Viral/genetics , HIV-1/drug effects , HIV-1/genetics , Humans , Macrophages/virology , Organophosphonates/adverse effects , Organophosphonates/pharmacology , Tenofovir
11.
AIDS Res Ther ; 7: 22, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20618951

ABSTRACT

BACKGROUND: At present, there is no effective vaccine or other approved product for the prevention of sexually transmitted human immunodeficiency virus type 1 (HIV-1) infection. It has been reported that women in resource-poor communities use vaginally applied citrus juices as topical microbicides. These easily accessible food products have historically been applied to prevent pregnancy and sexually transmitted diseases. The aim of this study was to evaluate the efficacy and cytotoxicity of these substances using an established topical microbicide testing algorithm. Freshly squeezed lemon and lime juice and household vinegar were tested in their original state or in pH neutralized form for efficacy and cytotoxicity in the CCR5-tropic cell-free entry and cell-associated transmission assays, CXCR4-tropic entry and fusion assays, and in a human PBMC-based anti-HIV-1 assay. These products were also tested for their effect on viability of cervico-vaginal cell lines, human cervical explant tissues, and beneficial Lactobacillus species. RESULTS: Natural lime and lemon juice and household vinegar demonstrated anti-HIV-1 activity and cytotoxicity in transformed cell lines. Neutralization of the products reduced both anti-HIV-1 activity and cytotoxicity, resulting in a low therapeutic window for both acidic and neutralized formulations. For the natural juices and vinegar, the IC50 was

12.
Antimicrob Agents Chemother ; 52(5): 1768-81, 2008 May.
Article in English | MEDLINE | ID: mdl-18316528

ABSTRACT

Topical microbicides are self-administered, prophylactic products for protection against sexually transmitted pathogens. A large number of compounds with known anti-human immunodeficiency virus type 1 (HIV-1) inhibitory activity have been proposed as candidate topical microbicides. To identify potential leads, an in vitro screening algorithm was developed to evaluate candidate microbicides in assays that assess inhibition of cell-associated and cell-free HIV-1 transmission, entry, and fusion. The algorithm advances compounds by evaluation in a series of defined assays that generate measurements of relative antiviral potency to determine advancement or failure. Initial testing consists of a dual determination of inhibitory activity in the CD4-dependent CCR5-tropic cell-associated transmission inhibition assay and in the CD4/CCR5-mediated HIV-1 entry assay. The activity is confirmed by repeat testing, and identified actives are advanced to secondary screens to determine their effect on transmission of CXCR4-tropic viruses in the presence or absence of CD4 and their ability to inhibit CXCR4- and CCR5-tropic envelope-mediated cell-to-cell fusion. In addition, confirmed active compounds are also evaluated in the presence of human seminal plasma, in assays incorporating a pH 4 to 7 transition, and for growth inhibition of relevant strains of lactobacilli. Leads may then be advanced for specialized testing, including determinations in human cervical explants and in peripheral blood mononuclear cells against primary HIV subtypes, combination testing with other inhibitors, and additional cytotoxicity assays. PRO 2000 and SPL7013 (the active component of VivaGel), two microbicide products currently being evaluated in human clinical trials, were tested in this in vitro algorithm and were shown to be highly active against CCR5- and CXCR4-tropic HIV-1 infection.


Subject(s)
Algorithms , Anti-HIV Agents/pharmacology , Anti-Infective Agents, Local/pharmacology , HIV-1/drug effects , Amides/pharmacology , Anilides/pharmacology , CCR5 Receptor Antagonists , CD4 Antigens/immunology , Cell Line , Drug Evaluation, Preclinical , Furans/pharmacology , HeLa Cells , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Naphthalenesulfonates/pharmacology , Polymers/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Thioamides
13.
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.

14.
Antiviral Res ; 136: 51-59, 2016 12.
Article in English | MEDLINE | ID: mdl-27825797

ABSTRACT

Camptothecin (CPT) is a natural product discovered to be active against various cancers through its ability to inhibit Topoisomerase I (TOP1). CPT analogs also have anti-HIV-1 (HIV) activity that was previously shown to be independent of TOP1 inhibition. We show that a cancer inactive CPT analog (O2-16) inhibits HIV infection by disrupting multimerization of the HIV protein Vif. Antiviral activity depended on the expression of the cellular viral restriction factor APOBEC3G (A3G) that, in the absence of functional Vif, has the ability to hypermutate HIV proviral DNA during reverse transcription. Our studies demonstrate that O2-16 has low cytotoxicity and inhibits Vif-dependent A3G degradation, enabling A3G packaging into HIV viral particles that results in A3G signature hypermutations in viral genomes. This antiviral activity was A3G-dependent and broadly neutralizing against sixteen HIV clinical isolates from groups M (subtypes A-G), N, and O as well as seven single and multi-drug resistant strains of HIV. Molecular modeling predicted binding near the PPLP motif crucial for Vif multimerization and activity. O2-16 also was active in blocking Vif degradation of APOBEC3F (A3F). We propose that CPT analogs not active against TOP1 have novel therapeutic potential as Vif antagonists that enable A3G-dependent hypermutation of HIV.


Subject(s)
APOBEC-3G Deaminase/metabolism , Camptothecin/analogs & derivatives , DNA Topoisomerases, Type I/metabolism , HIV-1/drug effects , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase/genetics , Camptothecin/pharmacology , Cell Line , Drug Resistance, Viral/genetics , Genome, Viral , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Humans , Models, Molecular , Mutation , Protein Binding , Protein Multimerization/drug effects , Virion/metabolism , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/chemistry
15.
Virology ; 484: 276-287, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26141568

ABSTRACT

APOBEC3G (A3G) is a cytidine deaminase that restricts HIV-1 replication by inducing G-to-A hypermutation in viral DNA; deamination-independent mechanisms are also implicated. HIV-1 Vif protein counteracts A3G by inducing its proteasomal degradation. Thus, the Vif-A3G axis is a potential therapeutic target. To identify compounds that inhibit Vif:A3G interaction, a 307,520 compound library was tested in a TR-FRET screen. Two identified compounds, redoxal and lomofungin, inhibited HIV-1 replication in peripheral blood mononuclear cells. Lomofungin activity was linked to A3G, but not pursued further due to cytotoxicity. Redoxal displayed A3G-dependent restriction, inhibiting viral replication by stabilizing A3G protein levels and increasing A3G in virions. A3G-independent activity was also detected. Treatment with uridine or orotate, intermediates of pyrimidine synthesis, diminished redoxal-induced stabilization of A3G and antiviral activity. These results identify redoxal as an inhibitor of HIV-1 replication and suggest its ability to inhibit pyrimidine biosynthesis suppresses viral replication by augmenting A3G antiviral activity.


Subject(s)
Aminobiphenyl Compounds/metabolism , Antiviral Agents/metabolism , Cytidine Deaminase/metabolism , HIV-1/immunology , HIV-1/physiology , Pyrimidines/biosynthesis , Virus Replication , APOBEC-3G Deaminase , Cells, Cultured , Enzyme Inhibitors/metabolism , Humans , Leukocytes, Mononuclear/virology , Phenazines/metabolism
16.
J Med Chem ; 57(12): 5270-81, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24856833

ABSTRACT

We previously described indole-containing compounds with the potential to inhibit HIV-1 fusion by targeting the hydrophobic pocket of transmembrane glycoprotein gp41. Here we report optimization and structure-activity relationship studies on the basic scaffold, defining the role of shape, contact surface area, and molecular properties. Thirty new compounds were evaluated in binding, cell-cell fusion, and viral replication assays. Below a 1 µM threshold, correlation between binding and biological activity was diminished, indicating an amphipathic requirement for activity in cells. The most active inhibitor 6j exhibited 0.6 µM binding affinity and 0.2 µM EC50 against cell-cell fusion and live virus replication and was active against T20 resistant strains. Twenty-two compounds with the same connectivity displayed a consensus pose in docking calculations, with rank order matching the biological activity. The work provides insight into requirements for small molecule inhibition of HIV-1 fusion and demonstrates a potent low molecular weight fusion inhibitor.


Subject(s)
Benzoates/chemistry , HIV Envelope Protein gp41/metabolism , HIV Fusion Inhibitors/chemistry , HIV-1/drug effects , Indoles/chemistry , Benzoates/chemical synthesis , Benzoates/pharmacology , Cell Fusion , Cell Line , Drug Resistance, Viral , Enfuvirtide , HIV Envelope Protein gp41/pharmacology , HIV Fusion Inhibitors/chemical synthesis , HIV Fusion Inhibitors/pharmacology , HIV-1/physiology , Humans , Indoles/chemical synthesis , Indoles/pharmacology , Molecular Docking Simulation , Peptide Fragments/pharmacology , Structure-Activity Relationship , Virus Replication/drug effects
17.
Antiviral Res ; 98(3): 365-72, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23602851

ABSTRACT

Research efforts on the human immunodeficiency virus (HIV) integrase have resulted in two approved drugs. However, co-infection of HIV with Mycobacterium tuberculosis and other microbial and viral agents has introduced added complications to this pandemic, requiring favorable drug-drug interaction profiles for antiviral therapeutics targeting HIV. Cytochrome P450 (CYP) and uridine 5'-diphospho-glucuronosyltransferase (UGT) are pivotal determining factors in the occurrence of adverse drug-drug interactions. For this reason, it is important that anti-HIV agents, such as integrase inhibitors, possess favorable profiles with respect to CYP and UGT. We have discovered a novel HIV integrase inhibitor (compound 1) that exhibits low nM antiviral activity against a diverse set of HIV-1 isolates, and against HIV-2 and the simian immunodeficiency virus (SIV). Compound 1 displays low in vitro cytotoxicity and its resistance and related drug susceptibility profiles are favorable. Data from in vitro studies revealed that compound 1 was not a substrate for UGT isoforms and that it was not an inhibitor or activator of key CYP isozymes.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Glucuronosyltransferase/metabolism , HIV Integrase Inhibitors/chemical synthesis , HIV-1/drug effects , Pyridines/chemical synthesis , Pyrrolidines/chemical synthesis , Drug Evaluation, Preclinical , Drug Resistance, Viral , HIV Integrase/genetics , HIV Integrase/metabolism , HIV Integrase Inhibitors/pharmacology , HIV-1/enzymology , HIV-2/drug effects , HIV-2/metabolism , HeLa Cells , Humans , Isoenzymes/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Microbial Sensitivity Tests , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Docking Simulation , Mutation , Pyridines/pharmacology , Pyrrolidines/pharmacology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/metabolism
18.
ChemMedChem ; 8(3): 426-32, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23361947

ABSTRACT

The development of drug resistance remains a critical problem for current HIV-1 antiviral therapies, creating a need for new inhibitors of HIV-1 replication. We previously reported on a novel anti-HIV-1 compound, N(2)-(phenoxyacetyl)-N-[4-(1-piperidinylcarbonyl)benzyl]glycinamide (14), that binds to the highly conserved phosphatidylinositol (4,5)-bisphosphate (PI(4,5)P(2)) binding pocket of the HIV-1 matrix (MA) protein. In this study, we re-evaluate the hits from the virtual screen used to identify compound 14 and test them directly in an HIV-1 replication assay using primary human peripheral blood mononuclear cells. This study resulted in the identification of three new compounds with antiviral activity; 2-(4-{[3-(4-fluorophenyl)-1,2,4-oxadiazol-5-yl]methyl})-1-piperazinyl)-N-(4-methylphenyl)acetamide (7), 3-(2-ethoxyphenyl)-5-[[4-(4-nitrophenyl)piperazin-1-yl]methyl]-1,2,4-oxadiazole (17), and N-[4-ethoxy-3-(1-piperidinylsulfonyl)phenyl]-2-(imidazo[2,1-b][1,3]thiazol-6-yl)acetamide (18), with compound 7 being the most potent of these hits. Mechanistic studies on 7 demonstrated that it directly interacts with and functions through HIV-1 MA. In accordance with our drug target, compound 7 competes with PI(4,5)P(2) for MA binding and, as a result, diminishes the production of new virus. Mutation of residues within the PI(4,5)P(2) binding site of MA decreased the antiviral effect of compound 7. Additionally, compound 7 displays a broadly neutralizing anti-HIV activity, with IC(50) values of 7.5-15.6 µM for the group M isolates tested. Taken together, these results point towards a novel chemical probe that can be used to more closely study the biological role of MA and could, through further optimization, lead to a new class of anti-HIV-1 therapeutics.


Subject(s)
Acetanilides/pharmacology , Anti-HIV Agents/pharmacology , HIV Antigens/chemistry , HIV-1/metabolism , Oxadiazoles/pharmacology , Phosphatidylinositol 4,5-Diphosphate/chemistry , Small Molecule Libraries/pharmacology , Virus Replication/drug effects , gag Gene Products, Human Immunodeficiency Virus/chemistry , Acetanilides/chemistry , Anti-HIV Agents/chemistry , Binding Sites , Cells, Cultured , HEK293 Cells , HIV Antigens/genetics , HIV Antigens/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Molecular Docking Simulation , Oxadiazoles/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Small Molecule Libraries/chemistry , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
19.
Vaccine ; 27(6): 857-63, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19084043

ABSTRACT

The elicitation of broadly cross-reactive HIV-1 neutralizing antibodies in humans remains a major challenge in developing a viable AIDS vaccine. We hypothesized that prolonged exposure to candidate vaccine immunogens could enhance the elicitation of such antibodies. In an attempt to develop HIV-1 vaccine immunogens with prolonged half-lives and increased stability, we constructed a fusion protein, gp41Fc, in which a truncated HIV-1 gp41(89.6) was fused to a human IgG(1) Fc. Gp41Fc is stable in solution, retains its antigenic structure and is highly immunogenic in rabbits. The serum titers reached 1:102,400 for the gp41Fc and 1:5,120 for gp140(89.6). Rabbit IgG neutralized diverse HIV-1 isolates and HIV-2, and the neutralization activity was attributed to gp41-specific IgG. The concentration of the gp41Fc in the serum correlated with the neutralization activity of rabbit IgG which recognized mostly conformation-independent epitopes on gp41 and predominantly bound to peptides derived from the gp41 immunodominant loop region. These results suggest that the prolonged half-life of gp41Fc in the serum may enhance the generation of cross-reactive neutralizing antibodies. Further research is needed to confirm and extend these results which may have implications for the development of vaccine immunogens with enhanced capability to elicit cross-reactive HIV-1-neutralizing antibodies.


Subject(s)
AIDS Vaccines/immunology , AIDS Vaccines/pharmacokinetics , Adjuvants, Immunologic/pharmacology , HIV Antibodies/blood , HIV Envelope Protein gp41/immunology , HIV-1/immunology , Immunoglobulin Fc Fragments/pharmacology , AIDS Vaccines/genetics , Adjuvants, Immunologic/genetics , Animals , Female , HIV Envelope Protein gp41/genetics , HIV-1/genetics , HIV-2/immunology , Half-Life , Humans , Immunoglobulin Fc Fragments/genetics , Neutralization Tests , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacokinetics
20.
J Biol Chem ; 283(49): 34045-52, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18809675

ABSTRACT

Entry inhibitors of human immunodeficiency virus, type 1 (HIV-1) have been the focus of much recent research. C34, a potent fusion inhibitor derived from the HR2 region of gp41, was engineered into a 1:1 human serum albumin conjugate through stable covalent attachment of a maleimido-C34 analog onto cysteine 34 of albumin. This bioconjugate, PC-1505, was designed to require less frequent dosing and less peptide than T-20 and was assessed for its antifusogenic activity both in vitro and in vivo in the SCID-hu Thy/Liv mouse model. PC-1505 was essentially equipotent to the original C34 peptide and to T-20 in vitro. In HIV-1-infected SCID-hu Thy/Liv mice, T-20 lost activity with infrequent dosing, whereas the antiviral potency of PC-1505 was sustained, and PC-1505 was active against T-20-resistant ("DIV") virus with a G36D substitution in gp41. The in vivo results are the direct result of a significantly improved pharmacokinetic profile for the C34 peptide following albumin conjugation. Contrary to previous reports that the gp41 NHR trimer is poorly accessible to C34 fused to protein cargoes of increasing size (Hamburger, A. E., Kim, S., Welch, B. D., and Kay, M. S. (2005) J. Biol. Chem. 280, 12567-12572), these results are the first demonstration of the capacity for a large, endogenous serum protein to gain unobstructed access to the transient gp41 intermediates that exist during the HIV fusion process, and it supports further development of albumin conjugation as a promising approach to inhibit HIV-1 entry.


Subject(s)
Albumins/chemistry , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV-1/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides/pharmacology , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Disease Models, Animal , Enfuvirtide , HIV Fusion Inhibitors/chemistry , Mice , Mice, SCID , Molecular Sequence Data , Peptides/chemistry , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL